BBAMEM-19-17R1 Title: Molecular basis f - Digital CSIC

89
Elsevier Editorial System(tm) for BBA - Biomembranes Manuscript Draft Manuscript Number: BBAMEM-19-17R1 Title: Molecular basis for the protective effects of low-density lipoprotein receptor-related protein 1 (LRP1)-derived peptides against LDL aggregation Article Type: Regular Paper Keywords: Lipoprotein aggregation; LRP1-derived peptides; ApoB-100; SMase, PLA2, atherosclerosis Corresponding Author: Dr. Concepción Vicenta Llorente, Corresponding Author's Institution: First Author: Aleyda Benitez Amaro, PhD student Order of Authors: Aleyda Benitez Amaro, PhD student; Chiara Pallara, PhD; Laura Nasarre; Andrea Rivas Urbina, PhD student; Sonia Benitez, PhD; Angela Vea, PhD student; Olga Bornachea, PhD; David de Gonzalo Calvo, PhD; Gabriel Serra Mir, PhD; Sandra Villegas, PhD; Roger Prades, PhD; Jose Luis Sanchez Quesada, Phd; Cristina Chiva, PhD; Eduard Sabido, PhD; Teresa Tarragó, PhD; Concepción Vicenta Llorente Abstract: Aggregated LDL is the first ligand reported to interact with the cluster II CR9 domain of low-density lipoprotein receptor-related protein 1 (LRP1). In particular, the C-terminal half of domain CR9, comprising the region Gly1127-Cys1140 exclusively recognizes aggregated LDL and it is crucial for aggregated LDL binding. Our aim was to study the effect of the sequence Gly1127-Cys1140 (named peptide LP3 and its retro-enantio version, named peptide DP3) on the structural characteristics of sphingomyelinase- (SMase) and phospholipase 2 (PLA2)- modified LDL particles. Turbidimetry, gel filtration chromatography (GFC) and tomography electronic microscopy (TEM) analysis showed that LP3 and DP3 peptides strongly inhibited SMase- and PLA2-induced LDL aggregation. Nondenaturing polyacrylamide gradient gel electrophoresis (GGE), agarose gel electrophoresis and high-performance thin-layer chromatography (HPTLC) indicated that LP3 and DP3 prevented SMase-induced alterations in LDL particle size, electric charge and phospholipid content, respectively, but not those induced by PLA2. Western blot analysis showed that LP3 and DP3 counteracted changes in ApoB-100 conformation induced by the two enzymes. LDL proteomics (LDL trypsin digestion followed by mass spectroscopy) and computational modeling methods evidenced that peptides protect ApoB-100 conformation due to their electrostatic interactions with a basic region of ApoB-100. These results demonstrate that LRP1- derived peptides are protective against LDL aggregation, even in conditions of extreme lipolysis, through their capacity to bind to ApoB- 100 regions critical to preserve ApoB-100 conformation. These results suggests that these LRP1(CR9) derived peptides could be promising tools to prevent LDL aggregation induced by the main proteolytic enzymes acting in the arterial intima. Response to Reviewers: ANSWERS TO REVIEWERS

Transcript of BBAMEM-19-17R1 Title: Molecular basis f - Digital CSIC

Elsevier Editorial System(tm) for BBA -

Biomembranes

Manuscript Draft

Manuscript Number: BBAMEM-19-17R1

Title: Molecular basis for the protective effects of low-density

lipoprotein receptor-related protein 1 (LRP1)-derived peptides against

LDL aggregation

Article Type: Regular Paper

Keywords: Lipoprotein aggregation; LRP1-derived peptides; ApoB-100;

SMase, PLA2, atherosclerosis

Corresponding Author: Dr. Concepción Vicenta Llorente,

Corresponding Author's Institution:

First Author: Aleyda Benitez Amaro, PhD student

Order of Authors: Aleyda Benitez Amaro, PhD student; Chiara Pallara, PhD;

Laura Nasarre; Andrea Rivas Urbina, PhD student; Sonia Benitez, PhD;

Angela Vea, PhD student; Olga Bornachea, PhD; David de Gonzalo Calvo,

PhD; Gabriel Serra Mir, PhD; Sandra Villegas, PhD; Roger Prades, PhD;

Jose Luis Sanchez Quesada, Phd; Cristina Chiva, PhD; Eduard Sabido, PhD;

Teresa Tarragó, PhD; Concepción Vicenta Llorente

Abstract: Aggregated LDL is the first ligand reported to interact with

the cluster II CR9 domain of low-density lipoprotein receptor-related

protein 1 (LRP1). In particular, the C-terminal half of domain CR9,

comprising the region Gly1127-Cys1140 exclusively recognizes aggregated

LDL and it is crucial for aggregated LDL binding. Our aim was to study

the effect of the sequence Gly1127-Cys1140 (named peptide LP3 and its

retro-enantio version, named peptide DP3) on the structural

characteristics of sphingomyelinase- (SMase) and phospholipase 2 (PLA2)-

modified LDL particles. Turbidimetry, gel filtration chromatography (GFC)

and tomography electronic microscopy (TEM) analysis showed that LP3 and

DP3 peptides strongly inhibited SMase- and PLA2-induced LDL aggregation.

Nondenaturing polyacrylamide gradient gel electrophoresis (GGE), agarose

gel electrophoresis and high-performance thin-layer chromatography

(HPTLC) indicated that LP3 and DP3 prevented SMase-induced alterations in

LDL particle size, electric charge and phospholipid content,

respectively, but not those induced by PLA2. Western blot analysis showed

that LP3 and DP3 counteracted changes in ApoB-100 conformation induced by

the two enzymes. LDL proteomics (LDL trypsin digestion followed by mass

spectroscopy) and computational modeling methods evidenced that peptides

protect ApoB-100 conformation due to their electrostatic interactions

with a basic region of ApoB-100. These results demonstrate that LRP1-

derived peptides are protective against LDL aggregation, even in

conditions of extreme lipolysis, through their capacity to bind to ApoB-

100 regions critical to preserve ApoB-100 conformation. These results

suggests that these LRP1(CR9) derived peptides could be promising tools

to prevent LDL aggregation induced by the main proteolytic enzymes acting

in the arterial intima.

Response to Reviewers: ANSWERS TO REVIEWERS

Manuscript No.: BBAMEM-19-17

Title: Molecular basis for the protective effects of low-density

lipoprotein receptor-related protein 1 (LRP1)-derived peptides against

LDL aggregation

Article Type: Regular Paper

Journal Title: BBA - Biomembranes

Corresponding Author: Dr. Vicenta Llorente Cortés

Submit Date: Jan 20, 2019

We thank the Reviewers for their valuable suggestions and critical

comments that have enhanced the quality of the manuscript.

Reviewer #1: The manuscript is a well-written report on the effect of two

peptides derived from LRP1 on the inhibition of enzyme-induced

aggregation of LDL. Overall the experiments are well designed and

executed, and the results are presented clearly showing that DP3 and LP3

peptides inhibit LDL aggregation.

It would help if the authors provide more detail on the rational of using

the retro-enantio peptides, it would help if this is included in the

first paragraph of the result section. This is relevant as the authors

clearly show that DP3 is more active than LP3, and an explanation in the

discussion would be welcome. Further, provide detail about the blocking

of the termini in the methods section (in particular explain "H" on the

N-terminus). This is important as DP3 is more active than LP3. This can

also be discussed in the discussion.

We thank the Reviewer by this comment that will help to understand the

rational to use retro-enantio peptides. Peptide drugs offer high

activity, high specificity and low toxicity, but are susceptible to

proteolytic degradation. A strategy to overcome this problem is to

produce peptides using non-natural or D-amino acids, which usually are

not recognized by plasma proteases. However, in most of the cases, the

all-D peptide version (enantio) of an active L-peptide is not active or

show very low activity because the opposite orientation of the amino

acids side-chains compared to the parent peptide, which challenges the

recognition of the enantio peptide by its target receptor. Nevertheless,

in the literature there are some examples with enantio versions of

peptides that do show activity (Soto C et al, Biochem Biophys Res Commun

1996; Chalifour RJ et al, J Biol Chem 2003). A more conservative strategy

to preserve peptide activity using D-amino acids consists in applying the

retro-enantio approach. The idea behind this approach is to

simultaneously invert the sequence and chirality of the peptide (Chorev M

et al, Trends Biotechnol 1995; Jameson BA et al, Nature 1994) In this

way, the amine bone of the retro-enantio version is shift one unit but

the orientation of the side chains remain the same:

Usually, using this approach the retro-enantio peptide retains the

activity of the parent peptide (Prades R et al, Angew Chem Int Ed 2015).

This explanation has been summarized in the first paragraph of Results

(part 3.1) and expanded in the Discussion section of the revised ms.

In the first paragraph of the result (part 3.1)_“Peptide drugs offer high

activity, high specificity and low toxicity, but are susceptible to

proteolytic degradation. A strategy to overcome this problem is to

produce peptides using non-natural or D-amino acids, which shows improved

stability. Although all-D peptides are not recognized by plasma

proteases, they may be less active than their corresponding all-L

peptides because of differences in the orientation of amino acid side-

chains. To preserve the orientation of the side-chains of the parent

peptide in a D-analogue, the D-amino acid residues can be linked in the

reverse order to that of the parent peptide in order to produce a retro-

enantio peptide. This approach emphasizes the maintenance of the

topological native orientation of the side-chains but not of the

backbone, where the amide bond is shift one unit (Chorev M et al, Trends

Biotechnol 1995). By using this approach, peptides usually preserve their

activity while notably improve their plasma stability. In the literature

there are examples where this approach has been successfully used (Prades

R et al, Angew Chem Int Ed 2015)”.

In the Discussion section we have added the following explanation (first

paragraph)_“we tested both the original LP3 version and its retro-enantio

version DP3. Although all-D peptides are not recognised by proteases, and

thus have higher stability, they may be less active than their

corresponding all-L peptides because of differences in the orientation of

amino acid side-chains. To preserve the orientation of the side-chains of

the parent L-peptide in a D-analogue, the retro-enantio approach (Soto C

et al, Biochem Biophys Res Commun 1996; Chalifour RJ et al, J Biol Chem

2003) has been used in this study. Here, we demonstrate that LP3 and its

derived retro-enantio version (DP3) per se have a high capacity to

inhibit LDL aggregation induced by SMase and PLA2. In this regard, the

preservation of the topological side-chain orientation of DP3 together

with a high stability in biological samples account for its notable

performance in the inhibition assays.”

In the reference section we have added three new references

Further, provide detail about the blocking of the termini in the methods

section (in particular explain "H" on the N-terminus). Peptides are

always written from N to C terminus (N->C). The peptides described in

this manuscript have an amide at their C-terminus and we have indicated

so by typing –NH2 at this termini. The way of indicating this could be

misleading for readers not familiar with peptide nomenclature, since

these could think that the peptide sequences is typed from C to N. To

clearly indicate the N and C terminus of the peptide we added the H- at

the N-terminal part of the sequence to indicate that it finalizes as

amine.

To avoid any confusion in this regard, this explanation has been included

in the Methods section of the revised ms_“Peptide sequences are written

from N to C, H- at the end terminus indicate amine ending, whereas –NH2

at the C terminus indicates amide”.

It is evidently shown that the two peptides are able to inhibit the SMase

and PLA2 induced LDL aggregation. Can the authors show that the peptides

associate with the LDL surface prior or after enzyme treatment?

Thanks to the Reviewer by this interesting question. Limited trypsin

proteolysis combined with Mass spectrometry-based proteomics was the

strategy used to analyze this key aspect mentioned by the Reviewer. This

strategy allows to identify the sequences of ApoB-100 in native LDL

protected by the peptide against trypsin-induced proteolysis. Five

sequences of ApoB-100 in native LDL were protected from proteolysis. The

peptide protection was obviously temporal, it was observed at 1 hour and

disappeared at 3 hours (Figure 7A). These results evidenced that the

peptide interacts with the LDL surface prior enzymatic treatment,

protecting certain sequences against proteolysis.

We have remarked this interesting point in the discussion section of the

revised manuscript _ “Limited trypsin proteolysis combined with Mass

spectrometry-based proteomics of ApoB-100/DP3 complexes evidenced that

the peptide interacts with the LDL surface prior enzymatic treatment”.

Is it possible that the peptides bind directly to the enzyme thereby

preventing LDL aggregation? This possibility should be discussed.

We have tested this possibility by exploring the effect of peptides on

SMase activity in the current incubation conditions. As shown in

Supplemental Figure 6, SMase activity, measured in relation to the amount

of phosphorylcholine generated, was similar in absence or presence of the

peptides, indicating that, although we cannot discard a potential

interaction between peptide and SMase, it is clear that this interaction

has not effect on SMase activity. This comment has been included in the

Discussion of the revised ms. as following: “Although we can not discard

a potential interaction between peptides and SMase, SMase activity assays

clearly show that peptides does not exert any effect on SMase functional

activity”.

The authors discuss the appearance of non-polar spots on the surface that

trigger aggregation (page 20). Is it possible that the peptides associate

with these spots (hydrophobic defects?), as has been shown for

amphipathic helices of apolipoproteins? (4F is an amphipathic peptide).

This has been reported in a manuscript by Liu et al, in which PL-C

treated LDL resulted in the recruitment of apolipoproteins, thereby

offering protection against aggregation. (Liu, H., Scraba, D.G., and

Ryan, R.O. 1993 Prevention of phospholipase-C induced aggregation of low

density lipoprotein by amphipathic apolipoproteins, FEBS Lett. 316, 27-

33.)

Thanks to the Reviewer for this interesting observation. As the Reviewer

comments, several peptides and proteins bearing amphipathic properties

have been reported to be able to block LDL aggregation by a direct

interaction with LDL phospholipid surface. In this respect, common

examples are 4F peptide (Nguyen SD et al, J Lipid Res 2015) or different

apolipoproteins (Liu H et al, FEBS Lett 1993). Despite the lack of

amphipathic properties in any of the LRP1-derived peptides, this

hypothesis was even considered as possible mechanism of action for the

peptides described herein.Thus, as a preliminary test, molecular ligand

docking was performed using a phospholipidic monolayer mimicking LDL

surface (Hevonoja T et al, Biochem Biophys Acta 2000) as receptor and DP3

peptide as ligand. The top ranked docking poses showed rather low binding

energy values (around -4 kcal/mol) and mostly unreasonable binding modes.

Despite their theoretical character, these results support that there is

not a direct association between LRP1-derived peptides and LDL

phospholipid surface. This explanation has been summarized in the Results

(part 3.7) and expanded in the Discussion section of the revised ms.

In Result (part 3.7)_”Molecular docking calculations were perfomed using

a phospholipidic monolayer mimicking LDL surface (Hevonoja T et al,

Biochem Biophys Acta 2000) as receptor and DP3 peptide as ligand. The top

ranked docking poses showed rather low binding energy values (around -4

kcal/mol) and mostly unreasonable binding modes. This theoretical

calculations support that peptides do not associate with LDL phospholipid

surface”.

This argumentation has been expanded in the Discussion section_ Two

different explanations about the protective effect of peptides against

SMase-induced LDL lipolysis have been explored. By one side, we have

explored whether there is a direct effect of peptides on SMase activity.

This possibility has been discarded since SMase activity assays clearly

show that peptides does not exert any effect on SMase functional

activity. Other possibility that has been explored is a potential

interaction of peptides with the LDL phospholipid surface. Several

peptides and proteins bearing amphipathic properties have been reported

to be able to block LDL aggregation by a direct interaction with LDL

phospholipid surface. In this respect, common examples are 4F peptide

(Nguyen SD et al, J Lipid Res 2015) or different apolipoproteins (Liu H

et al, FEBS Lett 1993). Despite the lack of amphipathic properties in

LRP1-derived peptides, this hypothesis has been considered as possible

mechanism of action for the peptides described herein. Theoretical

molecular docking calculations performed in the present study indicate a

lack of association between peptides and LDL phospholipid surface”

New references about this issue have been included in the Reference

section_ One new reference has been added to the reference section of the

revised ms.

Fig 1 legend. Indicate that turbidity was measured by the absorbance at

405 nm.

We have included this indication in the Figure 1 legend of the revised

ms.

Table S3, indicate that this is a molar ratio.

We have included this indication in Table S3 of the revised ms.

Reviewer #3: The manuscript is a comparative study among different

peptides and their effects on the aggregation of sphingomyelinase-

(SMase) and phospholipase 2 (PLA2)-modified LDL particles.

The manuscript is well-written. The study is interesting. The peptides

were succesfully synthetized by solid-phase peptide synthesis as reported

in table S1.

The manuscript is ready for publication.

We thank the Reviewer for his/her positive evaluation of our manuscript

Reviewer #4: In the manuscript entitled "Molecular basis for the

protective effects of low-density lipoprotein receptor-related protein 1

(LRP1)-derived peptides against LDL aggregation » the authors have

investigate the effect of two peptide (LP3 and DP3) on the structural

characteristics of SMase and PLA2 modified LDL particles. They showed by

turbidimetry, gel filtration chromatography and tomography electronic

microscopy (TEM) analysis that LP3 and DP3 peptides strongly inhibited

SMase- and PLA2-induced LDL aggregation. They showed that :

LRP1-derived peptides prevent SMase-induced alterations in LDL particle

size, electric charge and phospholipid content but do not prevent PLA2-

induced alterations in LDL particle size, electric charge and

phospholipid content. Their data demonstrate that LRP1-derived peptides

are protective against LDL aggregation, through their capacity to bind to

ApoB-100 regions critical to preserve ApoB-100 conformation.

The article is well written and easy to read. The authors have convincing

data demonstrating mechanism involved by these both peptides and provide

evidence that these peptides could be promising tools to prevent LDL

aggregation in the arterial intima. This work present interesting data

that could be published with some minor complementary informations listed

below:

1- In the discussion section, the authors should better explain why both

peptides exerted protection against conformational alterations of poB-100

induced by both SMases and PLA2 whereas they didn't counteract particle

size charge and phospholipids induced by PLA2.

We thank the Reviewer by this interesting comment. LRP1-derived peptides

preserved ApoB-100 conformation, sphyngomyelin (SM) content and LDL size

in SMase-LDL while in PLA2-LDL, peptides preserve only ApoB-100

conformation but not phosphatidylcholine (PC) content or LDL size. We

propose that the differential effect of LRP1-derived peptides in these

two modified LDLs could be explained by topological reasons. Although

sphingomyelin (SM), compared to phosphatidylcholine (PC), is a minor

phospholipid in LDL (185 versus 450 molecules/LDL), SM, the target

phospholipid for SMase, is localized in cholesterol-enriched

environments, while PC is associated with poor-cholesterol environment.

Surface cholesterol seems to be a key regulator of the process of

phospholipolysis (Hevonoja T et al, Biochem Biophys Acta 2000) and thus,

could modulate SM but not PC lipolysis. We propose that peptide

preservation of ApoB-100 conformation directly impacts on the maintenance

of the cholesterol-enriched environment structure since, according to our

molecular modeling studies, ApoB-100 sequences interacting with peptide

are those close to cholesterol in the LDL surface. Therefore, SM would be

protected against SMase phospholipolysis by its privilege of being part

of cholesterol-enriched domains, whose structure is likely dependent on

the maintenance of ApoB-100 conformation. In contrast, PC, belonging to

cholesterol-poor domains, seems to be unprotected against PLA2

independently of the status of ApoB-100 conformation.

These comments, complementary to those performed by Reviewer 1, have been

jointly integrated in the Discussion as following_ “Two different

explanations about the protective effect of peptides against SMase-

induced LDL lipolysis have been explored. By one side, we have explored

whether there is a direct effect of peptides on SMase activity. This

possibility has been discarded since SMase activity assays clearly show

that peptides does not exert any effect on SMase functional activity.

Other possibility that has been explored is a potential interaction of

peptides with the LDL phospholipid surface. Several peptides and proteins

bearing amphipathic properties have been reported to be able to block LDL

aggregation by a direct interaction with LDL phospholipid surface. In

this respect, common examples are 4F peptide (Nguyen SD et al, J Lipid

Res 2015) or different apolipoproteins (Liu H et al, FEBS Lett 1993).

Despite the lack of amphipathic properties in LRP1-derived peptides, this

hypothesis has been considered as possible mechanism of action for the

peptides described herein. Theoretical molecular docking calculations

performed in the present study indicate a lack of association between

peptides and LDL phospholipid surface. A key point that offer clues about

how peptides protect SM against SMase-induced hydrolysis is their

unability to protect PC against PLA2-induced hydrolysis. We propose that

the differential anti-phospholypolytic effects of the peptide on SM and

PC phospholipids could be explained by the particular topological

distribution of SM and PC on the LDL surface. Although SM, compared to

PC, is a minor phospholipid in LDL (185 versus 450 molecules/LDL), SM is

localized in cholesterol-enriched environments, key regulators of

phospholipolysis (Hevonoja T et al, Biochem Biophys Acta 2000). We

propose that peptide preservation of ApoB-100 conformation directly

impacts on the maintenance of the cholesterol-enriched environment

structure since, according to our molecular modeling studies, ApoB-100

sequences complexed with peptide are those close to cholesterol in the

LDL surface. Therefore, SM would be peptide-protected against

phospholipolysis by its privilege of being part of cholesterol-enriched

domains, whose structure would be preserved through peptide-ApoB-100

conformational stabilization. In contrast, PC, belonging to cholesterol-

poor domains, is unprotected against PLA2, independently of the status of

ApoB-100 conformational stabilization”.

2-The authors indicated at the end of the discussion that "LRP1-derived

peptides emerge as tools potentially useful to prevent atherosclerosis

and cardiovascular complications, in particular in individuals with

aggregation-prone LDL such as obese and diabetic patients. » This is a

very interesting point but the authors should discuss a little bit more

the possibility to use these peptides in a therapeutic way. How do they

envisage to deliver these peptides in the future? (Are these peptides

stable? Do they consider local delivery after a first cardiovascular

event to decrease recurrence risk as a possible strategy? Or systemic

delivery…?

We thank the Reviewer for these interesting comments that will indeed

increase the potential translational impact of our manuscript.

In the future, we are going to test the effect of the retro-enantio

peptide (increased stability) in in vivo models of atherosclerosis. We

are mainly interested in the capacity of DP3 to reduce LDL retention and

LDL-induced smooth muscle cell (SMC)-foam cell formation in the vascular

wall.

Our first option will be to inject DP3 subcutaneously (not focal delivery

at the moment) taking advantage that atherosclerotic-prone areas of the

vasculature in in vivo models of angioplasty-induced atherosclerosis have

increased endothelial permeability.

These comments have been included in the Discussion (part conclusions) of

the revised ms._“In the future, we are going to test the efficacy of the

retro-enantio peptide version (DP3) to reduce vascular LDL retention and

LDL-induced smooth muscle cell (SMC)-foam cell formation in in vivo

models of atherosclerosis. Our first option in the near future will be to

inject the peptide DP3 subcutaneously (not focal delivery at the moment)

in a murine model of angioplasty-induced atherosclerosis taking

advantage of the increased endothelial permeability the vasculature of in

vivo models of angioplasty-induced atherosclerosis. In a not too distant

future, we expect LRP1-derived peptides to emerge as tools potentially

useful to prevent atherosclerosis and cardiovascular complications, in

particular in individuals with aggregation-prone LDL as obese and

diabetic patients”.

Editor-in-Chief

BBA_Biomembranes

Editorial Office

Barcelona, 2019, April 9th

Dear Editor,

Enclose please find our article BBAMEM-19-17 (R1) entitled “Molecular basis for the protective effects of

low-density lipoprotein receptor-related protein 1 (LRP1)-derived peptides against LDL aggregation”

to be considered for publication in BBA_Biomembranes.

We have addressed, point-by-point, all the issues raised by the Reviewers. According to their suggestions,

we have modified Methods, Results and Discussion sections of the manuscript. As a result, we think that the

manuscript has been deeply improved. We have changed each section of the manuscript by including new

information that has highly increased the impact of reported results.

We hope that the manuscript will be now acceptable for publication in BBA Biomembranes.

All authors have read and approved submission of the manuscript and the manuscript has not been published

and is not being considered for publication elsewhere in whole or part in any language except as an abstract.

Roger Prades, Chiara Pallara and Teresa Tarragó are employees of Iproteos SL. Teresa Tarragó is

cofounder of Iproteos SL and member of its board of directors.

None of the rest of authors have any conflict of interest with the publication of the manuscript.

We thank you for your time and effort.

Sincerely

Dr. Vicenta Llorente Cortes, Lipids and Cardiovascular Pathology group, Biomedical Research Institute IIB

Sant Pau, IIBB-CSIC, Hospital de la Santa Creu i Sant Pau, Sant Antoni Mª Claret, 167, 08025 Barcelona.

Phone: +34 935565888, Fax: +34 935565559, E-mail: [email protected]; [email protected]

Cover Letter

BBAMEM-19-17 (R1)

1

ANSWERS TO REVIEWERS

Manuscript No.: BBAMEM-19-17

Title: Molecular basis for the protective effects of low-density lipoprotein receptor-

related protein 1 (LRP1)-derived peptides against LDL aggregation

Article Type: Regular Paper

Journal Title: BBA - Biomembranes

Corresponding Author: Dr. Vicenta Llorente Cortés

Submit Date: Jan 20, 2019

Editor-in-Chief

BBA_Biomembranes

Editorial Office

Barcelona, 2019, April 9th

Dear Editor,

Enclose please find our article BBAMEM-19-17 (R1) entitled “Molecular basis for the

protective effects of low-density lipoprotein receptor-related protein 1 (LRP1)-derived

peptides against LDL aggregation” to be considered for publication in BBA_Biomembranes.

We have addressed, point-by-point, all the issues raised by the Reviewers. According to

their suggestions, we have modified Methods, Results and Discussion sections of the

manuscript. As a result, we think that the manuscript has been deeply improved. We have

changed each section of the manuscript by including new information that has highly increased

the impact of reported results.

We hope that the manuscript will be now acceptable for publication in BBA Biomembranes.

All authors have read and approved submission of the manuscript and the manuscript has not

been published and is not being considered for publication elsewhere in whole or part in any

language except as an abstract.

Roger Prades, Chiara Pallara and Teresa Tarragó are employees of Iproteos SL. Teresa Tarragó

is cofounder of Iproteos SL and member of its board of directors.

None of the rest of authors have any conflict of interest with the publication of the manuscript.

We thank you for your time and effort.

Response to Reviewers

BBAMEM-19-17 (R1)

2

Sincerely

Dr. Vicenta Llorente Cortes, Lipids and Cardiovascular Pathology group, Biomedical Research

Institute IIB Sant Pau, IIBB-CSIC, Hospital de la Santa Creu i Sant Pau, Sant Antoni Mª Claret,

167, 08025 Barcelona. Phone: +34 935565888, Fax: +34 935565559, E-mail:

[email protected]; [email protected]

BBAMEM-19-17 (R1)

3

We thank the reviewer for the valuable suggestions and critical comments that have

enhanced the quality of the manuscript.

Reviewer #1: The manuscript is a well-written report on the effect of two peptides

derived from LRP1 on the inhibition of enzyme-induced aggregation of LDL. Overall the

experiments are well designed and executed, and the results are presented clearly

showing that DP3 and LP3 peptides inhibit LDL aggregation.

It would help if the authors provide more detail on the rational of using the retro-enantio

peptides, it would help if this is included in the first paragraph of the result section. This

is relevant as the authors clearly show that DP3 is more active than LP3, and an

explanation in the discussion would be welcome. Further, provide detail about the

blocking of the termini in the methods section (in particular explain "H" on the N-

terminus). This is important as DP3 is more active than LP3. This can also be

discussed in the discussion.

We thank the Reviewer by this comment that will help to understand the rational to use

retro-enantio peptides. Peptide drugs offer high activity, high specificity and low toxicity,

but are susceptible to proteolytic degradation. A strategy to overcome this problem is to

produce peptides using non-natural or D-amino acids, which usually are not recognized

by plasma proteases. However, in most of the cases, the all-D peptide version

(enantio) of an active L-peptide is not active or show very low activity because the

opposite orientation of the amino acids side-chains compared to the parent peptide,

which challenges the recognition of the enantio peptide by its target receptor.

Nevertheless, in the literature there are some examples with enantio versions of

peptides that do show activity (Soto C et al, Biochem Biophys Res Commun 1996;

Chalifour RJ et al, J Biol Chem 2003). A more conservative strategy to preserve

peptide activity using D-amino acids consists in applying the retro-enantio approach.

The idea behind this approach is to simultaneously invert the sequence and chirality of

the peptide (Chorev M et al, Trends Biotechnol 1995; Jameson BA et al, Nature 1994)

In this way, the amine bone of the retro-enantio version is shift one unit but the

orientation of the side chains remain the same:

Usually, using this approach the retro-enantio peptide retains the activity of the parent

peptide (Prades R et al, Angew Chem Int Ed 2015). This explanation has been

summarized in the first paragraph of Results (part 3.1) and expanded in the Discussion

section of the revised ms.

In the first paragraph of the result (part 3.1)_“Peptide drugs offer high activity, high

specificity and low toxicity, but are susceptible to proteolytic degradation. A

strategy to overcome this problem is to produce peptides using non-natural or

D-amino acids, which shows improved stability. Although all-D peptides are not

recognized by plasma proteases, they may be less active than their

corresponding all-L peptides because of differences in the orientation of amino

acid side-chains. To preserve the orientation of the side-chains of the parent

peptide in a D-analogue, the D-amino acid residues can be linked in the reverse

order to that of the parent peptide in order to produce a retro-enantio peptide.

This approach emphasizes the maintenance of the topological native orientation

of the side-chains but not of the backbone, where the amide bond is shift one

BBAMEM-19-17 (R1)

4

unit (Chorev M et al, Trends Biotechnol 1995). By using this approach, peptides

usually preserve their activity while notably improve their plasma stability. In the

literature there are examples where this approach has been successfully used

(Prades R et al, Angew Chem Int Ed 2015)”.

In the Discussion section we have added the following explanation (first

paragraph)_“we tested both the original LP3 version and its retro-enantio

version DP3. Although all-D peptides are not recognised by proteases, and thus

have higher stability, they may be less active than their corresponding all-L

peptides because of differences in the orientation of amino acid side-chains. To

preserve the orientation of the side-chains of the parent L-peptide in a D-

analogue, the retro-enantio approach (Soto C et al, Biochem Biophys Res

Commun 1996; Chalifour RJ et al, J Biol Chem 2003) has been used in this

study. Here, we demonstrate that LP3 and its derived retro-enantio version

(DP3) per se have a high capacity to inhibit LDL aggregation induced by SMase

and PLA2. In this regard, the preservation of the topological side-chain

orientation of DP3 together with a high stability in biological samples account for

its notable performance in the inhibition assays.”

In the reference section we have added three new references

Further, provide detail about the blocking of the termini in the methods section (in

particular explain "H" on the N-terminus). Peptides are always written from N to C

terminus (N->C). The peptides described in this manuscript have an amide at their C-

terminus and we have indicated so by typing –NH2 at this termini. The way of indicating

this could be misleading for readers not familiar with peptide nomenclature, since these

could think that the peptide sequences is typed from C to N. To clearly indicate the N

and C terminus of the peptide we added the H- at the N-terminal part of the sequence

to indicate that it finalizes as amine.

To avoid any confusion in this regard, this explanation has been included in the

Methods section of the revised ms_“Peptide sequences are written from N to C, H-

at the end terminus indicate amine ending, whereas –NH2 at the C terminus indicates

amide”.

It is evidently shown that the two peptides are able to inhibit the SMase and PLA2

induced LDL aggregation. Can the authors show that the peptides associate with the

LDL surface prior or after enzyme treatment?

Thanks to the Reviewer by this interesting question. Limited trypsin proteolysis

combined with Mass spectrometry-based proteomics was the strategy used to analyze

this key aspect mentioned by the Reviewer. This strategy allows to identify the

sequences of ApoB-100 in native LDL protected by the peptide against trypsin-induced

proteolysis. Five sequences of ApoB-100 in native LDL were protected from

proteolysis. The peptide protection was obviously temporal, it was observed at 1 hour

and disappeared at 3 hours (Figure 7A). These results evidenced that the peptide

interacts with the LDL surface prior enzymatic treatment, protecting certain sequences

against proteolysis.

We have remarked this interesting point in the discussion section of the revised

manuscript _ “Limited trypsin proteolysis combined with Mass spectrometry-based

BBAMEM-19-17 (R1)

5

proteomics of ApoB-100/DP3 complexes evidenced that the peptide interacts with the

LDL surface prior enzymatic treatment”.

Is it possible that the peptides bind directly to the enzyme thereby preventing LDL

aggregation? This possibility should be discussed.

We have tested this possibility by exploring the effect of peptides on SMase activity in

the current incubation conditions. As shown in Supplemental Figure 6, SMase activity,

measured in relation to the amount of phosphorylcholine generated, was similar in

absence or presence of the peptides, indicating that, although we cannot discard a

potential interaction between peptide and SMase, it is clear that this interaction has not

effect on SMase activity. This comment has been included in the Discussion of the

revised ms. as following: “Although we can not discard a potential interaction between

peptides and SMase, SMase activity assays clearly show that peptides does not exert

any effect on SMase functional activity”.

The authors discuss the appearance of non-polar spots on the surface that trigger

aggregation (page 20). Is it possible that the peptides associate with these spots

(hydrophobic defects?), as has been shown for amphipathic helices of apolipoproteins?

(4F is an amphipathic peptide). This has been reported in a manuscript by Liu et al, in

which PL-C treated LDL resulted in the recruitment of apolipoproteins, thereby offering

protection against aggregation. (Liu, H., Scraba, D.G., and Ryan, R.O. 1993 Prevention

of phospholipase-C induced aggregation of low density lipoprotein by amphipathic

apolipoproteins, FEBS Lett. 316, 27-33.)

Thanks to the Reviewer for this interesting observation. As the Reviewer comments,

several peptides and proteins bearing amphipathic properties have been reported to be

able to block LDL aggregation by a direct interaction with LDL phospholipid surface. In

this respect, common examples are 4F peptide (Nguyen SD et al, J Lipid Res 2015) or

different apolipoproteins (Liu H et al, FEBS Lett 1993). Despite the lack of amphipathic

properties in any of the LRP1-derived peptides, this hypothesis was even considered

as possible mechanism of action for the peptides described herein.Thus, as a

preliminary test, molecular ligand docking was performed using a phospholipidic

monolayer mimicking LDL surface (Hevonoja T et al, Biochem Biophys Acta 2000) as

receptor and DP3 peptide as ligand. The top ranked docking poses showed rather low

binding energy values (around -4 kcal/mol) and mostly unreasonable binding modes.

Despite their theoretical character, these results support that there is not a direct

association between LRP1-derived peptides and LDL phospholipid surface. This

explanation has been summarized in the Results (part 3.7) and expanded in the

Discussion section of the revised ms.

In Result (part 3.7)_”Molecular docking calculations were perfomed using a

phospholipidic monolayer mimicking LDL surface (Hevonoja T et al, Biochem Biophys

Acta 2000) as receptor and DP3 peptide as ligand. The top ranked docking poses

showed rather low binding energy values (around -4 kcal/mol) and mostly

unreasonable binding modes. This theoretical calculations support that peptides do not

associate with LDL phospholipid surface”.

This argumentation has been expanded in the Discussion section_ Two different

explanations about the protective effect of peptides against SMase-induced LDL

lipolysis have been explored. By one side, we have explored whether there is a direct

effect of peptides on SMase activity. This possibility has been discarded since SMase

activity assays clearly show that peptides does not exert any effect on SMase

functional activity. Other possibility that has been explored is a potential interaction of

BBAMEM-19-17 (R1)

6

peptides with the LDL phospholipid surface. Several peptides and proteins bearing

amphipathic properties have been reported to be able to block LDL aggregation by a

direct interaction with LDL phospholipid surface. In this respect, common examples are

4F peptide (Nguyen SD et al, J Lipid Res 2015) or different apolipoproteins (Liu H et al,

FEBS Lett 1993). Despite the lack of amphipathic properties in LRP1-derived peptides,

this hypothesis has been considered as possible mechanism of action for the peptides

described herein. Theoretical molecular docking calculations performed in the present

study indicate a lack of association between peptides and LDL phospholipid surface”

New references about this issue have been included in the Reference section_

One new reference has been added to the reference section of the revised ms.

Fig 1 legend. Indicate that turbidity was measured by the absorbance at 405 nm.

We have included this indication in the Figure 1 legend of the revised ms.

Table S3, indicate that this is a molar ratio.

We have included this indication in Table S3 of the revised ms.

Reviewer #3: The manuscript is a comparative study among different peptides and

their effects on the aggregation of sphingomyelinase- (SMase) and phospholipase 2

(PLA2)-modified LDL particles.

The manuscript is well-written. The study is interesting. The peptides were succesfully

synthetized by solid-phase peptide synthesis as reported in table S1.

The manuscript is ready for publication.

We thank the Reviewer for his/her positive evaluation of our manuscript

BBAMEM-19-17 (R1)

7

Reviewer #4: In the manuscript entitled "Molecular basis for the protective effects of

low-density lipoprotein receptor-related protein 1 (LRP1)-derived peptides against LDL

aggregation » the authors have investigate the effect of two peptide (LP3 and DP3) on

the structural characteristics of SMase and PLA2 modified LDL particles. They showed

by turbidimetry, gel filtration chromatography and tomography electronic microscopy

(TEM) analysis that LP3 and DP3 peptides strongly inhibited SMase- and PLA2-

induced LDL aggregation. They showed that :

LRP1-derived peptides prevent SMase-induced alterations in LDL particle size, electric

charge and phospholipid content but do not prevent PLA2-induced alterations in LDL

particle size, electric charge and phospholipid content. Their data demonstrate that

LRP1-derived peptides are protective against LDL aggregation, through their capacity

to bind to ApoB-100 regions critical to preserve ApoB-100 conformation.

The article is well written and easy to read. The authors have convincing data

demonstrating mechanism involved by these both peptides and provide evidence that

these peptides could be promising tools to prevent LDL aggregation in the arterial

intima. This work present interesting data that could be published with some minor

complementary informations listed below:

1- In the discussion section, the authors should better explain why both peptides

exerted protection against conformational alterations of poB-100 induced by both

SMases and PLA2 whereas they didn't counteract particle size charge and

phospholipids induced by PLA2.

We thank the Reviewer by this interesting comment. LRP1-derived peptides preserved

ApoB-100 conformation, sphyngomyelin (SM) content and LDL size in SMase-LDL

while in PLA2-LDL, peptides preserve only ApoB-100 conformation but not

phosphatidylcholine (PC) content or LDL size. We propose that the differential effect of

LRP1-derived peptides in these two modified LDLs could be explained by topological

reasons. Although sphingomyelin (SM), compared to phosphatidylcholine (PC), is a

minor phospholipid in LDL (185 versus 450 molecules/LDL), SM, the target

phospholipid for SMase, is localized in cholesterol-enriched environments, while PC is

associated with poor-cholesterol environment. Surface cholesterol seems to be a key

regulator of the process of phospholipolysis (Hevonoja T et al, Biochem Biophys Acta

2000) and thus, could modulate SM but not PC lipolysis. We propose that peptide

preservation of ApoB-100 conformation directly impacts on the maintenance of the

cholesterol-enriched environment structure since, according to our molecular modeling

studies, ApoB-100 sequences interacting with peptide are those close to cholesterol in

the LDL surface. Therefore, SM would be protected against SMase phospholipolysis by

its privilege of being part of cholesterol-enriched domains, whose structure is likely

dependent on the maintenance of ApoB-100 conformation. In contrast, PC, belonging

to cholesterol-poor domains, seems to be unprotected against PLA2 independently of

the status of ApoB-100 conformation.

These comments, complementary to those performed by Reviewer 1, have been

jointly integrated in the Discussion as following_ “Two different explanations about

the protective effect of peptides against SMase-induced LDL lipolysis have been

explored. By one side, we have explored whether there is a direct effect of peptides on

SMase activity. This possibility has been discarded since SMase activity assays clearly

show that peptides does not exert any effect on SMase functional activity. Other

possibility that has been explored is a potential interaction of peptides with the LDL

phospholipid surface. Several peptides and proteins bearing amphipathic properties

have been reported to be able to block LDL aggregation by a direct interaction with

BBAMEM-19-17 (R1)

8

LDL phospholipid surface. In this respect, common examples are 4F peptide (Nguyen

SD et al, J Lipid Res 2015) or different apolipoproteins (Liu H et al, FEBS Lett 1993).

Despite the lack of amphipathic properties in LRP1-derived peptides, this hypothesis

has been considered as possible mechanism of action for the peptides described

herein. Theoretical molecular docking calculations performed in the present study

indicate a lack of association between peptides and LDL phospholipid surface. A key

point that offer clues about how peptides protect SM against SMase-induced hydrolysis

is their unability to protect PC against PLA2-induced hydrolysis. We propose that the

differential anti-phospholypolytic effects of the peptide on SM and PC phospholipids

could be explained by the particular topological distribution of SM and PC on the LDL

surface. Although SM, compared to PC, is a minor phospholipid in LDL (185 versus

450 molecules/LDL), SM is localized in cholesterol-enriched environments, key

regulators of phospholipolysis (Hevonoja T et al, Biochem Biophys Acta 2000). We

propose that peptide preservation of ApoB-100 conformation directly impacts on the

maintenance of the cholesterol-enriched environment structure since, according to our

molecular modeling studies, ApoB-100 sequences complexed with peptide are those

close to cholesterol in the LDL surface. Therefore, SM would be peptide-protected

against phospholipolysis by its privilege of being part of cholesterol-enriched domains,

whose structure would be preserved through peptide-ApoB-100 conformational

stabilization. In contrast, PC, belonging to cholesterol-poor domains, is unprotected

against PLA2, independently of the status of ApoB-100 conformational stabilization”.

2-The authors indicated at the end of the discussion that "LRP1-derived peptides

emerge as tools potentially useful to prevent atherosclerosis and cardiovascular

complications, in particular in individuals with aggregation-prone LDL such as obese

and diabetic patients. » This is a very interesting point but the authors should discuss a

little bit more the possibility to use these peptides in a therapeutic way. How do they

envisage to deliver these peptides in the future? (Are these peptides stable? Do they

consider local delivery after a first cardiovascular event to decrease recurrence risk as

a possible strategy? Or systemic delivery…?

We thank the Reviewer for these interesting comments that will indeed increase the

potential translational impact of our manuscript.

In the future, we are going to test the effect of the retro-enantio peptide (increased

stability) in in vivo models of atherosclerosis. We are mainly interested in the capacity

of DP3 to reduce LDL retention and LDL-induced smooth muscle cell (SMC)-foam cell

formation in the vascular wall.

Our first option will be to inject DP3 subcutaneously (not focal delivery at the moment)

taking advantage that atherosclerotic-prone areas of the vasculature in in vivo models

of angioplasty-induced atherosclerosis have increased endothelial permeability.

These comments have been included in the Discussion (part conclusions) of the

revised ms._“In the future, we are going to test the efficacy of the retro-enantio peptide

version (DP3) to reduce vascular LDL retention and LDL-induced smooth muscle cell

(SMC)-foam cell formation in in vivo models of atherosclerosis. Our first option in the

near future will be to inject the peptide DP3 subcutaneously (not focal delivery at the

moment) in a murine model of angioplasty-induced atherosclerosis taking advantage

of the increased endothelial permeability the vasculature of in vivo models of

angioplasty-induced atherosclerosis. In a not too distant future, we expect LRP1-

derived peptides to emerge as tools potentially useful to prevent atherosclerosis and

cardiovascular complications, in particular in individuals with aggregation-prone LDL as

obese and diabetic patients”.

ApoB-100/DP3 complex

STABILIZATION OF ApoB-100 CONFORMATION

SMase PLA2

STABILIZATION OF CHOLESTEROL/SM

enriched environment

Unaltered PL Surface Unaltered LDL size

NO EFFECT ON PC-enriched/colesterol-poor

environment

Altered PL Surface Decreased LDL size

LDL aggregation

Figure 8. Schematic representation of the molecular basis for the protective effect of LRP1-derived peptides against SMase- and PLA2-induced

LDL aggregation. DP3 forms a complex with ApoB-100 and this molecular interaction stabilizes ApoB-100 conformation. ApoB-100 conformation

stabilization might guarantee the structural preservation of surface colesterol-enriched environments, where SM is located. Structural preservation

of cholesterol, a key regulator of phospholipolysis, would protect SM from SMase activity. As a result, LDL complexed with DP3 remains unaltered

when exposed to SMase. This scenario changes when LDL complexed with DP3 is exposed to PLA2. The target for PLA2 is PC, phospholipid

associated with poor-colesterol environments. Therefore, PC is unprotected against the attack of PLA2, that hydrolyzes PC producing lysoPC and

NEFA. Remarkably, LDL complexed with DP3 is protected against SMase and PLA2-induced aggregation even in conditions of extreme

phospholipolysis, indicating that the maintenance of ApoB-100 conformation is enough to prevent LDL aggregation.

Lyso-PC & NEFA

*Graphical Abstract (for review)

Highlights

LRP1-derived peptides prevent SMase-induced alterations in LDL particle size,

electric charge and phospholipid content.

LRP1-derived peptides do not prevent PLA2-induced alterations in LDL particle

size, electric charge and phospholipid content.

LRP1-derived peptides prevent lipolytic-induced LDL aggregation through

electrostatical interaction with basic ApoB-100 sequences key for preservation

of ApoB-100 conformation.

Highlights (for review)

Manuscript BBAMEM-19-17 (R1)

1

Molecular basis for the protective effects of low-density lipoprotein receptor-related protein 1

(LRP1)-derived peptides against LDL aggregation

Short title: LRP1-derived peptides and LDL aggregation

Aleyda Benitez-Amaroa,b, Chiara Pallarac, Laura Nasarrea, Andrea Rivas-Urbinad, Sonia Benitezd,

Angela Veaa, Olga Bornacheaa,b, David de Gonzalo-Calvoa,b,e, Gabriel Serra-Mirf, Sandra Villegasf,

Roger Pradesc, José Luís Sanchez-Quesadad,g, Cristina Chivah,i, Eduard Sabidoh,i, Teresa

Tarragóc, and Vicenta Llorente-Cortésa,b,e**

aGroup of Lipids and Cardiovascular Pathology. Biomedical Research Institute Sant Pau (IIB Sant

Pau), Hospital de la Santa Creu i Sant Pau. Barcelona. Spain.

bInstitute of Biomedical Research of Barcelona (IIBB). Spanish National Research Council (CSIC),

Barcelona, Spain.

cIproteos S.L., Barcelona Science Park (PCB), Barcelona, Spain.

dCardiovascular Biochemistry Group, Research Institute of the Hospital de Sant Pau (IIB Sant Pau),

Barcelona, Spain.

eCIBER enfermedades cardiovasculares (CIBERcv).

fProtein Design and Immunotherapy Group. Departament de Bioquímica i Biologia Molecular,

Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.

gCIBER diabetes y enfermedades metabólicas asociadas (CIBERdem)

hProteomics Unit, Centre de Regulació Genòmica, Barcelona Institute of Science and Technology,

Barcelona, Spain.

iUniversitat Pompeu Fabra, Barcelona, Spain.

**To whom correspondence should be addressed: Vicenta Llorente-Cortés, Lipids and

Cardiovascular Pathology group leader, ICCC Program, Biomedical Research Institute IIB Sant

Pau, Hospital de la Santa Creu i Sant Pau, Sant Antoni Mª Claret, 167, 08025 Barcelona. Phone:

+34 935565888, Fax: +34 935565559, E-mail: [email protected]; [email protected]

*REVISED Manuscript (text UNmarked)Click here to view linked References

Manuscript BBAMEM-19-17 (R1)

2

Abstract

Aggregated LDL is the first ligand reported to interact with the cluster II CR9 domain of low-density

lipoprotein receptor-related protein 1 (LRP1). In particular, the C-terminal half of domain CR9,

comprising the region Gly1127-Cys1140 exclusively recognizes aggregated LDL and it is crucial for

aggregated LDL binding. Our aim was to study the effect of the sequence Gly1127-Cys1140 (named

peptide LP3 and its retro-enantio version, named peptide DP3) on the structural characteristics of

sphingomyelinase- (SMase) and phospholipase 2 (PLA2)-modified LDL particles. Turbidimetry, gel

filtration chromatography (GFC) and transmission electronic microscopy (TEM) analysis showed

that LP3 and DP3 peptides strongly inhibited SMase- and PLA2-induced LDL aggregation.

Nondenaturing polyacrylamide gradient gel electrophoresis (GGE), agarose gel electrophoresis and

high-performance thin-layer chromatography (HPTLC) indicated that LP3 and DP3 prevented

SMase-induced alterations in LDL particle size, electric charge and phospholipid content,

respectively, but not those induced by PLA2. Western blot analysis showed that LP3 and DP3

counteracted changes in ApoB-100 conformation induced by the two enzymes. LDL proteomics

(LDL trypsin digestion followed by mass spectroscopy) and computational modeling methods

evidenced that peptides preserve ApoB-100 conformation due to their electrostatic interactions with

a basic region of ApoB-100. These results demonstrate that LRP1-derived peptides are protective

against LDL aggregation, even in conditions of extreme lipolysis, through their capacity to bind to

ApoB-100 regions critical for ApoB-100 conformational preservation. These results suggests that

these LRP1(CR9) derived peptides could be promising tools to prevent LDL aggregation induced by

the main proteolytic enzymes acting in the arterial intima.

Key words: Lipoprotein aggregation; LRP1-derived peptides; ApoB-100; SMase, PLA2,

atherosclerosis

Manuscript BBAMEM-19-17 (R1)

3

Abbreviations: agLDL, aggregated LDL; apo, apolipoprotein; CE, cholesteryl esters; FC, free

cholesterol; hcVSMC, human coronary vascular smooth muscle cells; GFC, gel filtration

chromatography; GGE, nondenaturing polyacrylamide gradient gel electrophoresis; HPTLC, high-

performance thin-layer chromatography; LC-MS/MS, liquid chromatography tandem-mass

spectrometry; LDL, low-density lipoprotein; LysoPC, lysophosphatidylcholine; LRP1, low-density

lipoprotein receptor-related protein 1; NEFA, nonesterified fatty acids; PC, phosphatidylcholine;

PLA2, phospholipase A2; PL, phospholipids; REMD, Replica Exchange Molecular Dynamics; SM,

sphingomyelin; SMase, sphingomyelinase; TEM, transmission electron microscopy

Manuscript BBAMEM-19-17 (R1)

4

1. Introduction

Ischemic heart disease is the first cause of death in Western countries, and myocardial infarction

accounts for about 50% of deaths from this disease. The Framingham study showed that

cardiovascular risk positively correlates with low-density lipoprotein (LDL)-cholesterol and inversely

with high-density lipoprotein (HDL)-cholesterol 1,2. High levels of LDL-cholesterol induce

alterations in the endothelium permeability, which in turn promote LDL influx into the arterial intima

3. Intimal LDL-cholesterol accumulation is a critical step in vascular cholesteryl ester (CE)

deposition, a process that increases the tendency of the atherosclerotic plaque to rupture, triggering

thrombosis and the development of ischemic cardiomyopathy 4-6. CEs in atherosclerotic plaques

are deposited both extra- and intra-cellularly. Extracellular deposition of LDL-CEs, a crucial initiating

event in atherosclerosis 7, is mediated by proteoglycans in the extracellular matrix of the arterial

intima. The electrostatic interaction between proteoglycans and LDL and the proteolytic/lipolytic

actions of enzymes on LDL are enhanced in the arterial intima and promote LDL retention and

aggregation 8,9. Two of the main enzymes acting on LDL are sphingomyelinase (SMase),

secreted by endothelial cells and macrophages 10,11 and phospholipase A2 (PLA2) 12,13. These

two enzymes play key roles in LDL aggregation in the arterial intima during atherogenesis 12-15.

Aggregated LDL (agLDL) has been detected and isolated from atherosclerotic plaques from animal

models and humans 11,16. Unlike native LDL, agLDL is a potent inducer of massive intracellular

CE accumulation both in macrophages 17-19 and human coronary vascular smooth muscle cells

(hcVSMCs) 20. In particular, in hcVSMCs, agLDL is actively taken up through the low-density

lipoprotein receptor-related protein 1 (LRP1) 21,22. The internalization of agLDL, in turn, induces

LRP1 expression 23,24, promoting a positive feedback loop that efficiently transforms hcVSMCs

into foam cells. We have also demonstrated that hcVSMC-foam cells synthesize and release high

amounts of tissue factor, which is crucial for the pro-thrombotic transformation of the vascular wall

and thus for the progression of atherosclerosis to thrombosis 25,26. The relevance of this

mechanism in atherosclerosis is evident since VSMCs are the main components of the vascular wall

Manuscript BBAMEM-19-17 (R1)

5

and more than 50% of foam cells previously considered to be monocyte-derived macrophages in

human atherosclerotic plaques originate from VSMCs 27. Together, these findings support the

notion that the generation of hVSMC-derived foam cells through LRP1-mediated agLDL uptake is a

key mechanism underlying cholesterol accumulation in vasculature susceptible to atherosclerosis.

Our group has identified LRP1 cluster II, in particular the region Gly1127-Cys1140 (peptide LP3: H-

GDNDSEDNSDEENC-NH2) that spans the C-terminal half of domain CR9, as pivotal for binding to

agLDL and subsequent internalization of this agLDL into human VSMCs 28. We hypothesize that

peptide LP3 or its retro-enantio version, peptide DP3, would counteract LDL aggregation. Using a

range of biochemical, biophysical and molecular experiments, here we demonstrate that LP3 and

DP3 peptides efficiently prevent LDL aggregation induced by the lipolytic enzymes SMase and

PLA2. Although LP3 and DP3 efficiently counteracted alterations in LDL particle size, charge and

SM loss induced by SMase, they were unable to prevent the alterations induced in the same

parameters by PLA2. However, both peptides protected ApoB-100 from conformational alterations

induced by SMase and PLA2. Proteomics, ab initio modeling and molecular dynamics showed that

LRP1-derived peptides establish stable electrostatic interactions with basic sequences of ApoB-100.

2. Material and Methods

2.1. Peptide sequences and synthesis

Peptide sequences are written from N to C, H- at the end terminus indicate amine ending, whereas

–NH2 at the C terminus indicates amide. The amino acid sequence of LP3 peptide (parent) is H-

GDNDSEDNSDEENC-NH2, DP3 is the retro-enantio version of LP3 without cysteine: H-

needsndesdndh-NH2, where the amino acid letter code in lowercase stands for amino acids with D-

chirality. As a negative control, we used an ApoB-100-based peptide coded as IP321 with the

sequence: H-RLTRKRGLK-NH2. We also compared the efficacy of LP3, DP3 and IP321 with the

previously described anti-atherosclerotic apoA-I mimetic, 4F. The latter is 18 amino acids long and

has the following sequence: Ac-DWFKAFYDKVAEKFKEAF-NH2 VSMCs 29. All peptides were

Manuscript BBAMEM-19-17 (R1)

6

synthesized by Iproteos by standard 9-fluorenylmethoxycarbonyl/tert-butyl (Fmoc/tBu) solid-phase

peptide synthesis. Syntheses were performed manually on a 100-µmols scale/each using Rink

amide ChemMatrix® resin. Peptide elongation and other solid-phase manipulations were done

manually in polypropylene syringes, each fitted with a polyethylene porous disk. Solvents and

soluble reagents were removed by suction. Washings between synthetic steps were done with

dimethylformamide and dichloromethane using 5 mL of solvents/g resin each time. Nα-Fmoc-

protected amino acids (4 equivalents) were coupled using 2-(1H-benzotriazole-1-yl)-1,1,3,3-

tetramethyluronium (TBTU, 4 equivalents), and N,N-diisopropylethylamine (8 equivalents). The

extent of the reaction was monitored using the Kaiser test (primary amines). During couplings, the

mixture was allowed to react with intermittent manual stirring. Fmoc group was removed by treating

the resin with 20% piperidine in DMF (3 mL/g resin). The peptides were cleaved from the resin using

a mixture of trifluoroacetic acid, triisopropylsilane and water (95:2.5:2.5). The crude products

obtained were purified in reverse phase using a semi-prep HPLC instrument equipped with a C18

column. The purity and identity of the synthesized peptides was assessed by HPLC, HPLC-MS and

MALDI-TOF analysis (Table S1).

2.2. Peptide stability in human serum and cell culture medium

The stability of the peptides was studied in human serum (Sigma-Aldrich). For the stability assay,

the peptides were dissolved in a mixture of 10% Hanks’ Balanced Solution Salt (HBSSx1) and

human serum. The final concentration of peptide was 200 mM VSMCs 30. The samples were

incubated at 37°C with orbital agitation for 24 h. At selected time points, serum aliquots were

extracted and the serum proteins of the mixture were then precipitated by adding methanol.

Samples were then centrifuged at 4°C, and the supernatant was filtered out and analyzed by HPLC.

Peptide stability during the assays was monitored using HPLC (UV detection at 220 nm).

2.3. LDL isolation and purification

Human LDL (d1.019-d1.063 g/mL) was obtained from pooled normolipemic human plasma by

sequential ultracentrifugation in a KBr density gradient. Briefly, very low density lipoproteins (VLDLs)

Manuscript BBAMEM-19-17 (R1)

7

were first discarded after spinning plasma at 36,000 rpm for 18 h at 4ºC using a fixed-angle rotor

(50.2 Ti, Beckman) mounted on an Optima L-100 XP ultracentrifuge (Beckman). Subsequently,

VLDL-free plasma was layered with 1.063 g/mL KBr solution and centrifuged at 36,000 rpm for 18 h

at 4ºC. LDLs were dialyzed against 0.02 M Trizma, 0.15 M NaCl, 1 mM EDTA, pH 7.5 for 18 h, and

then against normal saline for 2 h. Finally, isolated LDLs were filter-sterilized (0.22 μm Millex-GV

filter unit, Millipore). Protein concentration was determined using the BCA protein assay (Thermo

Scientific) and the cholesterol concentration with a commercial kit (IL test Cholesterol, Izasa).

2.4. Analysis of SMase activity in the presence and absence of peptides

The capacity of LP3 and DP3, IP321 and manumycin A (positive control) to inhibit

sphingomyelinase activity was measured using the Amplex Red Sphingomyelinase Assay Kit

(Molecular Probes Invitrogen Detection Technologies, #A12220), an assay that measures the

formation of phosphorylcholine. To measure SMase activity in the presence and absence of

peptides, DMSO volume and peptide concentrations were kept constant [10 M (molar ratio

peptide/ApoB= 4.7/1)] while SMase concentration was increased (0, 0.2, 0.4, 0.6, 0.8 and 1 mU/ml).

Each reaction contained 50 μM Amplex Red reagent, 1 U/mL HRP, 0,1 U/ml choline oxidase, 4 U/ml

of alkaline phosphatase, 0,25 mM sphingomyelin (diluted with Triton X-100 or with the Reaction

Buffer 1x). Reactions were incubated at 37ºC for 30 min. Fluorescence intensity was measured

using a fluorescence microplate reader (Victor 1420 multilabel counter) using excitation at 530 nm

and fluorescent detection at 590nm.

2.5. LDL modification by Sphingomyelinase (SMase) and Phospholipase A2 (PLA2) in

the presence and absence of peptides

Human LDL (1.44 mg/mL) were incubated with 40 U/L of Bacillus cereus SMase (Sigma-Aldrich,

Schnelldorf, Germany) or with 50 µg/L of type II secretory PLA2 from honey bee venom (Sigma-

Aldrich, Schnelldorf, Germany) in 20 mM Tris buffer (pH 7.0) containing 150 mM NaCl, 2 mM CaCl2,

and 2 mM MgCl2 at 37ºC. LDL incubation with SMase and PLA2 was performed at peptide

concentrations of 0, 2.5, 5, 7.7, 8.8, 10 µM peptide/ApoB-100 ratio :0/1, 1.2/1, 2.4/1, 3.5/1, 4.1/1,

Manuscript BBAMEM-19-17 (R1)

8

4.7/1) and 0, 4.2, 6.3, 10.0, 14.6, 20.8 µM peptide/ApoB-100 ratio (0/1, 2.0/1, 2.9/1, 4.7/1, 6.8/1,

9.8/1), respectively, for increased time periods. LDL lipolysis was stopped by addition of EDTA

(final concentration 10 mM).

2.6. Characterization of SMase- and PLA2-induced LDL alterations in the presence and

absence of peptides. A detailed information of the different procedures

2.6.1.LDL Composition_The LDL composition was measured by commercial methods adapted to a

Cobas 6000/c501 autoanalyzer (Roche Diagnostics, Indianapolis, IN, USA). Total cholesterol,

triglycerides and ApoB reagents were obtained from Roche Diagnostics. PLs, NEFAs, and free

cholesterol reagents were obtained from Wako Pure Chemicals (Tokyo Japan).

2.6.2.Turbidity_Sample turbidity (LDL aggregation monitorization) was determined by measuring

absorbance of 100 µl of LDLs (1.44 mg/mL ApoB) in a 96-well microplates at 405 nm.

2.6.3.Gel Filtration Chromatography (GFC)_To separate aggregated from non-aggregated LDL,

samples (50 µL at 1.44 mg/mL) were subjected to gel filtration chromatography (GFC) in two online

connected Superose 6 Increase 5/150 GL columns with an AKTA-FPLC system (GE Healthcare) at

a flow of 0.3 mL/min 31,32. The area under the curve of each peak was calculated using the

Unicorn 4.0 software (GE Healthcare).

2.6.4. Agarose gel electrophoresis_LDL (1.5 µL of LDL (1.44 mg/mL) was electrophoresed on 0.5%

agarose gels at 65V for 40 min followed by 90V for 15 min. Fixed gels were dehydrated and stained

with Coomassie Blue. Decolored gels were captured with the Bio-Rad GS-800 scanner.

2.6.5. SDS-PAGE and Western blot

ApoB-100 was detected by Western blot analysis after electrophoresis of LDL in 6% SDS-PAGE

gels. Nitrocellulose membranes were incubated with anti-ApoB-100 polyclonal antibodies (Roche

Diagnostics S.L., Ref: 3032574122, polyclonal anti-human ApoB-100, dilution 1: 5000) for 1 h at

Manuscript BBAMEM-19-17 (R1)

9

room temperature. After several washes, membranes were incubated with secondary anti-sheep

antibodies (dilution 1: 10000, Dako; Glostrup, Denmark). Bands were detected using ECL Prime

Western Blotting Detection Reagent (Amersham) and quantified by densitometry using a ChemiDoc

system and Quantity One software (Bio‐Rad, Hercules, CA, USA). The results are expressed as

arbitrary units of intensity.

2.6.6.TEM

TEM was performed as previously described 32. LDL was placed on a 400 mesh glow-discharge

carbon-coated grid, negatively stained with 2% potassium phosphotungstate, pH 7.0, and air dried.

Grids were placed in a Jeol 120-kV JEM-1400 (Jeol, Japan) transmission electron microscope. TEM

images were processed using ImageJ (Fiji) software. Briefly, contrast was enhanced, followed by

background subtraction.

2.6.7. Nondenaturing acrylamide gradient gel electrophoresis (GGE)

GGE was performed following Nichols et al., with small modifications 32. Two solutions at 2% and

16% were prepared by using a stock solution of acrylamide and bis-acrylamide (30% total, 5%

cross-linker) and mixed using 2 P-1 peristaltic pumps (Pharmacia). The different samples (5 μL at

1.44 mg/mL) were preincubated for 15 min with 10 μL Sudan black (0.1% [wt/vol]) in ethylene glycol

and 5 μL saccharose (50% [wt/vol]). Ten microliters of this mixture was electrophoresed at 4°C for

30 min at 20 V, 30 min at 70 V, and 4 h at 100 V. Bands were scanned by densitometry.

2.6.8. HPTLC

LDL (15 µg) was lipid-extracted with organic solvents as previously described by our group with

minimal modifications 25. Samples were applied to horizontal, one-dimensional high-performance

thin-layer chromatography (HPTLC) (Merck, HPTLC Silica Gel 60, 1.05641.0001). Phospholipids

were separated on HPTLC by chloroform:ethyl

acetate:acetone:isopropanol:ethanol:methanol:water:acetic acid (30:6:6:6:16:28:6:2); visualized by

Manuscript BBAMEM-19-17 (R1)

10

staining with 7.5% Cu-acetate (w/v), 2.5% CuSO4 (w/v), and 8% H3PO4 (v/v) in water. A mixture of

phospholipids (Sigma Ref pH-7) were also run as standards. The spots corresponding to the

different phospholipids were quantified by densitometry against the standard curve using a GS-800

Calibrated Densitometer (Bio-Rad).

2.7. Proteomics analyses of LDL samples

2.7.1. Sample preparation_LDL, LDL unexposed or exposed to SMase in the absence of peptide or

in the presence of DP3 or IP321) were directly subjected to limited trypsin digestion (1 µg) for 1 h or

3 h. In both cases, tryptic peptides were desalted using a C18 column and evaporated to dryness.

2.7.2. Mass spectrometry data acquisition_ LDL samples were analyzed using a LTQ-Orbitrap Velos

Pro mass spectrometer (Thermo Fisher Scientific, San Jose, CA, USA) coupled to an EasyLC

(Thermo Fisher Scientific (Proxeon), Odense, Denmark). Peptides were separated by reverse-

phase chromatography using a 25-cm column with an inner diameter of 75 μm, packed with 5 μm

C18 particles (Nikkyo Technos Co., Ltd. Japan). Chromatographic gradients started at 93% buffer A

and 7% buffer B and gradually increased to 65% buffer A / 35% buffer B in 60 min at a flow rate of

250 nl/min. The mass spectrometer was operated in positive ionization mode with nanospray

voltage set at 2.2 kV and source temperature at 250°C. Ultramark 1621 for the FT mass analyzer

was used for external calibration prior to the analyses, and an internal calibration was performed

using the background polysiloxane ion signal at m/z 445.1200. The acquisition was performed in

data-dependent acquisition mode, and full MS scans with 1 micro scans at a resolution of 60,000

were used over a mass range of m/z 350-2000 with detection in the Orbitrap. Auto gain control

(AGC) was set to 1E6, dynamic exclusion (60 seconds) and charge state filtering disqualifying singly

charged peptides was activated in each cycle of data-dependent acquisition analysis. After each

survey scan, the top ten most intense ions with multiple charged ions above a threshold ion count of

5000 were selected for fragmentation at normalized collision energy of 35%. Fragment ion spectra

produced via collision-induced dissociation (CID) were acquired in an Ion Trap apparatus. AGC set

Manuscript BBAMEM-19-17 (R1)

11

to 5e4, an isolation window of 2.0 m/z, activation time of 0.1ms and maximum injection time of 100

ms were used. All data were acquired with Xcalibur software v2.2.

2.7.3. Mass spectrometry data analysis

The spectra acquired for the LDL samples were analyzed using the Proteome Discoverer software

suite (v2.0, Thermo Fisher Scientific) and the Mascot search engine (v2.5, Matrix Science). The

data were searched against the Swiss-Prot human database (v.2017/10). At the MS1 level, a

precursor ion mass tolerance of 7 ppm was used, and up to three missed cleavages were allowed.

The fragment ion mass tolerance was set to 0.5 Da for MS2 spectra. Methionine oxidation and N-

terminal protein acetylation were defined as variable modifications, whereas carbamidomethylation

on cysteine residues was set as a fixed modification. False discovery rate (FDR) in peptide

identification was limited to a maximum of 5% using a decoy database. Quantitation data were

retrieved from the “Precursor ion area detector” node of Proteome Discoverer (v2.0) using 2 ppm

mass tolerance for the peptide extracted ion current (XIC). Finally, for each LDL sample, Skyline

software v4.1 was used to build a spectral library for the ApoB100 protein with the corresponding

identified spectra and to extract all its MS1 areas. Extracted ion chromatograms for the different LDL

samples have been deposited to Panorama with identified PXD011261; URL

(https://panoramaweb.org/j8CaUy.url).

2.8. Molecular modeling of peptide-LDL interactions

The structural model of ApoB-100 in complex with DP3 peptide was built using rigid body molecular

docking simulation and starting from the isolated ab initio models of both docking partners. First of

all, an exhaustive conformational sampling was performed on the peptide in order to identify the

most stable and populated conformational states, which were assumed to be the most favorable to

bind ApoB-100 protein. For that, on one hand, a 3D structure of DP3 was designed from scratch

and consequently used as input for Replica Exchange Molecular Dynamics (REMD) simulations in

implicit water using the AMBER ff12SB force field 33 and NAMD software 34. In each REMD

scheme, 16 independent 100 ns-long simulations (replicas) were performed simultaneously at

Manuscript BBAMEM-19-17 (R1)

12

temperatures ranging from 300 to 613 K. Finally, the representative structure of the most populated

cluster was extracted and used as input structure for the docking simulation. On the other hand, the

3D structure of ApoB-100 protein was built by ab initio protein modeling, since no crystal structure of

protein was available and very low sequence identity templates (ranging from 8 to 13%) were found

in the PDB. Indeed, this computational technique tends to require vast resources, thus only a small

domain of roughly 100 residues around the basic region believed to be responsible for LDL receptor

binding (3189-3261 residues) was selected for modeling. Consequently, a total of 10 ab initio

models were built using ROBETTA server 35 and finally used as input structures for the docking

simulation. Starting from each ApoB-100 protein structure and DP3 peptide previously obtained, 10

independent rigid-body docking simulations were performed using AutoDock Vina software fork

Smina 36 and setting the docking grid box around Apo-B100 3227IKFDKYKAEK3236 region. Finally,

all the docking results were merged and the lowest-energy poses were visually inspected.

2.9. Statistical analysis

Data are presented as mean±SD. One-way ANOVA followed by the Bonferroni or the Storey (q-

value) correction was performed for comparison of >2 groups. Student’s t test was performed for

comparison of 2 groups. P<0.05 was considered significant.

3. Results

3.1. DP3 shows higher stability than the parent peptide LP3 in human serum

Peptide drugs offer high activity, high specificity and low toxicity, but are susceptible to proteolytic

degradation. A strategy to overcome this problem is to produce peptides using non-natural or D-

amino acids, which shows improved stability. Although all-D peptides are not recognized by plasma

proteases, they may be less active than their corresponding all-L peptides because of differences in

the orientation of amino acid side-chains. To preserve the orientation of the side-chains of the

parent peptide in a D-analogue, the D-amino acid residues can be linked in the reverse order to that

of the parent peptide in order to produce a retro-enantio peptide. This approach emphasizes the

Manuscript BBAMEM-19-17 (R1)

13

maintenance of the topological native orientation of the side-chains but not of the backbone, where

the amide bond is shift one unit 37. By using this approach, peptides usually preserve their activity

while notably improve their plasma stability. In the literature there are examples where this approach

has been successfully used 30. LP3 and DP3 were synthesized by means of solid-phase peptide

synthesis using a Fmoc/tBu strategy. The purity of both peptides (≥98%) was assessed by HPLC

analysis (Supplemental Fig. S1). Other parameters indicative of the purity and identity of the

synthesized peptides were obtained by HPLC-MS and MALDI-TOF analysis (Supplemental Table

S1). To study the stability of the parent peptide (LP3), it was dissolved in a mixture of HBSS (x1)

and human serum. LP3 showed moderate stability with a calculated half-life of 8 h (Supplemental

Fig. S2A). To achieve an increase in the stability of LP3 for the purpose of in vivo studies, a retro-

enantio version of the peptide (DP3) was synthesized. This approach consisted of using all D-amino

acids and reversing the sequence of the parent peptide. This synthetic strategy has been shown to

be useful 38. As the retro-enantio version of the peptide is synthesized using D-amino acids, the

sequence is not recognized by serum proteases, thus extending its half-life considerably

(Supplemental Fig. S2B).

3.2. Time- and dose-response blocking effects of LP3 and DP3 on SMase- and PLA2- induced

LDL turbidity

To study the effects of the peptides on SMase-induced LDL aggregation, LDL was incubated with

SMase and PLA2 in the presence and absence of LP3, DP3 and IP321 (negative control) under the

experimental conditions detailed in Supplemental Fig. S3. The peptide concentrations and

peptide/ApoB-100 molar ratios tested in the aggregation of SMase-modified LDL (SMase-LDL) are

detailed in Supplemental Table S2. LP3 and DP3 reduced SMase-LDL aggregation at all the times

tested (4, 6 and 18 h) in a dose-dependent manner (Fig. 1A). DP3 showed a higher inhibitory

capacity than LP3 at concentrations of 5.0, 7.7 and 8.8 µM, and SMase-LDL aggregation was fully

inhibited by LP3 at 10 µM (peptide/ApoB-100 ratio: 4.7/1) and by DP3 at 7.7 µM (peptide/ApoB-100

ratio: 3.5/1). To study the effects of the peptidomimetics on the aggregation of PLA2-modified LDL,

Manuscript BBAMEM-19-17 (R1)

14

LDL was incubated with PLA2 in the presence or absence of LP3, DP3 and IP321 at the doses and

peptide/ApoB-100 ratios detailed in Supplemental Table S3. We did not detect any effect of PLA2 on

LDL aggregation at time points lower than 24 h (data not shown). LP3 and DP3 showed high

inhibitory activity on PLA2-LDL aggregation, although, as in the case of SMase, DP3 showed

outperformed LP3 at concentrations of 10 µM and 14.6 µM at 24, 30 and 36 h (Fig. 1B). PLA2-LDL

aggregation was fully inhibited by LP3 at a concentration of 20.83 µM (peptide/ApoB-100 ratio:

9.8/1) and by DP3 at 14.6 µM (peptide/ApoB-100 ratio: 6.8/1) at all tested times. IP321 did not

inhibit SMase- or PLA2-LDL aggregation at any dose or time tested (Fig. 1A,B). Given that

Apolipoprotein A-I peptidomimetic 4F has been reported to block SMase-LDL aggregation 29, we

compared the capacity of LP3, DP3 and 4F (10 M, ratio 4.7/1, 24 h) to inhibit SMase- and PLA2-

LDL aggregation. SMase-induced aggregation was almost fully blocked by LP3 (86.2%±1.8) and

DP3 (98.1%±1.4), but only partially blocked by 4F (55.7%±2.3). PLA2-LDL aggregation was partially

blocked by LP3 (59.6%±0.8) and 4F (45.8%±1.1) and almost completely inhibited by DP3

(84.4%±1.9) (Table 1). IP321 did not have any significant effect on SMase- or PLA2-LDL

aggregation. None of the peptides alter the turbidimetry values of untreated LDL (Supplemental Fig.

S4).

3.3. LP3 and DP3 decrease the percentage of aggregated particles in SMase-LDL and PLA2-

LDL

To separate aggregated from monomeric LDL particles, LDL samples were subjected to gel filtration

chromatography (GFC). GFC showed that LDL comprised mainly monomeric LDL (Fig. 2a). A small

percentage of residual LDL aggregates was detected in untreated LDL due to the fact that both

untreated and treated LDL were exposed to 37ºC for the same period (Supplemental Fig. S3). In

contrast to LDL, SMase-LDL (Fig. 2b) was composed mainly of LDL aggregates. LP3 (Fig. 2c) and

DP3 (Fig. 2d) partially prevented the formation of LDL aggregates, DP3 showing higher efficiency

than LP3. IP321 did not alter the GFC pattern observed in SMase-LDL (Fig. 2e). As observed with

SMase-LDL, PLA2-LDL (Fig. 2f) showed a large peak of LDL aggregates. LP3 (Fig. 2g), but

Manuscript BBAMEM-19-17 (R1)

15

specially DP3 (Fig. 2h), increased the percentage of monomeric LDL and decreased that of LDL

aggregates. IP321 did not cause any significant alteration in the pattern of PLA2-LDL (Fig. 2i).

LRP1-derived peptides did not alter per se the GFC pattern of LDL (Supplemental Fig. S5).

Quantification of peak areas showed that LP3 and DP3, but not IP321, conferred SMase-LDL and

PLA2-LDL with a GFC profile closer to that of LDL (Fig. 2j,k). TEM images of LDL size distribution

(Fig. 3) showed that LDL comprised mainly individual LDL particles (Fig. 3A). In contrast, SMase-

LDL (Fig. 3B) and PLA2-LDL (Fig. 3C) generated in the presence of IP321 or in the absence of

peptides were formed mostly by aggregated and fused LDL. In the presence of LP3 and DP3,

SMase-LDL (Fig. 3B) and PLA2-LDL (Fig. 3C) were composed of LDL particles with a wide variety

of diameter sizes, although most were in the range present in LDL.

3.4. LP3 and DP3 block the alterations in LDL phospholipid composition in SMase-LDL but

not in PLA2-LDL

Lipoprotein composition analysis (Table 2) revealed that exposure of LDL to SMase or PLA2 did not

alter cholesterol, triglyceride or ApoB content of LDL, either in the presence or absence of the

peptides. In contrast, phospholipid (PL) content of LDL was significantly reduced by both enzymes

(P<0.005). LP3 and DP3 efficiently prevented PL reduction in SMase-LDL (P<0.005). However,

peptides did not show any effect on PL depletion or on non-esterified fatty acid (NEFA) increase

induced by PLA2 treatment. To identify the PL species influenced by enzymatic treatment, HPTLC

was used to separate LDL PLs. This procedure revealed that the main PLs in the LDLs were L--

phosphatidylcholine (L--PC) and sphingomyelin (SM). L--PC levels were unaltered by SMase

treatment (Fig. 4A,B). In contrast, SM was removed from LDL by this treatment (Fig. 4A,C). LP3 and

DP3, but not IP321, efficiently protected LDL-SM against SMase-induced SM degradation (Fig.

4A,C). To examine whether the protective action of LRP1-derived peptides on LDL was due to their

capacity to inhibit SMase activity, we tested the effect of these compounds on the capacity of

SMase to promote SM lipolysis. SMase activity, measured in relation to the amount of

phosphorylcholine generated, was similar in the presence and absence peptides, thereby indicating

Manuscript BBAMEM-19-17 (R1)

16

that none of the peptides exerted direct effects on SMase activity (Supplemental Fig. S6). PLA2

completely removed L--PC from LDL (Fig. 4D,E), leading to a sharp increase in the L--LysoPC

content (Fig. 4D,F). PLA2 did not have any effect on the LDL-SM content (Fig. 4D,G). The peptides

were unable to counteract L--PC decay and the increase in L--LysoPC induced by PLA2 in LDL.

3.5. LP3 and DP3 block the alterations in the LDL electrophoretic profile of SMase-LDL but

not those of PLA2-LDL

To study the effects of the peptides on the electrophoretic mobility of SMase- and PLA2-LDL, we

used gradient gel electrophoresis (GGE) and agarose gels to analyze the modification of LDL in the

presence and absence of the peptides. In native GGE gels (4 h of electrophoresis), the position of

the LDL particle was observed to be determined by particle size. GGE gels showed that SMase

caused a strong loss of the band corresponding to LDL. The lack of this band in SMase-LDL is

attributed to the fact that the pore size of GGE gels precludes the entrance of the largest LDL

aggregates. The band loss induced by SMase was efficiently counteracted by LP3 and DP3 but not

by IP321 (Fig. 5A). In PLA2-LDL (Fig. 5B), GGE showed a minor band corresponding to LDL

(marked as band 1), an intensive band (marked as band 2) with higher electrophoretic mobility

(smaller size) than band 1, and a third band (marked as band 3), which was a minor band with lower

electrophoretic mobility (higher size) than band 1. LP3 and DP3 did not have any effect on the GGE

pattern of PLA2-LDL.

Agarose gels, where LDL runs according to its negative charge, showed that SMase-LDL had a

more diffuse band compared to the well-defined LDL band. SMase-LDL in the presence of LP3 and

DP3 also showed a defined LDL band (Fig. 5C). In contrast to SMase-LDL, PLA2-LDL showed a

single band with a higher negative electric charge than LDL combined with a degradation smear

pattern (Fig. 5D). LP3, and in particular DP3, slightly increased the electrophoretic mobility and

band intensity of PLA2-LDL while they diminished the degradation smear pattern. The peptides did

not cause any change in the electrophoretic mobility of untreated LDL (Supplemental Fig. S7).

Manuscript BBAMEM-19-17 (R1)

17

3.6. DP3 blocks the conformational changes of ApoB-100 in SMase-LDL and PLA2-LDL

We studied the potential conformational modifications of ApoB-100 by Western blot analysis.

Treatment of LDL with SMase (Fig. 6A) or PLA2 (Fig. 6B) increased the exposure of reactive ApoB-

100 epitopes to anti-ApoB-100 antibodies. Both LP3 and DP3 protected ApoB-100 against the

higher reactivity to anti-ApoB-100 antibodies induced by exposure to SMase and PLA2 (Fig. 6A,B).

IP321 had no effect on the increased ApoB-100 reactivity induced by the two enzymatic treatments.

3.7. LRP1-derived peptides electrostatically interact with specific lysine-enriched sequences

in ApoB-100 protein

Limited proteolysis combined with mass spectrometry-based proteomics (LC-MS/MS) evidenced

that DP3 protects specific sequences of ApoB-100 in LDL either by direct interaction or by

remodeling the protein structure (Fig. 7A). The protein/peptide complexes were subjected to limited

proteolysis with trypsin for 1 h and 3 h. The protection of protein sequences by the peptide was

observed by shotgun LC-MSMS analysis at short times (The whole data concerning LDL is available

via PanoramaPublic with identifier PXD011261). At 3-hours digestions, the effect disappeared. The

transient protection of the peptide can be explained by the proteolytic action that trypsin exerted on

the protein complex. Remarkably, most of the protected sequences were also protected in SMase-

LDL unexposed to peptides, suggesting that the aggregation process safeguards them from trypsin

digestion and that these specific sequences are likely to be those that play a major role in LDL

aggregation. The high coincidence between sequences protected by DP3 and those protected by

aggregation indicates that most DP3-protected sequences participate in LDL aggregation, thereby

explaining the high efficiency of LRP1-derived peptides to prevent LDL aggregation.

To further study the molecular basis for ApoB-100 recognition by DP3, peptide homology modeling

and molecular docking methods were used. First, to identify a potential binding site of DP3 peptide

on ApoB-100 protein structure, each ApoB-100 fragment protected by DP3 binding during trypsin

digestion was mapped on the overall ApoB-100 sequence. Virtually all the fragments corresponded

to lipid-associating domains 39, except for a highly positively charged sequence

(3227IKFDKYKAEK3236) located in the ApoB-100 C-terminal region and adjacent to the LDL receptor

Manuscript BBAMEM-19-17 (R1)

18

binding domain (3385RLTRKRGLK3393) 40-42. This sequence was thus assumed to be responsible

for DP3 peptide recognition and used as the binding site for the molecular docking calculations.

Finally, an energy-based rigid body docking experiment was performed between ApoB-100 protein

and DP3 peptide structures previously modeled alone by ab initio methods. Interestingly, we found

that one of the lowest-energy poses of the docking simulations accommodated DP3 in such an

orientation that ApoB-100 Lys3228, Lys3231 and Lys3233 (but not Lys3226 nor Lys3236) were deeply buried

upon peptide binding. This finding is in strong agreement with the trypsin digestion data described

above. Additionally, strong electrostatic interactions involving same specific key residues of the two

docking partners were also found. In this regard, two of the acidic residues in DP3 correspond to

those assumed to be responsible for the specific recognition between LRP1 module CR9 and

agLDL particles [PMID 25918169] (namely D-Glu3 and D-Glu9) established salt bridge contacts with

two positively charged ApoB-100 residues (namely Lys3229 and Lys3234), while the oxygen atom of

the DP3 D-Asn2 backbone and ApoB-100 Lys3232 were found to be involved in a hydrogen bond

favored by the positive ionic charge carried by the lysine side chain amine group (Fig. 7B).

4. Discussion

A previous study from our group demonstrated that LRP1, in particular the C-terminal half of domain

CR9 (from Gly1127 to Cys1140) named LP3, interacts with agLDL 28. In the current study, we tested

the effect of both the original LP3 version and its retro-enantio version DP3 on SMase and PLA2-

induced LDL aggregation.

In the present study, we observed that PLA2 required larger times and doses than SMase to induce

LDL aggregation. This finding is consistent with previous studies showing that LDL hydrolysis by

PLA2 is slower than that by SMase 43-45. The latter cleaves SM into phosphocholine and

ceramide, whereas PLA2 hydrolyzes the fatty acids in the sn-2 position of PC, generating free fatty

acids and lyso-PC molecules. When SM is lost, ceramide-enriched microdomains have been

proposed to act as non-polar spots at the LDL surface, thereby facilitating LDL aggregation through

hydrophobic associations between domains 44. Here, PLA2-induced PC degradation induces a

Manuscript BBAMEM-19-17 (R1)

19

dramatic increase in Lyso-L--PC and a strong decrease in LDL surface area, in line with previous

studies 44,46,47. In addition, SMase and PLA2 provoked higher accessibility of ApoB-100 epitopes

to anti-ApoB-100 antibodies, as showed by Western blot analysis of LDL. This observation would

suggest conformational alterations in ApoB-100 linked to phospholipolysis, in line with previous

results from other groups 48. GFC followed by TEM analysis evidenced that LDL treatment with

SMase induced aggregated and fused LDL particles, in agreement with previous studies 44and

that PLA2-LDL comprises a high variety of aggregate sizes, as previously shown 13.

Here, we demonstrate that LP3 and its derived retro-enantio version (DP3) per se have a high

capacity to inhibit LDL aggregation induced by SMase and PLA2. However, all experimental

approaches (turbidimetry, GFC, and TEM) used in this work demonstrate that DP3 outperforms

LP3. Although all-D peptides are not recognised by proteases, and thus have higher stability, they

may be less active than their corresponding all-L peptides because of differences in the orientation

of amino acid side-chains. To preserve the orientation of the side-chains of the parent L-peptide in a

D-analogue, the retro-enantio approach 49,50 has been used in this study. In this regard, the

preservation of the topological side-chain orientation of DP3 together with a high stability in

biological samples account for its notable performance in the inhibition assays. In addition,

comparison of the capacity of LRP1 (LP3 and DP3) and ApoA-1 (4F) peptides to inhibit LDL

aggregation revealed that at 10 µM (peptide/ApoB-100 ratio: 4.7:1), LP3 and DP3 almost completely

repressed SMase-induced LDL turbidimetry, while L-4F exerted only a partial effect. Previous

studies addressing the efficacy of 4F to inhibit LDL aggregation showed that a molar ratio of 20:1 (4-

fold higher) is required to fully block SMase-induced LDL aggregation 29. However, the

experimental conditions used for LDL aggregation by Nguyen SD et al. (shorter times, and different

procedures for incubation of LDL with L-4F) differed from those applied in our study and thus hinder

a direct comparison between results. To the best of our knowledge, the present study is the first to

report data on the efficacy of 4F to inhibit PLA2-induced LDL aggregation. Here we found that

comparing the inhibitory capacities of L-4F, DP3 and LP3 at 4.7:1 peptide:ApoB-100 ratio, L-4F

Manuscript BBAMEM-19-17 (R1)

20

showed a similar capacity than LP3 to inhibit PLA2-induced LDL aggregation but lower capacity than

DP3.

One key question is how LRP1-derived peptides inhibit LDL aggregation. An important observation

in this regard is that these peptides counteracted alterations in LDL particle size, charge and

phospholipids induced by SMase but not those induced by PLA2. Despite of this, LRP1-derived

peptides exerted protection against SMase and PLA2-induced conformational alterations of ApoB-

100 and against LDL aggregation. These results indicate that 1) peptide protection of ApoB-100

conformation occurrs even in the context of extreme lipolysis and, 2) preservation of ApoB-100

conformation seems to be a common mechanisms underlying LDL protection against SMase and

PLA2-induced LDL aggregation. But, how LRP1-derived peptides preserve ApoB-100

conformation???. In the current study, limited trypsin proteolysis combined with Mass spectrometry-

based proteomic studies of LDL/DP3 complexes evidenced that peptide DP3 interacts with the LDL

surface prior to enzymatic treatment, protecting certain ApoB-100 sequences against trypsin-

induced proteolysis. Most of the peptide-protected sequences in ApoB-100, correspond to lipid-

associated ApoB-100 domains in LDL. The protection that peptide confers to ApoB-100 can be

induced by direct interaction or by remodeling the protein structure. Moreover, a highly positively

charged sequence (3227IKFDKYKAEK3236) located in the ApoB-100 C-terminal region and adjacent to

the LDL receptor binding domain (3385RLTRKRGLK3393) 51-54 was assumed to be responsible for

DP3 peptide recognition and used as the binding site for the molecular docking calculations. In

addition, homology modeling and molecular dynamics methods combined with molecular docking

provided a crucial first insight into the mechanistic basis of LRP1 peptide recognition by ApoB-100

protein. We propose a structural model of DP3 peptide in complex with ApoB-100. This complex is

mainly formed by salt bridge contacts between two of the acidic residues in DP3 (namely D-Glu3

and D-Glu9) and two positively charged ApoB-100 residues (namely Lys3229 and Lys3234), while the

oxygen atom of the DP3 D-Asn2 backbone and ApoB-100 Lys3232 were found to be involved in a

hydrogen bond favored by the positive ionic charge carried by the lysine side chain amine group.

This model is in complete agreement with the trypsin-limited proteolysis data reported herein, as

well as with the study of the recognition between LRP1 module CR9 and aggregated LDL particles

Manuscript BBAMEM-19-17 (R1)

21

recently published 28. Previous studies have consistently demonstrated that conformational

alterations of ApoB-100 lead to ApoB-100 degradation, a key process for LDL aggregation

48,55,56. In this context, the capacity of LRP1-derived peptides to preserve ApoB-100

conformation explain the high potential of these peptides to protect LDL against LDL aggregation.

Other crucial question is the mechanism by which LRP1-derived peptides protect SM but not PC

from enzymatic hydrolysis. Two different mechanistical options for the protective effect of peptides

against SMase-induced LDL lipolysis have been explored. By one side, we have explored whether

there is a direct effect of peptides on SMase activity. This possibility has been discarded since

SMase activity assays clearly show that peptides does not exert any effect on SMase functional

activity. Other possibility that has been explored is a potential interaction of peptides with the LDL

phospholipid surface. Several peptides and proteins bearing amphipathic properties have been

reported to be able to block LDL aggregation by a direct interaction with LDL phospholipid surface.

In this respect, common examples are 4F peptide 29 or different apolipoproteins 57. Despite the

lack of amphipathic properties in LRP1-derived peptides, this hypothesis has been considered as

possible mechanism of action for the peptides described herein. Theoretical molecular docking

calculations performed in the present study indicate a lack of association between peptides and LDL

phospholipid surface. A key point offering clues about how peptides protect SM is their unability to

protect PC hydrolysis. On the basis of the differential topological distribution of SM and PC in the

LDL surface 46, we propose that the differential anti-phospholypolytic effects of the peptide on SM

and PC phospholipolysis may be determined, at least in part, by the particular imbrication of SM and

PC with free cholesterol on the LDL surface. Although SM, compared to PC, is a minor phospholipid

in LDL (185 versus 450 molecules/LDL), SM is localized in cholesterol-enriched environments,

considered key regulators of phospholipolysis 46. ApoB-100 conformational stabilization could

directly impact the maintenance of the cholesterol-enriched environment structure since, according

to our molecular modeling studies, ApoB-100 sequences complexed with peptide are those close to

cholesterol in the LDL surface. Therefore, SM could be protected by LRP1-derived peptides against

phospholipolysis by its privilege of being part of cholesterol-enriched domains, whose structure

Manuscript BBAMEM-19-17 (R1)

22

would be preserved through the formation of peptide-ApoB-100 complexes. In contrast, PC,

belonging to cholesterol-poor domains, would be unprotected against PLA2-induced lipolysis,

independently of the status of ApoB-100 conformation.

5. Conclusion

Here, we show that LRP1-derived peptides, LP3 and, in particular, its retro-enantio version (DP3),

efficiently preserve LDL against SMase or PLA2-induced LDL aggregation. LP3 and DP3 maintain

LDL particle size, charge, phospholipid composition and ApoB-100 conformation in LDL treated with

SMase, but only ApoB-100 conformation in LDL exposed to PLA2 (summarized in Figure 8,

Graphical abstract). LP3 and DP3 keep ApoB-100 conformation even under extreme lipolysis

through key electrostatic interactions with a highly positively charged sequence

(3227IKFDKYKAEK3236) located in the ApoB-100 C-terminal region. A recent study has shown that the

individual susceptibility of LDL to aggregation is associated with future cardiac deaths independently

of other classical risk factors 58. LRP(CR9) derived peptides protect LDL against the main

proteolytic enzymes contributing to LDL retention and aggregation in the arterial intima. In the next

future, we are going to test the efficacy of the retro-enantio peptide version (DP3) to reduce

vascular LDL retention and LDL-induced smooth muscle cell (SMC)-foam cell formation in in vivo

models of atherosclerosis. Our first option in the near future will be to inject the peptide DP3

subcutaneously (not focal delivery at the moment) in a murine model of angioplasty-induced

atherosclerosis. This model offers the advantage of increased endothelial permeability, crucial to

favor peptide arrival to vasculature. In a not too distant future, we expect LRP1-derived peptides to

emerge as tools potentially useful to prevent atherosclerosis and cardiovascular complications, in

particular in individuals with aggregation-prone LDL as obese and diabetic patients

DATA AVAILABILITY

Mass spectroscopy proteomics of LDL. Panorama Public PXD011261

https://panoramaweb.org/j8CaUy.url

Manuscript BBAMEM-19-17 (R1)

23

Conflict of interest

R.P., C.P., and T.T. are employees of Iproteos SL. T.T. is cofounder of Iproteos SL and member of

its board of directors.

Acknowledgments

The Ministry of Science and Innovation of Spain, in the framework of the State Plan of Scientific

and Technical Innovation Investigation 2013-2016, awarded funding to the project

“DEVELOPMENT OF AN INNOVATIVE THERAPY FOR THE TREATMENT OF THE

ATHEROSCLEROSIS THROUGH INHIBITION OF CHOLESTEROL VASCULAR

ACCUMULATION”, led by IPROTEOS SL with file number RTC-2016-5078-1. This project was also

financed by the Ministry of Economy, Industry and Competitiveness (MINECO) in the framework of

the Subprogram RETOS-COLABORACIÓN, 2016 call. The project is also co-financed by the

European Union with the objective to promoting the technological development, innovation and

quality research. Support was also received from SAF2017-89613R (to SV), co-financed by the

European Regional Development Fund (ERDF), the Fundació MARATÓ TV3 Project 201521-10 (to

VLl-C), FIS PI13/00364 and PI16/00471 (to JSQ) and FIS PI18/01584 (to VLl-C) from the Instituto

de Salud Carlos III (ISCIII) and co-financed with ERDFs. Support was also received from Ministerio

de Economía y Competitividad to DdG-C (IJCI-2016-29393). CIBER Diabetes y Enfermedades

Metabólicas Asociadas (CIBERDEM; CB07/08/0016 (JSQ) and CIBER Enfermedades

Cardiovasculares (CIBERCV; CB16/1100403 (DdG-C, VLl-C) are projects run by the Instituto de

Salud Carlos III (ISCIII). The CRG/UPF Proteomics Unit is member of ProteoRed PRB3 consortium

which is supported by grant PT17/0019 of the PE I+D+i 2013-2016 from the Instituto de Salud

Carlos III (ISCIII) and ERDF. We also acknowledge support from the Spanish Ministry of Economy

and Competitiveness, “Centro de Excelencia Severo Ochoa 2013-2017”, SEV-2012-0208, and

“Secretaria d’Universitats i Recerca del Departament d’Economia I Coneixement de la Generalitat

de Catalunya” (2017SGR595 to ES and 2017SGR946 to VLl-C. SB, JLS-Q, AR-U, DdG-C and VL-

C are members of the Group of Vascular Biology of the Spanish Society of Atherosclerosis (SEA).

Manuscript BBAMEM-19-17 (R1)

24

References

[1] Castelli WP, Anderson K, Wilson PWF, Levy D. Lipids and risk of coronary heart disease The Framingham Study. Ann Epidemiol. 1992; 2:23–28.

[2] Gordon T, Castelli WP, Hjortland MC, Kannel WB, Dawber TR, High density lipoprotein as a protective factor against coronary heart disease. Am J Med. 1977; 62:707–714.

[3] Guretzki H-J, Gerbitz K-D, Olgemöller B, Schleicher E. Atherogenic levels of low density lipoprotein alter the permeability and composition of the endothelial barrier. Atherosclerosis. 1994; 107:15–24.

[4] Aikawa M, Libby P. The vulnerable atherosclerotic plaque. Cardiovasc Pathol. 2004; 13 :125–138.

[5] Mauriello A, Sangiorgi GM, Virmani R, Trimarchi S, Holmes Jr DR, Kolodgie FD, Piepgras DG, Piperno G, Liotti D, Narula J, Righini P, Ippoliti A, Spagnoli LG. A pathobiologic link between risk factors profile and morphological markers of carotid instability. Atherosclerosis. 2010; 208: 572–580.

[6] Puri R, Nissen SE, Shao M, Elshazly MB, Kataoka Y, Kapadia SR, Tuzcu EM, Nicholls SJ. Non-HDL Cholesterol and Triglycerides: Implications for Coronary Atheroma Progression and Clinical Events., Arterioscler Thromb Vasc Biol. 2016; 36:2220-2228.

[7] Skålén K, Gustafsson M, Knutsen Rydberg E, Hultén LM, Wiklund, Innerarity TL, Boren J. Subendothelial retention of atherogenic lipoproteins in early atherosclerosis. Nature. 2002; 417:750-754.

[8] Öörni K, Pentikäinen MO, Ala-Korpela M, Kovanen PT. Aggregation, fusion, and vesicle formation of modified low density lipoprotein particles: molecular mechanisms and effects on matrix interactions. J Lipid Res. 2000; 41:1703–1714.

[9] Öörni K, Posio P, Ala-Korpela M, Jauhiainen M, Kovanen PT. Sphingomyelinase induces aggregation and fusion of small very low-density lipoprotein and intermediate-density lipoprotein particles and increases their retention to human arterial proteoglycans, Arterioscler Thromb Vasc Biol. 2005; 25:1678-1683

[10] Marathe S, Schissel SL, Yellin MJ, Beatini N, Mintzer R, Williams KJ, Tabas I. Human vascular endothelial cells are a rich and regulatable source of secretory sphingomyelinase. Implications for early atherogenesis and ceramide-mediated cell signaling. J Biol Chem. 1998; 273:4081-4088.

[11] Schissel SL, Schuchman EH, Williams KJ, Tabas I. Zn2+-stimulated Sphingomyelinase Is Secreted by Many Cell Types and Is a Product of the Acid Sphingomyelinase Gene. J Biol Chem. 1996;. 271:18431–18436.

[12] Aviram M, Maor I. Phospholipase A2-modified LDL is taken up at enhanced rate by macrophages. Biochem Biophys Res Commun. 1992; 185:465–472.

[13] Öörni K, Hakala JK, Annila A, Ala-Korpela M, Kovanen PT. Sphingomyelinase induces aggregation and fusion, but phospholipase A2 only aggregation, of low density lipoprotein (LDL) particles: Two distinct mechanisms leading to increased binding strength of LDL to human aortic proteoglycans. J Biol Chem. 1998; 273:29127-29134.

[14] Hakala JK, Öörni K, Pentikäinen MO, Hurt-Camejo E, Kovanen PT. Lipolysis of LDL by Human Secretory Phospholipase A Induces Particle Fusion and Enhances the Retention of LDL to Human Aortic Proteoglycans. Arterioscler Thromb Vasc Biol. 2001; 21:1053-1058.

[15] Tabas I, Li Y, Brocia RW, Xu SW, Swenson TL, Williams KJ. Lipoprotein lipase and sphingomyelinase synergistically enhance the association of atherogenic lipoproteins with smooth muscle cells and extracellular matrix. A possible mechanism for low density lipoprotein and lipoprotein(a) retention and macrophage foam cell formation, J Biol Chem. 1993; 268:20419-20432.

[16] Von Ballmoos MW, Dubler D, Mirlacher M, Cathomas G, Muser J, Biedermann BC. Increased apolipoprotein deposits in early atherosclerotic lesions distinguish symptomatic from

Manuscript BBAMEM-19-17 (R1)

25

asymptomatic patients. Arterioscler Thromb Vasc Biol. 2006; 26:359-364. [17] Khoo JC, Miller E, McLoughlin P, Steinberg D. Enhanced macrophage uptake of low density

lipoprotein after self-aggregation. Arterioscler Thromb Vasc Biol. 1988; 8:348-358. [18] Zhang WY, Ishii I, Kruth HS. Plasmin-mediated macrophage reversal of low density

lipoprotein aggregation. J Biol Chem. 2000; 275:33176-33183. [19] Kruth HS. Sequestration of aggregated low-density lipoproteins by macrophages. Curr Opin

Lipidol. 2002; 13:483-488. [20] Llorente-Cortés V, Martínez-González J, Badimon L. Esterified Cholesterol Accumulation

Induced by Aggregated LDL Uptake in Human Vascular Smooth Muscle Cells Is Reduced by HMG-CoA Reductase Inhibitors, Arterioscler Thromb Vasc Biol. 1998; 18:738-746.

[21] Llorente-Cortés V, Martínez-González J, Badimon L. LDL Receptor–Related Protein Mediates Uptake of Aggregated LDL in Human Vascular Smooth Muscle Cells. Arterioscler Thromb Vasc Biol. 2000; 20:1572-1579.

[22] Llorente-Cortés V, Otero-Viñas M, Hurt-Camejo E, Martínez-González J, Badimon L. Human coronary smooth muscle cells internalize versican-modified LDL through LDL receptor-related protein and LDL receptors. Arterioscler Thromb Vasc Biol. 2002; 22:387-393.

[23] Llorente-Cortés V, Otero-Viñas M, Sánchez S, Rodríguez C, Badimon L. Low-Density Lipoprotein Upregulates Low-Density Lipoprotein Receptor-Related Protein Expression in Vascular Smooth Muscle Cells. Circulation. 2002; 106:3104-3110

[24] Costales P, Aledo R, Vérnia S, Das A, Shah VH, Casado M, Badimon L, Llorente-Cortés V. Selective role of sterol regulatory element binding protein isoforms in aggregated LDL-induced vascular low density lipoprotein receptor-related protein-1 expression, Atherosclerosis. 2010; 213:458–468.

[25] Llorente-Cortés V, Otero-Viñas M, Camino-López S, Llampayas O, Badimon L. Aggregated low-density lipoprotein uptake induces membrane tissue factor procoagulant activity and microparticle release in human vascular smooth muscle cells, Circulation. 2004; 110:452-459.

[26] Camino‐ López S, Badimon L, GonzáLez A, Canals D, Peña E, Llorente‐ Cortés V. Aggregated low density lipoprotein induces tissue factor by inhibiting sphingomyelinase activity in human vascular smooth muscle cells. J Thromb Haemost. 2009; 7:2137–2146.

[27] Allahverdian S, Chehroudi AC, McManus BM, Abraham T, Francis GA. Contribution of intimal smooth muscle cells to cholesterol accumulation and macrophage-like cells in human atherosclerosis., Circulation.2014; 129:1551-1559.

[28] Costales P, Fuentes-Prior P, Castellano J, Revuelta-Lopez E, Corral-Rodríguez MÁ, Nasarre L, Badimon L, Llorente-Cortes V. Domain CR9 of Low Density Lipoprotein (LDL) Receptor-related Protein 1 (LRP1) Is Critical for Aggregated LDL-induced Foam Cell Formation from Human Vascular Smooth Muscle Cells. J Biol Chem. 2015; 290:14852–14865.

[29] Nguyen SD, Javanainen M, Rissanen S, Zhao H, Huusko J, Kivelä AM, Ylä-Herttuala S, Navab M, Fogelman AM, Vattulainen I, Kovanen PT, Öörni K. Apolipoprotein A-I mimetic peptide 4F blocks sphingomyelinase-induced LDL aggregation J Lipid Res. 2015; 56:1206–1221.

[30] Pujals S, Fernández-Carneado J, Ludevid MD, Giralt E. D-SAP: A new, noncytotoxic, and fully protease resistant cell-penetrating peptide. ChemMedChem. 2008; 3:296-301.

[31] Bancells C, Villegas S, Blanco FJ, Benítez S, Gállego I, Beloki L, Pérez-Cuellar M, Ordóñez-Llanos J, Sánchez-Quesada JL. Aggregated Electronegative Low Density Lipoprotein in Human Plasma Shows a High Tendency toward Phospholipolysis and Particle Fusion. J Biol Chem. 2010; 285:32425–32435.

[32] Martínez-Bujidos M, Rull A, González-Cura B, Pérez-Cuéllar M, Montoliu-Gaya L, Villegas S, Ordóñez-Llanos J, Sánchez-Quesada J.L. Clusterin/apolipoprotein J binds to aggregated LDL in human plasma and plays a protective role against LDL aggregation. FASEB J. 2014; 29:1688–1700.

[33] Case D, Cheatham TE 3rd, Darden T, Gohlke H, Luo R, Merz KM Jr, Inufriev A, Simmerling C, Wang B, Woods RJ. The Amber biomolecular simulation programs. J Comput Chem. 2005;26:1668-1688.

[34] Phillips JC, Braun R, Wang W, Gumbart J, Tajkhorshid E, Villa E, Chipot C, Skeel RD, Kalé

Manuscript BBAMEM-19-17 (R1)

26

L, Schulten K. Scalable molecular dynamics with NAMD. J Comput Chem. 2005;26:1781-1802.

[35] Kim DE, Chivian D, Baker D. Protein structure prediction and analysis using the Robetta server. Nucleic Acids Res. 2004;32:W526-W531.

[36] Koes DR, Baumgartner MP, Camacho CJ. Lessons learned in empirical scoring with smina from the CSAR 2011 benchmarking exercise. J Chem Inf Model. 2013; 53:1893-1904.

[37] Chorev M, Goodman M. Recent developments in retropeptides and proteins_An ongoing topochemical exploration. Trends Biotechnol. 1995;13:438-445.

[38] Prades R, Oller-Salvia B, Schwarzmaier SM, Selva J, Moros M, Balbi M, Grazú V, De La Fuente JM, Egea G, Plesnila N, Teixidõ M, Giralt E. Applying the retro-enantio approach to obtain a peptide capable of overcoming the blood-brain barrier. Angew. Chemie - Int. Ed. 2015; 54:3967-3972.

[39] Prassl R, Laggner P. Molecular structure of low density lipoprotein: current status and future challenges. Eur Biophys J. 2009;38:145-158.

[40] Segrest JP, Jones MK, De Loof H, Dashti N. Structure of Apolipoprotein B-100 in low density lipoproteins. J Lipid Res. 2001; 42:1346-1367.

[41] Yang CY, Chen SH, Gianturco SH, Bradley WA, Sparrow JT, Tanimura M, Li WH, Sparrow DA, DeLoof H, Rosseneu M, et al. Sequence, structure, receptor-binding domains and internal repeats of human apolipoprotein B-100. Nature. 1986;323:738-742.

[42] Hospattankar AV, Law SW, Lackner K, Brewer HB Jr. Identification of low density lipoprotein receptor binding domains of human apolipoprotein B-100: a proposed consensus LDL receptor binding sequence of apoB-100. Biochem Biophys Res Commun. 1986;139:1078-1085.[43] Schissel SL, Tweedie-Hardman J, Rapp JH, Graham G, Williams KJ, Tabas I. Rabbit aorta and human atherosclerotic lesions hydrolyze the sphingomyelin of retained low-density lipoprotein. Proposed role for arterial-wall sphingomyelinase in subendothelial retention and aggregation of atherogenic lipoproteins. J Clin Invest. 1996; 98:1455–1464.

[44] Sartipy P, Camejo G, Svensson L, Hurt-Camejo E. Phospholipase A2 modification of low density lipoproteins forms small high density particles with increased affinity for proteoglycans and glycosaminoglycans, J Biol Chem. 1999; 274:25913-25920.

[45] Oestvang J, Bonnefont-Rousselot D, Ninio E, Hakala JK, Johansen B, Anthonsen MW, Modification of LDL with human secretory phospholipase A2 or sphingomyelinase promotes its arachidonic acid-releasing propensity. J Lipid Res. 2004; 45:831–838.

[46] Hevonoja T, Pentikäinen MO, Hyvönen MT, Kovanen PT, Ala-Korpela M. Structure of low density lipoprotein (LDL) particles: Basis for understanding molecular changes in modified LDL. Biochim Biophys Acta - Mol. Cell Biol. Lipids. 2000; 1488:189–210.

[47] Benítez S, Camacho M, Arcelus R, Vila L, Bancells C, Ordóñez-Llanos J, Sánchez-Quesada JL. Increased lysophosphatidylcholine and non-esterified fatty acid content in LDL induces chemokine release in endothelial cells. Atherosclerosis. 2004; 177:299–305.

[48] Kleinman Y, Krul ES, Burnes M, Aronson W, Pfleger B, Schonfeld G. Lipolysis of LDL with phospholipase A2 alters the expression of selected apoB-100 epitopes and the interaction of LDL with cells, J Lipid Res. 1988; 29:729-743.

[49] Soto C, Kindy MS, Baumann M, Frangione B. Inhibition of Alzheimer amyloidosis by peptides that prevent beta-sheet conformation. Biochem Biophys Res Commun. 1996; 226:672–680.

[50] Chalifour RJ, McLaughlin RW, Lavoie L, Morissette C, Tremblay N, Boulé M, Sarazin P, Stéa D, Lacombe D, Tremblay P, Gervais F. Stereoselective interactions of peptide inhibitors with the beta-amyloid peptide. J Biol Chem. 2003;278:34874-34881.

[51] Sneck M, Nguyen SD, Pihlajamaa T, Yohannes G, Riekkola M-L, Milne R, Kovanen PT, Öörni K. Conformational changes of apoB-100 in SMase-modified LDL mediate formation of large aggregates at acidic pH, J. Lipid Res. 2012; 53:1832-1839.

[52] Bancells C, Benítez S, Ordóñez-Llanos J, Öörni K, Kovanen PT, Milne RW, Sánchez-Quesada JL. Immunochemical Analysis of the Electronegative LDL Subfraction Shows That Abnormal N-terminal Apolipoprotein B Conformation Is Involved in Increased Binding to Proteoglycans, J Biol Chem. 2011; 286:1125–1133.

[53] Liu H, Scraba DG, Ryan RO. Prevention of phospholipase-C induced aggregation of LDL by amphipathic apolipoproteins. FEBS Lett. 1993; 316:27–33.

Manuscript BBAMEM-19-17 (R1)

27

[54] Ruuth M, Nguyen SD, Vihervaara T, Hilvo M, Laajala TD, Kondadi PK, Gisterå A, Lähteenmäki H, Kittilä T, Huusko J, Uusitupa M, Schwab U, Savolainen MJ, Sinisalo J, Lokki M-L, Nieminen MS, Jula A, Perola M, Ylä-Herttula S, Rudel L, Öörni A, Baumann M, Baruch A, Laaksonen R, Ketelhuth DFJ, Aittokallio T, Jauhiainen M, Käkelä R, Borén J, Williams KJ, Kovanen PT, Öörni K. Susceptibility of low-density lipoprotein particles to aggregate depends on particle lipidome, is modifiable, and associates with future cardiovascular deaths. Eur Heart J. 2018; 39:2562-2573

Figure Legends

Fig.1. LRP1-derived peptides (LP3 and DP3) decrease SMase- and PLA2-induced LDL aggregation in a dose-dependent manner. LDL was used at 1.44 mg/mL ApoB. Peptides were tested at

increasing concentrations (from 2.5 to 10 M for SMase-LDL and from 4.16 to 20.83 M for PLA2-LDL). Aggregation of LDL was induced by SMase (A) or PLA2 (B) and monitored at the indicated times by measuring turbidimetry (absorbance at 405 nm). Data are mean ± SD of 2 experiments performed in duplicate. Unsighted bar errors are minor than symbol size.*P<0.005 versus SMase-

LDL + IP321; P<0.005 versus SMase-LDL + LP3.

Fig. 2. LRP1-derived peptides (LP3 and DP3) reduce the AUC of agLDL peak in SMase-LDL and PLA2-LDL. LDL was untreated (LDL) or treated with SMase (40 U/L) and PLA2 (50 µg/L) in the absence (w/o peptide) or presence of LP3, DP3 or IP321 (10 µM, peptide/ApoB-100 ratio: 4.7:1, 24 h). LDLs (0.5 mL at 0.4 mg/mL ApoB) were chromatographed at a flow rate of 0.5 ml/min using a Superose 6 column in a AKTA-FPLC system (GE Healthcare) as described in Material and Methods. Representative GFC of: a) non enzymatically-treated LDL; b) LDL incubated with SMase; c) LDL incubated with SMase in the presence of LP3; d) LDL incubated with SMase in the presence of DP3; e) LDL incubated with SMase in the presence of IP321; f) LDL incubated with PLA2; g) LDL incubated with PLA2 in the presence of LP3; h) LDL incubated with PLA2 in the presence of DP3; and i) LDL incubated with PLA2 in the presence of IP321. j) and k) Bar graphs showing the percentage of aggregated versus monomeric LDL in SMase-LDL and PLA2-LDL, respectively, calculated from the chromatograms as area under the curve as explained in Methods.

Fig. 3. LRP1-derived peptides (LP3 and DP3) diminish the proportion of large size LDL aggregates in SMase-LDL and PLA2-LDL. Untreated LDL (A) or LDL treated with SMase (40 U/L) (B) and PLA2 (50 µg/L) (C) in the presence of LP3, DP3 or IP321 (10 µM, peptide/ApoB-100 ratio: 4.7:1, 24 h) or in absence of peptides (w/o peptide). TEM was performed in a Jeol 120 kV JEM-1400 microscope in the LDL preparations treated as explained above for 24 h at 37ºC. Black bars, 500 nm.

Fig. 4. LRP1-derived peptides (LP3 and DP3) prevent SM depletion in SMase-LDL but not L--PC depletion in PLA2-LDL. LDL was untreated (LDL) or treated with SMase (40 U/L) and PLA2 (50 µg/L) in the absence (w/o peptide) or presence of LP3, DP3 or IP321 (10 µM, peptide/ApoB-100 ratio: 4.7:1, 24 h) and the phospholipid pattern was analyzed by HPTLC after lipid extraction. Representative HPTLC of LDL (A and D) and bar graphs (B, C, E, F and G) showing quantification of phospholipid bands. Results are expressed as percentage of PL relative to untreated LDL and

shown as mean ± SD of 3 independent experiments performed in triplicate. L--PC indicates L--

phosphatidylcholine; SM: sphingomyelin; L--lyso-PC: L--lysophosphatidylcholine. *P<0.005 versus LDL.

Fig. 5. LRP1-derived peptides (LP3 and DP3) counteract LDL alterations in size and electric charge induced by SMase but not those induced by PLA2. LDL was untreated (LDL) or treated with SMase (40 U/L) and PLA2 (50 µg/L) in the absence (w/o peptide) or presence of LP3, DP3 o IP321 (10 µM,

Manuscript BBAMEM-19-17 (R1)

28

ratio peptide/ApoB-100: 4.7:1, 24 hours). LDL electrophoretic mobility was analyzed by GGE (A and B) and agarose gels (C and D). In GGE, LDL (5 µg/well) was electrophoresed in 2-16% acrylamide native gradient gels. LDLs were prestained with 1% Sudan black. In agarose gel electrophoresis, LDL (2 µg/well) was electrophoresed in 0.5% agarose gels, which were dehydrated and stained with Coomassie Blue. Peptides were assayed at 10 µM (Ratio Peptide/ApoB= 4.7/1)

Fig. 6. LRP1-derived peptides (LP3 and DP3) counteract ApoB-100 conformational alterations induced by SMase and PLA2 in LDL. LDL was untreated (LDL) or treated with SMase (40 U/L) and PLA2 (50 µg/L) in the absence (w/o peptide) or presence of LP3, DP3 or IP321 (10 µM, peptide/ApoB-100 ratio: 4.7:1, 24 h). Representative western blot analysis of ApoB-100 in LDL exposed to SMase (A) or PLA2 (B). LDL (5 µg/well) was electrophoresed in 10% acrylamide gels under non-reducing conditions for 2 h at 100 V and transferred to nitrocellulose membranes, which were then incubated with anti-ApoB-100 antibodies. Fig. 7. DP3 protect ApoB-100 sequences against tryptic-limited proteolysis A) List of ApoB100 peptides that are protected by DP3 and by LDL aggregation as shown by tryptic-limited proteolysis coupled to mass spectrometry analysis. Green indicates signal detected, Yellow a poor signal, and Red no MS signal. B) Structural details of the molecular interaction between Apo-B100 protein and DP3 peptide. View of the ApoB-100 protein surface contacting the DP3 peptide: the basic residues of ApoB-100 exposed to trypsin digestion and buried deeply upon peptide binding highlighted in teal and violet, respectively. DP3 peptide acid residues responsible for ApoB-100 binding highlighted in yellow sticks.

Fig. 8. Schematic representation of the molecular basis for the protective effect of LRP1-derived peptides against SMase- and PLA

2-induced LDL aggregation. DP3 forms a complex with ApoB-100

and this molecular interaction stabilizes ApoB-100 conformation. ApoB-100 conformation stabilization could guarantee the structural preservation of surface colesterol-enriched environments, where SM is located. Structural preservation of cholesterol, a key regulator of phospholipolysis, would protect SM from SMase activity. As a result, LDL complexed with DP3 remains unaltered when exposed to SMase. This scenario changes when LDL complexed with DP3 is exposed to PLA

2. The target for PLA

2 is PC, phospholipid associated with poor-colesterol

environments. Therefore, PC is unprotected against the attack of PLA2, that hydrolyzes PC producing lyso-PC and NEFA. Remarkably, LDL complexed with DP3 is protected against SMase and PLA2-induced aggregation even in conditions of extreme phospholipolysis, indicating that the maintenance of ApoB-100 conformation is key in the prevention of LDL aggregation.

Manuscript BBAMEM-19-17 (R1)

1

Molecular basis for the protective effects of low-density lipoprotein receptor-related protein 1

(LRP1)-derived peptides against LDL aggregation

Short title: LRP1-derived peptides and LDL aggregation

Aleyda Benitez-Amaroa,b, Chiara Pallarac, Laura Nasarrea, Andrea Rivas-Urbinad, Sonia Benitezd,

Angela Veaa, Olga Bornacheaa,b, David de Gonzalo-Calvoa,b,e, Gabriel Serra-Mirf, Sandra Villegasf,

Roger Pradesc, José Luís Sanchez-Quesadad,g, Cristina Chivah,i, Eduard Sabidoh,i, Teresa

Tarragóc, and Vicenta Llorente-Cortésa,b,e**

aGroup of Lipids and Cardiovascular Pathology. Biomedical Research Institute Sant Pau (IIB Sant

Pau), Hospital de la Santa Creu i Sant Pau. Barcelona. Spain.

bInstitute of Biomedical Research of Barcelona (IIBB). Spanish National Research Council (CSIC),

Barcelona, Spain.

cIproteos S.L., Barcelona Science Park (PCB), Barcelona, Spain.

dCardiovascular Biochemistry Group, Research Institute of the Hospital de Sant Pau (IIB Sant Pau),

Barcelona, Spain.

eCIBER enfermedades cardiovasculares (CIBERcv).

fProtein Design and Immunotherapy Group. Departament de Bioquímica i Biologia Molecular,

Facultat de Biociències, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain.

gCIBER diabetes y enfermedades metabólicas asociadas (CIBERdem)

hProteomics Unit, Centre de Regulació Genòmica, Barcelona Institute of Science and Technology,

Barcelona, Spain.

iUniversitat Pompeu Fabra, Barcelona, Spain.

**To whom correspondence should be addressed: Vicenta Llorente-Cortés, Lipids and

Cardiovascular Pathology group leader, ICCC Program, Biomedical Research Institute IIB Sant

Pau, Hospital de la Santa Creu i Sant Pau, Sant Antoni Mª Claret, 167, 08025 Barcelona. Phone:

+34 935565888, Fax: +34 935565559, E-mail: [email protected]; [email protected]

Formatted: Spanish (Spain,

International Sort)

Formatted: English (United States)

*REVISED Manuscript (text with changes Marked)Click here to view linked References

Manuscript BBAMEM-19-17 (R1)

2

Abstract

Aggregated LDL is the first ligand reported to interact with the cluster II CR9 domain of low-density

lipoprotein receptor-related protein 1 (LRP1). In particular, the C-terminal half of domain CR9,

comprising the region Gly1127-Cys1140 exclusively recognizes aggregated LDL and it is crucial for

aggregated LDL binding. Our aim was to study the effect of the sequence Gly1127-Cys1140 (named

peptide LP3 and its retro-enantio version, named peptide DP3) on the structural characteristics of

sphingomyelinase- (SMase) and phospholipase 2 (PLA2)-modified LDL particles. Turbidimetry, gel

filtration chromatography (GFC) and transmission electronic microscopy (TEM) analysis showed

that LP3 and DP3 peptides strongly inhibited SMase- and PLA2-induced LDL aggregation.

Nondenaturing polyacrylamide gradient gel electrophoresis (GGE), agarose gel electrophoresis and

high-performance thin-layer chromatography (HPTLC) indicated that LP3 and DP3 prevented

SMase-induced alterations in LDL particle size, electric charge and phospholipid content,

respectively, but not those induced by PLA2. Western blot analysis showed that LP3 and DP3

counteracted changes in ApoB-100 conformation induced by the two enzymes. LDL proteomics

(LDL trypsin digestion followed by mass spectroscopy) and computational modeling methods

evidenced that peptides preserve ApoB-100 conformation due to their electrostatic interactions with

a basic region of ApoB-100. These results demonstrate that LRP1-derived peptides are protective

against LDL aggregation, even in conditions of extreme lipolysis, through their capacity to bind to

ApoB-100 regions critical for ApoB-100 conformational preservation. These results suggests that

these LRP1(CR9) derived peptides could be promising tools to prevent LDL aggregation induced by

the main proteolytic enzymes acting in the arterial intima.

Key words: Lipoprotein aggregation; LRP1-derived peptides; ApoB-100; SMase, PLA2,

atherosclerosis

Manuscript BBAMEM-19-17 (R1)

3

Abbreviations: agLDL, aggregated LDL; apo, apolipoprotein; CE, cholesteryl esters; FC, free

cholesterol; hcVSMC, human coronary vascular smooth muscle cells; GFC, gel filtration

chromatography; GGE, nondenaturing polyacrylamide gradient gel electrophoresis; HPTLC, high-

performance thin-layer chromatography; LC-MS/MS, liquid chromatography tandem-mass

spectrometry; LDL, low-density lipoprotein; LysoPC, lysophosphatidylcholine; LRP1, low-density

lipoprotein receptor-related protein 1; NEFA, nonesterified fatty acids; PC, phosphatidylcholine;

PLA2, phospholipase A2; PL, phospholipids; REMD, Replica Exchange Molecular Dynamics; SM,

sphingomyelin; SMase, sphingomyelinase; TEM, transmission electron microscopy

Manuscript BBAMEM-19-17 (R1)

4

1. Introduction

Ischemic heart disease is the first cause of death in Western countries, and myocardial infarction

accounts for about 50% of deaths from this disease. The Framingham study showed that

cardiovascular risk positively correlates with low-density lipoprotein (LDL)-cholesterol and inversely

with high-density lipoprotein (HDL)-cholesterol 1,2. High levels of LDL-cholesterol induce

alterations in the endothelium permeability, which in turn promote LDL influx into the arterial intima

3. Intimal LDL-cholesterol accumulation is a critical step in vascular cholesteryl ester (CE)

deposition, a process that increases the tendency of the atherosclerotic plaque to rupture, triggering

thrombosis and the development of ischemic cardiomyopathy 4-6. CEs in atherosclerotic plaques

are deposited both extra- and intra-cellularly. Extracellular deposition of LDL-CEs, a crucial initiating

event in atherosclerosis 7, is mediated by proteoglycans in the extracellular matrix of the arterial

intima. The electrostatic interaction between proteoglycans and LDL and the proteolytic/lipolytic

actions of enzymes on LDL are enhanced in the arterial intima and promote LDL retention and

aggregation 8,9. Two of the main enzymes acting on LDL are sphingomyelinase (SMase),

secreted by endothelial cells and macrophages 10,11 and phospholipase A2 (PLA2) 12,13. These

two enzymes play key roles in LDL aggregation in the arterial intima during atherogenesis 12-15.

Aggregated LDL (agLDL) has been detected and isolated from atherosclerotic plaques from animal

models and humans 11,16. Unlike native LDL, agLDL is a potent inducer of massive intracellular

CE accumulation both in macrophages 17-19 and human coronary vascular smooth muscle cells

(hcVSMCs) 20. In particular, in hcVSMCs, agLDL is actively taken up through the low-density

lipoprotein receptor-related protein 1 (LRP1) 21,22. The internalization of agLDL, in turn, induces

LRP1 expression 23,24, promoting a positive feedback loop that efficiently transforms hcVSMCs

into foam cells. We have also demonstrated that hcVSMC-foam cells synthesize and release high

amounts of tissue factor, which is crucial for the pro-thrombotic transformation of the vascular wall

and thus for the progression of atherosclerosis to thrombosis 25,26. The relevance of this

mechanism in atherosclerosis is evident since VSMCs are the main components of the vascular wall

Manuscript BBAMEM-19-17 (R1)

5

and more than 50% of foam cells previously considered to be monocyte-derived macrophages in

human atherosclerotic plaques originate from VSMCs 27. Together, these findings support the

notion that the generation of hVSMC-derived foam cells through LRP1-mediated agLDL uptake is a

key mechanism underlying cholesterol accumulation in vasculature susceptible to atherosclerosis.

Our group has identified LRP1 cluster II, in particular the region Gly1127-Cys1140 (peptide LP3: H-

GDNDSEDNSDEENC-NH2) that spans the C-terminal half of domain CR9, as pivotal for binding to

agLDL and subsequent internalization of this agLDL into human VSMCs 28. We hypothesize that

peptide LP3 or its retro-enantio version, peptide DP3, would counteract LDL aggregation. Using a

range of biochemical, biophysical and molecular experiments, here we demonstrate that LP3 and

DP3 peptides efficiently prevent LDL aggregation induced by the lipolytic enzymes SMase and

PLA2. Although LP3 and DP3 efficiently counteracted alterations in LDL particle size, charge and

SM loss induced by SMase, they were unable to prevent the alterations induced in the same

parameters by PLA2. However, both peptides protected ApoB-100 from conformational alterations

induced by SMase and PLA2. Proteomics, ab initio modeling and molecular dynamics showed that

LRP1-derived peptides establish stable electrostatic interactions with basic sequences of ApoB-100.

2. Material and Methods

2.1. Peptide sequences and synthesis

Peptide sequences are written from N to C, H- at the end terminus indicate amine ending, whereas

–NH2 at the C terminus indicates amide. The amino acid sequence of LP3 peptide (parent) is H-

GDNDSEDNSDEENC-NH2, DP3 is the retro-enantio version of LP3 without cysteine: H-

needsndesdndh-NH2, where the amino acid letter code in lowercase stands for amino acids with D-

chirality. As a negative control, we used an ApoB-100-based peptide coded as IP321 with the

sequence: H-RLTRKRGLK-NH2. We also compared the efficacy of LP3, DP3 and IP321 with the

previously described anti-atherosclerotic apoA-I mimetic, 4F. The latter is 18 amino acids long and

has the following sequence: Ac-DWFKAFYDKVAEKFKEAF-NH2 VSMCs 29. All peptides were

Manuscript BBAMEM-19-17 (R1)

6

synthesized by Iproteos by standard 9-fluorenylmethoxycarbonyl/tert-butyl (Fmoc/tBu) solid-phase

peptide synthesis. Syntheses were performed manually on a 100-µmols scale/each using Rink

amide ChemMatrix® resin. Peptide elongation and other solid-phase manipulations were done

manually in polypropylene syringes, each fitted with a polyethylene porous disk. Solvents and

soluble reagents were removed by suction. Washings between synthetic steps were done with

dimethylformamide and dichloromethane using 5 mL of solvents/g resin each time. Nα-Fmoc-

protected amino acids (4 equivalents) were coupled using 2-(1H-benzotriazole-1-yl)-1,1,3,3-

tetramethyluronium (TBTU, 4 equivalents), and N,N-diisopropylethylamine (8 equivalents). The

extent of the reaction was monitored using the Kaiser test (primary amines). During couplings, the

mixture was allowed to react with intermittent manual stirring. Fmoc group was removed by treating

the resin with 20% piperidine in DMF (3 mL/g resin). The peptides were cleaved from the resin using

a mixture of trifluoroacetic acid, triisopropylsilane and water (95:2.5:2.5). The crude products

obtained were purified in reverse phase using a semi-prep HPLC instrument equipped with a C18

column. The purity and identity of the synthesized peptides was assessed by HPLC, HPLC-MS and

MALDI-TOF analysis (Table S1).

2.2. Peptide stability in human serum and cell culture medium

The stability of the peptides was studied in human serum (Sigma-Aldrich). For the stability assay,

the peptides were dissolved in a mixture of 10% Hanks’ Balanced Solution Salt (HBSSx1) and

human serum. The final concentration of peptide was 200 mM VSMCs 30. The samples were

incubated at 37°C with orbital agitation for 24 h. At selected time points, serum aliquots were

extracted and the serum proteins of the mixture were then precipitated by adding methanol.

Samples were then centrifuged at 4°C, and the supernatant was filtered out and analyzed by HPLC.

Peptide stability during the assays was monitored using HPLC (UV detection at 220 nm).

2.3. LDL isolation and purification

Human LDL (d1.019-d1.063 g/mL) was obtained from pooled normolipemic human plasma by

sequential ultracentrifugation in a KBr density gradient. Briefly, very low density lipoproteins (VLDLs)

Manuscript BBAMEM-19-17 (R1)

7

were first discarded after spinning plasma at 36,000 rpm for 18 h at 4ºC using a fixed-angle rotor

(50.2 Ti, Beckman) mounted on an Optima L-100 XP ultracentrifuge (Beckman). Subsequently,

VLDL-free plasma was layered with 1.063 g/mL KBr solution and centrifuged at 36,000 rpm for 18 h

at 4ºC. LDLs were dialyzed against 0.02 M Trizma, 0.15 M NaCl, 1 mM EDTA, pH 7.5 for 18 h, and

then against normal saline for 2 h. Finally, isolated LDLs were filter-sterilized (0.22 μm Millex-GV

filter unit, Millipore). Protein concentration was determined using the BCA protein assay (Thermo

Scientific) and the cholesterol concentration with a commercial kit (IL test Cholesterol, Izasa).

2.4. Analysis of SMase activity in the presence and absence of peptides

The capacity of LP3 and DP3, IP321 and manumycin A (positive control) to inhibit

sphingomyelinase activity was measured using the Amplex Red Sphingomyelinase Assay Kit

(Molecular Probes Invitrogen Detection Technologies, #A12220), an assay that measures the

formation of phosphorylcholine. To measure SMase activity in the presence and absence of

peptides, DMSO volume and peptide concentrations were kept constant [10 M (molar ratio

peptide/ApoB= 4.7/1)] while SMase concentration was increased (0, 0.2, 0.4, 0.6, 0.8 and 1 mU/ml).

Each reaction contained 50 μM Amplex Red reagent, 1 U/mL HRP, 0,1 U/ml choline oxidase, 4 U/ml

of alkaline phosphatase, 0,25 mM sphingomyelin (diluted with Triton X-100 or with the Reaction

Buffer 1x). Reactions were incubated at 37ºC for 30 min. Fluorescence intensity was measured

using a fluorescence microplate reader (Victor 1420 multilabel counter) using excitation at 530 nm

and fluorescent detection at 590nm.

2.5. LDL modification by Sphingomyelinase (SMase) and Phospholipase A2 (PLA2) in

the presence and absence of peptides

Human LDL (1.44 mg/mL) were incubated with 40 U/L of Bacillus cereus SMase (Sigma-Aldrich,

Schnelldorf, Germany) or with 50 µg/L of type II secretory PLA2 from honey bee venom (Sigma-

Aldrich, Schnelldorf, Germany) in 20 mM Tris buffer (pH 7.0) containing 150 mM NaCl, 2 mM CaCl2,

and 2 mM MgCl2 at 37ºC. LDL incubation with SMase and PLA2 was performed at peptide

concentrations of 0, 2.5, 5, 7.7, 8.8, 10 µM peptide/ApoB-100 ratio :0/1, 1.2/1, 2.4/1, 3.5/1, 4.1/1,

Manuscript BBAMEM-19-17 (R1)

8

4.7/1) and 0, 4.2, 6.3, 10.0, 14.6, 20.8 µM peptide/ApoB-100 ratio (0/1, 2.0/1, 2.9/1, 4.7/1, 6.8/1,

9.8/1), respectively, for increased time periods. LDL lipolysis was stopped by addition of EDTA

(final concentration 10 mM).

2.6. Characterization of SMase- and PLA2-induced LDL alterations in the presence and

absence of peptides. A detailed information of the different procedures

2.6.1.LDL Composition_The LDL composition was measured by commercial methods adapted to a

Cobas 6000/c501 autoanalyzer (Roche Diagnostics, Indianapolis, IN, USA). Total cholesterol,

triglycerides and ApoB reagents were obtained from Roche Diagnostics. PLs, NEFAs, and free

cholesterol reagents were obtained from Wako Pure Chemicals (Tokyo Japan).

2.6.2.Turbidity_Sample turbidity (LDL aggregation monitorization) was determined by measuring

absorbance of 100 µl of LDLs (1.44 mg/mL ApoB) in a 96-well microplates at 405 nm.

2.6.3.Gel Filtration Chromatography (GFC)_To separate aggregated from non-aggregated LDL,

samples (50 µL at 1.44 mg/mL) were subjected to gel filtration chromatography (GFC) in two online

connected Superose 6 Increase 5/150 GL columns with an AKTA-FPLC system (GE Healthcare) at

a flow of 0.3 mL/min 31,32. The area under the curve of each peak was calculated using the

Unicorn 4.0 software (GE Healthcare).

2.6.4. Agarose gel electrophoresis_LDL (1.5 µL of LDL (1.44 mg/mL) was electrophoresed on 0.5%

agarose gels at 65V for 40 min followed by 90V for 15 min. Fixed gels were dehydrated and stained

with Coomassie Blue. Decolored gels were captured with the Bio-Rad GS-800 scanner.

2.6.5. SDS-PAGE and Western blot

ApoB-100 was detected by Western blot analysis after electrophoresis of LDL in 6% SDS-PAGE

gels. Nitrocellulose membranes were incubated with anti-ApoB-100 polyclonal antibodies (Roche

Diagnostics S.L., Ref: 3032574122, polyclonal anti-human ApoB-100, dilution 1: 5000) for 1 h at

Manuscript BBAMEM-19-17 (R1)

9

room temperature. After several washes, membranes were incubated with secondary anti-sheep

antibodies (dilution 1: 10000, Dako; Glostrup, Denmark). Bands were detected using ECL Prime

Western Blotting Detection Reagent (Amersham) and quantified by densitometry using a ChemiDoc

system and Quantity One software (Bio‐Rad, Hercules, CA, USA). The results are expressed as

arbitrary units of intensity.

2.6.6.TEM

TEM was performed as previously described 32. LDL was placed on a 400 mesh glow-discharge

carbon-coated grid, negatively stained with 2% potassium phosphotungstate, pH 7.0, and air dried.

Grids were placed in a Jeol 120-kV JEM-1400 (Jeol, Japan) transmission electron microscope. TEM

images were processed using ImageJ (Fiji) software. Briefly, contrast was enhanced, followed by

background subtraction.

2.6.7. Nondenaturing acrylamide gradient gel electrophoresis (GGE)

GGE was performed following Nichols et al., with small modifications 32. Two solutions at 2% and

16% were prepared by using a stock solution of acrylamide and bis-acrylamide (30% total, 5%

cross-linker) and mixed using 2 P-1 peristaltic pumps (Pharmacia). The different samples (5 μL at

1.44 mg/mL) were preincubated for 15 min with 10 μL Sudan black (0.1% [wt/vol]) in ethylene glycol

and 5 μL saccharose (50% [wt/vol]). Ten microliters of this mixture was electrophoresed at 4°C for

30 min at 20 V, 30 min at 70 V, and 4 h at 100 V. Bands were scanned by densitometry.

2.6.8. HPTLC

LDL (15 µg) was lipid-extracted with organic solvents as previously described by our group with

minimal modifications 25. Samples were applied to horizontal, one-dimensional high-performance

thin-layer chromatography (HPTLC) (Merck, HPTLC Silica Gel 60, 1.05641.0001). Phospholipids

were separated on HPTLC by chloroform:ethyl

acetate:acetone:isopropanol:ethanol:methanol:water:acetic acid (30:6:6:6:16:28:6:2); visualized by

Manuscript BBAMEM-19-17 (R1)

10

staining with 7.5% Cu-acetate (w/v), 2.5% CuSO4 (w/v), and 8% H3PO4 (v/v) in water. A mixture of

phospholipids (Sigma Ref pH-7) were also run as standards. The spots corresponding to the

different phospholipids were quantified by densitometry against the standard curve using a GS-800

Calibrated Densitometer (Bio-Rad).

2.7. Proteomics analyses of LDL samples

2.7.1. Sample preparation_LDL, LDL unexposed or exposed to SMase in the absence of peptide or

in the presence of DP3 or IP321) were directly subjected to limited trypsin digestion (1 µg) for 1 h or

3 h. In both cases, tryptic peptides were desalted using a C18 column and evaporated to dryness.

2.7.2. Mass spectrometry data acquisition_ LDL samples were analyzed using a LTQ-Orbitrap Velos

Pro mass spectrometer (Thermo Fisher Scientific, San Jose, CA, USA) coupled to an EasyLC

(Thermo Fisher Scientific (Proxeon), Odense, Denmark). Peptides were separated by reverse-

phase chromatography using a 25-cm column with an inner diameter of 75 μm, packed with 5 μm

C18 particles (Nikkyo Technos Co., Ltd. Japan). Chromatographic gradients started at 93% buffer A

and 7% buffer B and gradually increased to 65% buffer A / 35% buffer B in 60 min at a flow rate of

250 nl/min. The mass spectrometer was operated in positive ionization mode with nanospray

voltage set at 2.2 kV and source temperature at 250°C. Ultramark 1621 for the FT mass analyzer

was used for external calibration prior to the analyses, and an internal calibration was performed

using the background polysiloxane ion signal at m/z 445.1200. The acquisition was performed in

data-dependent acquisition mode, and full MS scans with 1 micro scans at a resolution of 60,000

were used over a mass range of m/z 350-2000 with detection in the Orbitrap. Auto gain control

(AGC) was set to 1E6, dynamic exclusion (60 seconds) and charge state filtering disqualifying singly

charged peptides was activated in each cycle of data-dependent acquisition analysis. After each

survey scan, the top ten most intense ions with multiple charged ions above a threshold ion count of

5000 were selected for fragmentation at normalized collision energy of 35%. Fragment ion spectra

produced via collision-induced dissociation (CID) were acquired in an Ion Trap apparatus. AGC set

Manuscript BBAMEM-19-17 (R1)

11

to 5e4, an isolation window of 2.0 m/z, activation time of 0.1ms and maximum injection time of 100

ms were used. All data were acquired with Xcalibur software v2.2.

2.7.3. Mass spectrometry data analysis

The spectra acquired for the LDL samples were analyzed using the Proteome Discoverer software

suite (v2.0, Thermo Fisher Scientific) and the Mascot search engine (v2.5, Matrix Science). The

data were searched against the Swiss-Prot human database (v.2017/10). At the MS1 level, a

precursor ion mass tolerance of 7 ppm was used, and up to three missed cleavages were allowed.

The fragment ion mass tolerance was set to 0.5 Da for MS2 spectra. Methionine oxidation and N-

terminal protein acetylation were defined as variable modifications, whereas carbamidomethylation

on cysteine residues was set as a fixed modification. False discovery rate (FDR) in peptide

identification was limited to a maximum of 5% using a decoy database. Quantitation data were

retrieved from the “Precursor ion area detector” node of Proteome Discoverer (v2.0) using 2 ppm

mass tolerance for the peptide extracted ion current (XIC). Finally, for each LDL sample, Skyline

software v4.1 was used to build a spectral library for the ApoB100 protein with the corresponding

identified spectra and to extract all its MS1 areas. Extracted ion chromatograms for the different LDL

samples have been deposited to Panorama with identified PXD011261; URL

(https://panoramaweb.org/j8CaUy.url).

2.8. Molecular modeling of peptide-LDL interactions

The structural model of ApoB-100 in complex with DP3 peptide was built using rigid body molecular

docking simulation and starting from the isolated ab initio models of both docking partners. First of

all, an exhaustive conformational sampling was performed on the peptide in order to identify the

most stable and populated conformational states, which were assumed to be the most favorable to

bind ApoB-100 protein. For that, on one hand, a 3D structure of DP3 was designed from scratch

and consequently used as input for Replica Exchange Molecular Dynamics (REMD) simulations in

implicit water using the AMBER ff12SB force field 33 and NAMD software 34. In each REMD

scheme, 16 independent 100 ns-long simulations (replicas) were performed simultaneously at

Manuscript BBAMEM-19-17 (R1)

12

temperatures ranging from 300 to 613 K. Finally, the representative structure of the most populated

cluster was extracted and used as input structure for the docking simulation. On the other hand, the

3D structure of ApoB-100 protein was built by ab initio protein modeling, since no crystal structure of

protein was available and very low sequence identity templates (ranging from 8 to 13%) were found

in the PDB. Indeed, this computational technique tends to require vast resources, thus only a small

domain of roughly 100 residues around the basic region believed to be responsible for LDL receptor

binding (3189-3261 residues) was selected for modeling. Consequently, a total of 10 ab initio

models were built using ROBETTA server 35 and finally used as input structures for the docking

simulation. Starting from each ApoB-100 protein structure and DP3 peptide previously obtained, 10

independent rigid-body docking simulations were performed using AutoDock Vina software fork

Smina 36 and setting the docking grid box around Apo-B100 3227IKFDKYKAEK3236 region. Finally,

all the docking results were merged and the lowest-energy poses were visually inspected.

2.9. Statistical analysis

Data are presented as mean±SD. One-way ANOVA followed by the Bonferroni or the Storey (q-

value) correction was performed for comparison of >2 groups. Student’s t test was performed for

comparison of 2 groups. P<0.05 was considered significant.

3. Results

3.1. DP3 shows higher stability than the parent peptide LP3 in human serum

Peptide drugs offer high activity, high specificity and low toxicity, but are susceptible to proteolytic

degradation. A strategy to overcome this problem is to produce peptides using non-natural or D-

amino acids, which shows improved stability. Although all-D peptides are not recognized by plasma

proteases, they may be less active than their corresponding all-L peptides because of differences in

the orientation of amino acid side-chains. To preserve the orientation of the side-chains of the

parent peptide in a D-analogue, the D-amino acid residues can be linked in the reverse order to that

of the parent peptide in order to produce a retro-enantio peptide. This approach emphasizes the

Formatted: Font: 11 pt

Manuscript BBAMEM-19-17 (R1)

13

maintenance of the topological native orientation of the side-chains but not of the backbone, where

the amide bond is shift one unit 37. By using this approach, peptides usually preserve their activity

while notably improve their plasma stability. In the literature there are examples where this approach

has been successfully used 30. LP3 and DP3 were synthesized by means of solid-phase peptide

synthesis using a Fmoc/tBu strategy. The purity of both peptides (≥98%) was assessed by HPLC

analysis (Supplemental Fig. S1). Other parameters indicative of the purity and identity of the

synthesized peptides were obtained by HPLC-MS and MALDI-TOF analysis (Supplemental Table

S1). To study the stability of the parent peptide (LP3), it was dissolved in a mixture of HBSS (x1)

and human serum. LP3 showed moderate stability with a calculated half-life of 8 h (Supplemental

Fig. S2A). To achieve an increase in the stability of LP3 for the purpose of in vivo studies, a retro-

enantio version of the peptide (DP3) was synthesized. This approach consisted of using all D-amino

acids and reversing the sequence of the parent peptide. This synthetic strategy has been shown to

be useful 3833. As the retro-enantio version of the peptide is synthesized using D-amino acids, the

sequence is not recognized by serum proteases, thus extending its half-life considerably

(Supplemental Fig. S2B).

3.2. Time- and dose-response blocking effects of LP3 and DP3 on SMase- and PLA2- induced

LDL turbidity

To study the effects of the peptides on SMase-induced LDL aggregation, LDL was incubated with

SMase and PLA2 in the presence and absence of LP3, DP3 and IP321 (negative control) under the

experimental conditions detailed in Supplemental Fig. S3. The peptide concentrations and

peptide/ApoB-100 molar ratios tested in the aggregation of SMase-modified LDL (SMase-LDL) are

detailed in Supplemental Table S2. LP3 and DP3 reduced SMase-LDL aggregation at all the times

tested (4, 6 and 18 h) in a dose-dependent manner (Fig. 1A). DP3 showed a higher inhibitory

capacity than LP3 at concentrations of 5.0, 7.7 and 8.8 µM, and SMase-LDL aggregation was fully

inhibited by LP3 at 10 µM (peptide/ApoB-100 ratio: 4.7/1) and by DP3 at 7.7 µM (peptide/ApoB-100

ratio: 3.5/1). To study the effects of the peptidomimetics on the aggregation of PLA2-modified LDL,

Formatted: English (United States)

Formatted: Font: 11 pt

Formatted: English (United States)

Manuscript BBAMEM-19-17 (R1)

14

LDL was incubated with PLA2 in the presence or absence of LP3, DP3 and IP321 at the doses and

peptide/ApoB-100 ratios detailed in Supplemental Table S3. We did not detect any effect of PLA2 on

LDL aggregation at time points lower than 24 h (data not shown). LP3 and DP3 showed high

inhibitory activity on PLA2-LDL aggregation, although, as in the case of SMase, DP3 showed

outperformed LP3 at concentrations of 10 µM and 14.6 µM at 24, 30 and 36 h (Fig. 1B). PLA2-LDL

aggregation was fully inhibited by LP3 at a concentration of 20.83 µM (peptide/ApoB-100 ratio:

9.8/1) and by DP3 at 14.6 µM (peptide/ApoB-100 ratio: 6.8/1) at all tested times. IP321 did not

inhibit SMase- or PLA2-LDL aggregation at any dose or time tested (Fig. 1A,B). Given that

Apolipoprotein A-I peptidomimetic 4F has been reported to block SMase-LDL aggregation 29, we

compared the capacity of LP3, DP3 and 4F (10 M, ratio 4.7/1, 24 h) to inhibit SMase- and PLA2-

LDL aggregation. SMase-induced aggregation was almost fully blocked by LP3 (86.2%±1.8) and

DP3 (98.1%±1.4), but only partially blocked by 4F (55.7%±2.3). PLA2-LDL aggregation was partially

blocked by LP3 (59.6%±0.8) and 4F (45.8%±1.1) and almost completely inhibited by DP3

(84.4%±1.9) (Table 1). IP321 did not have any significant effect on SMase- or PLA2-LDL

aggregation. None of the peptides alter the turbidimetry values of untreated LDL (Supplemental Fig.

S4).

3.3. LP3 and DP3 decrease the percentage of aggregated particles in SMase-LDL and PLA2-

LDL

To separate aggregated from monomeric LDL particles, LDL samples were subjected to gel filtration

chromatography (GFC). GFC showed that LDL comprised mainly monomeric LDL (Fig. 2a). A small

percentage of residual LDL aggregates was detected in untreated LDL due to the fact that both

untreated and treated LDL were exposed to 37ºC for the same period (Supplemental Fig. S3). In

contrast to LDL, SMase-LDL (Fig. 2b) was composed mainly of LDL aggregates. LP3 (Fig. 2c) and

DP3 (Fig. 2d) partially prevented the formation of LDL aggregates, DP3 showing higher efficiency

than LP3. IP321 did not alter the GFC pattern observed in SMase-LDL (Fig. 2e). As observed with

SMase-LDL, PLA2-LDL (Fig. 2f) showed a large peak of LDL aggregates. LP3 (Fig. 2g), but

Manuscript BBAMEM-19-17 (R1)

15

specially DP3 (Fig. 2h), increased the percentage of monomeric LDL and decreased that of LDL

aggregates. IP321 did not cause any significant alteration in the pattern of PLA2-LDL (Fig. 2i).

LRP1-derived peptides did not alter per se the GFC pattern of LDL (Supplemental Fig. S5).

Quantification of peak areas showed that LP3 and DP3, but not IP321, conferred SMase-LDL and

PLA2-LDL with a GFC profile closer to that of LDL (Fig. 2j,k). TEM images of LDL size distribution

(Fig. 3) showed that LDL comprised mainly individual LDL particles (Fig. 3A). In contrast, SMase-

LDL (Fig. 3B) and PLA2-LDL (Fig. 3C) generated in the presence of IP321 or in the absence of

peptides were formed mostly by aggregated and fused LDL. In the presence of LP3 and DP3,

SMase-LDL (Fig. 3B) and PLA2-LDL (Fig. 3C) were composed of LDL particles with a wide variety

of diameter sizes, although most were in the range present in LDL.

3.4. LP3 and DP3 block the alterations in LDL phospholipid composition in SMase-LDL but

not in PLA2-LDL

Lipoprotein composition analysis (Table 2) revealed that exposure of LDL to SMase or PLA2 did not

alter cholesterol, triglyceride or ApoB content of LDL, either in the presence or absence of the

peptides. In contrast, phospholipid (PL) content of LDL was significantly reduced by both enzymes

(P<0.005). LP3 and DP3 efficiently prevented PL reduction in SMase-LDL (P<0.005). However,

peptides did not show any effect on PL depletion or on non-esterified fatty acid (NEFA) increase

induced by PLA2 treatment. To identify the PL species influenced by enzymatic treatment, HPTLC

was used to separate LDL PLs. This procedure revealed that the main PLs in the LDLs were L--

phosphatidylcholine (L--PC) and sphingomyelin (SM). L--PC levels were unaltered by SMase

treatment (Fig. 4A,B). In contrast, SM was removed from LDL by this treatment (Fig. 4A,C). LP3 and

DP3, but not IP321, efficiently protected LDL-SM against SMase-induced SM degradation (Fig.

4A,C). To examine whether the protective action of LRP1-derived peptides on LDL was due to their

capacity to inhibit SMase activity, we tested the effect of these compounds on the capacity of

SMase to promote SM lipolysis. SMase activity, measured in relation to the amount of

phosphorylcholine generated, was similar in the presence and absence peptides, thereby indicating

Manuscript BBAMEM-19-17 (R1)

16

that none of the peptides exerted direct effects on SMase activity (Supplemental Fig. S6). PLA2

completely removed L--PC from LDL (Fig. 4D,E), leading to a sharp increase in the L--LysoPC

content (Fig. 4D,F). PLA2 did not have any effect on the LDL-SM content (Fig. 4D,G). The peptides

were unable to counteract L--PC decay and the increase in L--LysoPC induced by PLA2 in LDL.

3.5. LP3 and DP3 block the alterations in the LDL electrophoretic profile of SMase-LDL but

not those of PLA2-LDL

To study the effects of the peptides on the electrophoretic mobility of SMase- and PLA2-LDL, we

used gradient gel electrophoresis (GGE) and agarose gels to analyze the modification of LDL in the

presence and absence of the peptides. In native GGE gels (4 h of electrophoresis), the position of

the LDL particle was observed to be determined by particle size. GGE gels showed that SMase

caused a strong loss of the band corresponding to LDL. The lack of this band in SMase-LDL is

attributed to the fact that the pore size of GGE gels precludes the entrance of the largest LDL

aggregates. The band loss induced by SMase was efficiently counteracted by LP3 and DP3 but not

by IP321 (Fig. 5A). In PLA2-LDL (Fig. 5B), GGE showed a minor band corresponding to LDL

(marked as band 1), an intensive band (marked as band 2) with higher electrophoretic mobility

(smaller size) than band 1, and a third band (marked as band 3), which was a minor band with lower

electrophoretic mobility (higher size) than band 1. LP3 and DP3 did not have any effect on the GGE

pattern of PLA2-LDL.

Agarose gels, where LDL runs according to its negative charge, showed that SMase-LDL had a

more diffuse band compared to the well-defined LDL band. SMase-LDL in the presence of LP3 and

DP3 also showed a defined LDL band (Fig. 5C). In contrast to SMase-LDL, PLA2-LDL showed a

single band with a higher negative electric charge than LDL combined with a degradation smear

pattern (Fig. 5D). LP3, and in particular DP3, slightly increased the electrophoretic mobility and

band intensity of PLA2-LDL while they diminished the degradation smear pattern. The peptides did

not cause any change in the electrophoretic mobility of untreated LDL (Supplemental Fig. S7).

Manuscript BBAMEM-19-17 (R1)

17

3.6. DP3 blocks the conformational changes of ApoB-100 in SMase-LDL and PLA2-LDL

We studied the potential conformational modifications of ApoB-100 by Western blot analysis.

Treatment of LDL with SMase (Fig. 6A) or PLA2 (Fig. 6B) increased the exposure of reactive ApoB-

100 epitopes to anti-ApoB-100 antibodies. Both LP3 and DP3 protected ApoB-100 against the

higher reactivity to anti-ApoB-100 antibodies induced by exposure to SMase and PLA2 (Fig. 6A,B).

IP321 had no effect on the increased ApoB-100 reactivity induced by the two enzymatic treatments.

3.7. LRP1-derived peptides electrostatically interact with specific lysine-enriched sequences

in ApoB-100 protein

Limited proteolysis combined with mass spectrometry-based proteomics (LC-MS/MS) evidenced

that DP3 protects specific sequences of ApoB-100 in LDL either by direct interaction or by

remodeling the protein structure (Fig. 7A). The protein/peptide complexes were subjected to limited

proteolysis with trypsin for 1 h and 3 h. The protection of protein sequences by the peptide was

observed by shotgun LC-MSMS analysis at short times (The whole data concerning LDL is available

via PanoramaPublic with identifier PXD011261). At 3-hours digestions, the effect disappeared. The

transient protection of the peptide can be explained by the proteolytic action that trypsin exerted on

the protein complex. Remarkably, most of the protected sequences were also protected in SMase-

LDL unexposed to peptides, suggesting that the aggregation process safeguards them from trypsin

digestion and that these specific sequences are likely to be those that play a major role in LDL

aggregation. The high coincidence between sequences protected by DP3 and those protected by

aggregation indicates that most DP3-protected sequences participate in LDL aggregation, thereby

explaining the high efficiency of LRP1-derived peptides to prevent LDL aggregation.

To further study the molecular basis for ApoB-100 recognition by DP3, peptide homology modeling

and molecular docking methods were used. First, to identify a potential binding site of DP3 peptide

on ApoB-100 protein structure, each ApoB-100 fragment protected by DP3 binding during trypsin

digestion was mapped on the overall ApoB-100 sequence. Virtually all the fragments corresponded

to lipid-associating domains [PMID 18797861] 39, except for a highly positively charged sequence

(3227IKFDKYKAEK3236) located in the ApoB-100 C-terminal region and adjacent to the LDL receptor

Formatted: Highlight

Manuscript BBAMEM-19-17 (R1)

18

binding domain (3385RLTRKRGLK3393) 40-42. This sequence was thus assumed to be responsible

for DP3 peptide recognition and used as the binding site for the molecular docking calculations.

Finally, an energy-based rigid body docking experiment was performed between ApoB-100 protein

and DP3 peptide structures previously modeled alone by ab initio methods. Interestingly, we found

that one of the lowest-energy poses of the docking simulations accommodated DP3 in such an

orientation that ApoB-100 Lys3228, Lys3231 and Lys3233 (but not Lys3226 nor Lys3236) were deeply buried

upon peptide binding. This finding is in strong agreement with the trypsin digestion data described

above. Additionally, strong electrostatic interactions involving same specific key residues of the two

docking partners were also found. In this regard, two of the acidic residues in DP3 correspond to

those assumed to be responsible for the specific recognition between LRP1 module CR9 and

agLDL particles [PMID 25918169] (namely D-Glu3 and D-Glu9) established salt bridge contacts with

two positively charged ApoB-100 residues (namely Lys3229 and Lys3234), while the oxygen atom of

the DP3 D-Asn2 backbone and ApoB-100 Lys3232 were found to be involved in a hydrogen bond

favored by the positive ionic charge carried by the lysine side chain amine group (Fig. 7B).

4. Discussion

A previous study from our group demonstrated that LRP1, in particular the C-terminal half of domain

CR9 (from Gly1127 to Cys1140) named LP3, interacts with agLDL 28. In the current study, we tested

the effect of both the original LP3 version and its retro-enantio version DP3 on SMase and PLA2-

induced LDL aggregation.

In the present study, we observed that PLA2 required larger times and doses than SMase to induce

LDL aggregation. This finding is consistent with previous studies showing that LDL hydrolysis by

PLA2 is slower than that by SMase 34-3643-45. The latter cleaves SM into phosphocholine and

ceramide, whereas PLA2 hydrolyzes the fatty acids in the sn-2 position of PC, generating free fatty

acids and lyso-PC molecules. When SM is lost, ceramide-enriched microdomains have been

proposed to act as non-polar spots at the LDL surface, thereby facilitating LDL aggregation through

hydrophobic associations between domains 44. Here, PLA2-induced PC degradation induces a

Formatted: Subscript

Manuscript BBAMEM-19-17 (R1)

19

dramatic increase in Lyso-L--PC and a strong decrease in LDL surface area, in line with previous

studies 44,46,47. In addition, Here, Both SMase and PLA2 inducedprovoked higher accessibility of

ApoB-100 epitopes to anti-ApoB-100 antibodies, as showed by Western blot analysis of LDL. This

observation would suggest conformational alterations in ApoB-100 linked to phospholipolysis, in line

with previous results from other groups 48. GFC followed by TEM analysis evidenced that LDL

treatment with SMase induced aggregated and fused LDL particles, in agreement with previous

studies 3544. TEM also revealedand that PLA2-LDL comprises a high variety of aggregate sizes,

as previously shown 13.

Here, we demonstrate that LP3 and its derived retro-enantio version (DP3) per se have a high

capacity to inhibit LDL aggregation induced by SMase and PLA2. However, all experimental

approaches (turbidimetry, GFC, and TEM) used in this work demonstrate that DP3 outperforms

LP3. Although all-D peptides are not recognised by proteases, and thus have higher stability, they

may be less active than their corresponding all-L peptides because of differences in the orientation

of amino acid side-chains. To preserve the orientation of the side-chains of the parent L-peptide in a

D-analogue, the retro-enantio approach 49,50 has been used in this study. In this regard, the

preservation of the topological side-chain orientation of DP3 together with a high stability in

biological samples account for its notable performance in the inhibition assays. In addition,

comparison of the capacity of LRP1 (LP3 and DP3) and ApoA-1 (4F) peptides to inhibit LDL

aggregation revealed that at 10 µM (peptide/ApoB-100 ratio: 4.7:1), LP3 and DP3 almost completely

repressed SMase-induced LDL turbidimetry, while L-4F exerted only a partial effect. Previous

studies addressing the efficacy of 4F to inhibit LDL aggregation showed that a molar ratio of 20:1 (4-

fold higher) is required to fully block SMase-induced LDL aggregation 29. However, the

experimental conditions used for LDL aggregation by Nguyen SD et al. (shorter times, and different

procedures for incubation of LDL with L-4F) differed from those applied in our study and thus hinder

a direct comparison between results. To the best of our knowledge, the present study is the first to

report data on the efficacy of 4F to inhibit PLA2-induced LDL aggregation. Here we found that

comparing the inhibitory capacities of L-4F, DP3 and LP3 at 4.7:1 peptide:ApoB-100 ratio, L-4F

Formatted: Font: Italic

Manuscript BBAMEM-19-17 (R1)

20

showed a similar capacity than LP3 to inhibit PLA2-induced LDL aggregation but lower capacity than

DP3.

One key question is how LRP1-derived peptides inhibit LDL aggregation. An important observation

in this regard is that these peptides counteracted alterations in LDL particle size, charge and

phospholipids induced by SMase but not those induced by PLA2. Despite of this, LRP1-derived

peptides exerted protection against SMase and PLA2-induced conformational alterations of ApoB-

100 and against LDL aggregation. These results indicate that 1) peptide protection of ApoB-100

conformation occurreds even in the context of extreme lipolysis and, 2) preservation of ApoB-100

conformation seems to be a common mechanisms underlying LDL protection against SMase and

PLA2-induced LDL aggregation. But, how LRP1-derived peptides preserve ApoB-100

conformation???. In the current study, limited trypsin proteolysis combined with Mass spectrometry-

based proteomic studies of LDL/DP3 complexes evidenced that peptide DP3 interacts with the LDL

surface prior to enzymatic treatment, protecting certain ApoB-100 sequences against trypsin-

induced proteolysis. Most of the peptide-protected sequences in ApoB-100, correspond to lipid-

associated ApoB-100 domains in LDL. The protection that peptide confers to ApoB-100 can be

induced by direct interaction or by remodeling the protein structure. Moreover, a highly positively

charged sequence (3227IKFDKYKAEK3236) located in the ApoB-100 C-terminal region and adjacent to

the LDL receptor binding domain (3385RLTRKRGLK3393) 51-54 was assumed to be responsible for

DP3 peptide recognition and used as the binding site for the molecular docking calculations. In

addition, homology modeling and molecular dynamics methods combined with molecular docking

provided a crucial first insight into the mechanistic basis of LRP1 peptide recognition by ApoB-100

protein. We propose a structural model of DP3 peptide in complex with ApoB-100. This complex is

mainly formed by salt bridge contacts between two of the acidic residues in DP3 (namely D-Glu3

and D-Glu9) and two positively charged ApoB-100 residues (namely Lys3229 and Lys3234), while the

oxygen atom of the DP3 D-Asn2 backbone and ApoB-100 Lys3232 were found to be involved in a

hydrogen bond favored by the positive ionic charge carried by the lysine side chain amine group.

This model is in complete agreement with the trypsin-limited proteolysis data reported herein, as

well as with the study of the recognition between LRP1 module CR9 and aggregated LDL particles

Formatted: Not Superscript/ Subscript

Formatted: Font: Not Italic

Formatted: Font: Not Italic

Formatted: Font: Not Italic

Formatted: Font: Not Italic

Formatted: Font: Not Italic

Manuscript BBAMEM-19-17 (R1)

21

recently published 28. Previous studies have consistently demonstrated that conformational

alterations of ApoB-100 lead to ApoB-100 degradation, a key process for LDL aggregation

48,55,56. In this context, the capacity of LRP1-derived peptides to preserve ApoB-100

conformation explain the high potential of these peptides to protect LDL against LDL aggregation.

Other crucial question is the mechanism by which LRP1-derived peptides protect SM but not PC

from enzymatic hydrolysis. Two different mechanistical options for the protective effect of peptides

against SMase-induced LDL lipolysis have been explored. By one side, we have explored whether

there is a direct effect of peptides on SMase activity. This possibility has been discarded since

SMase activity assays clearly show that peptides does not exert any effect on SMase functional

activity. Other possibility that has been explored is a potential interaction of peptides with the LDL

phospholipid surface. Several peptides and proteins bearing amphipathic properties have been

reported to be able to block LDL aggregation by a direct interaction with LDL phospholipid surface.

In this respect, common examples are 4F peptide 29 or different apolipoproteins 57. Despite the

lack of amphipathic properties in LRP1-derived peptides, this hypothesis has been considered as

possible mechanism of action for the peptides described herein. Theoretical molecular docking

calculations performed in the present study indicate a lack of association between peptides and LDL

phospholipid surface. A key point offering clues about how peptides protect SM is their unability to

protect PC hydrolysis. On the basis of the differential topological distribution of SM and PC in the

LDL surface 46, we propose that the differential anti-phospholypolytic effects of the peptide on SM

and PC phospholipolysis may be determined, at least in part, by the particular imbrication of SM and

PC with free cholesterol on the LDL surface. Although SM, compared to PC, is a minor phospholipid

in LDL (185 versus 450 molecules/LDL), SM is localized in cholesterol-enriched environments,

considered key regulators of phospholipolysis 46. ApoB-100 conformational stabilization could

directly impact the maintenance of the cholesterol-enriched environment structure since, according

to our molecular modeling studies, ApoB-100 sequences complexed with peptide are those close to

cholesterol in the LDL surface. Therefore, SM could be protected by LRP1-derived peptides against

phospholipolysis by its privilege of being part of cholesterol-enriched domains, whose structure

Manuscript BBAMEM-19-17 (R1)

22

would be preserved through the formation of peptide-ApoB-100 complexes. In contrast, PC,

belonging to cholesterol-poor domains, would be unprotected against PLA2-induced lipolysis,

independently of the status of ApoB-100 conformation.

5. Conclusion

Here, we show that LRP1-derived peptides, LP3 and, in particular, its retro-enantio version (DP3),

efficiently preserve LDL against SMase or PLA2-induced LDL aggregation. LP3 and DP3 maintain

LDL particle size, charge, phospholipid composition and ApoB-100 conformation in LDL treated with

SMase, but only ApoB-100 conformation in LDL exposed to PLA2 (summarized in Figure 8,

Graphical abstract). LP3 and DP3 keep ApoB-100 conformation even under extreme lipolysis

through key electrostatic interactions with a highly positively charged sequence

(3227IKFDKYKAEK3236) located in the ApoB-100 C-terminal region. A recent study has shown that the

individual susceptibility of LDL to aggregation is associated with future cardiac deaths independently

of other classical risk factors 4258. LRP(CR9) derived peptides protect LDL against the main

proteolytic enzymes contributing to LDL retention and aggregation in the arterial intima. In the next

future, we are going to test the efficacy of the retro-enantio peptide version (DP3) to reduce

vascular LDL retention and LDL-induced smooth muscle cell (SMC)-foam cell formation in in vivo

models of atherosclerosis. Our first option in the near future will be to inject the peptide DP3

subcutaneously (not focal delivery at the moment) in a murine model of angioplasty-induced

atherosclerosis. This model offers the taking advantage of the increased endothelial permeability,

crucial to favor peptide arrival to vasculature. In a not too distant future, we expect LRP1-derived

peptides to emerge as tools potentially useful to prevent atherosclerosis and cardiovascular

complications, in particular in individuals with aggregation-prone LDL as obese and diabetic patients

DATA AVAILABILITY

Mass spectroscopy proteomics of LDL. Panorama Public PXD011261

https://panoramaweb.org/j8CaUy.url

Manuscript BBAMEM-19-17 (R1)

23

Conflict of interest

R.P., C.P., and T.T. are employees of Iproteos SL. T.T. is cofounder of Iproteos SL and member of

its board of directors.

Acknowledgments

The Ministry of Science and Innovation of Spain, in the framework of the State Plan of Scientific

and Technical Innovation Investigation 2013-2016, awarded funding to the project

“DEVELOPMENT OF AN INNOVATIVE THERAPY FOR THE TREATMENT OF THE

ATHEROSCLEROSIS THROUGH INHIBITION OF CHOLESTEROL VASCULAR

ACCUMULATION”, led by IPROTEOS SL with file number RTC-2016-5078-1. This project was also

financed by the Ministry of Economy, Industry and Competitiveness (MINECO) in the framework of

the Subprogram RETOS-COLABORACIÓN, 2016 call. The project is also co-financed by the

European Union with the objective to promoting the technological development, innovation and

quality research. Support was also received from SAF2017-89613R (to SV), co-financed by the

European Regional Development Fund (ERDF), the Fundació MARATÓ TV3 Project 201521-10 (to

VLl-C), FIS PI13/00364 and PI16/00471 (to JSQ) and FIS PI18/01584 (to VLl-C) from the Instituto

de Salud Carlos III (ISCIII) and co-financed with ERDFs. Support was also received from Ministerio

de Economía y Competitividad to DdG-C (IJCI-2016-29393). CIBER Diabetes y Enfermedades

Metabólicas Asociadas (CIBERDEM; CB07/08/0016 (JSQ) and CIBER Enfermedades

Cardiovasculares (CIBERCV; CB16/1100403 (DdG-C, VLl-C) are projects run by the Instituto de

Salud Carlos III (ISCIII). The CRG/UPF Proteomics Unit is member of ProteoRed PRB3 consortium

which is supported by grant PT17/0019 of the PE I+D+i 2013-2016 from the Instituto de Salud

Carlos III (ISCIII) and ERDF. We also acknowledge support from the Spanish Ministry of Economy

and Competitiveness, “Centro de Excelencia Severo Ochoa 2013-2017”, SEV-2012-0208, and

“Secretaria d’Universitats i Recerca del Departament d’Economia I Coneixement de la Generalitat

de Catalunya” (2017SGR595 to ES and 2017SGR946 to VLl-C. SB, JLS-Q, AR-U, DdG-C and VL-

C are members of the Group of Vascular Biology of the Spanish Society of Atherosclerosis (SEA).

Manuscript BBAMEM-19-17 (R1)

24

References

[1] Castelli WP, Anderson K, Wilson PWF, Levy D. Lipids and risk of coronary heart disease The Framingham Study. Ann Epidemiol. 1992; 2:23–28.

[2] Gordon T, Castelli WP, Hjortland MC, Kannel WB, Dawber TR, High density lipoprotein as a protective factor against coronary heart disease. Am J Med. 1977; 62:707–714.

[3] Guretzki H-J, Gerbitz K-D, Olgemöller B, Schleicher E. Atherogenic levels of low density lipoprotein alter the permeability and composition of the endothelial barrier. Atherosclerosis. 1994; 107:15–24.

[4] Aikawa M, Libby P. The vulnerable atherosclerotic plaque. Cardiovasc Pathol. 2004; 13 :125–138.

[5] Mauriello A, Sangiorgi GM, Virmani R, Trimarchi S, Holmes Jr DR, Kolodgie FD, Piepgras DG, Piperno G, Liotti D, Narula J, Righini P, Ippoliti A, Spagnoli LG. A pathobiologic link between risk factors profile and morphological markers of carotid instability. Atherosclerosis. 2010; 208: 572–580.

[6] Puri R, Nissen SE, Shao M, Elshazly MB, Kataoka Y, Kapadia SR, Tuzcu EM, Nicholls SJ. Non-HDL Cholesterol and Triglycerides: Implications for Coronary Atheroma Progression and Clinical Events., Arterioscler Thromb Vasc Biol. 2016; 36:2220-2228.

[7] Skålén K, Gustafsson M, Knutsen Rydberg E, Hultén LM, Wiklund, Innerarity TL, Boren J. Subendothelial retention of atherogenic lipoproteins in early atherosclerosis. Nature. 2002; 417:750-754.

[8] Öörni K, Pentikäinen MO, Ala-Korpela M, Kovanen PT. Aggregation, fusion, and vesicle formation of modified low density lipoprotein particles: molecular mechanisms and effects on matrix interactions. J Lipid Res. 2000; 41:1703–1714.

[9] Öörni K, Posio P, Ala-Korpela M, Jauhiainen M, Kovanen PT. Sphingomyelinase induces aggregation and fusion of small very low-density lipoprotein and intermediate-density lipoprotein particles and increases their retention to human arterial proteoglycans, Arterioscler Thromb Vasc Biol. 2005; 25:1678-1683

[10] Marathe S, Schissel SL, Yellin MJ, Beatini N, Mintzer R, Williams KJ, Tabas I. Human vascular endothelial cells are a rich and regulatable source of secretory sphingomyelinase. Implications for early atherogenesis and ceramide-mediated cell signaling. J Biol Chem. 1998; 273:4081-4088.

[11] Schissel SL, Schuchman EH, Williams KJ, Tabas I. Zn2+-stimulated Sphingomyelinase Is Secreted by Many Cell Types and Is a Product of the Acid Sphingomyelinase Gene. J Biol Chem. 1996;. 271:18431–18436.

[12] Aviram M, Maor I. Phospholipase A2-modified LDL is taken up at enhanced rate by macrophages. Biochem Biophys Res Commun. 1992; 185:465–472.

[13] Öörni K, Hakala JK, Annila A, Ala-Korpela M, Kovanen PT. Sphingomyelinase induces aggregation and fusion, but phospholipase A2 only aggregation, of low density lipoprotein (LDL) particles: Two distinct mechanisms leading to increased binding strength of LDL to human aortic proteoglycans. J Biol Chem. 1998; 273:29127-29134.

[14] Hakala JK, Öörni K, Pentikäinen MO, Hurt-Camejo E, Kovanen PT. Lipolysis of LDL by Human Secretory Phospholipase A Induces Particle Fusion and Enhances the Retention of LDL to Human Aortic Proteoglycans. Arterioscler Thromb Vasc Biol. 2001; 21:1053-1058.

[15] Tabas I, Li Y, Brocia RW, Xu SW, Swenson TL, Williams KJ. Lipoprotein lipase and sphingomyelinase synergistically enhance the association of atherogenic lipoproteins with smooth muscle cells and extracellular matrix. A possible mechanism for low density lipoprotein and lipoprotein(a) retention and macrophage foam cell formation, J Biol Chem. 1993; 268:20419-20432.

[16] Von Ballmoos MW, Dubler D, Mirlacher M, Cathomas G, Muser J, Biedermann BC. Increased apolipoprotein deposits in early atherosclerotic lesions distinguish symptomatic from

Formatted: Spanish (Spain,

International Sort)

Formatted: Spanish (Spain,

International Sort)

Manuscript BBAMEM-19-17 (R1)

25

asymptomatic patients. Arterioscler Thromb Vasc Biol. 2006; 26:359-364. [17] Khoo JC, Miller E, McLoughlin P, Steinberg D. Enhanced macrophage uptake of low density

lipoprotein after self-aggregation. Arterioscler Thromb Vasc Biol. 1988; 8:348-358. [18] Zhang WY, Ishii I, Kruth HS. Plasmin-mediated macrophage reversal of low density

lipoprotein aggregation. J Biol Chem. 2000; 275:33176-33183. [19] Kruth HS. Sequestration of aggregated low-density lipoproteins by macrophages. Curr Opin

Lipidol. 2002; 13:483-488. [20] Llorente-Cortés V, Martínez-González J, Badimon L. Esterified Cholesterol Accumulation

Induced by Aggregated LDL Uptake in Human Vascular Smooth Muscle Cells Is Reduced by HMG-CoA Reductase Inhibitors, Arterioscler Thromb Vasc Biol. 1998; 18:738-746.

[21] Llorente-Cortés V, Martínez-González J, Badimon L. LDL Receptor–Related Protein Mediates Uptake of Aggregated LDL in Human Vascular Smooth Muscle Cells. Arterioscler Thromb Vasc Biol. 2000; 20:1572-1579.

[22] Llorente-Cortés V, Otero-Viñas M, Hurt-Camejo E, Martínez-González J, Badimon L. Human coronary smooth muscle cells internalize versican-modified LDL through LDL receptor-related protein and LDL receptors. Arterioscler Thromb Vasc Biol. 2002; 22:387-393.

[23] Llorente-Cortés V, Otero-Viñas M, Sánchez S, Rodríguez C, Badimon L. Low-Density Lipoprotein Upregulates Low-Density Lipoprotein Receptor-Related Protein Expression in Vascular Smooth Muscle Cells. Circulation. 2002; 106:3104-3110

[24] Costales P, Aledo R, Vérnia S, Das A, Shah VH, Casado M, Badimon L, Llorente-Cortés V. Selective role of sterol regulatory element binding protein isoforms in aggregated LDL-induced vascular low density lipoprotein receptor-related protein-1 expression, Atherosclerosis. 2010; 213:458–468.

[25] Llorente-Cortés V, Otero-Viñas M, Camino-López S, Llampayas O, Badimon L. Aggregated low-density lipoprotein uptake induces membrane tissue factor procoagulant activity and microparticle release in human vascular smooth muscle cells, Circulation. 2004; 110:452-459.

[26] Camino‐ López S, Badimon L, GonzáLez A, Canals D, Peña E, Llorente‐ Cortés V. Aggregated low density lipoprotein induces tissue factor by inhibiting sphingomyelinase activity in human vascular smooth muscle cells. J Thromb Haemost. 2009; 7:2137–2146.

[27] Allahverdian S, Chehroudi AC, McManus BM, Abraham T, Francis GA. Contribution of intimal smooth muscle cells to cholesterol accumulation and macrophage-like cells in human atherosclerosis., Circulation.2014; 129:1551-1559.

[28] Costales P, Fuentes-Prior P, Castellano J, Revuelta-Lopez E, Corral-Rodríguez MÁ, Nasarre L, Badimon L, Llorente-Cortes V. Domain CR9 of Low Density Lipoprotein (LDL) Receptor-related Protein 1 (LRP1) Is Critical for Aggregated LDL-induced Foam Cell Formation from Human Vascular Smooth Muscle Cells. J Biol Chem. 2015; 290:14852–14865.

[29] Nguyen SD, Javanainen M, Rissanen S, Zhao H, Huusko J, Kivelä AM, Ylä-Herttuala S, Navab M, Fogelman AM, Vattulainen I, Kovanen PT, Öörni K. Apolipoprotein A-I mimetic peptide 4F blocks sphingomyelinase-induced LDL aggregation J Lipid Res. 2015; 56:1206–1221.

[30] Pujals S, Fernández-Carneado J, Ludevid MD, Giralt E. D-SAP: A new, noncytotoxic, and fully protease resistant cell-penetrating peptide. ChemMedChem. 2008; 3:296-301.

[31] Bancells C, Villegas S, Blanco FJ, Benítez S, Gállego I, Beloki L, Pérez-Cuellar M, Ordóñez-Llanos J, Sánchez-Quesada JL. Aggregated Electronegative Low Density Lipoprotein in Human Plasma Shows a High Tendency toward Phospholipolysis and Particle Fusion. J Biol Chem. 2010; 285:32425–32435.

[32] Martínez-Bujidos M, Rull A, González-Cura B, Pérez-Cuéllar M, Montoliu-Gaya L, Villegas S, Ordóñez-Llanos J, Sánchez-Quesada J.L. Clusterin/apolipoprotein J binds to aggregated LDL in human plasma and plays a protective role against LDL aggregation. FASEB J. 2014; 29:1688–1700.

[33] Case D, Cheatham TE 3rd, Darden T, Gohlke H, Luo R, Merz KM Jr, Inufriev A, Simmerling C, Wang B, Woods RJ. The Amber biomolecular simulation programs. J Comput Chem. 2005;26:1668-1688.

[34] Phillips JC, Braun R, Wang W, Gumbart J, Tajkhorshid E, Villa E, Chipot C, Skeel RD, Kalé

Formatted: English (United States)

Manuscript BBAMEM-19-17 (R1)

26

L, Schulten K. Scalable molecular dynamics with NAMD. J Comput Chem. 2005;26:1781-1802.

[35] Kim DE, Chivian D, Baker D. Protein structure prediction and analysis using the Robetta server. Nucleic Acids Res. 2004;32:W526-W531.

[36] Koes DR, Baumgartner MP, Camacho CJ. Lessons learned in empirical scoring with smina from the CSAR 2011 benchmarking exercise. J Chem Inf Model. 2013; 53:1893-1904.

[37] Chorev M, Goodman M. Recent developments in retropeptides and proteins_An ongoing topochemical exploration. Trends Biotechnol. 1995;13:438-445.

[383] Prades R, Oller-Salvia B, Schwarzmaier SM, Selva J, Moros M, Balbi M, Grazú V, De La Fuente JM, Egea G, Plesnila N, Teixidõ M, Giralt E. Applying the retro-enantio approach to obtain a peptide capable of overcoming the blood-brain barrier. Angew. Chemie - Int. Ed. 2015; 54:3967-3972.

[39] Prassl R, Laggner P. Molecular structure of low density lipoprotein: current status and future challenges. Eur Biophys J. 2009;38:145-158.

[40] Segrest JP, Jones MK, De Loof H, Dashti N. Structure of Apolipoprotein B-100 in low density lipoproteins. J Lipid Res. 2001; 42:1346-1367.

[41] Yang CY, Chen SH, Gianturco SH, Bradley WA, Sparrow JT, Tanimura M, Li WH, Sparrow DA, DeLoof H, Rosseneu M, et al. Sequence, structure, receptor-binding domains and internal repeats of human apolipoprotein B-100. Nature. 1986;323:738-742.

[42] Hospattankar AV, Law SW, Lackner K, Brewer HB Jr. Identification of low density lipoprotein receptor binding domains of human apolipoprotein B-100: a proposed consensus LDL receptor binding sequence of apoB-100. Biochem Biophys Res Commun. 1986;139:1078-1085.

[3443] Schissel SL, Tweedie-Hardman J, Rapp JH, Graham G, Williams KJ, Tabas I. Rabbit aorta and human atherosclerotic lesions hydrolyze the sphingomyelin of retained low-density lipoprotein. Proposed role for arterial-wall sphingomyelinase in subendothelial retention and aggregation of atherogenic lipoproteins. J Clin Invest. 1996; 98:1455–1464.

[3544] Sartipy P, Camejo G, Svensson L, Hurt-Camejo E. Phospholipase A2 modification of low density lipoproteins forms small high density particles with increased affinity for proteoglycans and glycosaminoglycans, J Biol Chem. 1999; 274:25913-25920.

[3645] Oestvang J, Bonnefont-Rousselot D, Ninio E, Hakala JK, Johansen B, Anthonsen MW, Modification of LDL with human secretory phospholipase A2 or sphingomyelinase promotes its arachidonic acid-releasing propensity. J Lipid Res. 2004; 45:831–838.

[3746] Hevonoja T, Pentikäinen MO, Hyvönen MT, Kovanen PT, Ala-Korpela M. Structure of low density lipoprotein (LDL) particles: Basis for understanding molecular changes in modified LDL. Biochim Biophys Acta - Mol. Cell Biol. Lipids. 2000; 1488:189–210.

[3847] Benítez S, Camacho M, Arcelus R, Vila L, Bancells C, Ordóñez-Llanos J, Sánchez-Quesada JL. Increased lysophosphatidylcholine and non-esterified fatty acid content in LDL induces chemokine release in endothelial cells. Atherosclerosis. 2004; 177:299–305.

[3948] Kleinman Y, Krul ES, Burnes M, Aronson W, Pfleger B, Schonfeld G. Lipolysis of LDL with phospholipase A2 alters the expression of selected apoB-100 epitopes and the interaction of LDL with cells, J Lipid Res. 1988; 29:729-743.

[49] Soto C, Kindy MS, Baumann M, Frangione B. Inhibition of Alzheimer amyloidosis by peptides that prevent beta-sheet conformation. Biochem Biophys Res Commun. 1996; 226:672–680.

[50] Chalifour RJ, McLaughlin RW, Lavoie L, Morissette C, Tremblay N, Boulé M, Sarazin P, Stéa D, Lacombe D, Tremblay P, Gervais F. Stereoselective interactions of peptide inhibitors with the beta-amyloid peptide. J Biol Chem. 2003;278:34874-34881.

[4051] Sneck M, Nguyen SD, Pihlajamaa T, Yohannes G, Riekkola M-L, Milne R, Kovanen PT, Öörni K. Conformational changes of apoB-100 in SMase-modified LDL mediate formation of large aggregates at acidic pH, J. Lipid Res. 2012; 53:1832-1839.

[5241] Bancells C, Benítez S, Ordóñez-Llanos J, Öörni K, Kovanen PT, Milne RW, Sánchez-Quesada JL. Immunochemical Analysis of the Electronegative LDL Subfraction Shows That Abnormal N-terminal Apolipoprotein B Conformation Is Involved in Increased Binding to Proteoglycans, J Biol Chem. 2011; 286:1125–1133.

[53] Liu H, Scraba DG, Ryan RO. Prevention of phospholipase-C induced aggregation of LDL by

Formatted: Indent: Left: 0 cm,

Hanging: 1.13 cm

Formatted: Font: (Default) Arial, 11

Formatted: Font: (Default) Arial

Formatted: Font: (Default) Arial

Formatted: Font: (Default) Arial, 11

Formatted: Check spelling and

grammar

Formatted: English (United States)

Formatted: Font: (Default) Arial

Formatted: English (United States)

Formatted: Font: (Default) Arial

Formatted: Check spelling and

grammar

Formatted: Spanish (Spain,

International Sort)

Formatted: Indent: Left: 0 cm,

Hanging: 1.13 cm

Formatted: Font: (Default) Arial,

Spanish (Spain, International Sort)

Formatted: Spanish (Spain,

International Sort)

Manuscript BBAMEM-19-17 (R1)

27

amphipathic apolipoproteins. FEBS Lett. 1993; 316:27–33. [4254] Ruuth M, Nguyen SD, Vihervaara T, Hilvo M, Laajala TD, Kondadi PK, Gisterå A,

Lähteenmäki H, Kittilä T, Huusko J, Uusitupa M, Schwab U, Savolainen MJ, Sinisalo J, Lokki M-L, Nieminen MS, Jula A, Perola M, Ylä-Herttula S, Rudel L, Öörni A, Baumann M, Baruch A, Laaksonen R, Ketelhuth DFJ, Aittokallio T, Jauhiainen M, Käkelä R, Borén J, Williams KJ, Kovanen PT, Öörni K. Susceptibility of low-density lipoprotein particles to aggregate depends on particle lipidome, is modifiable, and associates with future cardiovascular deaths. Eur Heart J. 2018; 39:2562-2573

Figure Legends

Fig.1. LRP1-derived peptides (LP3 and DP3) decrease SMase- and PLA2-induced LDL aggregation in a dose-dependent manner. LDL was used at 1.44 mg/mL ApoB. Peptides were tested at

increasing concentrations (from 2.5 to 10 M for SMase-LDL and from 4.16 to 20.83 M for PLA2-LDL). Aggregation of LDL was induced by SMase (A) or PLA2 (B) and monitored at the indicated times by measuring turbidimetry (absorbance at 405 nm). Data are mean ± SD of 2 experiments performed in duplicate. Unsighted bar errors are minor than symbol size.*P<0.005 versus SMase-

LDL + IP321; P<0.005 versus SMase-LDL + LP3.

Fig. 2. LRP1-derived peptides (LP3 and DP3) reduce the AUC of agLDL peak in SMase-LDL and PLA2-LDL. LDL was untreated (LDL) or treated with SMase (40 U/L) and PLA2 (50 µg/L) in the absence (w/o peptide) or presence of LP3, DP3 or IP321 (10 µM, peptide/ApoB-100 ratio: 4.7:1, 24 h). LDLs (0.5 mL at 0.4 mg/mL ApoB) were chromatographed at a flow rate of 0.5 ml/min using a Superose 6 column in a AKTA-FPLC system (GE Healthcare) as described in Material and Methods. Representative GFC of: a) non enzymatically-treated LDL; b) LDL incubated with SMase; c) LDL incubated with SMase in the presence of LP3; d) LDL incubated with SMase in the presence of DP3; e) LDL incubated with SMase in the presence of IP321; f) LDL incubated with PLA2; g) LDL incubated with PLA2 in the presence of LP3; h) LDL incubated with PLA2 in the presence of DP3; and i) LDL incubated with PLA2 in the presence of IP321. j) and k) Bar graphs showing the percentage of aggregated versus monomeric LDL in SMase-LDL and PLA2-LDL, respectively, calculated from the chromatograms as area under the curve as explained in Methods.

Fig. 3. LRP1-derived peptides (LP3 and DP3) diminish the proportion of large size LDL aggregates in SMase-LDL and PLA2-LDL. Untreated LDL (A) or LDL treated with SMase (40 U/L) (B) and PLA2 (50 µg/L) (C) in the presence of LP3, DP3 or IP321 (10 µM, peptide/ApoB-100 ratio: 4.7:1, 24 h) or in absence of peptides (w/o peptide). TEM was performed in a Jeol 120 kV JEM-1400 microscope in the LDL preparations treated as explained above for 24 h at 37ºC. Black bars, 500 nm.

Fig. 4. LRP1-derived peptides (LP3 and DP3) prevent SM depletion in SMase-LDL but not L--PC depletion in PLA2-LDL. LDL was untreated (LDL) or treated with SMase (40 U/L) and PLA2 (50 µg/L) in the absence (w/o peptide) or presence of LP3, DP3 or IP321 (10 µM, peptide/ApoB-100 ratio: 4.7:1, 24 h) and the phospholipid pattern was analyzed by HPTLC after lipid extraction. Representative HPTLC of LDL (A and D) and bar graphs (B, C, E, F and G) showing quantification of phospholipid bands. Results are expressed as percentage of PL relative to untreated LDL and

shown as mean ± SD of 3 independent experiments performed in triplicate. L--PC indicates L--

phosphatidylcholine; SM: sphingomyelin; L--lyso-PC: L--lysophosphatidylcholine. *P<0.005 versus LDL.

Fig. 5. LRP1-derived peptides (LP3 and DP3) counteract LDL alterations in size and electric charge induced by SMase but not those induced by PLA2. LDL was untreated (LDL) or treated with SMase

Manuscript BBAMEM-19-17 (R1)

28

(40 U/L) and PLA2 (50 µg/L) in the absence (w/o peptide) or presence of LP3, DP3 o IP321 (10 µM, ratio peptide/ApoB-100: 4.7:1, 24 hours). LDL electrophoretic mobility was analyzed by GGE (A and B) and agarose gels (C and D). In GGE, LDL (5 µg/well) was electrophoresed in 2-16% acrylamide native gradient gels. LDLs were prestained with 1% Sudan black. In agarose gel electrophoresis, LDL (2 µg/well) was electrophoresed in 0.5% agarose gels, which were dehydrated and stained with Coomassie Blue. Peptides were assayed at 10 µM (Ratio Peptide/ApoB= 4.7/1)

Fig. 6. LRP1-derived peptides (LP3 and DP3) counteract ApoB-100 conformational alterations induced by SMase and PLA2 in LDL. LDL was untreated (LDL) or treated with SMase (40 U/L) and PLA2 (50 µg/L) in the absence (w/o peptide) or presence of LP3, DP3 or IP321 (10 µM, peptide/ApoB-100 ratio: 4.7:1, 24 h). Representative western blot analysis of ApoB-100 in LDL exposed to SMase (A) or PLA2 (B). LDL (5 µg/well) was electrophoresed in 10% acrylamide gels under non-reducing conditions for 2 h at 100 V and transferred to nitrocellulose membranes, which were then incubated with anti-ApoB-100 antibodies. Fig. 7. DP3 protect ApoB-100 sequences against tryptic-limited proteolysis A) List of ApoB100 peptides that are protected by DP3 and by LDL aggregation as shown by tryptic-limited proteolysis coupled to mass spectrometry analysis. Green indicates signal detected, Yellow a poor signal, and Red no MS signal. B) Structural details of the molecular interaction between Apo-B100 protein and DP3 peptide. View of the ApoB-100 protein surface contacting the DP3 peptide: the basic residues of ApoB-100 exposed to trypsin digestion and buried deeply upon peptide binding highlighted in teal and violet, respectively. DP3 peptide acid residues responsible for ApoB-100 binding highlighted in yellow sticks.

Fig. 8. Fig. 8. Schematic representation of the molecular basis for the protective effect of LRP1-derived peptides against SMase- and PLA

2-induced LDL aggregation. DP3 forms a complex with

ApoB-100 and this molecular interaction stabilizes ApoB-100 conformation. ApoB-100 conformation stabilization could guarantee the structural preservation of surface colesterol-enriched environments, where SM is located. Structural preservation of cholesterol, a key regulator of phospholipolysis, would protect SM from SMase activity. As a result, LDL complexed with DP3 remains unaltered when exposed to SMase. This scenario changes when LDL complexed with DP3 is exposed to PLA

2. The target for PLA

2 is PC, phospholipid associated with poor-colesterol

environments. Therefore, PC is unprotected against the attack of PLA2, that hydrolyzes PC producing lyso-PC and NEFA. Remarkably, LDL complexed with DP3 is protected against SMase and PLA2-induced aggregation even in conditions of extreme phospholipolysis, indicating that the maintenance of ApoB-100 conformation is key in the prevention of LDL aggregation.

Formatted: Subscript

Formatted: English (United States)

Figure 1

C o n c e n tra t io n (u M )

Ab

so

rb

an

ce

(4

05

nm

)

0 2 4 6 8 1 0 1 2

0 .1 0

0 .1 5

0 .2 0

0 .2 5L P 3

D P 3

IP 3 2 1

C o n c e n tra t io n (u M )

Ab

so

rb

an

ce

(4

05

nm

)

0 2 4 6 8 1 0 1 2

0 .1

0 .2

0 .3

0 .4L P 3

D P 3

IP 3 2 1

C o n c e n tra t io n (u M )

Ab

so

rb

an

ce

(4

05

nm

)

0 2 4 6 8 1 0 1 2

0 .2

0 .4

0 .6

0 .8

1 .0L P 3

D P 3

IP 3 2 1

Time 4h

Time 8h

Time 18h

SMase-LDL

C o n c e n tra t io n (u M )

Ab

so

rb

an

ce

(4

05

nm

)

0 5 1 0 1 5 2 0 2 5

0 .0

0 .1

0 .2

0 .3

0 .4

0 .5L P 3

D P 3

IP 3 2 1

C o n c e n tra t io n (u M )

Ab

so

rb

an

ce

(4

05

nm

)

0 5 1 0 1 5 2 0 2 5

0 .0

0 .2

0 .4

0 .6

0 .8L P 3

D P 3

IP 3 2 1

C o n c e n tra t io n (u M )

Ab

so

rb

an

ce

(4

05

nm

)

0 5 1 0 1 5 2 0 2 5

0 .0

0 .2

0 .4

0 .6

0 .8

1 .0L P 3

D P 3

IP 3 2 1

PLA2-LDL

Time 24h

Time 30h

Time 36h

A B

*

*

*

* *

* *

* *

*

*

* * *

*

*

*

*

*

* *

* *

*

*

*

* *

* *

*

* *

*

* *

*

*

* *

Figure 1

Figure 2

1a001:1_UV

0.0

10.0

20.0

30.0

40.0

50.0

60.0

70.0

80.0

mAU

0.0 2.0 4.0 6.0 8.0 10.0 ml

Aggregated LDL

Monomeric LDL

3a001:1_UV

0

50

100

150

200

250

mAU

0.0 2.0 4.0 6.0 8.0 10.0 ml

Aggregated

Monomeric

4a001:1_UV

0

20

40

60

80

mAU

0.0 2.0 4.0 6.0 8.0 10.0 ml

Aggregated

Monomeric

5a001:1_UV

0.0

10.0

20.0

30.0

40.0

50.0

60.0

70.0

mAU

0.0 2.0 4.0 6.0 8.0 10.0 ml

Aggregated

Monomeric 7a001:1_UV

0

50

100

150

200

250

300

mAU

0.0 2.0 4.0 6.0 8.0 10.0 ml

Aggregated

Monomeric

8a001:1_UV

0

50

100

150

mAU

0.0 2.0 4.0 6.0 8.0 10.0 ml

Aggregated

Monomeric

9a001:1_UV

0.0

20.0

40.0

60.0

80.0

mAU

0.0 2.0 4.0 6.0 8.0 10.0 ml

Aggregated

Monomeric

10a001:1_UV

0.0

10.0

20.0

30.0

40.0

50.0

60.0

mAU

0.0 2.0 4.0 6.0 8.0 10.0 ml

Aggregated

Monomeric

12a001:1_UV

0

50

100

150

200

250

mAU

0.0 2.0 4.0 6.0 8.0 10.0 ml

Aggregated

Monomeric

w/o peptide LP3

DP3 IP321

SMase-LDL

LDL

DP3 IP321

PLA2-LDL

w/o peptide LP3

SMas

e-i

nd

uce

d L

DL

aggr

ega

tio

n

(%)

PLA

2-in

du

ced

LD

L ag

gre

gati

on

(

%)

a

b c

d e

f g

h i

SMase-LDL PLA2-LDL

0

20

40

60

80

100

Aggregated

Monomeric

0

20

40

60

80

100

Aggregated

Monomeric

j K

Figure 2

Figure 3

b

c d e

f

h

w/o peptide

LDL

LP3

SMase-LDL PLA2-LDL

DP3 IP321

w/o peptide LP3

DP3 IP321

A

B C

Figure 3

Figure 4

LDL LP3 DP3 IP321

SMase-LDL

LP3 DP3 IP321

PLA2-LDL

SM

L-a-PC

LDL

w/o

peptide

w/o

peptide

L-a-LysoPC

SM

le

ve

ls i

n P

LA

2-L

DL

(re

lativ

e t

o L

DL

)

LD

L

w/o

pept ide

LP

3

DP

3

IP321

0

2 0

4 0

6 0

8 0

1 0 0

1 2 0 L-a

-P

C l

ev

els

in

PL

A2

-L

DL

(re

lativ

e t

o L

DL

)

LD

L

w/o

pept ide

LP

3

DP

3

IP321

0

2 0

4 0

6 0

8 0

1 0 0

1 2 0

L-a

-ly

so

PC

le

ve

ls i

n P

LA

2-L

DL

(re

lativ

e t

o L

DL

)

LD

L

w/o

pept ide

LP

3

DP

3

IP321

0

1

2

3

4

5

6

L-a

-P

C l

ev

els

in

SM

as

e-L

DL

(re

lativ

e t

o L

DL

)

LD

L

w/o

pept ide

LP

3

DP

3

IP321

0

2 0

4 0

6 0

8 0

1 0 0

1 2 0

SM

le

ve

ls i

n S

Ma

se

-L

DL

(re

lativ

e t

o L

DL

)

LD

L

w/o

pept ide

LP

3

DP

3

IP321

0

2 0

4 0

6 0

8 0

1 0 0

1 2 0

SM

L-a-PC

SMase-LDL SMase-LDL

PLA2-LDL

PLA2-LDL

PLA2-LDL

* *

* * * *

* * * *

A B C

D E F

G

Figure 4

Figure 5

GGE

Agarose gel

w/o P LP3 DP3 IP321

SMase-LDL

LP3 DP3 IP321

PLA2-LDL

-

+

LDL LDL

w/o P LP3 DP3 IP321

SMase-LDL

LDL LP3 DP3 IP321

PLA2-LDL

-

+

-

+

-

+

LDL LDL

LDL LDL

1 2

3

w/o P

w/o P LDL

A B

C D

Figure 5

Figure 6

LDL LP3 DP3 IP321

SMase-LDL

LDL LP3 DP3 IP321

PLA2-LDL

ApoB-100 ApoB-100

250 kDa

250 kDa

Mw Mw

A B

w/o

peptide w/o

peptide

Figure 6

Figure 7 A

B

Peptide Start End Status Protein nat

iveL

DL

nat

iveL

DL

+ D

P3

Smas

e-LD

L

Smas

e-LD

L +

DP

3

YELKLAIPEGK 128 138 protected sp|P04114|APOB_HUMAN 1h

3h

YGMVAQVTQTLKLEDTPK 275 292 protected sp|P04114|APOB_HUMAN 1h

3h

LEDTPKINSR 287 296 protected sp|P04114|APOB_HUMAN 1h

3h

IKFDKYKAEK 3227 3236 protected sp|P04114|APOB_HUMAN 1h

3h

FDKYKAEK 3229 3236 protected sp|P04114|APOB_HUMAN 1h

3h

Figure 7

ApoB-100/DP3 complex

STABILIZATION OF ApoB-100 CONFORMATION

SMase PLA2

STABILIZATION OF CHOLESTEROL/SM

enriched environment

Unaltered PL Surface Unaltered LDL size

NO EFFECT ON PC-enriched/colesterol-poor

environment

Altered PL Surface Decreased LDL size

LDL aggregation

Lyso-PC & NEFA

Figure 8 Figure 8

Supplemental Tables

Table S1. HPLC chromatography peptide characteristics

Peptide Retention time (min) Purity (%) Calculated mass (Da) Found Mass (Da)

LP3 4,9 99 1542 1543 [M+H]+

DP3 3,1 98 1481 1482 [M+H]+

IP321 3,3 98 1126 1127 [M+H]+

HPLC spectra were recorded in Waters Alliance 2695 separation module equipped with a 2998

photodiode array detector, a Sunfire C18 column (100 mm x 4.6 mm, 3.5 µm, 100 Å, Waters)

and Empower software (Waters); flow rate = 1 mL/min; gradient 0-100% B in 8 min (A = 0.045%

trifuloroacetic acid in water, and B = 0.036% trifluoroacetic acid in acetonitrile), detection at 220

nm. HPLC-MS were recorded in a Waters Alliance 2695 equipped with a 2998 photodiode array

detector, ESI-MS micromass ZQ, a Sunfire C18 column (2.1 mm x 100 mm, 3.5 µm, 100 Å,

Waters) and Masslynx software (Waters); flow rate = 0.3 mL/min; gradient 0-100% B in 8 min (A

= 0.1% formic acid in water, and B = 0.07% formic acid in acetonitrile).

Table S2. Concentration and ratio of peptides used in SMase-induced LDL

turbidimetry experiments

Concentration (M) Peptide/ApoB-100 Molar Ratio

10 4.7/1

8.75 4.1/1

7.7 3.5/1

5 2.4/1

2.5 1.2/1

Table S3. Concentration and ratio of peptides used in PLA2-induced LDL

turbidimetry experiments

Concentration (uM) Peptide/ApoB-100 Molar Ratio

20.83 9.8/1

14.60 6.8/1

10 4.7/1

6.25 2.9/1

4.16 2.0/1

Supplementary Material (for online publication)Click here to download Supplementary Material (for online publication): BBA Supplemental Tables and Figures R1.docx

Figure S1

Figure S1. HPLC trace of LP3 (A) and DP3 (B). Each compound was analyzed using a C18 analytical HPLC column (Sunfire, Waters).

Flow rate 1 mL/min; Gradient 0-100% a-b in 8 min (a= 0.045% trifluoroacetic acid in water and b 0.036% trifluoroacetic acid in acetonitrile).

Detection wavelength 220 nm. The peptide shows purity above 98%. The observed mass (HPLC-MS, not shown) conforms to the mass of

the target compound.

A B

Figure S2

Figure S2. Percentage of peptide LP3 (A) and of DP3 (B) versus incubation time in human serum. Experiment was performed in triplicate. Data are shown

as the mean ± SD

1) Generation of Normolipemic Pool of humanplasma

2) LDL isolation

3) LDL modification by SMase and PLA2 with and w/o peptides

4) hVSMCc are exposed for 2h with modified LDL

Turbidimetry(Absorbance 405 nm)

37ºC18 hours

Lipid extraction and TLC to quantify

intracelular CE/FC ratio

Confocal MicroscopyBODIPY

Figure S3

Figure S3. Schematic representation of the experimental procedure

Figure S4

Figure S4. LRP1-derived peptides does not increase LDL turbidimetry. LDL at 1.44 mg/ml apoB was exposed to peptides (10 M,

Ratio Peptide/ApoB= 4.7/1) for 24 hours. Aggregation of LDL was monitored by measuring absorbance at 405 nm. Data are mean ±

SD of 4 experiments performed in duplicate. 5

Ab

so

rba

nc

e(4

05

nm

)

0

0,1

0,2

0,3

0,4

0,5

w/opeptide

LP3 DP3 IP321

1a001:1_UV

0.0

10.0

20.0

30.0

40.0

50.0

60.0

70.0

80.0

mAU

0.0 2.0 4.0 6.0 8.0 10.0 ml

LDL + buffer

2a001:1_UV

0.0

10.0

20.0

30.0

40.0

50.0

60.0

70.0

mAU

0.0 2.0 4.0 6.0 8.0 10.0 ml

LDL + DP3

Figure S5

Figure S5. Gel filtration chromatography (GFC) of LDL (0.5 mL at 0.4 mg/mL ApoB) unexposed or exposed to DP3 (10 M, Ratio

Peptide/ApoB= 4.7/1) for 24 hours. LDLs were chromatographed at a flow rate of 0.5 ml/min using a Superose 6 column in a AKTA-FPLC

system (GE Healthcare) as described in Methods.

Figure S6

Figure S6. Effect of LP3, DP3, IP321 and Manumycin A on SMase activity. SMase activity was measured by the Amplex Red assay

based on the SMase-induced formation of phosphorylcholine. The results are shown as mean ± SD of three independent experiments

performed in triplicate. *P<0.005 control (w/o peptide)

5 3 0 -5 9 0 n m

S M a s e (m U /m L )

SM

as

e a

cti

vit

y (

a.u

.)

0 .0 0 .2 0 .4 0 .6 0 .8 1 .0 1 .2

0

2 0 0 0 0

4 0 0 0 0

6 0 0 0 0

8 0 0 0 0w /o p e p tid e

L P 3

D P 3

IP 3 2 1

M a n u m y c in

* * * * *

Figure S7

LDL

-

+

LP3 DP3 IP321

Figure S7. Agarose gel electrophoresis of LDL (2 g/well) exposed to LP3, DP3 or IP321 (10 M, Ratio Peptide/ApoB= 4.7/1) for 18 hours.

LDLs were electrophoresed in 0.5% agarose gels that were dehydrated and stained with Coomassie Blue.

AUTHOR DECLARATION TEMPLATE

We wish to confirm that there are no known conflicts of interest associated with this publication

entitled “Molecular basis for the protective effects of low-density lipoprotein receptor-related

protein 1 (LRP1)-derived peptides against LDL aggregation” and there has been no significant

financial support for this work that could have influenced its outcome.

We state that Roger Prades, Chiara Pallara and Teresa Tarragó are employees of Iproteos SL.

Teresa Tarragó is cofounder of Iproteos SL and member of its board of directors.

We confirm that the manuscript has been read and approved by all named authors and that there

are no other persons who satisfied the criteria for authorship but are not listed.

We further confirm that the order of authors listed in the manuscript has been approved by all of

us.

We confirm that we have given due consideration to the protection of intellectual property

associated with this work and that there are no impediments to publication, including the timing of

publication, with respect to intellectual property. In so doing we confirm that we have followed the

regulations of our institutions concerning intellectual property.

We further confirm that any aspect of the work covered in this manuscript that has involved human

patients has been conducted with the ethical approval of all relevant bodies.

We understand that the Corresponding Author is the sole contact for the Editorial process

(including Editorial Manager and direct communications with the office). He/she is responsible for

communicating with the other authors about progress, submissions of revisions and final approval

of proofs.

We confirm that we have provided a current, correct email address which is accessible by the

Corresponding Author and which has been configured to accept email from VICENTA LLORENTE

CORTES ([email protected])

Signed by the corresponding author in the name of the rest of authors

Vicenta Llorente Cortes

*Conflict of Interest