Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
1.
ACS Biomater Sci Eng ; 10(5): 3242-3254, 2024 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-38632852

RESUMO

Osteoarthritis is characterized by enzymatic breakdown of the articular cartilage via the disruption of chondrocyte homeostasis, ultimately resulting in the destruction of the articular surface. Decades of research have highlighted the importance of inflammation in osteoarthritis progression, with inflammatory cytokines shifting resident chondrocytes into a pro-catabolic state. Inflammation can result in poor outcomes for cells implanted for cartilage regeneration. Therefore, a method to promote the growth of new cartilage and protect the implanted cells from the pro-inflammatory cytokines found in the joint space is required. In this study, we fabricate two gel types: polymer network hydrogels composed of chondroitin sulfate and hyaluronic acid, glycosaminoglycans (GAGs) known for their anti-inflammatory and prochondrogenic activity, and interpenetrating networks of GAGs and collagen I. Compared to a collagen-only hydrogel, which does not provide an anti-inflammatory stimulus, chondrocytes in GAG hydrogels result in reduced production of pro-inflammatory cytokines and enzymes as well as preservation of collagen II and aggrecan expression. Overall, GAG-based hydrogels have the potential to promote cartilage regeneration under pro-inflammatory conditions. Further, the data have implications for the use of GAGs to generally support tissue engineering in pro-inflammatory environments.


Assuntos
Condrócitos , Sulfatos de Condroitina , Ácido Hialurônico , Hidrogéis , Inflamação , Hidrogéis/química , Hidrogéis/farmacologia , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Ácido Hialurônico/química , Ácido Hialurônico/farmacologia , Sulfatos de Condroitina/farmacologia , Sulfatos de Condroitina/química , Inflamação/metabolismo , Inflamação/tratamento farmacológico , Inflamação/patologia , Animais , Cartilagem Articular/metabolismo , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/patologia , Citocinas/metabolismo , Agrecanas/metabolismo , Engenharia Tecidual/métodos , Osteoartrite/patologia , Osteoartrite/tratamento farmacológico , Osteoartrite/metabolismo
2.
Pharmaceutics ; 16(2)2024 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-38399234

RESUMO

The popularity of Glycosaminoglycans (GAGs) in drug delivery systems has grown as their innate ability to sequester and release charged molecules makes them adept in the controlled release of therapeutics. However, peptide therapeutics have been relegated to synthetic, polymeric systems, despite their high specificity and efficacy as therapeutics because they are rapidly degraded in vivo when not encapsulated. We present a GAG-based nanoparticle system for the easy encapsulation of cationic peptides, which offers control over particle diameter, peptide release behavior, and swelling behavior, as well as protection from proteolytic degradation, using a singular, organic polymer and no covalent linkages. These nanoparticles can encapsulate cargo with a particle diameter range spanning 130-220 nm and can be tuned to release cargo over a pH range of 4.5 to neutral through the modulation of the degree of sulfation and the molecular weight of the GAG. This particle system also confers better in vitro performance than the unencapsulated peptide via protection from enzymatic degradation. This method provides a facile way to protect therapeutic peptides via the inclusion of the presented binding sequence and can likely be expanded to larger, more diverse cargo as well, abrogating the complexity of previously demonstrated systems while offering broader tunability.

3.
Pharmaceutics ; 16(2)2024 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-38399285

RESUMO

Despite advances in breast cancer treatment, there remains a need for local management of noninvasive, low-grade ductal carcinoma in situ (DCIS). These focal lesions are well suited for local intraductal treatment. Intraductal administration supported target site drug retention, improved efficacy, and reduced systemic exposure. Here, we used a poly(N-isopropyl acrylamide, pNIPAM) nanoparticle delivery system loaded with cytotoxic piplartine and an MAPKAP Kinase 2 inhibitor (YARA) for this purpose. For tumor environment targeting, a collagen-binding peptide SILY (RRANAALKAGELYKSILYGSG-hydrazide) was attached to pNIPAM nanoparticles, and the nanoparticle diameter, zeta potential, drug loading, and release were assessed. The system was evaluated for cytotoxicity in a 2D cell culture and 3D spheroids. In vivo efficacy was evaluated using a chemical carcinogenesis model in female Sprague-Dawley rats. Nanoparticle delivery significantly reduced the IC50 of piplartine (4.9 times) compared to the drug in solution. The combination of piplartine and YARA in nanoparticles further reduced the piplartine IC50 (~15 times). Treatment with these nanoparticles decreased the in vivo tumor incidence (5.2 times). Notably, the concentration of piplartine in mammary glands treated with nanoparticles (35.3 ± 22.4 µg/mL) was substantially higher than in plasma (0.7 ± 0.05 µg/mL), demonstrating targeted drug retention. These results indicate that our nanocarrier system effectively reduced tumor development with low systemic exposure.

4.
Adv Healthc Mater ; : e2302500, 2023 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-38069833

RESUMO

Conductive biomaterials may capture native or exogenous bioelectric signaling, but incorporation of conductive moieties is limited by cytotoxicity, poor injectability, or insufficient stimulation. Microgel annealed scaffolds are promising as hydrogel-based materials due to their inherent void space that facilitates cell migration and proliferation better than nanoporous bulk hydrogels. Conductive microgels are generated from poly(ethylene) glycol (PEG and poly(3,4-ethylenedioxythiophene) polystyrene sulfonate (PEDOT: PSS) to explore the interplay of void volume and conductivity on myogenic differentiation. PEDOT: PSS increases microgel conductivity two-fold while maintaining stiffness, annealing strength, and viability of associated myoblastic cells. C2C12 myoblasts exhibit increases in the late-stage differentiation marker myosin heavy chain as a function of both porosity and conductivity. Myogenin, an earlier marker, is influenced only by porosity. Human skeletal muscle-derived cells exhibit increased Myod1, insulin like growth factor-1, and insulin-like growth factor binding protein 2 at earlier time points on conductive microgel scaffolds compared to non-conductive scaffolds. They also secrete more vascular endothelial growth factor at early time points and express factors that led to macrophage polarization patterns observe during muscle repair. These data indicate that conductivity aids myogenic differentiation of myogenic cell lines and primary cells, motivating the need for future translational studies to promote muscle repair.

5.
ACS Biomater Sci Eng ; 9(12): 6805-6820, 2023 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-37982792

RESUMO

In this study, we developed a novel hybrid collagen-binding nanocarrier for potential intraductal administration and local breast cancer treatment. The particles were formed by the encapsulation of nanostructured lipid carriers (NLCs) containing the cytotoxic drug paclitaxel within a shell of poly(N-isopropylacrylamide) (pNIPAM), and were functionalized with SILY, a peptide that binds to collagen type I (which is overexpressed in the mammary tumor microenvironment) to improve local retention and selectivity. The encapsulation of the NLCs in the pNIPAM shell increased nanoparticle size by approximately 140 nm, and after purification, a homogeneous system of hybrid nanoparticles (∼96%) was obtained. The nanoparticles exhibited high loading efficiency (<76%) and were capable of prolonging paclitaxel release for up to 120 h. SILY-modified nanoparticles showed the ability to bind to collagen-coated surfaces and naturally elaborated collagen. Hybrid nanoparticles presented cytotoxicity up to 3.7-fold higher than pNIPAM-only nanoparticles on mammary tumor cells cultured in monolayers. In spheroids, the increase in cytotoxicity was up to 1.8-fold. Compared to lipid nanoparticles, the hybrid nanoparticle modified with SILY increased the viability of nontumor breast cells by up to 1.59-fold in a coculture model, suggesting the effectiveness and safety of the system.


Assuntos
Antineoplásicos , Neoplasias da Mama , Nanopartículas , Humanos , Feminino , Paclitaxel/farmacologia , Neoplasias da Mama/tratamento farmacológico , Portadores de Fármacos/uso terapêutico , Antineoplásicos/uso terapêutico , Microambiente Tumoral
6.
J Control Release ; 362: 591-605, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37660990

RESUMO

Atopic dermatitis (AD) is a chronic inflammatory skin disorder that lacks ideal long-term treatment options due to a series of side effects, such as skin atrophy, related to the most common treatment prescribed to manage moderate-to-severe AD. In this study, a cell-penetrating MK2 inhibitor peptide YARA (YARAAARQARAKALNRQGLVAA) was loaded into hollow thermo-responsive pNIPAM nanoparticles (NP), which were further incorporated into chitosan hydrogels (H-NP-YARA) to promote local drug delivery, improve moisture and the anti-inflammatory activity. The NPs exhibited high loading efficiency (>50%) and the hydrogel remained porous following NP incorporation as observed by scanning electron microscopy (SEM). Both nanoparticles and hydrogels were able to improve the release of YARA and sustained release to up to 120 h. The hydrogels and NPs delivered 2 and 4-fold more YARA into viable skin layers of porcine skin in vitro at 12 h post-application than the non-encapsulated compound in intact and impaired barrier conditions. Furthermore, the YARA-loaded NPs (NP-YARA) and H-NP-YARA treatment decreased the levels of inflammatory cytokines up to 20 time-fold compared with the non-treated group of human keratinocytes under inflammatory conditions. Consistent with the results in cell culture, the loading of YARA in NP reduced the levels of IL-1ß, IL-6, and TNF-α up to 3.3 times in an ex vivo skin culture model after induction of inflammation. A further decrease of up to 17 times-fold was observed with H-NP-YARA treatment compared to the drug in solution. Our data collectively suggest that chitosan hydrogel containing YARA-loaded nanoparticles is a promising new formulation for the topical treatment of AD.


Assuntos
Quitosana , Dermatite Atópica , Nanopartículas , Animais , Suínos , Humanos , Dermatite Atópica/tratamento farmacológico , Quitosana/química , Hidrogéis/uso terapêutico , Peptídeos/uso terapêutico
7.
bioRxiv ; 2023 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-37577583

RESUMO

Bioelectricity is an understudied phenomenon to guide tissue homeostasis and regeneration. Conductive biomaterials may capture native or exogenous bioelectric signaling, but incorporation of conductive moieties is limited by cytotoxicity, poor injectability, or insufficient stimulation. Microgel annealed scaffolds are promising as hydrogel-based materials due to their inherent void space that facilitates cell migration and proliferation better than nanoporous bulk hydrogels. We generated conductive microgels from poly(ethylene) glycol and poly(3,4-ethylenedioxythiophene) polystyrene sulfonate (PEDOT:PSS) to explore the interplay of void volume and conductivity on myogenic differentiation. PEDOT:PSS increased microgel conductivity over 2-fold while maintaining stiffness, annealing strength, and viability of associated myoblastic cells. C2C12 myoblasts exhibited increases in the late-stage differentiation marker myosin heavy chain as a function of both porosity and conductivity. Myogenin, an earlier marker, was influenced only by porosity. Human skeletal muscle derived cells exhibited increased Myod1 , IGF-1, and IGFBP-2 at earlier timepoints on conductive microgel scaffolds compared to non-conductive scaffolds. They also secreted higher levels of VEGF at early timepoints and expressed factors that led to macrophage polarization patterns observed during muscle repair. These data indicate that conductivity aids myogenic differentiation of myogenic cell lines and primary cells, motivating the need for future translational studies to promote muscle repair.

8.
Bioact Mater ; 28: 467-479, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37408799

RESUMO

Implantable vascular devices are widely used in clinical treatments for various vascular diseases. However, current approved clinical implantable vascular devices generally have high failure rates primarily due to their surface lacking inherent functional endothelium. Here, inspired by the pathological mechanisms of vascular device failure and physiological functions of native endothelium, we developed a new generation of bioactive parylene (poly(p-xylylene))-based conformal coating to address these challenges of the vascular devices. This coating used a polyethylene glycol (PEG) linker to introduce an endothelial progenitor cell (EPC) specific binding ligand LXW7 (cGRGDdvc) onto the vascular devices for preventing platelet adhesion and selectively capturing endogenous EPCs. Also, we confirmed the long-term stability and function of this coating in human serum. Using two vascular disease-related large animal models, a porcine carotid artery interposition model and a porcine carotid artery-jugular vein arteriovenous graft model, we demonstrated that this coating enabled rapid generation of self-renewable "living" endothelium on the blood contacting surface of the expanded polytetrafluoroethylene (ePTFE) grafts after implantation. We expect this easy-to-apply conformal coating will present a promising avenue to engineer surface properties of "off-the-shelf" implantable vascular devices for long-lasting performance in the clinical settings.

9.
Lab Chip ; 23(13): 3050-3061, 2023 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-37278194

RESUMO

A feature of severe COVID-19 is the onset of an acute and intense systemic inflammatory response referred to as the "cytokine storm". The cytokine storm is characterized by high serum levels of inflammatory cytokines and the subsequent transport of inflammatory cells to damaging levels in vital organs (e.g., myocarditis). Immune trafficking and its effect on underlying tissues (e.g., myocardium) are challenging to observe at a high spatial and temporal resolution in mouse models. In this study, we created a vascularized organ-on-a-chip system to mimic cytokine storm-like conditions and tested the effectiveness of a novel multivalent selectin-targeting carbohydrate conjugate (composed of DS - dermatan sulfate and IkL - a selectin-binding peptide, termed DS-IkL) in blocking infiltration of polymorphonuclear leukocytes (PMN). Our data shows that cytokine storm-like conditions induce endothelial cells to produce additional inflammatory cytokines and facilitate infiltration of PMNs into tissue. Treatment of tissues with DS-IkL (60 µM) reduced PMN accumulation in the tissue by >50%. We then created cytokine storm-like conditions in a vascularized cardiac tissue-chip and found that PMN infiltration increases the spontaneous beating rate of the cardiac tissue, and this effect is eliminated by treatment with DS-IkL (60 µM). In summary, we demonstrate the utility of an organ-on-a-chip platform to mimic COVID-19 related cytokine storm and that blocking leukocyte infiltration with DS-IkL could be a viable strategy to mitigate associated cardiac complications.


Assuntos
COVID-19 , Neutrófilos , Camundongos , Animais , Cardiotoxicidade , Células Endoteliais , Sistemas Microfisiológicos , Citocinas
10.
Front Pharmacol ; 14: 1125209, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36937891

RESUMO

The lack of vascularization associated with deep burns delays the construction of wound beds, increases the risks of infection, and leads to the formation of hypertrophic scars or disfigurement. To address this challenge, we have fabricated a multi-functional pro-angiogenic molecule by grafting integrin αvß3 ligand LXW7 and collagen-binding peptide (SILY) to a dermatan sulfate (DS) glycosaminoglycan backbone, named LXW7-DS-SILY (LDS), and further employed this to functionalize collagen-based Integra scaffolds. Using a large deep burn wound model in C57/BLK6 mice (8-10 weeks old, 26-32g, n = 39), we demonstrated that LDS-modified collagen-based Integra scaffolds loaded with endothelial cells (ECs) accelerate wound healing rate, re-epithelialization, vascularization, and collagen deposition. Specifically, a 2 cm × 3 cm full-thickness skin burn wound was created 48 h after the burn, and then wounds were treated with four groups of different dressing scaffolds, including Integra + ECs, Integra + LDS, and Integra + LDS + ECs with Integra-only as the control. Digital photos were taken for wound healing measurement on post-treatment days 1, 7, 14, 21, 28, and 35. Post-treatment photos revealed that treatment with the Intgera + LDS + ECs scaffold exhibited a higher wound healing rate in the proliferation phase. Histology results showed significantly increased re-epithelialization, increased collagen deposition, increased thin and mixed collagen fiber content, increased angiogenesis, and shorter wound length within the Integra + LDS + ECs group at Day 35. On Day 14, the Integra + LDS + ECs group showed the same trend. The relative proportions of collagen changed from Day 14 to Day 35 in the Integra + LDS + ECs and Integra + ECs groups demonstrated decreased thick collagen fiber deposition and greater thin and mixed collagen fiber deposition. LDS-modified Integra scaffolds represent a promising novel treatment to accelerate deep burn wound healing, thereby potentially reducing the morbidity associated with open burn wounds. These scaffolds can also potentially reduce the need for autografting and morbidity in patients with already limited areas of harvestable skin.

11.
Colloids Surf B Biointerfaces ; 222: 113112, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36599186

RESUMO

Therapeutic peptides capable of reducing inflammation via inhibition of the MAP kinase 2 pathway have the potential to reduce inflammation in atopic dermatitis by suppressing secretion of inflammatory cytokines by resident keratinocytes. One of the biggest hurdles to the use of therapeutic peptides, however, is their rapid degradation by intrinsic proteases and peptidases found in serum. Here we introduce a new nanoparticle technology that enhances and extends the bioactivity of a MAP KAP kinase 2 inhibitor peptide (MK2i) via electrostatic complexation with Dermatan sulfate (DS), a glycosaminoglycan, and explore their properties under various conditions. DS-MK2i nanoparticles can be made using electrospray ionization or sonication and vortexing with no stabilizing polymers or crosslinking. Average particle diameter, polydispersity index, and zeta potential were measured over a pH range of 2.5-11.5, in increments of 0.5, in water and at physiological ionic strength. Both particle types were shown to be shelf stable, robust, and behave differently in response to pH. They are also significantly more effective at suppressing cytokine secretion in inflamed, human keratinocytes than peptide alone in the presence of serum, providing a facile method of protecting peptides for therapeutic delivery in conditions such as atopic dermatitis, and abrogating the need for serum-starvation in in vitro testing.


Assuntos
Dermatite Atópica , Nanopartículas , Humanos , Dermatite Atópica/tratamento farmacológico , Glicosaminoglicanos , Peptídeos/química , Nanopartículas/química , Inflamação
12.
J Biomed Mater Res A ; 111(5): 596-608, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36680496

RESUMO

Electrically conductive biomaterials direct cell behavior by capitalizing on the effect of bioelectricity in tissue homeostasis and healing. Many studies have leveraged conductive biomaterials to influence cells and improve tissue healing, even in the absence of external stimulation. However, most studies using electroactive materials neglect characterizing how the inclusion of conductive additives affects the material's mechanical properties, and the interplay between substrate electrical and mechanical properties on cell behavior is poorly understood. Furthermore, mechanisms dictating how electrically conductive materials affect cell behavior in the absence of external stimulation are not explicit. In this study, we developed a mechanically and electrically tunable conductive hydrogel using agarose and the conductive polymer PEDOT:PSS. Under certain conditions, we observed that the hydrogel physical and electrical properties were decoupled. We then seeded human mesenchymal stromal cells (MSCs) onto the hydrogels and observed enhanced adhesion and spreading of MSCs on conductive substrates, regardless of the hydrogel mechanical properties, and despite the gels having no cell-binding sites. To explain this observation, we measured protein interaction with the gels and found that charged proteins adsorbed significantly more to conductive hydrogels. These data demonstrate that conductivity promotes cell adhesion, likely by facilitating increased adsorption of proteins associated with cell binding, providing a better understanding of the mechanism of action of electrically conductive materials.


Assuntos
Hidrogéis , Células-Tronco Mesenquimais , Humanos , Sefarose , Hidrogéis/química , Adsorção , Materiais Biocompatíveis/química , Condutividade Elétrica
13.
Bioact Mater ; 20: 179-193, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35663336

RESUMO

Significant progress has been made in designing bone materials capable of directing endogenous cells to promote vascularized bone regeneration. However, current strategies lack regulation of the specific endogenous cell populations for vascularized bone regeneration, thus leading to adverse tissue formation and decreased regenerative efficiency. Here, we engineered a biomaterial to regulate endogenous cell adhesion and promote vascularized bone regeneration. The biomaterial works by presenting two synthetic ligands, LLP2A and LXW7, explicitly targeting integrins α4ß1 and αvß3, respectively, expressed on the surfaces of the cells related to bone formation and vascularization, such as mesenchymal stem cells (MSCs), osteoblasts, endothelial progenitor cells (EPCs), and endothelial cells (ECs). In vitro, the LLP2A/LXW7 modified biomaterial improved the adhesion of MSCs, osteoblasts, EPCs, and ECs via integrin α4ß1 and αvß3, respectively. In an adult rat calvarial bone defect model, the LLP2A/LXW7 modified biomaterial enhanced bone formation and vascularization by synergistically regulating endogenous cells with osteogenic and angiogenic potentials, such as DLX5+ cells, osteocalcin+ cells, CD34+/CD45- cells and CD31+ cells. In a fetal sheep spinal bone defect model, the LLP2A/LXW7 modified biomaterial augmented bone formation and vascularization without any adverse effects. This innovative biomaterial offers an off-the-shelf, easy-to-use, and biologically safe product suitable for vascularized bone regeneration in both fetal and adult disease environments.

14.
Proteoglycan Res ; 1(2)2023 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38884098

RESUMO

Many endothelial complications, whether from surgical or pathological origins, can result in the denudation of the endothelial layer and the exposure of collagen. Exposure of collagen results in the activation of platelets, leading to thrombotic and inflammatory cascades that ultimately result in vessel stenosis. We have previously reported the use of peptide-GAG compounds to target exposed collagen following endothelial injury. In this paper we optimize the spacer sequence of our collagen binding peptide to increase its conjugation to GAG backbones and increase the peptide-GAG collagen binding affinity by increasing peptide C-terminal cationic charge. Furthermore, we demonstrate the use of these molecules to inhibit platelet activation through collagen blocking, as well as their localization to exposed vascular collagen following systemic delivery. Altogether, optimization of peptide sequence and linkage chemistry can allow for increased conjugation and function, having implications for glycoconjugate use in other clinical applications.

15.
Theranostics ; 12(13): 6021-6037, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35966577

RESUMO

Although stem cell-derived extracellular vesicles (EVs) have remarkable therapeutic potential for various diseases, the therapeutic efficacy of EVs is limited due to their degradation and rapid diffusion after administration, hindering their translational applications. Here, we developed a new generation of collagen-binding EVs, by chemically conjugating a collagen-binding peptide SILY to EVs (SILY-EVs), which were designed to bind to collagen in the extracellular matrix (ECM) and form an EV-ECM complex to improve EVs' in situ retention and therapeutic efficacy after transplantation. Methods: SILY was conjugated to the surface of mesenchymal stem/stromal cell (MSC)-derived EVs by using click chemistry to construct SILY-EVs. Nanoparticle tracking analysis (NTA), ExoView analysis, cryogenic electron microscopy (cryo-EM) and western-blot analysis were used to characterize the SILY-EVs. Fluorescence imaging (FLI), MTS assay, ELISA and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) were used to evaluate the collagen binding and biological functions of SILY-EVs in vitro. In a mouse hind limb ischemia model, the in vivo imaging system (IVIS), laser doppler perfusion imaging (LDPI), micro-CT, FLI and RT-qPCR were used to determine the SILY-EV retention, inflammatory response, blood perfusion, gene expression, and tissue regeneration. Results:In vitro, the SILY conjugation significantly enhanced EV adhesion to the collagen surface and did not alter the EVs' biological functions. In the mouse hind limb ischemia model, SILY-EVs presented longer in situ retention, suppressed inflammatory responses, and significantly augmented muscle regeneration and vascularization, compared to the unmodified EVs. Conclusion: With the broad distribution of collagen in various tissues and organs, SILY-EVs hold promise to improve the therapeutic efficacy of EV-mediated treatment in a wide range of diseases and disorders. Moreover, SILY-EVs possess the potential to functionalize collagen-based biomaterials and deliver therapeutic agents for regenerative medicine applications.


Assuntos
Vesículas Extracelulares , Células-Tronco Mesenquimais , Animais , Modelos Animais de Doenças , Vesículas Extracelulares/metabolismo , Isquemia/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Células-Tronco , Cicatrização
16.
Am J Physiol Cell Physiol ; 322(4): C754-C761, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35235426

RESUMO

Proteoglycans play a crucial role in proper tissue morphology and function throughout the body that is defined by a combination of their core protein and the attached glycosaminoglycan chains. Although they serve a myriad of roles, the functions of extracellular proteoglycans can be generally sorted into four categories: modulation of tissue mechanical properties, regulation and protection of the extracellular matrix, sequestering of proteins, and regulation of cell signaling. The loss of proteoglycans can result in significant tissue dysfunction, ranging from poor mechanical properties to uncontrolled inflammation. Because of the key roles they play in proper tissue function and due to their complex synthesis, the past two decades have seen significant research into the development of proteoglycan mimetic molecules to recapitulate the function of proteoglycans for therapeutic and tissue engineering applications. These strategies have ranged from semisynthetic graft copolymers to recombinant proteoglycan domains synthesized by genetically engineered cells. In this review, we highlight some of the important functions of extracellular proteoglycans, as well as the strategies developed to recapitulate these functions.


Assuntos
Proteoglicanas , Engenharia Tecidual , Matriz Extracelular/metabolismo , Glicosaminoglicanos/metabolismo , Proteoglicanas/metabolismo
17.
ACS Biomater Sci Eng ; 8(3): 1247-1257, 2022 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-35133126

RESUMO

Adding chondroitin sulfate (CS) to collagen scaffolds has been shown to improve the outcomes for articular cartilage tissue engineering. Instead of physical entrapment or chemical crosslinking of CS within a scaffold, this study investigated the use of CS with attached collagen-binding peptides (termed CS-SILY). This method better recapitulates the aspects of native cartilage while retaining CS within a collagen type I and II blend (Col I/II) hydrogel. CS retention, average fibril diameter, and mechanical properties were altered by varying the number of SILY peptides attached to the CS backbone. When mesenchymal stromal cells (MSCs) were encapsulated within the scaffolds, the addition of CS-SILY molecules resulted in higher sulfated glycosaminoglycan production, and these results suggest that CS-SILY promotes MSC differentiation into chondrocytes. Taken together, our study shows the promise of adding a CS-SILY molecule to a Col I/II hydrogel with encapsulated MSCs to promote cartilage repair.


Assuntos
Cartilagem Articular , Engenharia Tecidual , Cartilagem Articular/metabolismo , Células Cultivadas , Sulfatos de Condroitina/química , Sulfatos de Condroitina/metabolismo , Colágeno , Colágeno Tipo I , Hidrogéis/química , Engenharia Tecidual/métodos
18.
Cardiovasc Res ; 118(1): 267-281, 2022 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-33125066

RESUMO

AIMS: One of the hallmarks of myocardial infarction (MI) is excessive inflammation. During an inflammatory insult, damaged endothelial cells shed their glycocalyx, a carbohydrate-rich layer on the cell surface which provides a regulatory interface to immune cell adhesion. Selectin-mediated neutrophilia occurs as a result of endothelial injury and inflammation. We recently designed a novel selectin-targeting glycocalyx mimetic (termed DS-IkL) capable of binding inflamed endothelial cells. This study examines the capacity of DS-IkL to limit neutrophil binding and platelet activation on inflamed endothelial cells, as well as the cardioprotective effects of DS-IkL after acute myocardial infarction. METHODS AND RESULTS: In vitro, DS-IkL diminished neutrophil interactions with both recombinant selectin and inflamed endothelial cells, and limited platelet activation on inflamed endothelial cells. Our data demonstrated that DS-IkL localized to regions of vascular inflammation in vivo after 45 min of left anterior descending coronary artery ligation-induced MI. Further, findings from this study show DS-IkL treatment had short- and long-term cardioprotective effects after ischaemia/reperfusion of the left anterior descending coronary artery. Mice treated with DS-IkL immediately after ischaemia/reperfusion and 24 h later exhibited reduced neutrophil extravasation, macrophage accumulation, fibroblast and endothelial cell proliferation, and fibrosis compared to saline controls. CONCLUSIONS: Our findings suggest that DS-IkL has great therapeutic potential after MI by limiting reperfusion injury induced by the immune response.


Assuntos
Anti-Inflamatórios/farmacologia , Selectina E/metabolismo , Células Endoteliais/efeitos dos fármacos , Infarto do Miocárdio/prevenção & controle , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Ativação de Neutrófilo/efeitos dos fármacos , Infiltração de Neutrófilos/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Animais , Adesão Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Feminino , Fibrose , Humanos , Masculino , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/imunologia , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Traumatismo por Reperfusão Miocárdica/imunologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/imunologia , Miocárdio/metabolismo , Miocárdio/patologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Ativação Plaquetária/efeitos dos fármacos , Transdução de Sinais
19.
Bioact Mater ; 10: 460-473, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34901560

RESUMO

Diabetic ischemic wound treatment remains a critical clinical challenge. Neovascularization plays a significant role in wound healing during all stages of the tissue repair process. Strategies that enhance angiogenesis and neovascularization and improve ischemic pathology may promote the healing of poor wounds, particularly diabetic wounds in highly ischemic conditions. We previously identified a cyclic peptide LXW7 that specifically binds to integrin αvß3 on endothelial progenitor cells (EPCs) and endothelial cells (ECs), activates vascular endothelial growth factor (VEGF) receptors, and promotes EC growth and maturation. In this study, we designed and synthesized a multi-functional pro-angiogenic molecule by grafting LXW7 and collagen-binding peptides (SILY) to a dermatan sulfate (DS) glycosaminoglycan backbone, named LXW7-DS-SILY, and further employed this multi-functional molecule to functionalize collagen-based extracellular matrix (ECM) scaffolds. We confirmed that LXW7-DS-SILY modification significantly promoted EPC attachment and growth on the ECM scaffolds in vitro and supported EPC survival in vivo in the ischemic environment. When applied in an established Zucker Diabetic Fatty (ZDF) rat ischemic skin flap model, LXW7-DS-SILY-functionalized ECM scaffolds loaded with EPCs significantly improved wound healing, enhanced neovascularization and modulated collagen fibrillogenesis in the ischemic environment. Altogether, this study provides a promising novel treatment to accelerate diabetic ischemic wound healing, thereby reducing limb amputation and mortality of diabetic patients.

20.
Mater Sci Eng R Rep ; 1462021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34483486

RESUMO

Hydrogels have been widely investigated in biomedical fields due to their similar physical and biochemical properties to the extracellular matrix (ECM). Collagen and hyaluronic acid (HA) are the main components of the ECM in many tissues. As a result, hydrogels prepared from collagen and HA hold inherent advantages in mimicking the structure and function of the native ECM. Numerous studies have focused on the development of collagen and HA hydrogels and their biomedical applications. In this extensive review, we provide a summary and analysis of the sources, features, and modifications of collagen and HA. Specifically, we highlight the fabrication, properties, and potential biomedical applications as well as promising commercialization of hydrogels based on these two natural polymers.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...