Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
J Proteome Res ; 20(1): 888-894, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33251806

RESUMO

Skeletal muscle myofibers have differential protein expression resulting in functionally distinct slow- and fast-twitch types. While certain protein classes are well-characterized, the depth of all proteins involved in this process is unknown. We utilized the Human Protein Atlas (HPA) and the HPASubC tool to classify mosaic expression patterns of staining across 49,600 unique tissue microarray (TMA) images using a visual proteomic approach. We identified 2164 proteins with potential mosaic expression, of which 1605 were categorized as "likely" or "real." This list included both well-known fiber-type-specific and novel proteins. A comparison of the 1605 mosaic proteins with a mass spectrometry (MS)-derived proteomic dataset of single human muscle fibers led to the assignment of 111 proteins to fiber types. We additionally used a multiplexed immunohistochemistry approach, a multiplexed RNA-ISH approach, and STRING v11 to further assign or suggest fiber types of newly characterized mosaic proteins. This visual proteomic analysis of mature skeletal muscle myofibers greatly expands the known repertoire of twitch-type-specific proteins.


Assuntos
Fibras Musculares de Contração Lenta , Doenças Musculares , Humanos , Fibras Musculares de Contração Rápida , Músculo Esquelético , Proteômica
2.
Am J Hum Genet ; 94(6): 854-69, 2014 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-24857694

RESUMO

QT interval variation is assumed to arise from variation in repolarization as evidenced from rare Na- and K-channel mutations in Mendelian QT prolongation syndromes. However, in the general population, common noncoding variants at a chromosome 1q locus are the most common genetic regulators of QT interval variation. In this study, we use multiple human genetic, molecular genetic, and cellular assays to identify a functional variant underlying trait association: a noncoding polymorphism (rs7539120) that maps within an enhancer of NOS1AP and affects cardiac function by increasing NOS1AP transcript expression. We further localized NOS1AP to cardiomyocyte intercalated discs (IDs) and demonstrate that overexpression of NOS1AP in cardiomyocytes leads to altered cellular electrophysiology. We advance the hypothesis that NOS1AP affects cardiac electrical conductance and coupling and thereby regulates the QT interval through propagation defects. As further evidence of an important role for propagation variation affecting QT interval in humans, we show that common polymorphisms mapping near a specific set of 170 genes encoding ID proteins are significantly enriched for association with the QT interval, as compared to genome-wide markers. These results suggest that focused studies of proteins within the cardiomyocyte ID are likely to provide insights into QT prolongation and its associated disorders.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Síndrome do QT Longo/genética , Miócitos Cardíacos/metabolismo , Locos de Características Quantitativas , Animais , Estudos de Coortes , Eletrocardiografia , Regulação da Expressão Gênica , Estudo de Associação Genômica Ampla , Genótipo , Células HEK293 , Humanos , Lentivirus/genética , Camundongos , Fenótipo , Polimorfismo de Nucleotídeo Único , Peixe-Zebra/embriologia , Peixe-Zebra/genética
3.
J Immunol ; 192(4): 1806-14, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24403532

RESUMO

Sepsis is a major cause of mortality, and dysregulation of the immune response plays a central role in this syndrome. H2S, a recently discovered gaso-transmitter, is endogenously generated by many cell types, regulating a number of physiologic processes and pathophysiologic conditions. We report that H2S increased survival after experimental sepsis induced by cecal ligation and puncture (CLP) in mice. Exogenous H2S decreased the systemic inflammatory response, reduced apoptosis in the spleen, and accelerated bacterial eradication. We found that C/EBP homologous protein 10 (CHOP), a mediator of the endoplasmic reticulum stress response, was elevated in several organs after CLP, and its expression was inhibited by H2S treatment. Using CHOP-knockout (KO) mice, we demonstrated for the first time, to our knowledge, that genetic deletion of Chop increased survival after LPS injection or CLP. CHOP-KO mice displayed diminished splenic caspase-3 activation and apoptosis, decreased cytokine production, and augmented bacterial clearance. Furthermore, septic CHOP-KO mice treated with H2S showed no additive survival benefit compared with septic CHOP-KO mice. Finally, we showed that H2S inhibited CHOP expression in macrophages by a mechanism involving Nrf2 activation. In conclusion, our findings show a protective effect of H2S treatment afforded, at least partially, by inhibition of CHOP expression. The data reveal a major negative role for the transcription factor CHOP in overall survival during sepsis and suggest a new target for clinical intervention, as well potential strategies for treatment.


Assuntos
Bactérias/imunologia , Sulfeto de Hidrogênio/metabolismo , Sepse/metabolismo , Fator de Transcrição CHOP/antagonistas & inibidores , Animais , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Ceco/cirurgia , Citocinas/biossíntese , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Ativação Enzimática , Lipopolissacarídeos , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 2 Relacionado a NF-E2/metabolismo , Sepse/tratamento farmacológico , Baço/efeitos dos fármacos , Sobrevida , Fator de Transcrição CHOP/biossíntese , Fator de Transcrição CHOP/genética
4.
Mol Ther ; 22(4): 752-61, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24434934

RESUMO

Fragile skin, susceptible to decubitus ulcers and incidental trauma, is a problem particularly for the elderly and for those with spinal cord injury. Here, we present a simple approach to strengthen the skin by the topical delivery of keratinocyte growth factor-1 (KGF-1) DNA. In initial feasibility studies with the novel minimalized, antibiotic-free DNA expression vector, NTC8385-VA1, the reporter genes luciferase and enhanced green fluorescent protein were delivered. Transfection was documented when luciferase expression significantly increased after transfection. Microscopic imaging of enhanced green fluorescent protein-transfected skin showed green fluorescence in hair follicles, hair shafts, and dermal and superficial epithelial cells. With KGF-1 transfection, KGF-1 mRNA level and protein production were documented with quantitative reverse transcriptase-polymerase chain reaction and immunohistochemistry, respectively. Epithelial thickness of the transfected skin in the KGF group was significantly increased compared with the control vector group (26 ± 2 versus 16 ± 4 µm) at 48 hours (P = 0.045). Dermal thickness tended to be increased in the KGF group (255 ± 36 versus 162 ± 16 µm) at 120 hours (P = 0.057). Biomechanical assessment showed that the KGF-1-treated skin was significantly stronger than control vector-transfected skin. These findings indicate that topically delivered KGF-1 DNA plasmid can increase epithelial thickness and strength, demonstrating the potential of this approach to restore compromised skin.


Assuntos
Fator 7 de Crescimento de Fibroblastos/genética , Técnicas de Transferência de Genes , Terapia Genética , Anormalidades da Pele/genética , Administração Tópica , Animais , DNA/administração & dosagem , DNA/genética , Fator 7 de Crescimento de Fibroblastos/administração & dosagem , Humanos , Camundongos , Plasmídeos/administração & dosagem , Anormalidades da Pele/terapia , Cicatrização/genética
5.
Proc Natl Acad Sci U S A ; 109(26): 10504-9, 2012 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-22699503

RESUMO

Infarction occurs when myocardial perfusion is interrupted for prolonged periods of time. Short episodes of ischemia and reperfusion protect against tissue injury when the heart is subjected to a subsequent prolonged ischemic episode, a phenomenon known as ischemic preconditioning (IPC). Hypoxia-inducible factor 1 (HIF-1) is a transcription factor that mediates adaptive responses to hypoxia/ischemia and is required for IPC. In this study, we performed a cellular and molecular characterization of the role of HIF-1 in IPC. We analyzed mice with knockout of HIF-1α or HIF-1ß in Tie2(+) lineage cells, which include bone marrow (BM) and vascular endothelial cells, compared with control littermates. Hearts were subjected to 30 min of ischemia and 120 min of reperfusion, either as ex vivo Langendorff preparations or by in situ occlusion of the left anterior descending artery. The IPC stimulus consisted of two cycles of 5-min ischemia and 5-min reperfusion. Mice lacking HIF-1α or HIF-1ß in Tie2(+) lineage cells showed complete absence of protection induced by IPC, whereas significant protection was induced by adenosine infusion. Treatment of mice with a HIF-1 inhibitor (digoxin or acriflavine) 4 h before Langendorff perfusion resulted in loss of IPC, as did administration of acriflavine directly into the perfusate immediately before IPC. We conclude that HIF-1 activity in endothelial cells is required for acute IPC. Expression and dimerization of the HIF-1α and HIF-1ß subunits is required, suggesting that the heterodimer is functioning as a transcriptional activator, despite the acute nature of the response.


Assuntos
Fator 1 Induzível por Hipóxia/genética , Precondicionamento Isquêmico , Transcrição Gênica , Animais , Sequência de Bases , Linhagem da Célula , Primers do DNA , Camundongos , Camundongos Knockout
6.
Proc Natl Acad Sci U S A ; 109(14): E841-50, 2012 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-22403061

RESUMO

Chronic systemic hypertension causes cardiac pressure overload leading to increased myocardial O(2) consumption. Hypoxia-inducible factor 1 (HIF-1) is a master regulator of O(2) homeostasis. Mouse embryos lacking expression of the O(2)-regulated HIF-1α subunit die at midgestation with severe cardiac malformations and vascular regression. Here we report that Hif1a(f/f);Tie2-Cre conditional knockout mice, which lack HIF-1α expression only in Tie2(+) lineage cells, develop normally, but when subjected to pressure overload induced by transaortic constriction (TAC), they manifest rapid cardiac decompensation, which is accompanied by excess cardiac fibrosis and myocardial hypertrophy, decreased myocardial capillary density, increased myocardial hypoxia and apoptosis, and increased TGF-ß signaling through both canonical and noncanonical pathways that activate SMAD2/3 and ERK1/2, respectively, within endothelial cells of cardiac blood vessels. TAC also induces dilatation of the proximal aorta through enhanced TGF-ß signaling in Hif1a(f/f);Tie2-Cre mice. Inhibition of TGF-ß signaling by treatment with neutralizing antibody or pharmacologic inhibition of MEK-ERK signaling prevented TAC-induced contractile dysfunction and pathological remodeling. Thus, HIF-1 plays a critical protective role in the adaptation of the heart and aorta to pressure overload by negatively regulating TGF-ß signaling in endothelial cells. Treatment of wild-type mice with digoxin, which inhibits HIF-1α synthesis, resulted in rapid cardiac failure after TAC. Although digoxin has been used for decades as an inotropic agent to treat heart failure, it does not improve survival, suggesting that the countertherapeutic effects of digoxin observed in the TAC mouse model may have clinical relevance.


Assuntos
Aorta/fisiopatologia , Endotélio Vascular/metabolismo , Coração/fisiopatologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/antagonistas & inibidores , Animais , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Camundongos , Camundongos Knockout , Pressão , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo
7.
Circ Res ; 110(12): 1596-603, 2012 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-22518031

RESUMO

RATIONALE: Mitochondria are semiautonomous cellular organelles with their own genome, which not only supply energy but also participate in cell death pathways. MicroRNAs (miRNAs) are usually 19 to 25 nt long, noncoding RNAs, involved in posttranscriptional gene regulation by binding to the 3'-untranslated regions of target mRNA, which impact on diverse cellular processes. OBJECTIVE: To determine if nuclear miRNAs translocate into the mitochondria and regulate mitochondrial function with possible pathophysiological implications in cardiac myocytes. METHODS AND RESULTS: We find that miR-181c is encoded in the nucleus, assembled in the cytoplasm, and finally translocated into the mitochondria of cardiac myocytes. Immunoprecipitation of Argonaute 2 from the mitochondrial fraction indicates binding of cytochrome c oxidase subunit 1 (mt-COX1) mRNA from the mitochondrial genome with miR-181c. Also, a luciferase reporter construct shows that mi-181c binds to the 3'UTR of mt-COX1. To study whether miR-181c regulates mt-COX1, we overexpressed precursor miR-181c (or a scrambled sequence) in primary cultures of neonatal rat ventricular myocytes. Overexpression of miR-181c did not change mt-COX1 mRNA but significantly decreased mt-COX1 protein, suggesting that miR-181c is primarily a translational regulator of mt-COX1. In addition to altering mt-COX1, overexpression of miR-181c results in increased mt-COX2 mRNA and protein content, with an increase in both mitochondrial respiration and reactive oxygen species generation in neonatal rat ventricular myocytes. Thus, our data show for the first time that miR-181c can enter and target the mitochondrial genome, ultimately causing electron transport chain complex IV remodeling and mitochondrial dysfunction. CONCLUSIONS: Nuclear miR-181c translocates into the mitochondria and regulates mitochondrial genome expression. This unique observation may open a new dimension to our understanding of mitochondrial dynamics and the role of miRNA in mitochondrial dysfunction.


Assuntos
Núcleo Celular/genética , Núcleo Celular/metabolismo , Genoma Mitocondrial/genética , MicroRNAs/fisiologia , Mitocôndrias Cardíacas/genética , Mitocôndrias Cardíacas/metabolismo , Miócitos Cardíacos/fisiologia , Animais , Células Cultivadas , Masculino , Ratos , Ratos Sprague-Dawley
8.
Proc Natl Acad Sci U S A ; 108(52): 20976-81, 2011 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-22171002

RESUMO

Neovascularization is a critical determinant of wound-healing outcomes for deep burn injuries. We hypothesize that dextran-based hydrogels can serve as instructive scaffolds to promote neovascularization and skin regeneration in third-degree burn wounds. Dextran hydrogels are soft and pliable, offering opportunities to improve the management of burn wound treatment. We first developed a procedure to treat burn wounds on mice with dextran hydrogels. In this procedure, we followed clinical practice of wound excision to remove full-thickness burned skin, and then covered the wound with the dextran hydrogel and a dressing layer. Our procedure allows the hydrogel to remain intact and securely in place during the entire healing period, thus offering opportunities to simplify the management of burn wound treatment. A 3-week comparative study indicated that dextran hydrogel promoted dermal regeneration with complete skin appendages. The hydrogel scaffold facilitated early inflammatory cell infiltration that led to its rapid degradation, promoting the infiltration of angiogenic cells into the healing wounds. Endothelial cells homed into the hydrogel scaffolds to enable neovascularization by day 7, resulting in an increased blood flow significantly greater than treated and untreated controls. By day 21, burn wounds treated with hydrogel developed a mature epithelial structure with hair follicles and sebaceous glands. After 5 weeks of treatment, the hydrogel scaffolds promoted new hair growth and epidermal morphology and thickness similar to normal mouse skin. Collectively, our evidence shows that customized dextran-based hydrogel alone, with no additional growth factors, cytokines, or cells, promoted remarkable neovascularization and skin regeneration and may lead to novel treatments for dermal wounds.


Assuntos
Queimaduras/terapia , Dextranos/uso terapêutico , Hidrogel de Polietilenoglicol-Dimetacrilato/uso terapêutico , Neovascularização Fisiológica/fisiologia , Fenômenos Fisiológicos da Pele , Alicerces Teciduais , Cicatrização/fisiologia , Análise de Variância , Animais , Camundongos , Microscopia Eletrônica de Varredura
9.
Rheumatology (Oxford) ; 52(8): 1448-53, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23598443

RESUMO

OBJECTIVE: Cardiac neonatal lupus (cardiac-NL), initiated by surface binding of anti-Ro60 autoantibodies to apoptotic cardiocytes during development, activates the urokinase plasminogen activator/urokinase plasminogen activator receptor (uPA/uPAR) system. Subsequent accumulation of apoptotic cells and plasmin generation facilitates increased binding of anti-Ro60 by disrupting and cleaving circulating ß2-glycoprotein I (ß2GPI) thereby eliminating its protective effect. The association of soluble levels of components of the uPA/uPAR system with cardiac-NL was examined. METHODS: Levels of the uPA/uPAR system were assessed by ELISA in cord blood and immunohistological evaluation of autopsies. RESULTS: uPA, uPAR and plasminogen levels were each significantly higher in cord blood from cardiac-NL (n = 35) compared with non-cardiac-NL (n = 26) anti-Ro-exposed neonates: 3.3 ± 0.1 vs 1.9 ± 0.05 ng/ml (P < 0.0001), 6.6 ± 0.3 vs 2.1 ± 0.2 ng/ml (P < 0.0001) and 435 ± 34 vs 220 ± 19 ng/ml (P < 0.0001), respectively. In three twin pairs discordant for cardiac-NL, the twin with cardiac-NL had higher levels of uPA, uPAR and plasminogen than the unaffected twin (3.1 ± 0.1 vs 1.9 ± 0.05 ng/ml; P = 0.0086, 6.2 ± 1.4 vs 2.2 ± 0.7 ng/ml; P = 0.147 and 412 ± 61 vs 260 ± 27 ng/ml; P = 0.152, respectively). Immunohistological evaluation of three hearts from fetuses dying with cardiac-NL revealed macrophages and giant cells expressing uPA and plasminogen in the septal region. CONCLUSION: Increased soluble uPA, uPAR and plasminogen in cord blood and expression in affected tissue of fetuses with cardiac-NL supports the hypothesis that fetal cardiac injury is in part mediated by plasmin generation initiated by anti-Ro binding to the apoptotic cardiocyte.


Assuntos
Sangue Fetal/imunologia , Fibrinolisina/imunologia , Cardiopatias/imunologia , Lúpus Eritematoso Sistêmico/congênito , Receptores de Ativador de Plasminogênio Tipo Uroquinase/imunologia , Ribonucleoproteínas/imunologia , Anticorpos Antinucleares/imunologia , Anticorpos Antinucleares/metabolismo , Biomarcadores , Estudos de Casos e Controles , Ensaio de Imunoadsorção Enzimática , Feminino , Fibrinolisina/metabolismo , Cardiopatias/mortalidade , Cardiopatias/patologia , Humanos , Imuno-Histoquímica , Recém-Nascido , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/mortalidade , Lúpus Eritematoso Sistêmico/patologia , Macrófagos/imunologia , Miócitos Cardíacos/imunologia , Miócitos Cardíacos/metabolismo , Gravidez , Receptores de Ativador de Plasminogênio Tipo Uroquinase/sangue , Valores de Referência , Ribonucleoproteínas/metabolismo , Taxa de Sobrevida , Ativador de Plasminogênio Tipo Uroquinase/sangue , Ativador de Plasminogênio Tipo Uroquinase/imunologia
10.
Blood ; 118(3): 804-15, 2011 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-21527523

RESUMO

Understanding the role of the extracellular matrix (ECM) in vascular morphogenesis has been possible using natural ECMs as in vitro models to study the underlying molecular mechanisms. However, little is known about vascular morphogenesis in synthetic matrices where properties can be tuned toward both the basic understanding of tubulogenesis in modular environments and as a clinically relevant alternative to natural materials for regenerative medicine. We investigated synthetic, tunable hyaluronic acid (HA) hydrogels and determined both the adhesion and degradation parameters that enable human endothelial colony-forming cells (ECFCs) to form efficient vascular networks. Entrapped ECFCs underwent tubulogenesis dependent on the cellular interactions with the HA hydrogel during each stage of vascular morphogenesis. Vacuole and lumen formed through integrins α(5)ß(1) and α(V)ß(3), while branching and sprouting were enabled by HA hydrogel degradation. Vascular networks formed within HA hydrogels containing ECFCs anastomosed with the host's circulation and supported blood flow in the hydrogel after transplantation. Collectively, we show that the signaling pathways of vascular morphogenesis of ECFCs can be precisely regulated in a synthetic matrix, resulting in a functional microvasculature useful for the study of 3-dimensional vascular biology and toward a range of vascular disorders and approaches in tissue regeneration.


Assuntos
Células Endoteliais/citologia , Células Endoteliais/fisiologia , Microvasos/crescimento & desenvolvimento , Neovascularização Fisiológica/fisiologia , Engenharia Tecidual/métodos , Prótese Vascular , Adesão Celular/fisiologia , Técnicas de Cultura de Células/métodos , Matriz Extracelular/fisiologia , Sangue Fetal/citologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato , Microvasos/citologia , Microvasos/fisiologia , Morfogênese/fisiologia
11.
J Biol Chem ; 286(35): 30444-30454, 2011 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-21730058

RESUMO

Autoimmune associated congenital heart block (CHB) may result from pathogenic cross-talk between inflammatory and profibrosing pathways. Incubation of macrophages with immune complexes (IC) composed of Ro60, a target of the pathologic maternal autoantibodies necessary for CHB, hY3 ssRNA, and affinity-purified anti-Ro60 antibody induces the Toll-like receptor 7 (TLR7)-dependent generation of supernatants that provoke a fibrosing phenotype in human fetal cardiac fibroblasts. We show herein that these cells are a major source of TGFß and that endothelin-1 (ET-1) is one of the key components responsible for the profibrosing effects generated by stimulated macrophages. Supernatants from macrophages incubated with IC induced the fibroblast secretion of TGFß, which was inhibited by treating the macrophages with an antagonist of TLR7. Under the same conditions, the induced fibroblast secretion of TGFß was decreased by inhibitors of the ET-1 receptors ETa or ETb or by an anti-ET-1 antibody but not by an isotype control. Exogenous ET-1 induced a profibrosing phenotype, whereas fibroblasts transfected with either ETa or ETb siRNA were unresponsive to the profibrosing effects of the IC-generated macrophage supernatants. Immunohistochemistry of the hearts from two fetuses dying with CHB revealed the presence of ET-1-producing mononuclear cells in the septal region in areas of calcification and fibrosis. In conclusion, these data support a novel role of ET-1 in linking TLR7 inflammatory signaling to subsequent fibrosis and provide new insight in considering therapeutics for CHB.


Assuntos
Anticorpos/química , Endotelina-1/fisiologia , Fibrose/metabolismo , Bloqueio Cardíaco/congênito , Inflamação , Ribonucleoproteínas/química , Receptor 7 Toll-Like/metabolismo , Autoimunidade , Feminino , Fibroblastos/metabolismo , Citometria de Fluxo , Bloqueio Cardíaco/metabolismo , Humanos , Leucócitos Mononucleares/citologia , Macrófagos/citologia , Macrófagos/metabolismo , Fator de Crescimento Transformador beta/metabolismo
12.
J Immunol ; 185(11): 6999-7006, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20962257

RESUMO

Platelets recruit leukocytes and mediate interactions between leukocytes and endothelial cells. Platelets have been long described as markers of transplant rejection, but the contribution of platelets to transplant rejection has not been critically examined. We demonstrate in this study that following T cell initiation of allograft rejection, platelets contribute to T cell recruitment and increased plasma inflammatory mediators and accelerate T cell-meditated skin graft rejection. Prior work from our laboratory has shown that platelets secrete glutamate when activated, which then induces platelet thromboxane production by signaling through platelet-expressed ionotropic glutamate receptors. Glutamate receptor antagonists therefore represent, to our knowledge, novel inhibitors of platelet-accelerated inflammation. We have found that plasma glutamate is increased in mice that receive skin grafts and that mice treated with glutamate receptor antagonists have improved graft survival and decreased plasma thromboxane, platelet factor 4 (CXCL4), and IFN-γ. Taken together, our work now demonstrates that subsequent to T cell initiation of skin graft rejection, platelets contribute to further T cell recruitment and that by blunting glutamate-mediated platelet activation, graft survival is improved.


Assuntos
Plaquetas/imunologia , Plaquetas/metabolismo , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/metabolismo , Receptores de Glutamato/fisiologia , Animais , Movimento Celular/imunologia , Rejeição de Enxerto/patologia , Sobrevivência de Enxerto/imunologia , Interferon gama/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos C57BL , Camundongos Nus , Ativação Plaquetária/imunologia , Receptores de Glutamato/biossíntese , Receptores de Glutamato/metabolismo , Transplante de Pele/imunologia , Transplante de Pele/patologia , Linfócitos T/imunologia , Linfócitos T/patologia , Tromboxanos/biossíntese , Regulação para Cima/imunologia
13.
Proc Natl Acad Sci U S A ; 106(44): 18769-74, 2009 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-19841279

RESUMO

Diabetes is a major risk factor for ischemic disease. Treatment options for diabetic patients with peripheral arterial disease when revascularization is not possible are limited, resulting in a high incidence of limb amputation. We evaluated the therapeutic potential of AdCA5, an adenovirus encoding a constitutively active form of HIF-1alpha, in a diabetic model of critical limb ischemia. Diabetic db/db and nondiabetic db/+ mice were subjected to unilateral femoral artery ligation. Limb perfusion, tissue viability, and motor function were more severely impaired in db/db mice. Intramuscular injection of AdCA5 into the ischemic limb of db/db mice increased the recovery of limb perfusion and function, reduced tissue necrosis, rescued the diabetes-associated impairment of circulating angiogenic cells, enhanced endothelial nitric oxide synthase activation, and increased vessel density and luminal area in the ischemic limb.


Assuntos
Adenoviridae/genética , Diabetes Mellitus Experimental/complicações , Extremidades/irrigação sanguínea , Terapia Genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/uso terapêutico , Isquemia/terapia , Animais , Diabetes Mellitus Experimental/enzimologia , Diabetes Mellitus Experimental/patologia , Extremidades/patologia , Artéria Femoral/cirurgia , Humanos , Imuno-Histoquímica , Isquemia/complicações , Isquemia/enzimologia , Isquemia/patologia , Ligadura , Camundongos , Músculo Esquelético/enzimologia , Músculo Esquelético/patologia , Neovascularização Fisiológica , Óxido Nítrico Sintase Tipo III/metabolismo , Transdução de Sinais
14.
Physiol Genomics ; 43(12): 726-38, 2011 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-21427359

RESUMO

OBJECTIVE: Evidence supports an antilipotoxic role for leptin in preventing inappropriate peripheral tissue lipid deposition. Obese, leptin-deficient mice develop left ventricular (LV) hypertrophy and myocardial steatosis with increased apoptosis and decreased longevity. Here we investigated the cardiac effects of caloric restriction versus leptin repletion in obese leptin-deficient (ob/ob) mice. METHODS: Echocardiography was performed on 7 mo old C57BL/6 wild-type mice (WT) and ob/ob mice fed ad libitum, leptin-repleted (LR-ob/ob), or calorie-restricted (CR-ob/ob) for 4 wk. Ventricular tissue was examined by electron microscopy (EM), triglyceride (TAG) content, oil red O staining, mitochondrial coupling assay, and microarray expression profiling. RESULTS: LR and CR-ob/ob mice showed decreased body and heart weight, and LV wall thickness compared with ad libitum ob/ob mice. LV fractional shortening was decreased in ad libitum ob/ob mice, but restored to WT in LR and CR groups. However, myocardial lipid content by EM and TAG analysis revealed persistent cardiac steatosis in the CR-ob/ob group. Although CR restored mitochondrial coupling to WT levels, PPARα was suppressed and genes associated with oxidative stress and cell death were upregulated in CR-ob/ob animals. In contrast, LR eliminated cardiac steatosis, normalized mitochondrial coupling, and restored PGC1α and PPARα expression, while inducing core genes involved in glycerolipid/free fatty acid (GL/FFA) cycling, a thermogenic pathway that can reduce intracellular lipids. CONCLUSIONS: Thus, CR in the absence of leptin fails to normalize cardiac steatosis. GL/FFA cycling may be, at least in part, leptin-dependent and a key pathway that protects the heart from lipid accumulation.


Assuntos
Restrição Calórica/métodos , Hipertrofia Ventricular Esquerda/patologia , Leptina/deficiência , Lipídeos/análise , Miocárdio/química , PPAR alfa/metabolismo , Análise de Variância , Animais , Apoptose/genética , Peso Corporal , Ecocardiografia , Perfilação da Expressão Gênica , Hipertrofia Ventricular Esquerda/etiologia , Leptina/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Análise em Microsséries , Microscopia Eletrônica , Miocárdio/patologia , Estresse Oxidativo/genética
15.
Skelet Muscle ; 11(1): 13, 2021 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-34001262

RESUMO

BACKGROUND: Skeletal muscle myofibers can be separated into functionally distinct cell types that differ in gene and protein expression. Current single cell expression data is generally based upon single nucleus RNA, rather than whole myofiber material. We examined if a whole-cell flow sorting approach could be applied to perform single cell RNA-seq (scRNA-seq) in a single muscle type. METHODS: We performed deep, whole cell, scRNA-seq on intact and fragmented skeletal myofibers from the mouse fast-twitch flexor digitorum brevis muscle utilizing a flow-gated method of large cell isolation. We performed deep sequencing of 763 intact and fragmented myofibers. RESULTS: Quality control metrics across the different gates indicated only 171 of these cells were optimal, with a median read count of 239,252 and an average of 12,098 transcripts per cell. scRNA-seq identified three clusters of myofibers (a slow/fast 2A cluster and two fast 2X clusters). Comparison to a public skeletal nuclear RNA-seq dataset demonstrated a diversity in transcript abundance by method. RISH validated multiple genes across fast and slow twitch skeletal muscle types. CONCLUSION: This study introduces and validates a method to isolate intact skeletal muscle myofibers to generate deep expression patterns and expands the known repertoire of fiber-type-specific genes.


Assuntos
Músculo Esquelético , Doenças Musculares , Animais , Separação Celular , , Camundongos , Análise de Sequência de RNA
16.
J Mol Cell Cardiol ; 49(1): 5-15, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20153750

RESUMO

The ubiquitin-proteasome system plays an important role in regulating muscle mass. Inducible immunoproteasome subunits LMP-2 and LMP-7 are constitutively expressed in the heart; however, their regulation and functions are poorly understood. We here investigated the hypothesis that immunoproteasomes regulate cardiac muscle mass in diabetic mice. Type 1 diabetes was induced in wildtype mice by streptozotocin. After hyperglycemia developed, insulin and the proteasome inhibitor epoxomicin were used to treat diabetic mice for 6weeks. Isolated mouse hearts were perfused with control or high glucose solution. Catalytic proteasome beta-subunits and proteolytic activities were analyzed in the heart by immunoblotting and fluorogenic peptide degradation assays, respectively. Insulin and epoxomicin blocked loss of heart weight and improved cardiac function in diabetic mice. LMP-7 and its corresponding chymotryptic-like proteasome activity were increased in diabetic hearts and high glucose-treated hearts. Myosin heavy chain protein was decreased in diabetic hearts, which was largely reversed by epoxomicin. High glucose decreased LMP-2 protein levels in perfused hearts. In diabetic hearts, LMP-2 expression was downregulated whereas expression of the phosphatase and tensin homologue deleted on chromosome ten (PTEN) and the muscle atrophy F-box were upregulated. Moreover, mice with muscle-specific knockout of PTEN gene demonstrated increased cardiac muscle mass, while mice with LMP-2 deficiency demonstrated PTEN accumulation, muscle mass loss, and contractile impairment in the heart. Therefore, we concluded that high glucose regulates immunoproteasome subunits and modifies proteasome activities in the heart, and that dysregulated immunoproteasome subunits may mediate loss of cardiac muscle mass in experimental diabetic mice.


Assuntos
Diabetes Mellitus/metabolismo , Miocárdio/metabolismo , Animais , Cisteína Endopeptidases , Glucose/metabolismo , Coração/fisiologia , Hiperglicemia/metabolismo , Insulina/metabolismo , Insulina/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Cadeias Pesadas de Miosina/metabolismo , PTEN Fosfo-Hidrolase , Complexo de Endopeptidases do Proteassoma/metabolismo , Estreptozocina/metabolismo , Ubiquitina/metabolismo
17.
Circ Res ; 102(7): 777-85, 2008 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-18296616

RESUMO

Platelets recruit leukocytes and mediate interactions between leukocytes and endothelial cells. Most studies examining this important platelet immune function have focused on the development of atherosclerosis, but similar mechanisms may contribute to acute and chronic vascular lesions in transplants. Platelets have been described as markers of transplant rejection, but little investigation has critically examined a role for platelets in transplant vasculopathy and, in particular, alloantibody-mediated transplant rejection. We now demonstrate using a skin transplant model that alloantibody indirectly induces platelet activation and rolling in vivo. Repeated IgG2a alloantibody injections result in sustained platelet-endothelial interactions and vascular pathology, including von Willebrand factor release, small platelet thrombi, and complement deposition. Maintenance of continued platelet-endothelial interactions are dependent on complement activation. Furthermore, we demonstrate that platelets recruit leukocytes to sites of alloantibody deposition and sustain leukocyte-endothelial cell interactions in vivo. Taken together, our model demonstrates an important role for platelets in alloantibody induced transplant rejection.


Assuntos
Plaquetas/patologia , Comunicação Celular/imunologia , Endotélio Vascular/patologia , Isoanticorpos/imunologia , Complexo Principal de Histocompatibilidade/imunologia , Animais , Plaquetas/imunologia , Endotélio Vascular/imunologia , Rejeição de Enxerto/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Imunoglobulina G/administração & dosagem , Imunoglobulina G/imunologia , Imunoglobulina G/farmacologia , Isoanticorpos/administração & dosagem , Leucócitos/imunologia , Leucócitos/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Modelos Animais , Ativação Plaquetária/imunologia , Transplante de Pele/imunologia , Transplante de Pele/patologia
18.
Circ Res ; 101(10): 1001-8, 2007 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-17885215

RESUMO

Fractalkine (FKN) is a membrane-bound chemokine that can be released by proteolysis to produce soluble FKN (s-FKN). FKN and its receptor, CX3CR1, are believed to be important factors in atherosclerosis and may play a role in acute inflammatory responses. Although FKN is expressed on endothelial cells (ECs), CX3CR1 is reported to reside mainly on certain leukocyte populations. RT-PCR and Western blotting demonstrated that both human coronary artery and umbilical vein ECs expressed CX3CR1 mRNA and protein. Confocal microscopy showed that CX3CR1 was located at the cell membrane and to a lesser extent in the cytoplasm. Following exposure of both types of ECs to hypoxia and reoxygenation, FKN expression increased rapidly and s-FKN was shed into the culture medium. The addition of s-FKN protein to cultured ECs resulted in a dose-dependent increase in intercellular adhesion molecule (ICAM)-1 mRNA. Perfusion of mouse hearts with s-FKN protein increased expression of ICAM-1 protein in vascular endothelium. Transfection of ECs with CX3CR1-interfering RNA to knockdown the receptor resulted in decreased ICAM-1 expression after s-FKN stimulation. In addition, when ECs were stimulated with s-FKN, greater adhesion of human neutrophils to the ECs was observed. This increase was ICAM-1 dependent and was blocked by CX3CR1 knockdown. Following exposure to s-FKN, ECs exhibited increased phosphorylation of Jak2 and Stat5 and the ICAM-1 expression induced by s-FKN was blocked by silencing of Stat5 with small interfering RNA. Vascular ECs express both FKN and its receptor CX3CR1. s-FKN is shed from ECs following hypoxia/reoxygenation and acts through CX3CR1 on ECs to increase ICAM-1 expression and promote neutrophil adhesion through activation of the Jak-Stat5 pathway.


Assuntos
Quimiocina CX3CL1/metabolismo , Endotélio Vascular/metabolismo , Hipóxia/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Receptores de Quimiocinas/metabolismo , Animais , Receptor 1 de Quimiocina CX3C , Adesão Celular/efeitos dos fármacos , Adesão Celular/imunologia , Células Cultivadas , Quimiocina CX3CL1/farmacologia , Vasos Coronários/citologia , Endotélio Vascular/citologia , Endotélio Vascular/imunologia , Humanos , Hipóxia/imunologia , Janus Quinase 2/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/citologia , Interferência de RNA , Receptores de Quimiocinas/genética , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Solubilidade , Veias Umbilicais/citologia , Regulação para Cima/fisiologia
20.
Cardiovasc Res ; 77(3): 463-70, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18006459

RESUMO

AIMS: We investigated whether hypoxia-inducible factor 1 alpha (HIF-1 alpha) plays a role in the acute phase of ischaemic preconditioning (IPC). METHODS AND RESULTS: Hearts from wild-type (WT) mice and mice heterozygous for a null allele at the locus encoding HIF-1 alpha (HET) were subjected to IPC (10-min ischaemia/5 min reperfusion, or two cycles of 5 min ischaemia/5 min reperfusion), followed by 30 min ischaemia and reperfusion. Left ventricular-developed pressure, heart rate, and coronary flow rate were measured continuously. Apoptosis and infarct size were assessed by TUNEL assay, cleaved caspase 3 immunohistochemistry, and triphenyltetrazolium chloride staining. Production of reactive oxygen species (ROS) in isolated cardiac mitochondria was measured by a chemiluminescence assay. The phosphatase and tensin homologue (PTEN) and AKT (protein kinase B) were analysed by immunoblot assay. IPC improved functional recovery and limited infarct size and apoptosis after prolonged ischaemia-reperfusion in WT hearts, but not in HET hearts. Mitochondrial ROS production, PTEN oxidation, and AKT phosphorylation were impaired in HET hearts. WT and HET hearts were protected by adenosine, which acts via an ROS-independent mechanism. CONCLUSION: HIF-1 alpha is required for IPC-induced mitochondrial ROS production and myocardial protection against ischaemia-reperfusion injury.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Precondicionamento Isquêmico Miocárdico , Infarto do Miocárdio/prevenção & controle , Adenosina/farmacologia , Animais , Apoptose , Masculino , Camundongos , Traumatismo por Reperfusão Miocárdica/prevenção & controle , PTEN Fosfo-Hidrolase/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa