ArticlePublisher preview available

Enhanced Targeted Repair of Vascular Injury by Apoptotic‐Cell‐Mimicking Nanovesicles Engineered with P‐Selectin Binding Peptide

Wiley
Advanced Functional Materials
Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract and Figures

Modulating inflammation is crucial for repairing vascular injury. Phagocytosis of apoptotic cells represents an effective mechanism for attenuating inflammation and improving regeneration during natural healing. However, strategies for repairing vascular injuries using biomaterials derived from apoptotic cells are still undeveloped. Herein, apoptotic body‐mimetic nanovesicles (ApoNVs) derived from rat adipose‐derived mesenchymal stem cells (rASCs) are prepared using a one‐step extrusion method. ApoNVs inherit the unique anti‐inflammatory and pro‐regenerative properties of the parental apoptotic rASCs, as evidenced by enhanced M2 polarization of macrophages and promoted endothelial cell proliferation and migration following treatment with ApoNVs. Moreover, ApoNVs enhance the contractile phenotype of vascular smooth muscle cells through the mediation of ApoNVs‐induced repolarized macrophages. After engineering ApoNVs with P‐selectin binding peptide (ApoNVs‐PBP), their ability to target injured artery increased nearly sevenfold compared to unmodified ApoNVs. In a rat wire‐mediated femoral artery injury model, ApoNVs‐PBP effectively suppress inflammation and significantly reduce blood flow velocity and neointimal hyperplasia at the injury site. ApoNVs exhibit similar therapeutic effects, though to a lesser extent. This study provides strong evidence validating the targeted delivery of ApoNVs as an innovative approach for repairing vascular injury and highlights their potential in treating other inflammatory diseases.
Regulatory effects of ApoNVs on macrophages, HUVECs and HAVSMCs. a) Schematic illustration of the experimental design used to assess the effect of ApoNVs on macrophages, HUVECs and HAVSMCs. b) Representative fluorescence images showing the cellular uptake of DiD‐labeled ApoNVs (red) by macrophages. The cytoskeleton F‐actin was stained with YF 488‐Phalloidin (green) and the nuclei were stained with DAPI (blue). c) Flow cytometry analysis of the expression of the inflammatory M1 marker CD86 and the anti‐inflammatory M2 marker CD206 in macrophages treated with or without ApoNVs. d,e) NO (d) and TNF‐α (e) production by macrophages with or without ApoNVs treatment. f) Representative fluorescence images showing the cellular uptake of DiD‐labeled ApoNVs (red) by HUVECs. The cytoskeleton F‐actin was stained with YF 488‐Phalloidin (green), and the nuclei were stained with DAPI (blue). g) HUVEC proliferation with or without ApoNV treatment. h,i) Tube formation of HUVECs with or without ApoNV treatment. j,k) Chemotactic migration of HUVECs with or without ApoNV treatment. l) Representative fluorescence images showing the cellular uptake of DiD‐labeled ApoNVs (red) by HAVSMCs. The cytoskeleton F‐actin was stained with YF 488‐Phalloidin (green) and the nuclei were stained with DAPI (blue). m) HAVSMC proliferation with or without ApoNV treatment. n,o) Chemotactic migration of HAVSMCs with or without ApoNV treatment. Data are presented as the mean ± standard error of the mean (SEM) (n = 3 per group). Statistical significance was determined by one‐way ANOVA with Tukey's test (d, e) and an independent unpaired two‐tailed Student's t test (g, i, k, m, o). *p < 0.05, **p < 0.01, ***p < 0.001 and ns: not significant.
… 
This content is subject to copyright. Terms and conditions apply.
RESEARCH ARTICLE
www.afm-journal.de
Enhanced Targeted Repair of Vascular Injury by
Apoptotic-Cell-Mimicking Nanovesicles Engineered with
P-Selectin Binding Peptide
Ruixin Zhang, Shunshun Yan, Shichun Li, Yu Shi, Yueyue Yang, Junwu Liu, Zixuan Dong,
Ting Wang, Jingxin Yue, Quhan Cheng, Ye Wan, Su Zhang, Shanshan Kang, Deling Kong,*
Kai Wang,* and Xiaoling Fu*
Modulating inflammation is crucial for repairing vascular injury. Phagocytosis
of apoptotic cells represents an effective mechanism for attenuating
inflammation and improving regeneration during natural healing. However,
strategies for repairing vascular injuries using biomaterials derived from
apoptotic cells are still undeveloped. Herein, apoptotic body-mimetic
nanovesicles (ApoNVs) derived from rat adipose-derived mesenchymal stem
cells (rASCs) are prepared using a one-step extrusion method. ApoNVs inherit
the unique anti-inflammatory and pro-regenerative properties of the parental
apoptotic rASCs, as evidenced by enhanced M2 polarization of macrophages
and promoted endothelial cell proliferation and migration following treatment
with ApoNVs. Moreover, ApoNVs enhance the contractile phenotype of
vascular smooth muscle cells through the mediation of ApoNVs-induced
repolarized macrophages. After engineering ApoNVs with P-selectin binding
peptide (ApoNVs-PBP), their ability to target injured artery increased nearly
sevenfold compared to unmodified ApoNVs. In a rat wire-mediated femoral
artery injury model, ApoNVs-PBP effectively suppress inflammation and
significantly reduce blood flow velocity and neointimal hyperplasia at the
injury site. ApoNVs exhibit similar therapeutic effects, though to a lesser
extent. This study provides strong evidence validating the targeted delivery of
ApoNVs as an innovative approach for repairing vascular injury and highlights
their potential in treating other inflammatory diseases.
R. Zhang, Y. Shi, Y. Yang, J. Liu, J. Yue, Q. Cheng, Y. Wan, S. Zhang,
S. Kang, D. Kong, K. Wang
State Key Laboratory of Medicinal Chemical Biology
Key Laboratory of Bioactive Materials for the Ministry of Education
College of Life Sciences
Nankai University
Tianjin , P. R. China
E-mail: kongdeling@nankai.edu.cn;@nankai.edu.cn
S. Yan, S. Li, Z. Dong, X. Fu
School of Biomedical Sciences and Engineering
South China University of Technology
Guangzhou International Campus
Guangzhou , P. R. China
E-mail: msxlfu@scut.edu.cn
The ORCID identification number(s) for the author(s) of this article
can be found under https://doi.org/./adfm.
DOI: 10.1002/adfm.202405574
1. Introduction
Cardiovascular disease remains the lead-
ing cause of death worldwide, with approx-
imately . million deaths attributed to
it in .[]It is estimated that around
. million people suer from coro-
nary heart disease, and both the preva-
lence and severity of the disease continue
to rise.[]Late-stage coronary heart dis-
ease can lead to coronary artery occlu-
sion, and treatment options usually in-
volve percutaneous coronary intervention
(PCI)[]with vascular injury as an almost
inevitable consequence.[]Vascular injury
caused by PCI further triggers a series
of cellular and molecular events that in-
crease the risk of thrombus formation,
restenosis, and even reemergence of arterial
occlusion.[]Research has shown that the
restenosis rate is approximately –%,[]
significantly aecting the clinical safety
and eectiveness of PCI.[]Without timely
and eective interventions, thrombosis and
restenosis can progress to secondary my-
ocardial infarction or stroke.[]Drug-eluting
T. Wa ng
Tianjin Key Laboratory of Urban Transport Emission Research
College of Environmental Science and Engineering
Nankai University
Tianjin , P. R. China
X. Fu
National Engineering Research Center for Tissue Restoration and
Reconstruction and Innovation Center for Tissue Restoration and
Reconstruction
Guangzhou , P. R. China
X. Fu
Laboratory of Biomedical Engineering of Guangdong Province
South China University of Technology
Guangzhou , P. R. China
Adv. Funct. Mater. 2024,34,  ©  Wiley-VCH GmbH
2405574 (1 of 22)
... A cellpenetrating peptide has been utilized to modify apoEVs for the treatment of ischemic stroke, yielding promising results [20]. A research team successfully conjugated P-selectin binding peptide to apoEVs and confirmed that the targeting ability of apoEVs to injured arteries significantly increased [21]. Taking these inspirations, we propose that the strategic integration of PMSC-apo-EVs' inherent bone regulatory ability with engineering Graphical Abstract technologies could potentially instigate a paradigm shift in the therapeutic conundrum of HO. ...
Article
Full-text available
Heterotopic Ossification (HO), refers to pathological extra skeletal bone formation, and there are currently no reliable methods except surgery to reverse these unexpected calcified tissues. Apoptotic vesicles (ApoEVs) are membrane-bound vesicles released by apoptotic cells, which are involved in metabolism regulation and intercellular communication. Due to its superior trauma-healing ability, the hard palate mucosa is expected to become an essential resource for tissue engineering. This work presents a minimally invasive nanotherapy based on an engineered apoEV. Briefly, apoEVs were extracted from hard palate mucosa and engineered with bone-targeting peptide SDSSD to treat HO. This engineered apoEV not only can achieve directed localization of heterotopic bones but also has the compelling dual function of promoting osteoclastic differentiation while inhibiting osteogenic differentiation. The underlying mechanism involves the activation of Hippo and Notch pathways, as well as the regulation of pyrimidine metabolism. We envision that this engineered apoEV may be a feasible and effective strategy for reversing HO. Graphical Abstract
Article
Full-text available
In the development of vascular tissue engineering, particularly in the case of small diameter vessels, one of the key obstacles is the blockage of these veins once they enter the in vivo environment. One of the contributing factors to this problem is the aberrant proliferation and migration of vascular smooth muscle cells (VSMCs) from the media layer of the artery to the interior of the channel. Two distinct phenotypes have been identified for smooth muscle cells, namely synthetic and contractile. Since the synthetic phenotype plays an essential role in the unusual growth and migration, the aim of this study was to convert the synthetic phenotype into the con-tractile one, which is a solution to prevent the abnormal growth of VSMCs. To achieve this goal, these cells were subjected to electrical signals, using a 1000 μA sinusoidal stimulation at 10 Hz for four days, with 20 min duration per 24 h. The morphological transformations and changes in the expression of vimentin, nestin, and β-actin proteins were then studied using ICC and flow cytometry assays. Also, the expression of VSMC specific markers such as smooth muscle myosin heavy chain (SMMHC) and smooth muscle alpha-actin (α-SMA) were evaluated using RT-PCR test. In the final phase of this study, the sheep decellularized vessel was employed as a scaffold for seeding these cells. Based on the results, electrical stimulation resulted in some morphological alterations in VSMCs. Furthermore, the observed reductions in the expression levels of vimentin, nestin and β-actin proteins and increase in the expression of SMMHC and α-SMA markers showed that it is possible to convert the synthetic phenotype to the contractile one using the studied regime of electrical stimulation. Finally, it can be concluded that electrical stimulation can significantly affect the phenotype of VSMCs, as demonstrated in this study.
Article
Full-text available
Vascular injury is central to the pathogenesis and progression of cardiovascular diseases, however, fostering alternative strategies to alleviate vascular injury remains a persisting challenge. Given the central role of cell‐derived nitric oxide (NO) in modulating the endogenous repair of vascular injury, NO‐generating proteolipid nanovesicles (PLV‐NO) are designed that recapitulate the cell‐mimicking functions for vascular repair and replacement. Specifically, the proteolipid nanovesicles (PLV) are versatilely fabricated using membrane proteins derived from different types of cells, followed by the incorporation of NO‐generating nanozymes capable of catalyzing endogenous donors to produce NO. Taking two vascular injury models, two types of PLV‐NO are tailored to meet the individual requirements of targeted diseases using platelet membrane proteins and endothelial membrane proteins, respectively. The platelet‐based PLV‐NO (pPLV‐NO) demonstrates its efficacy in targeted repair of a vascular endothelium injury model through systemic delivery. On the other hand, the endothelial cell (EC)‐based PLV‐NO (ePLV‐NO) exhibits suppression of thrombosis when modified onto a locally transplanted small‐diameter vascular graft (SDVG). The versatile design of PLV‐NO may enable a promising therapeutic option for various vascular injury‐evoked cardiovascular diseases.
Article
Full-text available
Apoptosis of endothelial cells prompts the release of apoptotic exosome-like vesicles (ApoExos), subtype extracellular vesicles secreted by apoptotic cells after caspase-3 activation. ApoExos are different from both apoptotic bodies and classical exosomes in their protein and nucleic acid contents and functions. In contrast to classical apoptotic bodies, ApoExos induce immunogenic responses that can be maladaptive when not tightly regulated. In the present study, we elucidated the mechanisms by which ApoExos are internalized by endothelial cells, which leads to shared specific and functional mRNAs of importance to endothelial function. Using flow cytometry and confocal microscopy, we revealed that ApoExos were actively internalized by endothelial cells. SiRNA-induced inhibition of classical endocytosis pathways with pharmacological inhibitors showed that ApoExos were internalized via phosphatidylserine-dependent macropinocytosis independently of classical endocytosis pathways. An electron microscopy analysis revealed that ApoExos increased the macropinocytosis rate in endothelial cells, setting in motion a positive feedback loop that increased the amount of internalized ApoExos. Deep sequencing of total RNA revealed that ApoExos possessed a unique protein-coding RNA profile, with PCSK5 being the most abundant mRNA. Internalization of ApoExos by cells led to the transfer of this RNA content from the ApoExos to cells. Specifically, PCSK5 mRNA was transferred to cells that had taken up ApoExos, and these cells subsequently expressed PCSK5. Collectively, our findings suggest that macropinocytosis is an effective entry pathway for the delivery of RNAs carried by ApoExos and that these RNAs are functionally expressed by the endothelial cells that internalize them. As ApoExos express a specific mRNA signature, these results suggest new avenues to understand how ApoExos produced at sites of vascular injury impact vascular function.
Article
Full-text available
Mesenchymal stromal cells (MSCs) have been extensively tested for the treatment of numerous clinical conditions and have demonstrated good safety but mixed efficacy. Although this outcome can be attributed in part to the heterogeneity of cell preparations, the lack of mechanistic understanding and tools to establish cell pharmacokinetics and pharmacodynamics, as well as the poorly defined criteria for patient stratification, have hampered the design of informative clinical trials. We and others have demonstrated that MSCs can rapidly undergo apoptosis after their infusion. Apoptotic MSCs are phagocytosed by monocytes/macrophages that are then reprogrammed to become anti-inflammatory cells. MSC apoptosis occurs when the cells are injected into patients who harbor activated cytotoxic T or NK cells. Therefore, the activation state of cytotoxic T or NK cells can be used as a biomarker to predict clinical responses to MSC treatment. Building on a large body of preexisting data, an alternative view on the mechanism of MSCs is that an inflammation-dependent MSC secretome is largely responsible for their immunomodulatory activity. We will discuss how these different mechanisms can coexist and are instructed by two different types of MSC “licensing”: one that is cell-contact dependent and the second that is mediated by inflammatory cytokines. The varied and complex mechanisms by which MSCs can orchestrate inflammatory responses and how this function is specifically driven by inflammation support a physiological role for tissue stroma in tissue homeostasis, and it acts as a sensor of damage and initiator of tissue repair by reprogramming the inflammatory environment.
Article
Full-text available
Background: Nitric oxide is a chemical agent produced by endothelial cells in a healthy blood vessel, inhibiting the overgrowth of vascular smooth muscle cells and regulating vessel tone. Liposomes are biocompatible and biodegradable drug carriers with a similar structure to cell bilayer phospholipid membrane, that can be used as useful nitric oxide carriers in vascular grafts. Method: Using a custom-designed apparatus, the sheep carotid arteries were decellularized while still maintaining important components of the vascular extracellular matrix (ECM), allowing them to be used as small-diameter vascular grafts. A Chemical signal of sodium nitrite was applied to control smooth muscle cells' behavior under static and dynamic cell culture conditions. The thin-film hydration approach was used to create nano-liposomes, which were then used as sodium nitrite carriers to control the drug release rate and enhance the amount of drug-loaded into the liposomes. Results: The ratio of 80:20:2 for DPPC: Cholesterol: PEG was determined as the optimum formulation of the liposome structure with high drug encapsulation efficiency (98%) and optimum drug release rate (the drug release rate was 40%, 65%, and 83% after 24, 48 and 72 hours, respectively). MTT assay results showed an improvement in endothelial cells proliferation in the presence of Nano-liposomal Sodium Nitrite (LNS) at the concentration of 0.5μg/ml. Using a suitable concentration of liposomal sodium nitrite (0.5 μg/ml) put onto the constructed scaffold resulted in the controllable development of smooth muscle cells in the experiment. The culture of smooth muscle cells in a pulsatile perfusion bioreactor indicated that in the presence of synthesized liposomal sodium nitrite, the overgrowth of smooth muscle cells was inhibited in dynamic cell culture conditions. The mechanical properties of ECM graft were measured, and a multi-scale model with an accuracy of 83% was proposed to predict mechanical properties successfully. Conclusion: The liposomal drug-loaded small-diameter vascular graft can prevent the overgrowth of SMCs and the formation of intimal hyperplasia in the graft. Aside from that, the effect of LNS on endothelial has the potential to stimulate endothelial cell proliferation and re-endothelialization.
Article
Full-text available
The modulation of inflammatory responses plays an important role in the pathobiology of cardiac failure. In a natural healing process, the ingestion of apoptotic cells and their apoptotic bodies by macrophages in a focal lesion result in resolution of inflammation and regeneration. However, therapeutic strategies to enhance this natural healing process using apoptotic cell‐derived biomaterials have not yet been established. In this study, apoptotic bodies‐mimetic nanovesicles derived from apoptotic fibroblasts (ApoNVs) conjugated with dextran and ischemic cardiac homing peptide (CHP) (ApoNV‐DCs) for ischemia‐reperfusion (IR)‐injured heart treatment are developed. Intravenously injected ApoNV‐DCs actively targeted the ischemic myocardium via conjugation with CHP, and are selectively phagocytosed by macrophages in an infarcted myocardium via conjugation with dextran. ApoNV‐DCs polarized macrophages from the M1 to M2 phenotype, resulting in the attenuation of inflammation. Four weeks after injection, ApoNV‐DCs attenuated cardiac remodeling, preserved blood vessels, and prevented cardiac function exacerbation in IR‐injured hearts. Taken together, the findings may open a new avenue for immunomodulation using targeted delivery of anti‐inflammatory nanovesicles that can be universally applied for various inflammatory diseases.
Article
Full-text available
Promotion of angiogenesis and restoration of the blood flow in the ischemic penumbra is an effective treatment for patients with ischemic stroke (IS). Radix astragali-safflower (AS), a classic herbal pair for accelerating blood circulation and dispersing blood stasis, has been used for thousands of years to treat patients with IS in China. Even so, the mechanism of the treatment of IS by AS is still undecipherable. In the current study, network pharmacology was firstly employed to unveil the mechanism of AS in treating IS, which showed that AS might promote angiogenesis associated with PTGS2 silence. Middle cerebral artery occlusion/reperfusion (MCAO/R) model rats were then used as the experimental animals to verify the prediction result. The experimental results revealed that treatment with AS improved the cerebral infarct volume, neurological damage, and cerebral histopathological damage; inhibited cell apoptosis; increased the contents of PDGF-BB, EPO, and TGF-β1; and reduced the levels of PF4, Ang-2, and TIMP-1 in serum. Immunohistochemical staining demonstrated that the expression of PTGS2 was dramatically increased in the hippocampus and cerebral cortex of rats with MCAO/R, and this trend was reversed by the treatment of AS. Immunofluorescent staining expressed that AS reversed the down-regulation of VEGF and further promoted the expression of CD31, which indicated that AS promoted angiogenesis in MCAO/R rats. The abnormal protein or mRNA expression of PTGS2, PGI2, bFGF, TSP-1, and VEGF in the penumbra were transposed by AS or Celecoxib (an inhibitor of PTGS2). In conclusion, the protective mechanism of AS for IS promoted angiogenesis and was involved with PTGS2 silence.
Article
Coronary artery revascularisation can be performed surgically or percutaneously. Surgery is associated with higher procedural risk and longer recovery than percutaneous interventions, but with long-term reduction of recurrent cardiac events. For many patients with obstructive coronary artery disease in need of revascularisation, surgical or percutaneous intervention is indicated on the basis of clinical and anatomical reasons or personal preferences. Medical therapy is a crucial accompaniment to coronary revascularisation, and data suggest that, in some subsets of patients, medical therapy alone might achieve similar results to coronary revascularisation. Most revascularisation data are based on prevalently White, non-elderly, male populations in high-income countries; robust data in women, older adults, and racial and other minorities, and from low-income and middle-income countries, are urgently needed.
Article
Apoptotic vesicles (ApoVs) hold great promise for inflammatory regulation and tissue repair. However, little effort has been dedicated to developing ApoV-based drug delivery platforms, while the insufficient targeting capability of ApoVs also limits their clinical applications. This work presents a platform architecture that integrates apoptosis induction, drug loading, and functionalized proteome regulation, followed by targeting modification, enabling the creation of an apoptotic vesicle delivery system to treat ischemic stroke. Briefly, α-mangostin (α-M) was utilized to induce mesenchymal stem cell (MSC) apoptosis while being loaded onto MSC-derived ApoVs as an anti-oxidant and anti-inflammatory agent for cerebral ischemia/reperfusion injury. Matrix metalloproteinase activatable cell-penetrating peptide (MAP), a microenvironment-responsive targeting peptide, was modified on the surface of ApoVs to obtain the MAP-functionalized α-M-loaded ApoVs. Such engineered ApoVs targeted the injured ischemic brain after systemic injection and achieved an enhanced neuroprotective activity due to the synergistic effect of ApoVs and α-M. The internal protein payloads of ApoVs, upon α-M activation, were found engaged in regulating immunological response, angiogenesis, and cell proliferation, all of which contributed to the therapeutic effects of ApoVs. The findings provide a universal framework for creating ApoV-based therapeutic drug delivery systems for the amelioration of inflammatory diseases and demonstrate the potential of MSC-derived ApoVs to treat neural injury.