Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Curr Pathobiol Rep ; 9(4): 93-105, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34900402

RESUMEN

Coronavirus disease 2019 (COVID-19) is an infectious disease caused by the SARS-CoV-2 betacoronavirus and has taken over 761,426 American lives as of the date of publication and will likely result in long-term, if not permanent, tissue damage for countless patients. COVID-19 presents with diverse and multisystemic pathologic processes, including a hyperinflammatory response, acute respiratory distress syndrome (ARDS), vascular injury, microangiopathy, tissue fibrosis, angiogenesis, and widespread thrombosis across multiple organs, including the lungs, heart, kidney, liver, and brain. C-X-C chemokines contribute to these pathologies by attracting inflammatory mediators, the disruption of endothelial cell integrity and function, and the initiation and propagation of the cytokine storm. Among these, CXCL10 is recognized as a critical contributor to the hyperinflammatory state and poor prognosis in COVID-19. CXCL10 is also known to regulate growth factor-induced fibrosis, and recent evidence suggests the CXCL10-CXCR3 signaling system may be vital in targeting convergent pro-inflammatory and pro-fibrotic pathways. This review will explore the mechanistic role of CXCL10 and related chemokines in fibrotic complications associated with COVID-19 and the potential of CXCL10-targeted therapeutics for early intervention and long-term treatment of COVID-19-induced fibrosis.

2.
Clin Exp Ophthalmol ; 49(1): 60-69, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33426793

RESUMEN

BACKGROUND: Glaucoma filtration surgery (GFS) is limited by subconjunctival, episcleral and scleral fibrosis sealing the trabeculectomy and scarring the filtering bleb. Mitomycin-C (MMC) is commonly applied intraoperatively to the subconjunctival and/or intrascleral space to reduce scarring and promotes GFS success but is associated with postoperative scleral melting and bleb leaks. IP-10 peptide (IP-10p), an ELR-negative CXC chemokine mimetic and inhibitor of fibroblast function, may be an alternative or adjunct to current postoperative GFS treatments. This study sought to determine if IP-10p produces histological changes in tissue remodelling, vascularity and fibrosis that enhance bleb survival after GFS. METHODS: Rabbits underwent tube-assisted filtration surgery on the right eye with either: (a) IP-10p injected into bleb at time of surgery and postoperative days 2, 4 and 7, (b) intraoperative MMC or (c) intraoperative MMC plus IP-10p injected into bleb at time of surgery and postoperative days 2, 4 and 7. Left contralateral eyes were treated with balanced salt solution (BSS). RESULTS: IP-10p-treated blebs demonstrated reduced collagen deposition, cellularity and overall reduction of scar formation compared to BSS-control. Bleb vascularity was reduced compared to BSS-control and MMC treatment groups. Additionally, IP-10p/MMC treated eyes demonstrated an increased number of conjunctival goblet cells in bleb histology compared to the dramatic loss seen with MMC treatment alone. CONCLUSIONS: This study demonstrates that IP-10p significantly reduces histological scarring compared to BSS or MMC alone, does not damage the conjunctiva to the extent of current standards, and may be an alternative or adjunct to MMC for those undergoing GFS.


Asunto(s)
Cirugía Filtrante , Glaucoma , Trabeculectomía , Animales , Conjuntiva/patología , Modelos Animales de Enfermedad , Fibrosis , Glaucoma/patología , Glaucoma/cirugía , Presión Intraocular , Mitomicina , Conejos , Cicatrización de Heridas
3.
Transl Vis Sci Technol ; 9(11): 23, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33150049

RESUMEN

Purpose: Mitomycin C is routinely applied during trabeculectomy surgeries to enhance bleb survival after glaucoma filtration surgery. The current approach involves placing cellulose sponges soaked in mitomycin C at a standard concentration onto bare sclera for a predetermined duration, which varies among surgeons. The purpose of this study was to compare the effects of sponge-applied versus intra-Tenon injection of mitomycin C during modified trabeculectomy. Methods: Two groups of five New Zealand White rabbits underwent glaucoma filtration surgery with either preoperative intra-Tenon injection of mitomycin C or intraoperative application of mitomycin C using a cellulose sponge. Postoperative intraocular pressure was recorded weekly, and eyes were enucleated and sent for pathological examination and histological analysis. Results: An intra-Tenon injection of mitomycin C resulted in decreased intraocular pressure measurements and bleb vascularity compared to the controls but increased levels compared to the sponge-applied group. Collagen deposition and cellularity were reduced and the goblet cell population was increased in the intra-Tenon injection group. Conclusions: This study shows that an intra-Tenon injection can be an effective method for administering mitomycin C compared to the standard-of-care approach of mitomycin C being sponge applied onto bare sclera. Mitomycin C injection led to a greater reduction in intraocular pressure and inhibition of fibroblasts. The associated goblet cell population that can lead to increased mitomycin C toxicity-related morbidity was minimized with the intra-Tenon injection compared to the sponge-applied MMC treatment. Therefore, patients with ocular surface disease may benefit from an intra-Tenon injection. Translational Relevance: This project provides a direct, qualitative assessment in an animal model of common techniques within glaucoma filtration surgery for drug delivery to improve surgical success.


Asunto(s)
Trabeculectomía , Animales , Humanos , Presión Intraocular , Mitomicina , Conejos , Esclerótica , Tonometría Ocular
4.
PLoS One ; 15(10): e0240986, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33095822

RESUMEN

Fibrosis is a chronic disease with heterogeneous clinical presentation, rate of progression, and occurrence of comorbidities. Systemic sclerosis (scleroderma, SSc) is a rare rheumatic autoimmune disease that encompasses several aspects of fibrosis, including highly variable fibrotic manifestation and rate of progression. The development of effective treatments is limited by these variabilities. The fibrotic response is characterized by both chronic inflammation and extracellular remodeling. Therefore, there is a need for improved understanding of which inflammation-related genes contribute to the ongoing turnover of extracellular matrix that accompanies disease. We have developed a multi-tiered method using Naïve Bayes modeling that is capable of predicting level of disease and clinical assessment of patients based on expression of a curated 60-gene panel that profiles inflammation and extracellular matrix production in the fibrotic disease state. Our novel modeling design, incorporating global and parametric-based methods, was highly accurate in distinguishing between severity groups, highlighting the importance of these genes in disease. We refined this gene set to a 12-gene index that can accurately identify SSc patient disease state subsets and informs knowledge of the central regulatory pathways in disease progression.


Asunto(s)
Matriz Extracelular/genética , Fibrosis , Perfilación de la Expresión Génica , Inflamación/genética , Esclerodermia Sistémica/genética , Factores de Edad , Algoritmos , Teorema de Bayes , Estudios de Casos y Controles , Fibrosis/genética , Humanos , Inflamación/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Modelos Biológicos , Piel/patología
5.
Sci Rep ; 9(1): 14050, 2019 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31575880

RESUMEN

A polarized macrophage response into inflammatory (M1) or regenerative/anti-inflammatory (M2) phenotypes is critical in host response to multiple intracellular bacterial infections. Ehrlichia is an obligate Gram-negative intracellular bacterium that causes human monocytic ehrlichiosis (HME): a febrile illness that may progress to fatal sepsis with multi-organ failure. We have shown that liver injury and Ehrlichia-induced sepsis occur due to dysregulated inflammation. Here, we investigated the contribution of macrophages to Ehrlichia-induced sepsis using murine models of mild and fatal ehrlichiosis. Lethally-infected mice showed accumulation of M1 macrophages (iNOS-positive) in the liver. In contrast, non-lethally infected mice showed polarization of M2 macrophages and their accumulation in peritoneum, but not in the liver. Predominance of M1 macrophages in lethally-infected mice was associated with expansion of IL-17-producing T, NK, and NKT cells. Consistent with the in vivo data, infection of bone marrow-derived macrophages (BMM) with lethal Ehrlichia polarized M0 macrophages into M1 phenotype under an mTORC1-dependent manner, while infection with non-lethal Ehrlichia polarized these cells into M2 types. This work highlights that mTORC1-mediated polarization of macrophages towards M1 phenotype may contribute to induction of pathogenic immune responses during fatal ehrlichiosis. Targeting mTORC1 pathway may provide a novel aproach for treatment of HME.


Asunto(s)
Ehrlichiosis/inmunología , Hígado/inmunología , Macrófagos/patología , Diana Mecanicista del Complejo 1 de la Rapamicina/fisiología , Animales , Ehrlichia , Ehrlichiosis/patología , Femenino , Hígado/patología , Macrófagos/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Endogámicos C57BL
6.
Front Pharmacol ; 9: 507, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29867506

RESUMEN

Tissue engineered scaffolds for adipose restoration/repair has significantly evolved in recent years. Patients requiring soft tissue reconstruction, caused by defects or pathology, require biomaterials that will restore void volume with new functional tissue. The gold standard of autologous fat grafting (AFG) is not a reliable option. This review focuses on the latest therapeutic strategies for the treatment of adipose tissue defects using biomolecule formulations and delivery, and specifically engineered biomaterials. Additionally, the clinical need for reliable off-the-shelf therapies, animal models, and challenges facing current technologies are discussed.

7.
PLoS One ; 13(6): e0199314, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29924864

RESUMEN

Progression of systemic scleroderma (SSc), a chronic connective tissue disease that causes a fibrotic phenotype, is highly heterogeneous amongst patients and difficult to accurately diagnose. To meet this clinical need, we developed a novel three-layer classification model, which analyses gene expression profiles from SSc skin biopsies to diagnose SSc severity. Two SSc skin biopsy microarray datasets were obtained from Gene Expression Omnibus. The skin scores obtained from the original papers were used to further categorize the data into subgroups of low (<18) and high (≥18) severity. Data was pre-processed for normalization, background correction, centering and scaling. A two-layered cross-validation scheme was employed to objectively evaluate the performance of classification models of unobserved data. Three classification models were used: support vector machine, random forest, and naive Bayes in combination with feature selection methods to improve performance accuracy. For both input datasets, random forest classifier combined with correlation-based feature selection (CFS) method and naive Bayes combined with CFS or support vector machine based recursive feature elimination method yielded the best results. Additionally, we performed a principal component analysis to show that low and high severity groups are readily separable by gene expression signatures. Ultimately, we found that our novel classification prediction model produced global gene signatures that significantly correlated with skin scores. This study represents the first report comparing the performance of various classification prediction models for gene signatures from SSc patients, using current clinical diagnostic factors. In summary, our three-classification model system is a powerful tool for elucidating gene signatures from SSc skin biopsies and can also be used to develop a prognostic gene signature for SSc and other fibrotic disorders.


Asunto(s)
Perfilación de la Expresión Génica , Modelos Genéticos , Esclerodermia Sistémica/diagnóstico , Esclerodermia Sistémica/genética , Índice de Severidad de la Enfermedad , Algoritmos , Biopsia , Femenino , Humanos , Masculino , Persona de Mediana Edad , Oncostatina M/genética , Análisis de Componente Principal , Esclerodermia Sistémica/patología , Transducción de Señal/genética
8.
Am J Pathol ; 188(7): 1676-1692, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29758183

RESUMEN

The muscle-specific ubiquitin ligase atrogin-1 (MAFbx) has been identified as a critical regulator of pathologic and physiological cardiac hypertrophy; it regulates these processes by ubiquitinating transcription factors [nuclear factor of activated T-cells and forkhead box O (FoxO) 1/3]. However, the role of atrogin-1 in regulating transcription factors in aging has not previously been described. Atrogin-1 cardiomyocyte-specific transgenic (Tg+) adult mice (α-major histocompatibility complex promoter driven) have normal cardiac function and size. Herein, we demonstrate that 18-month-old atrogin-1 Tg+ hearts exhibit significantly increased anterior wall thickness without functional impairment versus wild-type mice. Histologic analysis at 18 months revealed atrogin-1 Tg+ mice had significantly less fibrosis and significantly greater nuclei and cardiomyocyte cross-sectional analysis. Furthermore, by real-time quantitative PCR, atrogin-1 Tg+ had increased Col 6a4, 6a5, 6a6, matrix metalloproteinase 8 (Mmp8), and Mmp9 mRNA, suggesting a role for atrogin-1 in regulating collagen deposits and MMP-8 and MMP-9. Because atrogin-1 Tg+ mice exhibited significantly less collagen deposition and protein levels, enhanced Mmp8 and Mmp9 mRNA may offer one mechanism by which collagen levels are kept in check in the aged atrogin-1 Tg+ heart. In addition, atrogin-1 Tg+ hearts showed enhanced FoxO1/3 activity. The present study shows a novel link between atrogin-1-mediated regulation of FoxO1/3 activity and reduced collagen deposition and fibrosis in the aged heart. Therefore, targeting FoxO1/3 activity via the muscle-specific atrogin-1 ubiquitin ligase may offer a muscle-specific method to modulate aging-related cardiac fibrosis.


Asunto(s)
Envejecimiento , Cardiomegalia/prevención & control , Fenómenos Fisiológicos Cardiovasculares , Fibrosis/prevención & control , Proteínas Musculares/metabolismo , Proteínas Ligasas SKP Cullina F-box/metabolismo , Animales , Cardiomegalia/etiología , Cardiomegalia/metabolismo , Estudios Transversales , Fibrosis/etiología , Fibrosis/metabolismo , Ratones , Ratones Transgénicos , Proteínas Musculares/genética , Proteínas Ligasas SKP Cullina F-box/genética , Transducción de Señal
9.
Stem Cell Res Ther ; 8(1): 193, 2017 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-28874184

RESUMEN

BACKGROUND: Transplantation of mesenchymal stem cells (MSC) has been proposed to improve wound healing. However, as these cells only transiently survive in the implantation site, the mechanisms underlying this beneficial healing response are associated with restorative paracrine effects of MSC matricellular factors on resident stromal cells. However, this requires that the recipient has a robust reservoir of viable cells. Here, we examine the influence of MSCs on the behavior of cotransplanted fibroblasts, in a manner to provide augmented cellular reserve to debilitated individuals, specifically focusing on matrix remodeling following in-vivo wounding. METHODS: Using a Hylan-A dermal filler hydrogel containing collagen I and tenascin-C for delivery and increased survival of transplanted cells, we find that cotransplantation of MSCs with fibroblasts reduces scarring. RESULTS: Transplanted xenogeneic MSCs augmented fibroblast proliferation, migration, and extracellular matrix deposition critical for wound closure, and reduced inflammation following wounding. MSCs also corrected matrix remodeling by CXCR3-deficient fibroblasts which otherwise led to hypertrophic scarring. This effect was superior to MSC or fibroblast transplantation alone. CONCLUSIONS: Taken together, these data suggest that MSCs, even if eventually rejected, transplanted with fibroblasts normalize matrix regeneration during healing. The current study provides insight into cellular therapies as a viable method for antifibrotic treatment and demonstrates that even transiently engrafted cells can have a long-term impact via matrix modulation and education of other tissue cells.


Asunto(s)
Cicatriz Hipertrófica/prevención & control , Fibroblastos/trasplante , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Herida Quirúrgica/terapia , Cicatrización de Heridas , Animales , Comunicación Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Celulosa/administración & dosificación , Cicatriz Hipertrófica/metabolismo , Técnicas de Cocultivo , Combinación de Medicamentos , Matriz Extracelular/metabolismo , Matriz Extracelular/ultraestructura , Femenino , Fibroblastos/citología , Fibroblastos/inmunología , Fibroblastos/metabolismo , Eliminación de Gen , Expresión Génica , Compuestos de Hexametonio/administración & dosificación , Ácido Hialurónico/administración & dosificación , Ácido Hialurónico/análogos & derivados , Masculino , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Cultivo Primario de Células , Receptores CXCR3/deficiencia , Receptores CXCR3/genética , Piel/lesiones , Piel/metabolismo , Herida Quirúrgica/metabolismo , Herida Quirúrgica/patología , Tantalio/administración & dosificación , Trombina/administración & dosificación , Cicatrización de Heridas/efectos de los fármacos
10.
Biomaterials ; 112: 95-107, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27760399

RESUMEN

The present study tests the hypothesis that transient, early-stage shifts in macrophage polarization at the tissue-implant interface from a pro-inflammatory (M1) to an anti-inflammatory/regulatory (M2) phenotype mitigates the host inflammatory reaction against a non-degradable polypropylene mesh material and improves implant integration downstream. To address this hypothesis, a nanometer-thickness coating capable of releasing IL-4 (an M2 polarizing cytokine) from an implant surface at early stages of the host response has been developed. Results of XPS, ATR-FTIR and Alcian blue staining confirmed the presence of a uniform, conformal coating consisting of chitosan and dermatan sulfate. Immunolabeling showed uniform loading of IL-4 throughout the surface of the implant. ELISA assays revealed that the amount and release time of IL-4 from coated implants were tunable based upon the number of coating bilayers and that release followed a power law dependence profile. In-vitro macrophage culture assays showed that implants coated with IL-4 promoted polarization to an M2 phenotype, demonstrating maintenance of IL-4 bioactivity following processing and sterilization. Finally, in-vivo studies showed that mice with IL-4 coated implants had increased percentages of M2 macrophages and decreased percentages of M1 macrophages at the tissue-implant interface during early stages of the host response. These changes were correlated with diminished formation of fibrotic capsule surrounding the implant and improved tissue integration downstream. The results of this study demonstrate a versatile cytokine delivery system for shifting early-stage macrophage polarization at the tissue-implant interface of a non-degradable material and suggest that modulation of the innate immune reaction at early stages of the host response may represent a preferred strategy for promoting biomaterial integration and success.


Asunto(s)
Interfase Hueso-Implante , Materiales Biocompatibles Revestidos/síntesis química , Interleucina-4/administración & dosificación , Interleucina-4/química , Macrófagos/citología , Macrófagos/inmunología , Prótesis e Implantes , Animales , Polaridad Celular/efectos de los fármacos , Polaridad Celular/inmunología , Células Cultivadas , Implantes de Medicamentos/administración & dosificación , Implantes de Medicamentos/química , Femenino , Interleucina-4/inmunología , Macrófagos/efectos de los fármacos , Ensayo de Materiales , Ratones , Ratones Endogámicos C57BL
11.
Surg Infect (Larchmt) ; 18(2): 119-127, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27788042

RESUMEN

BACKGROUND: Infection is the most common complication in burn-injured patients and is believed to contribute to the hypertrophic scarring frequently observed in such injury. Pseudomonas aeruginosa is a common pathogen in burn wound infection. We examined the effect of local probiotic therapy with Lactobacillus plantarum on the severity of the scarring following burn wounding and infection with P. aeruginosa in a rabbit model. METHODS: Full-thickness burn wounds were inoculated with control vehicle or L. plantarum; wounds were then challenged with bioluminescent P. aeruginosa. The time course of the ensuing infection was monitored by quantification of the emitted light. After allowing wounds to contract to near completion, they were harvested and analyzed for markers of scar formation. RESULTS: Application of L. plantarum curtailed both the severity and the length of the pseudomonal infection. Probiotic therapy significantly reduced both Type I collagen mRNA concentrations and total collagen protein accumulation in infected wounds, consistent with reduced scarring. Surprisingly, the probiotic showed a nearly equivalent effect in uninfected wounds. Masson's trichrome staining confirmed these findings histologically. CONCLUSIONS: Lactobacillus plantarum shows exciting potential as a therapeutic agent to both counteract burn wound infection and to alleviate scarring even in the absence of infection.


Asunto(s)
Quemaduras/fisiopatología , Cicatriz/tratamiento farmacológico , Lactobacillus plantarum , Probióticos/uso terapéutico , Cicatrización de Heridas/efectos de los fármacos , Animales , Cicatriz/prevención & control , Colágeno/metabolismo , Modelos Animales de Enfermedad , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/prevención & control , Pseudomonas aeruginosa , Conejos , Infección de Heridas/tratamiento farmacológico , Infección de Heridas/microbiología , Infección de Heridas/prevención & control
12.
Cell Transplant ; 26(1): 103-113, 2017 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-27452449

RESUMEN

Mesenchymal stem cells (MSCs) remain of great interest in regenerative medicine because of their ability to home to sites of injury, differentiate into a variety of relevant lineages, and modulate inflammation and angiogenesis through paracrine activity. Many studies have found that despite the promise of MSC therapy, cell survival upon implant is highly limited and greatly reduces the therapeutic utility of MSCs. The matrikine tenascin C, a protein expressed often at the edges of a healing wound, contains unique EGF-like repeats that are able to bind EGFR at low affinities and induce downstream prosurvival signaling without inducing receptor internalization. In this study, we utilized tenascin C in a collagen/GAG-based polymer (TPolymer) that has been shown to be beneficial for skin wound healing, incorporating human MSCs into the polymer prior to application to mouse punch biopsy wound beds. We found that the TPolymer was able to promote MSC survival for 21 days in vivo, leading to associated improvements in wound healing such as dermal maturation and collagen content. This was most marked in a model of hypertrophic scarring, in which the scar formation was limited. This approach also reduced the inflammatory response in the wound bed, limiting CD3e+ cell invasion by approximately 50% in the early wound-healing process, while increasing the numbers of endothelial cells during the first week of wound healing as well. Ultimately, this matrikine-based approach to improving MSC survival may be of great use across a variety of cell therapies utilizing matrices as delivery vehicles for cells.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas/métodos , Polímeros/química , Tenascina/química , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Colágeno/metabolismo , Matriz Extracelular/metabolismo , Femenino , Citometría de Flujo , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Polietilenglicoles/química , Piel/metabolismo , Piel/patología , Enfermedades de la Piel/metabolismo , Enfermedades de la Piel/patología , Tenascina/farmacología , Cicatrización de Heridas/efectos de los fármacos
13.
Am J Pathol ; 186(8): 1989-2007, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27321750

RESUMEN

The Bcl2-associated anthanogene (BAG) 3 protein is a member of the BAG family of cochaperones, which supports multiple critical cellular processes, including critical structural roles supporting desmin and interactions with heat shock proteins and ubiquitin ligases intimately involved in protein quality control. The missense mutation P209L in exon 3 results in a primarily cardiac phenotype leading to skeletal muscle and cardiac complications. At least 10 other Bag3 mutations have been reported, nine resulting in a dilated cardiomyopathy for which no specific therapy is available. We generated αMHC-human Bag3 P209L transgenic mice and characterized the progressive cardiac phenotype in vivo to investigate its utility in modeling human disease, understand the underlying molecular mechanisms, and identify potential therapeutic targets. We identified a progressive heart failure by echocardiography and Doppler analysis and the presence of pre-amyloid oligomers at 1 year. Paralleling the pathogenesis of neurodegenerative diseases (eg, Parkinson disease), pre-amyloid oligomers-associated alterations in cardiac mitochondrial dynamics, haploinsufficiency of wild-type BAG3, and activation of p38 signaling were identified. Unexpectedly, increased numbers of activated cardiac fibroblasts were identified in Bag3 P209L Tg+ hearts without increased fibrosis. Together, these findings point to a previously undescribed therapeutic target that may have application to mutation-induced myofibrillar myopathies as well as other common causes of heart failure that commonly harbor misfolded proteins.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Reguladoras de la Apoptosis/genética , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/fisiopatología , Miocitos Cardíacos/patología , Animales , Western Blotting , Ecocardiografía , Técnica del Anticuerpo Fluorescente , Haploinsuficiencia , Insuficiencia Cardíaca/patología , Humanos , Etiquetado Corte-Fin in Situ , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Transgénicos , Mitocondrias/patología , Mutación Missense , Reacción en Cadena en Tiempo Real de la Polimerasa
14.
Wound Repair Regen ; 24(2): 204-14, 2016 03.
Artículo en Inglés | MEDLINE | ID: mdl-26969517

RESUMEN

Pericytes have generally been considered in the context of stabilizing vessels, ensuring the blood barriers, and regulating the flow through capillaries. However, new reports suggest that pericytes may function at critical times to either drive healing with minimal scarring or, perversely, contribute to fibrosis and ongoing scar formation. Beneficially, pericytes probably drive much of the vascular involution that occurs during the transition from the regenerative to the resolution phases of healing. Pathologically, pericytes can assume a fibrotic phenotype and promote scarring. This perspective will discuss pericyte involvement in wound repair and the relationship pericytes form with the parenchymal cells of the skin. We will further evaluate the role pericytes may have in disease progression in relation to chronic wounds and fibrosis.


Asunto(s)
Pericitos/fisiología , Medicina Regenerativa , Cicatrización de Heridas/fisiología , Animales , Enfermedad Crónica/terapia , Cicatriz/prevención & control , Medicina Basada en la Evidencia , Fibrosis/patología , Fibrosis/terapia , Humanos , Pericitos/citología , Medicina Regenerativa/tendencias , Piel/patología , Heridas y Lesiones/patología , Heridas y Lesiones/terapia
15.
Matrix Biol ; 49: 25-36, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26278492

RESUMEN

The process of repair of wounded skin involves intricate orchestration not only between the epidermal and dermal compartments but also between the resident and immigrant cells and the local microenvironment. Only now are we beginning to appreciate the complex roles played by the matrix in directing the outcome of the repair processes, and how this impacts the signals from the various cells. Recent findings speak of dynamic and reciprocal interactions that occurs among the matrix, growth factors, and cells that underlies this integrated process. Further confounding this integration are the physiologic and pathologic situations that directly alter the matrix to impart at least part of the dysrepair that occurs. These topics will be discussed with a call for innovative model systems of direct relevance to the human situation.


Asunto(s)
Matriz Extracelular/metabolismo , Piel/lesiones , Cicatrización de Heridas , Animales , Microambiente Celular , Modelos Animales de Enfermedad , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo
16.
Cardiovasc Diabetol ; 14: 97, 2015 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-26242235

RESUMEN

BACKGROUND: In diabetes mellitus the morbidity and mortality of cardiovascular disease is increased and represents an important independent mechanism by which heart disease is exacerbated. The pathogenesis of diabetic cardiomyopathy involves the enhanced activation of PPAR transcription factors, including PPARα, and to a lesser degree PPARß and PPARγ1. How these transcription factors are regulated in the heart is largely unknown. Recent studies have described post-translational ubiquitination of PPARs as ways in which PPAR activity is inhibited in cancer. However, specific mechanisms in the heart have not previously been described. Recent studies have implicated the muscle-specific ubiquitin ligase muscle ring finger-2 (MuRF2) in inhibiting the nuclear transcription factor SRF. Initial studies of MuRF2-/- hearts revealed enhanced PPAR activity, leading to the hypothesis that MuRF2 regulates PPAR activity by post-translational ubiquitination. METHODS: MuRF2-/- mice were challenged with a 26-week 60% fat diet designed to simulate obesity-mediated insulin resistance and diabetic cardiomyopathy. Mice were followed by conscious echocardiography, blood glucose, tissue triglyceride, glycogen levels, immunoblot analysis of intracellular signaling, heart and skeletal muscle morphometrics, and PPARα, PPARß, and PPARγ1-regulated mRNA expression. RESULTS: MuRF2 protein levels increase ~20% during the development of diabetic cardiomyopathy induced by high fat diet. Compared to littermate wildtype hearts, MuRF2-/- hearts exhibit an exaggerated diabetic cardiomyopathy, characterized by an early onset systolic dysfunction, larger left ventricular mass, and higher heart weight. MuRF2-/- hearts had significantly increased PPARα- and PPARγ1-regulated gene expression by RT-qPCR, consistent with MuRF2's regulation of these transcription factors in vivo. Mechanistically, MuRF2 mono-ubiquitinated PPARα and PPARγ1 in vitro, consistent with its non-degradatory role in diabetic cardiomyopathy. However, increasing MuRF2:PPARγ1 (>5:1) beyond physiological levels drove poly-ubiquitin-mediated degradation of PPARγ1 in vitro, indicating large MuRF2 increases may lead to PPAR degradation if found in other disease states. CONCLUSIONS: Mutations in MuRF2 have been described to contribute to the severity of familial hypertrophic cardiomyopathy. The present study suggests that the lack of MuRF2, as found in these patients, can result in an exaggerated diabetic cardiomyopathy. These studies also identify MuRF2 as the first ubiquitin ligase to regulate cardiac PPARα and PPARγ1 activities in vivo via post-translational modification without degradation.


Asunto(s)
Cardiomiopatías/prevención & control , Dieta Alta en Grasa , Proteínas Musculares/metabolismo , Miocardio/enzimología , Obesidad/etiología , PPAR gamma/metabolismo , Aumento de Peso , Animales , Cardiomiopatías/enzimología , Cardiomiopatías/etiología , Cardiomiopatías/genética , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Genotipo , Resistencia a la Insulina , Masculino , Ratones Noqueados , Proteínas Musculares/deficiencia , Proteínas Musculares/genética , Obesidad/enzimología , Obesidad/genética , PPAR gamma/genética , Fenotipo , ARN Mensajero/metabolismo , Transducción de Señal , Factores de Tiempo , Ubiquitinación
17.
BMC Endocr Disord ; 15: 36, 2015 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-26215257

RESUMEN

BACKGROUND: The pathogenesis of diabetic cardiomyopathy (DCM) involves the enhanced activation of peroxisome proliferator activating receptor (PPAR) transcription factors, including the most prominent isoform in the heart, PPARα. In cancer cells and adipocytes, post-translational modification of PPARs have been identified, including ligand-dependent degradation of PPARs by specific ubiquitin ligases. However, the regulation of PPARs in cardiomyocytes and heart have not previously been identified. We recently identified that muscle ring finger-1 (MuRF1) and MuRF2 differentially inhibit PPAR activities by mono-ubiquitination, leading to the hypothesis that MuRF3 may regulate PPAR activity in vivo to regulate DCM. METHODS: MuRF3-/- mice were challenged with 26 weeks 60% high fat diet to induce insulin resistance and DCM. Conscious echocardiography, blood glucose, tissue triglyceride, glycogen levels, immunoblot analysis of intracellular signaling, heart and skeletal muscle morphometrics, and PPARα, PPARß, and PPARγ1 activities were assayed. RESULTS: MuRF3-/- mice exhibited a premature systolic heart failure by 6 weeks high fat diet (vs. 12 weeks in MuRF3+/+). MuRF3-/- mice weighed significantly less than sibling-matched wildtype mice after 26 weeks HFD. These differences may be largely due to resistance to fat accumulation, as MRI analysis revealed MuRF3-/- mice had significantly less fat mass, but not lean body mass. In vitro ubiquitination assays identified MuRF3 mono-ubiquitinated PPARα and PPARγ1, but not PPARß. CONCLUSIONS: These findings suggest that MuRF3 helps stabilize cardiac PPARα and PPARγ1 in vivo to support resistance to the development of DCM. MuRF3 also plays an unexpected role in regulating fat storage despite being found only in striated muscle.


Asunto(s)
Cardiomiopatías Diabéticas/genética , Dieta Alta en Grasa/efectos adversos , Insuficiencia Cardíaca Sistólica/genética , Proteínas Musculares/genética , Miocitos Cardíacos/metabolismo , Tejido Adiposo , Animales , Composición Corporal , Peso Corporal , Cardiomiopatías Diabéticas/etiología , Cardiomiopatías Diabéticas/metabolismo , Insuficiencia Cardíaca Sistólica/etiología , Insuficiencia Cardíaca Sistólica/metabolismo , Técnicas In Vitro , Ratones , Ratones Noqueados , Proteínas Musculares/metabolismo , PPAR alfa/metabolismo , PPAR gamma/metabolismo , PPAR-beta/metabolismo , Ubiquitinación
18.
J Mol Cell Cardiol ; 77: 86-101, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25257914

RESUMEN

The cell-permeant peptide inhibitor of MAPKAP kinase 2 (MK2), MMI-0100, inhibits MK2 and downstream fibrosis and inflammation. Recent studies have demonstrated that MMI-0100 reduces intimal hyperplasia in a mouse vein graft model, pulmonary fibrosis in a murine bleomycin-induced model and development of adhesions in conjunction with abdominal surgery. MK2 is critical to the pathogenesis of ischemic heart injury as MK2(-/-) mice are resistant to ischemic remodeling. Therefore, we tested the hypothesis that inhibiting MK2 with MMI-0100 would protect the heart after acute myocardial infarction (AMI) in vivo. AMI was induced by placing a permanent LAD coronary ligation. When MMI-0100 peptide was given 30 min after permanent LAD coronary artery ligation, the resulting fibrosis was reduced/prevented ~50% at a 2 week time point, with a corresponding improvement in cardiac function and decrease in left ventricular dilation. In cultured cardiomyocytes and fibroblasts, MMI-0100 inhibited MK2 to reduce cardiomyocyte caspase 3/7 activity, while enhancing primary cardiac fibroblast caspase 3/7 activity, which may explain MMI-0100's salvage of cardiac function and anti-fibrotic effects in vivo. These findings suggest that therapeutic inhibition of MK2 after acute MI, using rationally-designed cell-permeant peptides, inhibits cardiac fibrosis and maintains cardiac function by mechanisms that involve inhibiting cardiomyocyte apoptosis, while enhancing primary cardiac fibroblast cell death.


Asunto(s)
Fibroblastos/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Infarto del Miocardio/tratamiento farmacológico , Miocitos Cardíacos/efectos de los fármacos , Péptidos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Animales , Apoptosis , Línea Celular , Fibroblastos/enzimología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones Endogámicos C57BL , Miocardio/patología , Miocitos Cardíacos/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Fibrosis Pulmonar/prevención & control
19.
Tissue Eng Part A ; 19(17-18): 1972-83, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23541003

RESUMEN

Multipotential stromal cells/mesenchymal stem cells (MSCs) are attractive candidates for regenerative therapy due to the ability of these cells to differentiate and positively influence neighboring cells. However, on implantation for wound reconstruction, these cells are lost as they are challenged by nonspecific inflammation signals generated in the wound environment and in response to any implanted foreign body. We have previously shown that sustained and surface-restricted epidermal growth factor receptor (EGFR) signaling by a tethered form of its prototypal ligand EGF enhances survival of MSC in the presence of death cytokines such as FasL, serum deprivation, and low oxygen in vitro. This was proposed to be due to the plasma membrane restriction of EGFR signaling. Interestingly, during wound repair, an extracellular matrix (ECM) component Tenascin-C (TNC) containing EGF-like repeats (EGFL) and fibronectin-like repeats (FNL) is upregulated. A few of the 14 EGFL on each of the 6 arms, especially the 14th, bind as low-affinity/high-avidity ligands to EGFR causing sustained surface-restricted EGFR signaling. We queried whether signaling by this physiologically relevant EGFR matrikine also protects MSCs from FasL-induced death. MSCs grown on TNC and Collagen I (as TNC by itself is antiadhesive) displayed a survival advantage in the presence of FasL. TNC neither sequestered nor neutralized FasL; rather, the effects of survival were via cell signaling. This survival was dependent on TNC activating EGFR and downstream pathways of Erk and Akt through EGFL; to a much lesser extent, the FNL of TNC also contributed to survival. Taken together, these results suggest that providing MSCs with a nonimmunogenic naturally occurring ECM moiety such as TNC enhances their survival in the presence of death factors, and this advantage occurs via signaling through EGFR primarily and integrins only to a minor extent. This matrix component is proposed to supplement MSC delivery on the scaffolds to provide a survival advantage against death upon in vivo implantation.


Asunto(s)
Citocinas/farmacología , Proteína Ligando Fas/farmacología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Multipotentes/citología , Células Madre Multipotentes/efectos de los fármacos , Tenascina/farmacología , Apoptosis/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Receptores ErbB/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos
20.
Cell Transplant ; 21(5): 919-31, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22236446

RESUMEN

In skin, the regeneration of the ontogenically distinct mesenchymal and epithelial compartments must proceed in a coordinated manner orchestrated by extracellular signaling networks. We have recently found that the switch from regeneration to remodeling during repair is modulated by chemokines that bind CXCR3 receptor. If this signaling is disrupted wounds continue to be active, resulting in a chronic hypercellular and hypertrophic state characterized by an immature matrix composition. As healing is masterminded in large part by fibroblasts and their synthesis of the extracellular matrix, the question arose as to whether this ongoing scarring can be modulated by transplanted fibroblasts. We examined wounds in the CXCR3-/- mouse scarring model. These wounds exhibited a significant delay in healing in all areas compared to young and aged wild-type mice. Full-thickness wounds were transplanted with fibroblasts derived from newborn CXCR3-/- or wild-type mice. The transplanted fibroblasts were labeled with fluorescent dye (CM-DiI) and suspended in hyaluronic acid gel; by 30 days, these transplanted cells comprised some 30% of the dermal stromal cells regardless of the host or source of transplanted cells. Wild-type fibroblasts transplanted into CXCR3-/- mice wounds reversed the delay and dysfunction previously seen in CXCR3-/- wounds; this correction was not noted with transplanted CXCR3-/- fibroblasts. Additionally, transplant of CXCR3-/- cells into wounds in wild-type animals did not adversely affect those wounds. The transplanted fibroblasts exhibited strong survival and migration patterns and led to an increase in tensile strength. Expression of matrix proteins and collagen in CXCR3-/- wounds transplanted with wild-type fibroblasts resembled normal wild-type healing, and the wound matrix in wild-type mice transplanted with CXCR3-/- cells also presented a mature matrix. These suggest that the major determinant of healing versus scarring lies with the nature of the matrix. These findings have intriguing implications for rational cellular interventions aimed at promoting wound healing via cell therapy.


Asunto(s)
Cicatriz/prevención & control , Fibroblastos/trasplante , Receptores CXCR3/metabolismo , Cicatrización de Heridas , Actinas/metabolismo , Animales , Animales Recién Nacidos , Colágeno/metabolismo , Fibroblastos/citología , Colorantes Fluorescentes/química , Ácido Hialurónico/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Receptores CXCR3/deficiencia , Receptores CXCR3/genética , Transducción de Señal , Piel/patología , Resistencia a la Tracción , Factores de Tiempo , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...