Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Adv Wound Care (New Rochelle) ; 11(6): 330-359, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34128387

RESUMO

Significance: Nonhealing wounds are an ever-growing global pandemic, with mortality rates and management costs exceeding many common cancers. Although our understanding of the molecular and cellular factors driving wound healing continues to grow, standards for diagnosing and evaluating wounds remain largely subjective and experiential, whereas therapeutic strategies fail to consistently achieve closure and clinicians are challenged to deliver individualized care protocols. There is a need to apply precision medicine practices to wound care by developing evidence-based approaches, which are predictive, prescriptive, and personalized. Recent Advances: Recent developments in "advanced" wound diagnostics, namely biomarkers (proteases, acute phase reactants, volatile emissions, and more) and imaging systems (ultrasound, autofluorescence, spectral imaging, and optical coherence tomography), have begun to revolutionize our understanding of the molecular wound landscape and usher in a modern age of therapeutic strategies. Herein, biomarkers and imaging systems with the greatest evidence to support their potential clinical utility are reviewed. Critical Issues: Although many potential biomarkers have been identified and several imaging systems have been or are being developed, more high-quality randomized controlled trials are necessary to elucidate the currently questionable role that these tools are playing in altering healing dynamics or predicting wound closure within the clinical setting. Future Directions: The literature supports the need for the development of effective point-of-care wound assessment tools, such as a platform diagnostic array that is capable of measuring multiple biomarkers at once. These, along with advances in telemedicine, synthetic biology, and "smart" wearables, will pave the way for the transformation of wound care into a precision medicine. Clinical Trial Registration number: NCT03148977.


Assuntos
Medicina de Precisão , Cicatrização , Diagnóstico por Imagem/métodos , Ensaios Clínicos Controlados Aleatórios como Assunto
2.
PLoS One ; 11(7): e0159598, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27459729

RESUMO

Debridement, the removal of diseased, nonviable tissue, is critical for clinicians to readily assess wound status and prepare the wound bed for advanced therapeutics or downstream active healing. Removing necrotic slough and eschar through surgical or mechanical methods is less specific and may be painful for patients. Enzymatic debridement agents, such as Clostridial collagenase, selectively and painlessly degrade devitalized tissue. In addition to its debriding activities, highly-purified Clostridial collagenase actively promotes healing, and our past studies reveal that extracellular matrices digested with this enzyme yield peptides that activate cellular migratory, proliferative and angiogenic responses to injury in vitro, and promote wound closure in vivo. Intriguingly, while collagenase Santyl® ointment, a sterile preparation containing Clostridial collagenases and other non-specific proteases, is a well-accepted enzymatic debridement agent, its role as an active healing entity has never been established. Based on our previous studies of pure Clostridial collagenase, we now ask whether the mixture of enzymes contained within Santyl® produces matrix-derived peptides that promote cellular injury responses in vitro and stimulate wound closure in vivo. Here, we identify novel collagen fragments, along with collagen-associated peptides derived from thrombospondin-1, multimerin-1, fibronectin, TGFß-induced protein ig-h3 and tenascin-C, generated from Santyl® collagenase-digested human dermal capillary endothelial and fibroblastic matrices, which increase cell proliferation and angiogenic remodeling in vitro by 50-100% over controls. Using an established model of impaired healing, we further demonstrate a specific dose of collagenase from Santyl® ointment, as well as the newly-identified and chemically-synthesized ECM-derived peptides significantly increase wound re-epithelialization by 60-100% over saline-treated controls. These results not only confirm and extend our earlier studies using purified collagenase- and matrix-derived peptides to stimulate healing in vitro and in vivo, but these Santyl®-generated, matrix-derived peptides may also represent exciting new opportunities for creating advanced wound healing therapies that are enabled by enzymatic debridement and potentially go beyond debridement.


Assuntos
Colagenases/metabolismo , Matriz Extracelular/metabolismo , Peptídeos/metabolismo , Sequência de Aminoácidos , Animais , Desbridamento , Fibroblastos/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Camundongos , Colagenase Microbiana/metabolismo , Modelos Animais , Peptídeos/química , Proteólise , Regeneração , Cicatrização
3.
J Transl Med ; 14(1): 197, 2016 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-27369317

RESUMO

BACKGROUND: Non-healing wounds are a major global health concern and account for the majority of non-traumatic limb amputations worldwide. However, compared to standard care practices, few advanced therapeutics effectively resolve these injuries stemming from cardiovascular disease, aging, and diabetes-related vasculopathies. While matrix turnover is disrupted in these injuries, debriding enzymes may promote healing by releasing matrix fragments that induce cell migration, proliferation, and morphogenesis, and plasma products may also stimulate these processes. Thus, we created matrix- and plasma-derived peptides, Comb1 and UN3, which induce cellular injury responses in vitro, and accelerate healing in rodent models of non-healing wounds. However, the effects of these peptides in non-healing wounds in diabetes are not known. Here, we interrogated whether these peptides stimulate healing in a diabetic porcine model highly reminiscent of human healing impairments in type 1 and type 2-diabetes. METHODS: After 3-6 weeks of streptozotocin-induced diabetes, full-thickness wounds were surgically created on the backs of adult female Yorkshire swine under general anesthesia. Comb1 and UN3 peptides or sterile saline (negative control) were administered to wounds daily for 3-7 days. Following sacrifice, wound tissues were harvested, and quantitative histological and immunohistochemical analyses were performed for wound closure, angiogenesis and granulation tissue deposition, along with quantitative molecular analyses of factors critical for angiogenesis, epithelialization, and dermal matrix remodeling. RESULTS: Comb1 and UN3 significantly increase re-epithelialization and angiogenesis in diabetic porcine wounds, compared to saline-treated controls. Additionally, fluorescein-conjugated Comb1 labels keratinocytes, fibroblasts, and vascular endothelial cells in porcine wounds, and Far western blotting reveals these cell populations express multiple fluorescein-Comb1-interacting proteins in vitro. Further, peptide treatment increases mRNA expression of several pro-angiogenic, epithelializing, and matrix-remodeling factors, importantly including balanced inductions in matrix metalloproteinase-2, -9, and tissue inhibitor of metalloproteinases-1, lending further insight into their mechanisms. CONCLUSIONS: Comb1 and UN3 stimulate wound resolution in diabetic Yorkshire swine through upregulation of multiple reparative growth factors and cytokines, especially matrix metalloproteinases and inhibitors that may aid in reversing the proteolytic imbalance characteristic of chronically inflamed non-healing wounds. Together, these peptides should have great therapeutic potential for all patients in need of healing, regardless of injury etiology.


Assuntos
Diabetes Mellitus Experimental/patologia , Matriz Extracelular/química , Especificidade de Órgãos/efeitos dos fármacos , Peptídeos/sangue , Peptídeos/farmacologia , Cicatrização/efeitos dos fármacos , Adulto , Indutores da Angiogênese/metabolismo , Animais , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Quimiotaxia/efeitos dos fármacos , Fator de Crescimento Epidérmico/metabolismo , Feminino , Fluoresceína-5-Isotiocianato/metabolismo , Humanos , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Sus scrofa
4.
Methods Mol Biol ; 1430: 221-9, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27172957

RESUMO

Microvascular endothelial cell-mural cell interactions are instrumental in modulating both physiological and pathologic angiogenesis. Pericyte-endothelial cell communication through direct physical associations and secreted effectors comprises a bidirectional signal array that regulates vascular maturation and integrity. As endothelial cell proliferation, migration, and morphogenesis are key elements of vascular growth and remodeling during angiogenesis, we have developed novel preclinical systems for studying the roles of endothelial-mural cell dynamics on cell cycle entry and angiogenic activity in vitro. These coculture models not only enable evaluation of endothelial cell-pericyte "cross talk" but also allow for the quantitative analysis of both heterotypic contact-dependent and contact-independent cell cycle progression in either cell population, as well as angiogenic sprouting in three-dimensional vascular networks. Cells actively proliferating in two-dimensional assays can be labeled via incorporation of 5-ethynyl-2'-deoxyuridine (EdU) into their DNA. Additionally, each cell population can be vitally labeled with a variety of cell-specific and/or membrane-permeant lipophilic dyes prior to coculture, such as DiO, or through immunofluorescence of mural or endothelial cell-specific markers after cellular fixation and/or permeabilization. Ultimately, this experimental approach can be used to investigate cellular contact-dependent and soluble mechanisms mediating mural-endothelial cell interactions, which may be instrumental in microvascular development and remodeling in vivo.


Assuntos
Células Endoteliais/citologia , Endotélio Vascular/citologia , Neovascularização Fisiológica , Pericitos/citologia , Comunicação Celular , Ciclo Celular , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Desoxiuridina/análogos & derivados , Desoxiuridina/metabolismo , Humanos , Neovascularização Patológica
5.
Stem Cells Transl Med ; 4(5): 459-67, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25769654

RESUMO

Diabetic retinopathy is characterized by progressive vascular dropout with subsequent vision loss. We have recently shown that an intravitreal injection of adipose-derived stem cells (ASCs) can stabilize the retinal microvasculature, enabling repair and regeneration of damaged capillary beds in vivo. Because an understanding of ASC status from healthy versus diseased donors will be important as autologous cellular therapies are developed for unmet clinical needs, we took advantage of the hyperglycemic Akimba mouse as a preclinical in vivo model of diabetic retinopathy in an effort aimed at evaluating therapeutic efficacy of adipose-derived stem cells (mASCs) derived either from healthy, nondiabetic or from diabetic mice. To these ends, Akimba mice received intravitreal injections of media conditioned by mASCs or mASCs themselves, subsequent to development of substantial retinal capillary dropout. mASCs from healthy mice were more effective than diabetic mASCs in protecting the diabetic retina from further vascular dropout. Engrafted ASCs were found to preferentially associate with the retinal vasculature. Conditioned medium was unable to recapitulate the vasoprotection seen with injected ASCs. In vitro diabetic ASCs showed decreased proliferation and increased apoptosis compared with healthy mASCs. Diabetic ASCs also secreted less vasoprotective factors than healthy mASCs, as determined by high-throughput enzyme-linked immunosorbent assay. Our findings suggest that diabetic ASCs are functionally impaired compared with healthy ASCs and support the utility of an allogeneic injection of ASCs versus autologous or conditioned media approaches in the treatment of diabetic retinopathy.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Diabetes Mellitus Experimental/terapia , Retinopatia Diabética/terapia , Transplante de Células-Tronco , Adipócitos/citologia , Animais , Meios de Cultivo Condicionados , Diabetes Mellitus Experimental/patologia , Retinopatia Diabética/patologia , Modelos Animais de Doenças , Camundongos , Células-Tronco/citologia
6.
Transl Res ; 163(4): 296-306, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24530608

RESUMO

Pericytes are mural cells of the microcirculation that have been shown to play key roles in regulating microvascular morphogenesis and stability throughout each tissue bed and organ system assessed. Of note, recent work has revealed that pericytes share several characteristics with mesenchymal- and adipose-derived stem cells, suggesting there may be lineage-related connections among bona fide pericytes and these vascular "progenitors," which can assume a perivascular position in association with endothelial cells. Hence, pericyte identity as a mediator of vascular remodeling may be confounded by its close relationships with its progenitors or pluripotent cell counterparts and yet demonstrates their potential utility as cell-based therapies for unmet clinical needs. Crucial to the development of such therapies is a comprehensive understanding of the origin and fate regulating these related cell types as well as the unveiling of the molecular mechanisms by which pericytes and endothelial cells communicate. Such mechanistic inputs, which disrupt normal cellular crosstalk during disease inception and progression, offer opportunities for intervention and are discussed in the context of the vasculopathies accompanying tumor growth, diabetes, and fibrosis.


Assuntos
Células Endoteliais/fisiologia , Pericitos/fisiologia , Transplante de Células-Tronco/tendências , Animais , Células Endoteliais/citologia , Humanos , Pericitos/citologia , Medicina Regenerativa/tendências
7.
PLoS One ; 8(5): e65691, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23741506

RESUMO

BACKGROUND: Retinal vasculopathies, including diabetic retinopathy (DR), threaten the vision of over 100 million people. Retinal pericytes are critical for microvascular control, supporting retinal endothelial cells via direct contact and paracrine mechanisms. With pericyte death or loss, endothelial dysfunction ensues, resulting in hypoxic insult, pathologic angiogenesis, and ultimately blindness. Adipose-derived stem cells (ASCs) differentiate into pericytes, suggesting they may be useful as a protective and regenerative cellular therapy for retinal vascular disease. In this study, we examine the ability of ASCs to differentiate into pericytes that can stabilize retinal vessels in multiple pre-clinical models of retinal vasculopathy. METHODOLOGY/PRINCIPAL FINDINGS: We found that ASCs express pericyte-specific markers in vitro. When injected intravitreally into the murine eye subjected to oxygen-induced retinopathy (OIR), ASCs were capable of migrating to and integrating with the retinal vasculature. Integrated ASCs maintained marker expression and pericyte-like morphology in vivo for at least 2 months. ASCs injected after OIR vessel destabilization and ablation enhanced vessel regrowth (16% reduction in avascular area). ASCs injected intravitreally before OIR vessel destabilization prevented retinal capillary dropout (53% reduction). Treatment of ASCs with transforming growth factor beta (TGF-ß1) enhanced hASC pericyte function, in a manner similar to native retinal pericytes, with increased marker expression of smooth muscle actin, cellular contractility, endothelial stabilization, and microvascular protection in OIR. Finally, injected ASCs prevented capillary loss in the diabetic retinopathic Akimba mouse (79% reduction 2 months after injection). CONCLUSIONS/SIGNIFICANCE: ASC-derived pericytes can integrate with retinal vasculature, adopting both pericyte morphology and marker expression, and provide functional vascular protection in multiple murine models of retinal vasculopathy. The pericyte phenotype demonstrated by ASCs is enhanced with TGF-ß1 treatment, as seen with native retinal pericytes. ASCs may represent an innovative cellular therapy for protection against and repair of DR and other retinal vascular diseases.


Assuntos
Adipócitos/metabolismo , Neovascularização Patológica/metabolismo , Pericitos/metabolismo , Retina/metabolismo , Retina/patologia , Células-Tronco/metabolismo , Adipócitos/citologia , Animais , Retinopatia Diabética/metabolismo , Retinopatia Diabética/patologia , Humanos , Camundongos , Oxigênio/efeitos adversos , Pericitos/citologia , Vasos Retinianos/metabolismo , Vasos Retinianos/patologia , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Fator de Crescimento Transformador beta1/farmacologia
9.
PLoS One ; 7(2): e32146, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22384158

RESUMO

Previous work in our laboratory has described several pro-angiogenic short peptides derived from endothelial extracellular matrices degraded by bacterial collagenase. Here we tested whether these peptides could stimulate wound healing in vivo. Our experiments demonstrated that a peptide created as combination of fragments of tenascin X and fibrillin 1 (comb1) applied into cranial dermal wounds created in mice treated with cyclophosphamide to impair wound healing, can improve the rate of wound closure. Furthermore, we identify and characterize a novel peptide (UN3) created and modified from two naturally-occurring peptides, which are present in human platelet-rich plasma. In vitro testing of UN3 demonstrates that it causes a 50% increase in endothelial proliferation, 250% increase in angiogenic response and a tripling of epithelial cell migration in response to injury. Results of in vivo experiments where comb1 and UN3 peptides were added together to cranial wounds in cyclophosphamide-treated mice leads to improvement of wound vascularization as shown by an increase of the number of blood vessels present in the wound beds. Application of the peptides markedly promotes cellular responses to injury and essentially restores wound healing dynamics to those of normal, acute wounds in the absence of cyclophosphamide impairment. Our current work is aimed at understanding the mechanisms underlying the stimulatory effects of these peptides as well as identification of the cellular receptors mediating these effects.


Assuntos
Matriz Extracelular/metabolismo , Plasma Rico em Plaquetas/metabolismo , Pele/patologia , Cicatrização , Animais , Plaquetas/citologia , Bovinos , Movimento Celular , Proliferação de Células , Células Cultivadas , Cromatografia em Gel , Cromatografia por Troca Iônica/métodos , Ciclofosfamida/farmacologia , Células Endoteliais/citologia , Fibrilina-1 , Fibrilinas , Humanos , Queratinócitos/citologia , Proteínas dos Microfilamentos/metabolismo , Neovascularização Patológica , Peptídeos/química , Inibidores da Síntese de Proteínas/farmacologia , Tenascina/metabolismo
10.
J Biol Chem ; 287(16): 13182-93, 2012 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-22367208

RESUMO

Calpains are calcium-regulated cysteine proteases that have been implicated in the regulation of cell death pathways. Here, we used our calpain-1 null mouse model to evaluate the function of calpain-1 in neural degeneration following a rodent model of traumatic brain injury. In vivo, calpain-1 null mice show significantly less neural degeneration and apoptosis and a smaller contusion 3 days post-injury than wild type littermates. Protection from traumatic brain injury corroborated with the resistance of calpain-1 neurons to apoptosis induced by oxidative stress. Biochemical analysis revealed that caspase-3 activation, extracellular calcium entry, mitochondrial membrane permeability, and release of apoptosis-inducing factor from mitochondria are partially blocked in the calpain-1 null neurons. These findings suggest that the calpain-1 knock-out mice may serve as a useful model system for neuronal protection and apoptosis in traumatic brain injury and other neurodegenerative disorders in which oxidative stress plays a role.


Assuntos
Apoptose/fisiologia , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Calpaína/genética , Calpaína/metabolismo , Estresse Oxidativo/fisiologia , Animais , Fator de Indução de Apoptose/metabolismo , Cálcio/metabolismo , Caspase 3/metabolismo , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Membranas Mitocondriais/metabolismo , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Neurônios/metabolismo , Neurônios/patologia , RNA Interferente Pequeno/genética
11.
Arterioscler Thromb Vasc Biol ; 31(10): 2181-92, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21757656

RESUMO

OBJECTIVE: The aim of our study is to determine the cellular and molecular origin for the pericytes in infantile hemangioma (IH) and their functional role in the formation of pathological blood vessels. METHODS AND RESULTS: Here we show that IH-derived stem cells (HemSCs) form pericyte-like cells. With in vitro studies, we demonstrate that HemSC-to-pericyte differentiation depends on direct contact with endothelial cells. JAGGED1 expressed ectopically in fibroblasts was sufficient to induce HemSCs to acquire a pericyte-like phenotype, indicating a critical role for JAGGED1. In vivo, we blocked pericyte differentiation with recombinant JAGGED1, and we observed reduced formation of blood vessels, with an evident lack of pericytes. Silencing JAGGED1 in the endothelial cells reduced blood vessel formation and resulted in a paucity of pericytes. CONCLUSIONS: Our data show that endothelial JAGGED1 controls HemSC-to-pericyte differentiation in a murine model of IH and suggests that pericytes have a fundamental role in formation of blood vessels in IH.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Transdiferenciação Celular , Hemangioma/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas de Membrana/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Células-Tronco Neoplásicas/metabolismo , Neovascularização Patológica/metabolismo , Pericitos/metabolismo , Transdução de Sinais , Animais , Proteínas de Ligação ao Cálcio/genética , Comunicação Celular , Células Cultivadas , Técnicas de Cocultura , Células Endoteliais/metabolismo , Hemangioma/irrigação sanguínea , Hemangioma/genética , Hemangioma/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteína Jagged-1 , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Nus , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Miócitos de Músculo Liso/transplante , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/transplante , Neovascularização Patológica/genética , Pericitos/patologia , Pericitos/transplante , Interferência de RNA , Proteínas Recombinantes/metabolismo , Proteínas Serrate-Jagged , Fatores de Tempo , Transfecção
12.
Curr Diab Rep ; 11(4): 253-64, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21611764

RESUMO

Patients struggling with diabetes are at elevated risks for several sight-threatening diseases, including proliferative diabetic retinopathy (DR). DR manifests in two stages: first, the retinal microvasculature is compromised and capillary degeneration occurs; subsequently, an over-compensatory angiogenic response is initiated. Early changes in the retinal microcirculation include disruptions in blood flow, thickening of basement membrane, eventual loss of mural cells, and the genesis of acellular capillaries. Endothelial apoptosis and capillary dropout lead to a hypoxic inner retina, alterations in growth factors, and upregulation of inflammatory mediators. With disease progression, pathologic angiogenesis generates abnormal preretinal microvessels. Current therapies, which include panretinal photocoagulation and vitrectomy, have remained unaltered for several decades. With several exciting preclinical advances, emergent technologies and innovative cellular targets may offer newfound hope for developing "next-generation" interventional or preventive clinical approaches that will significantly advance current standards of care and clinical outcomes.


Assuntos
Retinopatia Diabética/patologia , Microvasos/patologia , Neovascularização Patológica/fisiopatologia , Inibidores da Angiogênese/uso terapêutico , Animais , Retinopatia Diabética/tratamento farmacológico , Retinopatia Diabética/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Humanos , Microvasos/metabolismo , Neovascularização Patológica/metabolismo , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/metabolismo
13.
Wound Repair Regen ; 19(2): 134-48, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21362080

RESUMO

Here, we define dynamic reciprocity (DR) as an ongoing, bidirectional interaction among cells and their surrounding microenvironment. In this review, we posit that DR is especially meaningful during wound healing as the DR-driven biochemical, biophysical, and cellular responses to injury play pivotal roles in regulating tissue regenerative responses. Such cell-extracellular matrix interactions not only guide and regulate cellular morphology, but also cellular differentiation, migration, proliferation, and survival during tissue development, including, e.g., embryogenesis, angiogenesis, as well as during pathologic processes including cancer, diabetes, hypertension, and chronic wound healing. Herein, we examine DR within the wound microenvironment while considering specific examples across acute and chronic wound healing. This review also considers how a number of hypotheses that attempt to explain chronic wound pathophysiology may be understood within the DR framework. The implications of applying the principles of DR to optimize wound care practice and future development of innovative wound healing therapeutics are also briefly considered.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Cicatrização/fisiologia , Animais , Biofilmes , Comunicação Celular , Movimento Celular , Proliferação de Células , Doença Crônica , Pé Diabético/fisiopatologia , Matriz Extracelular/fisiologia , Hemostasia/fisiologia , Humanos , Inflamação/fisiopatologia , Integrinas/fisiologia , Metaloproteinases da Matriz/fisiologia , Regeneração/fisiologia , Úlcera Varicosa/fisiopatologia , Ferimentos e Lesões/metabolismo
14.
Microvasc Res ; 80(3): 339-48, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20709086

RESUMO

Pericytes surround capillary endothelial cells and exert contractile forces modulating microvascular tone and endothelial growth. We previously described pericyte contractile phenotype to be Rho GTPase- and α-smooth muscle actin (αSMA)-dependent. However, mechanisms mediating adhesion-dependent shape changes and contractile force transduction remain largely equivocal. We now report that the neutral cysteine protease, calpain, modulates pericyte contractility and cellular stiffness via talin, an integrin-binding and F-actin associating protein. Digital imaging and quantitative analyses of living cells reveal significant perturbations in contractile force transduction detected via deformation of silicone substrata, as well as perturbations of mechanical stiffness in cellular contractile subdomains quantified via atomic force microscope (AFM)-enabled nanoindentation. Pericytes overexpressing GFP-tagged talin show significantly enhanced contractility (~two-fold), which is mitigated when either the calpain-cleavage resistant mutant talin L432G or vinculin are expressed. Moreover, the cell-penetrating, calpain-specific inhibitor termed CALPASTAT reverses talin-enhanced, but not Rho GTP-dependent, contractility. Interestingly, our analysis revealed that CALPASTAT, but not its inactive mutant, alters contractile cell-driven substrata deformations while increasing mechanical stiffness of subcellular contractile regions of these pericytes. Altogether, our results reveal that calpain-dependent cleavage of talin modulates cell contractile dynamics, which in pericytes may prove instrumental in controlling normal capillary function or microvascular pathophysiology.


Assuntos
Calpaína/metabolismo , Forma Celular , Mecanotransdução Celular , Microvasos/metabolismo , Pericitos/metabolismo , Vasos Retinianos/metabolismo , Talina/metabolismo , Animais , Calpaína/antagonistas & inibidores , Calpaína/genética , Bovinos , Células Cultivadas , Inibidores de Cisteína Proteinase/farmacologia , Relação Dose-Resposta a Droga , Elasticidade , Camundongos , Microscopia de Força Atômica , Microvasos/efeitos dos fármacos , Mutação , Pericitos/efeitos dos fármacos , Fenótipo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Vasos Retinianos/efeitos dos fármacos , Talina/genética , Fatores de Tempo , Transfecção , Vinculina/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
15.
J Oncol ; 2010: 132641, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20445741

RESUMO

Angiogenesis is a vital process resulting in the formation of new blood vessels. It is normally a highly regulated process that occurs during human development, reproduction, and wound repair. However, angiogenesis can also become a fundamental pathogenic process found in cancer and several other diseases. To date, the inhibition of angiogenesis has been researched at both the bench and the bedside. While several studies have found moderate improvements when treating with angiogenesis inhibitors, greater success is being seen when the inhibition of angiogenesis is combined with other traditional forms of available therapy. This review summarizes several important angiogenic factors, examines new research and ongoing clinical trials for such factors, and attempts to explain how this new knowledge may be applied in the fight against cancer and other angiogenic-related diseases.

16.
J Biol Chem ; 285(23): 18060-71, 2010 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-20360004

RESUMO

The Rac exchange factor Tiam1 is involved in diverse cell functions and signaling pathways through multiple protein interactions, raising the question of how signaling and functional specificity are achieved. We have shown that Tiam1 interactions with different scaffold proteins activate different Rac-dependent pathways by recruiting specific Rac effector proteins, and reasoned that there must be regulatory mechanisms governing each interaction. Fibroblasts express at least two Tiam1-interacting proteins, insulin receptor substrate protein 53 kDa (IRSp53) and spinophilin. We used fluorescent resonance energy transfer (FRET) to measure localized Rac activation associated with IRSp53 and spinophilin complexes in individual fibroblasts to test this hypothesis. Pervanadate or platelet-derived growth factor induced localized Rac activation dependent on Tiam1 and IRSp53. Forskolin or epinephrine induced localized Rac activation dependent on Tiam1 and spinophilin. In spinophilin-deficient cells, Tiam1 co-localized with IRSp53 in response to pervanadate or platelet-derived growth factor. In IRSp53-deficient cells, Tiam1 co-localized with spinophilin in response to forskolin or epinephrine. Total cellular levels of activated Rac were affected only in cells with exogenous Tiam1, and were primarily increased in the membrane fraction. Downstream effects of Rac activation were also stimulus and scaffold-specific. Cell ruffling, spreading, and cell adhesion were dependent on IRSp53, but not spinophilin. Epinephrine decreased IRSp53-dependent adhesion and increased cell migration in a Rac and spinophilin-dependent fashion. These results support the idea that Tiam1 interactions with different scaffold proteins couple distinct upstream signals to localized Rac activation and specific downstream pathways, and suggest that manipulating Tiam1-scaffold interactions can modulate Rac-dependent cellular behaviors.


Assuntos
Regulação da Expressão Gênica , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas dos Microfilamentos/química , Proteínas do Tecido Nervoso/química , Proteínas rac de Ligação ao GTP/metabolismo , Actinas/química , Animais , Adesão Celular , Movimento Celular , Epinefrina/farmacologia , Fibroblastos/metabolismo , Transferência Ressonante de Energia de Fluorescência/métodos , Humanos , Camundongos , Células NIH 3T3 , Proteína 1 Indutora de Invasão e Metástase de Linfoma de Células T
17.
Microvasc Res ; 77(3): 281-8, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19323981

RESUMO

Endothelial cell migration and proliferation, central steps in both physiologic and pathologic angiogenesis, require cytoskeletal-dependent remodeling, which is, in large part, achieved by the dynamic regulation of the beta-actin network. Specifically, the beta-actin network has previously been shown to be (i) enriched in regions of highly motile cytoplasm, and (ii) modulated by its isoactin-specific barbed-end capping protein, beta cap73. We hypothesize that regulated over-expression of beta cap73 could disrupt angiogenesis by capping beta-actin-filament assembly thus inhibiting the incipient cellular migration and microvascular morphogenesis that ensues. Indeed, upon infection of capillary endothelial cells (cEC) with an adenovirus encoding the full-length beta cap73 (Ad-beta cap73), there is a robust cellular rounding response that occurs concomitantly with cytoskeletal disruption, as visualized with immunofluorescence microscopy. Further, we demonstrate that over-expression of Ad-beta cap73 inhibits cEC migration in wound healing studies. Quantitative in vitro angiogenesis assays reveal that Ad-beta cap73 not only prevents endothelial cells from forming capillary-like networks, but also induces the collapse of preformed endothelial tubes. In testing whether Ad-beta cap73 impairs angiogenic events by inducing anoikis/apoptosis, we demonstrate that beta cap73 infection activates a caspase-3-mediated cell death response as observed by quantitative Western blotting and immunofluorescence analyses. Altogether, these findings suggest that endothelial-specific targeting and beta cap73 over-expression may represent an innovative therapeutic approach capable of abrogating pathologic angiogenesis.


Assuntos
Inibidores da Angiogênese/metabolismo , Proteína de Capeamento de Actina CapZ/metabolismo , Citoesqueleto/fisiologia , Endotélio Vascular/metabolismo , Neovascularização Patológica/metabolismo , Inibidores da Angiogênese/genética , Animais , Anoikis/fisiologia , Proteína de Capeamento de Actina CapZ/genética , Capilares/metabolismo , Capilares/patologia , Bovinos , Movimento Celular/fisiologia , Células Cultivadas , Citoesqueleto/patologia , Endotélio Vascular/patologia , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Neovascularização Patológica/patologia , Neovascularização Patológica/fisiopatologia , RNA Mensageiro , Transdução de Sinais , Cicatrização
18.
J Invest Dermatol ; 128(10): 2498-507, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18528437

RESUMO

The link between loss of cell-cell adhesion, the activation of cell migration, and the behavior of intraepithelial (IE) tumor cells during the early stages of skin cancer progression is not well understood. The current study characterized the migratory behavior of a squamous cell carcinoma cell line (HaCaT-II-4) upon E-cadherin suppression in both 2D, monolayer cultures and within human skin equivalents that mimic premalignant disease. The migratory behavior of tumor cells was first analyzed in 3D tissue context by developing a model that mimics transepithelial tumor cell migration. We show that loss of cell adhesion enabled migration of single, IE tumor cells between normal keratinocytes as a prerequisite for stromal invasion. To further understand this migratory behavior, E-cadherin-deficient cells were analyzed in 2D, monolayer cultures and displayed altered cytoarchitecture and enhanced membrane protrusive activity that was associated with circumferential actin organization and induction of the nonmuscle, beta actin isoform. These features were associated with increased motility and random, individual cell migration in response to scrape-wounding. Thus, loss of E-cadherin-mediated adhesion led to the acquisition of phenotypic properties that augmented cell motility and directed the transition from the precancer to cancer in skin-like tissues.


Assuntos
Caderinas/metabolismo , Carcinoma de Células Escamosas/fisiopatologia , Movimento Celular , Citoesqueleto/ultraestrutura , Neoplasias Cutâneas/fisiopatologia , Pele/fisiopatologia , Citoesqueleto de Actina/ultraestrutura , Actinas/metabolismo , Caderinas/deficiência , Carcinoma de Células Escamosas/patologia , Adesão Celular , Linhagem Celular , Membrana Celular/ultraestrutura , Progressão da Doença , Epitélio/fisiopatologia , Humanos , Pele/lesões , Pele/patologia , Neoplasias Cutâneas/patologia , Fatores de Tempo , Engenharia Tecidual
19.
Lasers Surg Med ; 39(9): 706-15, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17960752

RESUMO

BACKGROUND: Low levels of laser or non-coherent light, termed low-level light therapy (LLLT) have been reported to accelerate some phases of wound healing, but its clinical use remains controversial. METHODS: A full thickness dorsal excisional wound in mice was treated with a single exposure to light of various wavelengths and fluences 30 minutes after wounding. Wound areas were measured until complete healing and immunofluorescence staining of tissue samples was carried out. RESULTS: Wound healing was significantly stimulated in BALB/c and SKH1 hairless mice but not in C57BL/6 mice. Illuminated wounds started to contract while control wounds initially expanded for the first 24 hours. We found a biphasic dose-response curve for fluence of 635-nm light with a maximum positive effect at 2 J/cm(2). Eight hundred twenty nanometer was found to be the best wavelength tested compared to 635, 670, and 720 nm. We found no difference between non-coherent 635+/-15-nm light from a lamp and coherent 633-nm light from a He/Ne laser. LLLT increased the number of alpha-smooth muscle actin (SMA)-positive cells at the wound edge. CONCLUSION: LLLT stimulates wound contraction in susceptible mouse strains but the mechanism remains uncertain.


Assuntos
Terapia com Luz de Baixa Intensidade , Cicatrização/efeitos da radiação , Análise de Variância , Animais , Relação Dose-Resposta à Radiação , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos
20.
Microvasc Res ; 66(3): 190-6, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14609524

RESUMO

To define the role of transforming growth factor-beta 1 (TGF-beta 1) in modulating pericyte contractile phenotype, we have ablated the TGF-beta signaling pathway by infection with a retrovirus bearing a TGF-beta type II receptor with a truncated C-terminal intracellular kinase domain (DNT beta RII). While TGF-beta 1 blocks pericyte proliferation and induces the expression of vascular smooth muscle contractile proteins in wild-type pericytes, DNT beta RII-bearing pericytes are neither growth inhibited by TGF-beta 1 nor do they accumulate alpha-smooth muscle actin (alpha-SMA) mRNA or protein. TGF-beta 1 induces expression of the myogenic transcription factor myf-5 and the cyclin-dependent kinase inhibitor p27; we show that these signaling pathways are disrupted in the DNT beta RII-bearing pericytes. These observations demonstrate that the TGF-beta 1 signaling pathway controls pericyte growth state and contractile phenotype.


Assuntos
Proteínas de Ligação a DNA , Regulação da Expressão Gênica , Pericitos/metabolismo , Retina/metabolismo , Transdução de Sinais , Transativadores , Fator de Crescimento Transformador beta/metabolismo , Actinas/metabolismo , Animais , Western Blotting , Bovinos , Diferenciação Celular , Divisão Celular , Células Cultivadas , Primers do DNA/farmacologia , Técnica Indireta de Fluorescência para Anticorpo , Genes Dominantes , Camundongos , Proteínas dos Microfilamentos/metabolismo , Microscopia de Fluorescência , Proteínas Musculares/metabolismo , Músculo Liso/metabolismo , Músculo Liso Vascular/citologia , Mutação , Fator Regulador Miogênico 5 , Fenótipo , Proteínas Serina-Treonina Quinases , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Retina/citologia , Retroviridae/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Crescimento Transformador beta1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA