Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Lab Invest ; 103(6): 100123, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36849037

RESUMEN

Age-related macular degeneration (AMD) is a leading cause of irreversible central vision loss in the elderly. The pathology of neovascular age-related macular degeneration (nAMD), also known as wet AMD, is associated with an abnormal blood vessel growth in the eye and involves an imbalance of proangiogenic and antiangiogenic factors. Thrombospondin (TSP)-1 and TSP-2 are endogenous matricellular proteins that inhibit angiogenesis. TSP-1 is significantly diminished in eyes with AMD, although the mechanisms involved in its reduction are unknown. Granzyme B (GzmB) is a serine protease with an increased extracellular activity in the outer retina and choroid of human eyes with nAMD-related choroidal neovascularization (CNV). This study investigated whether TSP-1 and TSP-2 are GzmB substrates using in silico and cell-free cleavage assays and explored the relationship between GzmB and TSP-1 in human eyes with nAMD-related CNV and the effect of GzmB on TSP-1 in retinal pigment epithelial culture and an explant choroid sprouting assay (CSA). In this study, TSP-1 and TSP-2 were identified as GzmB substrates. Cell-free cleavage assays substantiated the GzmB proteolysis of TSP-1 and TSP-2 by showing dose-dependent and time-dependent cleavage products. TSP-1 and TSP-2 proteolysis were hindered by the inhibition of GzmB. In the retinal pigment epithelium and choroid of human eyes with CNV, we observed a significant inverse correlation between TSP-1 and GzmB, as indicated by lower TSP-1 and higher GzmB immunoreactivity. In CSA, the vascular sprouting area increased significantly with GzmB treatment and reduced significantly with TSP-1 treatment. Western blot showed significantly reduced expression of TSP-1 in GzmB-treated retinal pigment epithelial cell culture and CSA supernatant compared with that in controls. Together, our findings suggest that the proteolysis of antiangiogenic factors such as TSP-1 by extracellular GzmB might represent a mechanism through which GzmB may contribute to nAMD-related CNV. Future studies are needed to investigate whether pharmacologic inhibition of extracellular GzmB can mitigate nAMD-related CNV by preserving intact TSP-1.


Asunto(s)
Neovascularización Coroidal , Degeneración Macular , Humanos , Anciano , Trombospondina 1/metabolismo , Granzimas/metabolismo , Proteolisis , Degeneración Macular/complicaciones , Degeneración Macular/metabolismo , Degeneración Macular/patología , Neovascularización Coroidal/tratamiento farmacológico , Neovascularización Coroidal/etiología , Neovascularización Coroidal/metabolismo
2.
Br J Dermatol ; 189(3): 279-291, 2023 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-36652225

RESUMEN

BACKGROUND: Granzyme K (GzmK) is a serine protease with minimal presence in healthy tissues while abundant in inflamed tissues. Initially thought to play an exclusive role in immune-mediated cell death, extracellular GzmK can also promote inflammation. OBJECTIVES: To evaluate the role of GzmK in the pathogenesis of atopic dermatitis (AD), the most common inflammatory skin disease. METHODS: A panel of human AD and control samples was analysed to determine if GzmK is elevated. Next, to determine a pathological role for GzmK in AD-like skin inflammation, oxazolone-induced dermatitis was induced in GzmK-/- and wild-type (WT) mice. RESULTS: In human lesional AD samples, there was an increase in the number of GzmK+ cells compared with healthy controls. GzmK-/- mice exhibited reduced overall disease severity characterized by reductions in scaling, erosions and erythema. Surprisingly, the presence of GzmK did not notably increase the overall pro-inflammatory response or epidermal barrier permeability in WT mice; rather, GzmK impaired angiogenesis, increased microvascular damage and microhaemorrhage. Mechanistically, GzmK contributed to vessel damage through cleavage of syndecan-1, a key structural component of the glycocalyx, which coats the luminal surface of vascular endothelia. CONCLUSIONS: GzmK may provide a potential therapeutic target for skin conditions associated with persistent inflammation, vasculitis and pathological angiogenesis.


Asunto(s)
Dermatitis Atópica , Granzimas , Animales , Humanos , Ratones , Dermatitis Atópica/patología , Epidermis/metabolismo , Granzimas/metabolismo , Inflamación , Piel/patología
3.
Front Pharmacol ; 13: 980742, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36204224

RESUMEN

Age-related ocular diseases are the leading cause of blindness in developed countries and constitute a sizable socioeconomic burden worldwide. Age-related macular degeneration (AMD) and Fuchs endothelial corneal dystrophy (FECD) are some of the most common age-related diseases of the retina and cornea, respectively. AMD is characterized by a breakdown of the retinal pigment epithelial monolayer, which maintains retinal homeostasis, leading to retinal degeneration, while FECD is characterized by degeneration of the corneal endothelial monolayer, which maintains corneal hydration status, leading to corneal edema. Both AMD and FECD pathogenesis are characterized by disorganized local extracellular matrix (ECM) and toxic protein deposits, with both processes linked to aberrant protease activity. Granzyme B (GrB) is a serine protease traditionally known for immune-mediated initiation of apoptosis; however, it is now recognized that GrB is expressed by a variety of immune and non-immune cells and aberrant extracellular localization of GrB substantially contributes to various age-related pathologies through dysregulated cleavage of ECM, tight junction, and adherens junction proteins. Despite growing recognition of GrB involvement in multiple age-related pathologies, its role in AMD and FECD remains poorly understood. This review summarizes the pathophysiology of, and similarities between AMD and FECD, outlines the current knowledge of the role of GrB in AMD and FECD, as well as hypothesizes putative contributions of GrB to AMD and FECD pathogenesis and highlights the therapeutic potential of pharmacologically inhibiting GrB as an adjunctive treatment for AMD and FECD.

4.
Sci Rep ; 12(1): 12622, 2022 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-35871073

RESUMEN

Pressure injuries, also known as pressure ulcers, are regions of localized damage to the skin and/or underlying tissue. Repeated rounds of ischemia-reperfusion (I/R) have a major causative role for tissue damage in pressure injury. Ischemia prevents oxygen/nutrient supply, and restoration of blood flow induces a burst of reactive oxygen species that damages blood vessels, surrounding tissues and can halt blood flow return. Minimizing the consequences of repeated I/R is expected to provide a protective effect against pressure injury. Sulfaphenazole (SP), an off patent sulfonamide antibiotic, is a potent CYP 2C6 and CYP 2C9 inhibitor, functioning to decrease post-ischemic vascular dysfunction and increase blood flow. The therapeutic effect of SP on pressure injury was therefore investigated in apolipoprotein E knockout mice, a model of aging susceptible to ischemic injury, which were subjected to repeated rounds of I/R-induced skin injury. SP reduced overall severity, improved wound closure and increased wound tensile strength compared to vehicle-treated controls. Saliently, SP restored tissue perfusion in and around the wound rapidly to pre-injury levels, decreased tissue hypoxia, and reduced both inflammation and fibrosis. SP also demonstrated bactericidal activity through enhanced M1 macrophage activity. The efficacy of SP in reducing thermal injury severity was also demonstrated. SP is therefore a potential therapeutic option for pressure injury and other ischemic skin injuries.


Asunto(s)
Úlcera por Presión , Daño por Reperfusión , Sulfafenazol , Animales , Ratones , Isquemia , Perfusión , Especies Reactivas de Oxígeno , Daño por Reperfusión/tratamiento farmacológico , Sulfafenazol/farmacología
5.
Gastroenterology ; 162(3): 877-889.e7, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34861219

RESUMEN

BACKGROUND & AIMS: Excessive shedding of apoptotic enterocytes into the intestinal lumen is observed in inflammatory bowel disease and is correlated with disease relapse. Based on their cytolytic capacity and surveillance behavior, we investigated whether intraepithelial lymphocytes expressing the γδ T cell receptor (γδ IELs) are actively involved in the shedding of enterocytes into the lumen. METHODS: Intravital microscopy was performed on GFP γδ T cell reporter mice treated with intraperitoneal lipopolysaccharide (10 mg/kg) for 90 minutes to induce tumor necrosis factor-mediated apoptosis. Cell shedding in various knockout or transgenic mice in the presence or absence of blocking antibody was quantified by immunostaining for ZO-1 funnels and cleaved caspase-3 (CC3). Granzyme A and granzyme B release from ex vivo-stimulated γδ IELs was quantified by enzyme-linked immunosorbent assay. Immunostaining for γδ T cell receptor and CC3 was performed on duodenal and ileal biopsies from controls and patients with Crohn's disease. RESULTS: Intravital microscopy of lipopolysaccharide-treated mice revealed that γδ IELs make extended contact with shedding enterocytes. These prolonged interactions require CD103 engagement by E-cadherin, and CD103 knockout or blockade significantly reduced lipopolysaccharide-induced shedding. Furthermore, we found that granzymes A and B, but not perforin, are required for cell shedding. These extracellular granzymes are released by γδ IELs both constitutively and after CD103/E-cadherin ligation. Moreover, we found that the frequency of γδ IEL localization to CC3-positive enterocytes is increased in Crohn's disease biopsies compared with healthy controls. CONCLUSIONS: Our results uncover a previously unrecognized role for γδ IELs in facilitating tumor necrosis factor-mediated shedding of apoptotic enterocytes via CD103-mediated extracellular granzyme release.


Asunto(s)
Antígenos CD/metabolismo , Enfermedad de Crohn/metabolismo , Enterocitos/fisiología , Granzimas/metabolismo , Cadenas alfa de Integrinas/metabolismo , Linfocitos Intraepiteliales/fisiología , Adolescente , Adulto , Animales , Antígenos CD/genética , Apoptosis , Cadherinas/metabolismo , Caspasa 3/metabolismo , Enfermedad de Crohn/patología , Duodeno/patología , Enterocitos/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Íleon/patología , Cadenas alfa de Integrinas/genética , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Linfocitos Intraepiteliales/enzimología , Linfocitos Intraepiteliales/patología , Microscopía Intravital , Yeyuno/inmunología , Yeyuno/patología , Lipopolisacáridos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Adulto Joven
6.
NPJ Aging Mech Dis ; 7(1): 6, 2021 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-33674592

RESUMEN

Pressure injuries (PIs), also known as bedsores or pressure ulcers, are a major cause of death and morbidity in the elderly. The serine protease, Granzyme B (GzmB), contributes to skin aging and impaired wound healing. Aging is a major risk factor for PIs; thus, the role of GzmB in PI pathogenesis was investigated. GzmB levels in human PI tissue and wound fluids were markedly elevated. A causative role for GzmB was assessed in GzmB knockout (GzmB-/-) and wild-type (WT) mice using a murine model of PI. An apolipoprotein E knockout (ApoE-/-) model of aging and vascular dysfunction was also utilized to assess GzmB in a relevant age-related model better resembling tissue perfusion in the elderly. PI severity displayed no difference between young GzmB-/- and WT mice. However, in aged mice, PI severity was reduced in mice lacking GzmB. Mechanistically, GzmB increased vascular wall inflammation and impaired extracellular matrix remodeling. Together, GzmB is an important contributor to age-dependent impaired PI healing.

7.
Nat Commun ; 12(1): 302, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33436591

RESUMEN

Pemphigoid diseases refer to a group of severe autoimmune skin blistering diseases characterized by subepidermal blistering and loss of dermal-epidermal adhesion induced by autoantibody and immune cell infiltrate at the dermal-epidermal junction and upper dermis. Here, we explore the role of the immune cell-secreted serine protease, granzyme B, in pemphigoid disease pathogenesis using three independent murine models. In all models, granzyme B knockout or topical pharmacological inhibition significantly reduces total blistering area compared to controls. In vivo and in vitro studies show that granzyme B contributes to blistering by degrading key anchoring proteins in the dermal-epidermal junction that are necessary for dermal-epidermal adhesion. Further, granzyme B mediates IL-8/macrophage inflammatory protein-2 secretion, lesional neutrophil infiltration, and lesional neutrophil elastase activity. Clinically, granzyme B is elevated and abundant in human pemphigoid disease blister fluids and lesional skin. Collectively, granzyme B is a potential therapeutic target in pemphigoid diseases.


Asunto(s)
Enfermedades Autoinmunes/enzimología , Enfermedades Autoinmunes/patología , Granzimas/antagonistas & inhibidores , Granzimas/metabolismo , Animales , Autoantígenos/metabolismo , Vesícula , Quimiocina CXCL2/metabolismo , Factores Quimiotácticos/farmacología , Modelos Animales de Enfermedad , Epidermólisis Ampollosa/enzimología , Epidermólisis Ampollosa/patología , Humanos , Inflamación/patología , Integrina alfa6/metabolismo , Interleucina-8/metabolismo , Infiltración Neutrófila/efectos de los fármacos , Colágenos no Fibrilares/metabolismo , Penfigoide Ampolloso/enzimología , Penfigoide Ampolloso/patología , Índice de Severidad de la Enfermedad , Colágeno Tipo XVII
8.
J Invest Dermatol ; 141(1): 36-47, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32504614

RESUMEN

Atopic dermatitis (AD) is the most common inflammatory skin condition. Skin barrier dysfunction is of major importance in AD because it facilitates allergen sensitization and systemic allergic responses. Long regarded as a pro-apoptotic protease, emerging studies indicate granzyme B (GzmB) to have extracellular roles involving the proteolytic cleavage of extracellular matrix, cell adhesion proteins, and basement membrane proteins. Minimally expressed in normal skin, GzmB is elevated in AD and is positively correlated with disease severity and pruritus. We hypothesized that GzmB contributes to AD through extracellular protein cleavage. A causative role for GzmB was assessed in an oxazolone-induced murine model of dermatitis, comparing GzmB-/- mice with wild-type mice, showing significant reductions in inflammation, epidermal thickness, and lesion formation in GzmB-/- mice. Topical administration of a small-molecule GzmB inhibitor reduced disease severity compared with vehicle-treated controls. Mechanistically, GzmB impaired epithelial barrier function through E-cadherin and FLG cleavage. GzmB proteolytic activity contributes to impaired epidermal barrier function and represents a valid therapeutic target for AD.


Asunto(s)
Cadherinas/metabolismo , Dermatitis Atópica/metabolismo , Granzimas/metabolismo , Oxazolona/efectos adversos , Proteínas S100/metabolismo , Animales , Dermatitis Atópica/inducido químicamente , Dermatitis Atópica/patología , Epidermis/metabolismo , Matriz Extracelular/metabolismo , Proteínas Filagrina , Humanos
9.
Cell Signal ; 76: 109804, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33035645

RESUMEN

Chronic inflammation and impaired wound healing play important roles in the pathophysiology of cardiovascular diseases. Moreover, the aberrant secretion of proteases plays a critical role in pathological tissue remodeling in chronic inflammatory conditions. Human Granzymes (Granule secreted enzymes - Gzms) comprise a family of five (GzmA, B, H, K, M) cell-secreted serine proteases. Although each unique in function and substrate specificities, Gzms were originally thought to share redundant, intracellular roles in cytotoxic lymphocyte-induced cell death. However, an abundance of evidence has challenged this dogma. It is now recognized, that individual Gzms exhibit unique substrate repertoires and functions both intracellularly and extracellularly. In the extracellular milieu, Gzms contribute to inflammation, vascular dysfunction and permeability, reduced cell adhesion, release of matrix-sequestered growth factors, receptor activation, and extracellular matrix cleavage. Despite these recent findings, the non-cytotoxic functions of Gzms in the context of cardiovascular disease pathogenesis remain poorly understood. Minimally detected in tissues and bodily fluids of normal individuals, GzmB is elevated in patients with acute coronary syndromes, coronary artery disease, and myocardial infarction. Pre-clinical animal models have exemplified the importance of GzmB in atherosclerosis, aortic aneurysm, and cardiac fibrosis as animals deficient in GzmB exhibit reduced tissue remodeling, improved disease phenotypes and increased survival. Although a role for GzmB in cardiovascular disease is described, further work to elucidate the mechanisms that underpin the remaining human Gzms activity in cardiovascular disease is necessary. The present review provides a summary of the pre-clinical and clinical evidence, as well as emerging areas of research pertaining to Gzms in tissue remodeling and cardiovascular disease.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Granzimas/fisiología , Animales , Humanos
10.
Front Immunol ; 11: 574, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32318066

RESUMEN

Granzymes are a family of serine proteases first shown to be intracellular initiators of immune-mediated cell death in target pathogenic cells. In addition to its intracellular role, Granzyme B (GzmB) has important extracellular functions in immune regulation and extracellular matrix (ECM) degradation. Verified substrates of extracellular GzmB activity include tight junctional and ECM proteins. Interestingly, little is known about the activity of GzmB in the outer human retina, a tissue in which the degradation of the tight junctional contacts of retinal pigment epithelial (RPE) cells and within the external limiting membrane, as well as remodeling of the ECM in Bruch's membrane, cause the breakdown of the blood-retinal barrier and slowing of metabolite transport between neuroretina and choroidal blood supply. Such pathological changes in outer retina signal early events in the development of age-related macular degeneration (AMD), a multifactorial, chronic inflammatory eye disease. This study is the first to focus on the distribution of GzmB in the outer retina of the healthy and diseased post-mortem human eye. Our results revealed that GzmB is present in RPE and choroidal mast cells. More immunoreactive cells are present in older (>65 years) compared to younger (<55 years) donor eyes, and choroidal immunoreactive cells are more numerous in eyes with choroidal neovascularization (CNV), while RPE immunoreactive cells are more numerous in eyes with soft drusen, an early AMD event. In vitro studies demonstrated that RPE-derived tight junctional and ECM proteins are cleaved by exogenous GzmB stimulation. These results suggest that the increased presence of GzmB immunoreactive cells in outer retina of older (healthy) eyes as well as in diseased eyes with CNV (from AMD) and eyes with soft drusen exacerbate ECM remodeling in the Bruch's membrane and degradation of the blood-retinal barrier. Currently there are no treatments that prevent remodeling of the Bruch's membrane and/or the loss of function of the outer blood-retinal barrier, known to promote early AMD changes, such as drusen deposition, RPE dysfunction and pro-inflammation. Specific inhibitors of GzmB, already in preclinical studies for non-ocular diseases, may provide new strategies to stop these early events associated with the development of AMD.


Asunto(s)
Coroides/enzimología , Neovascularización Coroidal/enzimología , Matriz Extracelular/enzimología , Granzimas/metabolismo , Epitelio Pigmentado de la Retina/enzimología , Adulto , Anciano , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Humanos , Masculino , Mastocitos/enzimología , Persona de Mediana Edad , Retina/enzimología , Uniones Estrechas/metabolismo
11.
Sci Rep ; 9(1): 10027, 2019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-31296909

RESUMEN

Wildfire smoke induces acute pulmonary distress and is of particular concern to risk groups such as the sick and elderly. Wood smoke (WS) contains many of the same toxic compounds as those found in cigarette smoke (CS) including polycyclic aromatic hydrocarbons, carbon monoxide, and free radicals. CS is a well-established risk factor for respiratory diseases such as asthma and COPD. Limited studies investigating the biological effects of WS on the airway epithelium have been performed. Using a cell culture-based model, we assessed the effects of a WS-infused solution on alveolar epithelial barrier function, cell migration, and survival. The average geometric mean of particles in the WS was 178 nm. GC/MS analysis of the WS solution identified phenolic and cellulosic compounds. WS exposure resulted in a significant reduction in barrier function, which peaked after 24 hours of continuous exposure. The junctional protein E-cadherin showed a prominent reduction in response to increasing concentrations of WS. Furthermore, WS significantly repressed cell migration following injury to the cell monolayer. There was no difference in cell viability following WS exposure. Mechanistically, WS exposure induced activation of the p44/42, but not p38, MAPK signaling pathway, and inhibition of p44/42 phosphorylation prevented the disruption of barrier function and loss of E-cadherin staining. Thus, WS may contribute to the breakdown of alveolar structure and function through a p44/42 MAPK-dependent pathway and may lead to the development and/or exacerbation of respiratory pathologies with chronic exposure.


Asunto(s)
Células Epiteliales Alveolares/patología , Epitelio/fisiopatología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Humo/efectos adversos , Uniones Estrechas/patología , Células A549 , Células Epiteliales Alveolares/efectos de los fármacos , Cadherinas/metabolismo , Línea Celular , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Enfermedades Pulmonares/inducido químicamente , Uniones Estrechas/efectos de los fármacos , Incendios Forestales
12.
Proc Natl Acad Sci U S A ; 116(10): 4518-4527, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30787191

RESUMEN

Podocalyxin (Podxl) is broadly expressed on the luminal face of most blood vessels in adult vertebrates, yet its function on these cells is poorly defined. In the present study, we identified specific functions for Podxl in maintaining endothelial barrier function. Using electrical cell substrate impedance sensing and live imaging, we found that, in the absence of Podxl, human umbilical vein endothelial cells fail to form an efficient barrier when plated on several extracellular matrix substrates. In addition, these monolayers lack adherens junctions and focal adhesions and display a disorganized cortical actin cytoskeleton. Thus, Podxl has a key role in promoting the appropriate endothelial morphogenesis required to form functional barriers. This conclusion is further supported by analyses of mutant mice in which we conditionally deleted a floxed allele of Podxl in vascular endothelial cells (vECs) using Tie2Cre mice (PodxlΔTie2Cre). Although we did not detect substantially altered permeability in naïve mice, systemic priming with lipopolysaccharide (LPS) selectively disrupted the blood-brain barrier (BBB) in PodxlΔTie2Cre mice. To study the potential consequence of this BBB breach, we used a selective agonist (TFLLR-NH2) of the protease-activated receptor-1 (PAR-1), a thrombin receptor expressed by vECs, neuronal cells, and glial cells. In response to systemic administration of TFLLR-NH2, LPS-primed PodxlΔTie2Cre mice become completely immobilized for a 5-min period, coinciding with severely dampened neuroelectric activity. We conclude that Podxl expression by CNS tissue vECs is essential for BBB maintenance under inflammatory conditions.


Asunto(s)
Barrera Hematoencefálica , Inflamación/metabolismo , Sialoglicoproteínas/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Morfogénesis
13.
J Invest Dermatol ; 139(4): 930-939, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30395844

RESUMEN

Granzyme K (GzmK), traditionally described as a pro-apoptotic, granule-secreted serine protease, has been proposed to promote inflammation. Found at low levels in the plasma of healthy individuals, GzmK is markedly elevated in response to sepsis and infection. In this study we investigated the role of GzmK in inflammation and remodeling in response to thermal injury. In human burn tissue, GzmK was elevated compared with normal skin, with expression predominantly found in macrophages. GzmK was expressed and secreted by cultured human classically activated macrophages. To assess the role of GzmK in response to skin wounding, wild-type or GzmK-/- mice were subjected to grade 2 thermal injury. GzmK-/- mice exhibited improved wound closure, matrix organization, and tensile strength compared with wild-type mice. Reduced proinflammatory IL-6, ICAM-1, VCAM-1, and MCP-1 expressions were observed at 3 days after injury. Additionally, GzmK induced IL-6 expression in keratinocytes and skin fibroblasts that was dependent on PAR-1 activation. Re-epithelialization showed the greatest degree of improvement of all healing parameters, suggesting that keratinocytes are sensitive to GzmK-mediated proteolysis. In support, keratinocytes, but not skin fibroblasts, exposed to GzmK showed impaired wound healing in vitro. In summary, GzmK influences wound healing by augmenting inflammation and impeding epithelialization.


Asunto(s)
Quemaduras/genética , Regulación de la Expresión Génica , Granzimas/genética , Inflamación/genética , Macrófagos/metabolismo , Repitelización/fisiología , Animales , Quemaduras/metabolismo , Quemaduras/patología , Citocinas/biosíntesis , Citocinas/genética , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibroblastos/patología , Granzimas/biosíntesis , Humanos , Inmunohistoquímica , Inflamación/metabolismo , Inflamación/patología , Queratinocitos/metabolismo , Queratinocitos/patología , Macrófagos/patología , Ratones , ARN/genética
14.
Compr Physiol ; 9(1): 75-125, 2018 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-30549015

RESUMEN

Cardiovascular disease leading to heart failure (HF) remains a leading cause of morbidity and mortality worldwide. Improved pharmacological and interventional coronary procedures have led to improved outcomes following acute myocardial infarction. This success has translated into an unforeseen increased incidence in HF. This review summarizes the signaling pathways implicated in the transition to HF following cardiac injury. In addition, we provide an update on cell death signaling and discuss recent advances in cardiac fibrosis as an independent event leading to HF. Finally, we discuss cell-based therapies and their possible use to avert the deteriorating nature of HF. © 2019 American Physiological Society. Compr Physiol 9:75-125, 2019.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Medicina Regenerativa/métodos , Transducción de Señal , Ingeniería de Tejidos/métodos , Animales , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/terapia , Humanos , Miocardio/metabolismo , Miocardio/patología
15.
Exp Mol Med ; 50(5): 1-11, 2018 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-29849046

RESUMEN

Granzyme B (GzmB) is a serine protease that has long been thought to function exclusively in lymphocyte-mediated apoptosis. In recent years, this paradigm has been revisited due to the recognition that GzmB accumulates in the extracellular milieu in many autoimmune and chronic inflammatory disorders, and contributes to impaired tissue remodeling due to the cleavage of extracellular matrix proteins. Knockout studies suggest that GzmB-mediated cleavage of decorin (DCN) contributes to impaired collagen fibrillogenesis and remodeling. As DCN is anti-fibrotic and contributes to reduced hypertrophic scarring, GzmB-induced DCN cleavage could play a role in wound healing following burn injury. In the present study, a novel, gel-formulated, first-in-class small-molecule inhibitor of GzmB, VTI-1002, was assessed in a murine model of impaired, diabetic burn wound healing. VTI-1002 exhibited high specificity, potency, and target selectivity. Gel-formulated VTI-1002 was able to penetrate the stratum corneum and was retained in the skin with minimal systemic absorption. Daily topical administration of VTI-1002 gel for 30 days following thermal injury showed significantly accelerated wound closure, increased DCN protein levels, and collagen organization that was translated into significantly increased wound tensile strength compared to controls. Overall, VTI-1002 gel was well-tolerated in vivo and no adverse events were observed. Topical application of VTI-1002 represents a novel therapeutic approach for the treatment of cutaneous burn wounds.


Asunto(s)
Quemaduras/patología , Granzimas/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/administración & dosificación , Bibliotecas de Moléculas Pequeñas/farmacología , Cicatrización de Heridas/efectos de los fármacos , Administración Tópica , Animales , Cicatriz/patología , Diabetes Mellitus Experimental/patología , Modelos Animales de Enfermedad , Granzimas/metabolismo , Masculino , Ratones Endogámicos C57BL
16.
Sci Rep ; 8(1): 9690, 2018 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-29946113

RESUMEN

In healthy skin, epidermis and dermis are anchored together at the dermal-epidermal junction (DEJ), a specialized basement membrane pivotal for skin integrity and function. However, increased inflammation in the DEJ is associated with the disruption and separation of this junction and sub-epidermal blistering. Granzyme B (GzmB) is a serine protease secreted by immune cells. Dysregulated inflammation may lead to increased GzmB accumulation and proteolysis in the extracellular milieu. Although elevated GzmB is observed at the level of the DEJ in inflammatory and blistering skin conditions, the present study is the first to explore GzmB in the context of DEJ degradation in autoimmune sub-epidermal blistering. In the present study, GzmB induced separation of the DEJ in healthy human skin. Subsequently, α6/ß4 integrin, collagen VII, and collagen XVII were identified as extracellular substrates for GzmB through western blot, and specific cleavage sites were identified by mass spectrometry. In human bullous pemphigoid, dermatitis herpetiformis, and epidermolysis bullosa acquisita, GzmB was elevated at the DEJ when compared to healthy samples, while α6/ß4 integrin, collagen VII, and collagen XVII were reduced or absent in the area of blistering. In summary, our results suggest that regardless of the initial causation of sub-epidermal blistering, GzmB activity is a common final pathway that could be amenable to a single targeted treatment approach.


Asunto(s)
Epidermis/metabolismo , Granzimas/metabolismo , Piel/metabolismo , Autoantígenos/metabolismo , Dermatitis Herpetiforme/metabolismo , Dermis/metabolismo , Humanos , Inmunohistoquímica , Técnicas In Vitro , Colágenos no Fibrilares/metabolismo , Penfigoide Ampolloso/metabolismo , Espectrometría de Masas en Tándem , Colágeno Tipo XVII
17.
Sci Rep ; 7(1): 15857, 2017 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-29158532

RESUMEN

Decorin (DCN) is a small-leucine rich proteoglycan that mediates collagen fibrillogenesis, organization, and tensile strength. Adventitial DCN is reduced in abdominal aortic aneurysm (AAA) resulting in vessel wall instability thereby predisposing the vessel to rupture. Recombinant DCN fusion protein CAR-DCN was engineered with an extended C-terminus comprised of CAR homing peptide that recognizes inflamed blood vessels and penetrates deep into the vessel wall. In the present study, the role of systemically-administered CAR-DCN in AAA progression and rupture was assessed in a murine model. Apolipoprotein E knockout (ApoE-KO) mice were infused with angiotensin II (AngII) for 28 days to induce AAA formation. CAR-DCN or vehicle was administrated systemically until day 15. Mortality due to AAA rupture was significantly reduced in CAR-DCN-treated mice compared to controls. Although the prevalence of AAA was similar between vehicle and CAR-DCN groups, the severity of AAA in the CAR-DCN group was significantly reduced. Histological analysis revealed that CAR-DCN treatment significantly increased DCN and collagen levels within the aortic wall as compared to vehicle controls. Taken together, these results suggest that CAR-DCN treatment attenuates the formation and rupture of Ang II-induced AAA in mice by reinforcing the aortic wall.


Asunto(s)
Aneurisma de la Aorta Abdominal/genética , Apolipoproteínas E/genética , Decorina/genética , Proteínas Recombinantes/genética , Angiotensina II/genética , Animales , Aneurisma de la Aorta Abdominal/inducido químicamente , Aneurisma de la Aorta Abdominal/fisiopatología , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Noqueados , Proteínas Recombinantes/administración & dosificación
18.
Oncotarget ; 7(48): 78516-78531, 2016 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-27705938

RESUMEN

The incidence of heart failure with concomitant cardiac fibrosis is very high in developed countries. Fibroblast activation in heart is causal to cardiac fibrosis as they convert to hypersynthetic cardiac myofibroblasts. There is no known treatment for cardiac fibrosis. Myofibroblasts contribute to the inappropriate remodeling of the myocardial interstitium, which leads to reduced cardiac function and ultimately heart failure. Elevated levels of autophagy have been linked to stress-induced ventricular remodeling and other cardiac diseases. Previously, we had shown that TGF-ß1 treatment of human atrial fibroblasts both induced autophagy and enhanced the fibrogenic response supporting a linkage between the myofibroblast phenotype and autophagy. We now demonstrate that with in vitro culture of primary rat cardiac fibroblasts, inhibition of autophagy represses fibroblast to myofibroblast phenoconversion. Culturing unpassaged cardiac fibroblasts for 72 hours on plastic tissue culture plates is associated with elevated α-smooth muscle actin (α-SMA) expression. This activation parallels increased microtubule-associated protein 1A/1B-light chain 3 (LC-3ß II) protein expression. Inhibition of autophagy with bafilomycin-A1 (Baf-A1) and chloroquine (CQ) in cardiac fibroblasts significantly reduces α-SMA and extracellular domain A fibronectin (ED-A FN) protein vs untreated controls. Myofibroblast cell migration and contractility were significantly reduced following inhibition of autophagy. These data support the possibility of a causal link between cardiac fibroblast-to-myofibroblast phenoconversion and autophagy.


Asunto(s)
Autofagia/efectos de los fármacos , Cardiomiopatías/prevención & control , Cloroquina/farmacología , Fibroblastos/efectos de los fármacos , Macrólidos/farmacología , Miocardio/patología , Miofibroblastos/efectos de los fármacos , Actinas/metabolismo , Animales , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Fibroblastos/metabolismo , Fibroblastos/patología , Fibronectinas/metabolismo , Fibrosis , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Miocardio/metabolismo , Miofibroblastos/metabolismo , Miofibroblastos/patología , Fenotipo , Fosforilación , Cultivo Primario de Células , Ratas Sprague-Dawley , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
Biochim Biophys Acta ; 1863(6 Pt A): 1261-8, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27039037

RESUMEN

Inappropriate cardiac interstitial remodeling is mediated by activated phenoconverted myofibroblasts. The synthesis of matrix proteins by these cells is triggered by both chemical and mechanical stimuli. Ski is a repressor of TGFß1/Smad signaling and has been described as possessing anti-fibrotic properties within the myocardium. We hypothesized that overexpression of Ski in myofibroblasts will induce an apoptotic response, which may either be supported or opposed by autophagic flux. We used primary myofibroblasts (activated fibroblasts) which were sourced from whole heart preparations that were only passaged once. We found that overexpression of Ski results in distinct morphological and biochemical changes within primary cardiac myofibroblasts associated with apoptosis. Ski treatment was associated with the expression of pro-apoptotic factors such as Bax, caspase-7, and -9. Our results indicate that Ski triggers a pro-death mechanism in primary rat cardiac myofibroblasts that is mediated through the intrinsic apoptotic pathway. Myofibroblast survival is prolonged by an autophagic response, as the dataset indicate that apoptosis is hastened when autophagy is inhibited. We suggest that the apoptotic death response of myofibroblasts is working in parallel with the previously observed anti-fibrotic properties of Ski within this cell type. As myofibroblasts are the sole mediators of matrix expansion in heart failure, we suggest that Ski, or a putative Ski-mimetic, may induce graded apoptosis in myofibroblasts within the failing heart and may be a novel therapeutic approach towards controlling cardiac fibrosis. Future studies are needed to examine the potential effects of Ski overexpression on other cell types in the heart.


Asunto(s)
Apoptosis/fisiología , Autofagia/fisiología , Miofibroblastos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Actinas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Autofagia/efectos de los fármacos , Autofagia/genética , Western Blotting , Caspasas/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Expresión Génica , Macrólidos/farmacología , Masculino , Microscopía Confocal , Miofibroblastos/citología , Miofibroblastos/efectos de los fármacos , Proteínas Proto-Oncogénicas/genética , Ratas Sprague-Dawley , Estaurosporina/farmacología , Factores de Tiempo , Transfección , Vimentina/metabolismo
20.
Am J Physiol Heart Circ Physiol ; 310(2): H239-49, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26566727

RESUMEN

In cardiac wound healing following myocardial infarction (MI), relatively inactive resident cardiac fibroblasts phenoconvert to hypersynthetic/secretory myofibroblasts that produce large quantities of extracellular matrix (ECM) and fibrillar collagen proteins. Our laboratory and others have identified TGFß1 as being a persistent stimulus in the chronic and inappropriate wound healing phase that is marked by hypertrophic scarring and eventual stiffening of the entire myocardium, ultimately leading to the pathogenesis of heart failure following MI. Ski is a potent negative regulator of TGFß/Smad signaling with known antifibrotic effects. Conversely, Scleraxis is a potent profibrotic basic helix-loop-helix transcription factor that stimulates fibrillar collagen expression. We hypothesize that TGFß1 induces Scleraxis expression by a novel Smad-independent pathway. Our data support the hypothesis that Scleraxis expression is induced by TGFß1 through a Smad-independent pathway in the cardiac myofibroblast. Specifically, we demonstrate that TGFß1 stimulates p42/44 (Erk1/2) kinases, which leads to increased Scleraxis expression. Inhibition of MEK1/2 using U0126 led to a sequential temporal reduction of phospho-p42/44 and subsequent Scleraxis expression. We also found that adenoviral Ski expression in primary myofibroblasts caused a significant repression of endogenous Scleraxis expression at both the mRNA and protein levels. Thus we have identified a novel TGFß1-driven, Smad-independent, signaling cascade that may play an important role in regulating the fibrotic response in activated cardiac myofibroblasts following cardiac injury.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Miocitos Cardíacos/metabolismo , Proteínas Smad/fisiología , Factor de Crecimiento Transformador beta1/fisiología , Células 3T3 , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Butadienos/farmacología , Células COS , Cardiomegalia/metabolismo , Cardiomegalia/patología , Chlorocebus aethiops , Fibrosis/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Nitrilos/farmacología , Cultivo Primario de Células , Proteínas Proto-Oncogénicas/farmacología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...