Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 155
Filtrar
2.
J Biol Chem ; 287(48): 40391-9, 2012 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-23027874

RESUMO

BACKGROUND: IL-18 induces profibrotic changes in TECs independent of TGF-ß1 activity. RESULTS: IL-18 stimulates the TLR4 promoter via AP-1 activation to increase TLR4 expression in TECs and stimulates profibrotic changes in TECs through increased TLR4 expression/signaling. CONCLUSION: The profibrotic effect of IL-18 in TECs is mediated through stimulation of TLR4 expression via activation of AP-1. SIGNIFICANCE: This represents a novel fibrotic signaling pathway in TECs independent of TGF-ß1. IL-18 is an important mediator of obstruction-induced renal fibrosis and tubular epithelial cell injury independent of TGF-ß1 activity. We sought to determine whether the profibrotic effect of IL-18 is mediated through Toll-like receptor 4 (TLR4). Male C57BL6 wild type and mice transgenic for human IL-18-binding protein were subjected to left unilateral ureteral obstruction versus sham operation. The kidneys were harvested 1 week postoperatively and analyzed for IL-18 production and TLR4 expression. In a separate arm, renal tubular epithelial cells (HK-2) were directly stimulated with IL-18 in the presence or absence of a TLR4 agonist, TLR4 antagonist, or TLR4 siRNA knockdown. Cell lysates were analyzed for TLR4, α-smooth muscle actin, and E-cadherin expression. TLR4 promotor activity, as well as AP-1 activation and the effect of AP-1 knockdown on TLR4 expression, was evaluated in HK-2 cells in response to IL-18 stimulation. The results demonstrate that IL-18 induces TLR4 expression during unilateral ureteral obstruction and induces TLR4 expression in HK-2 cells via AP-1 activation. Inhibition of TLR4 or knockdown of TLR4 gene expression in turn prevents IL-18-induced profibrotic changes in HK-2 cells. These results suggest that IL-18 induces profibrotic changes in tubular epithelial cells via increased TLR4 expression/signaling.


Assuntos
Interleucina-18/metabolismo , Nefropatias/metabolismo , Nefropatias/patologia , Regiões Promotoras Genéticas , Receptor 4 Toll-Like/genética , Regulação para Cima , Animais , Linhagem Celular , Células Epiteliais/metabolismo , Fibrose , Humanos , Nefropatias/genética , Túbulos Renais/citologia , Túbulos Renais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptor 4 Toll-Like/metabolismo
3.
J Surg Res ; 177(2): 330-3, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22703984

RESUMO

INTRODUCTION: Mesenchymal stem cell (MSC) therapy improves cardiac function after ischemia/reperfusion injury, but its effectiveness is limited by MSC survival in hypoxic environments. Toll-like receptor 4 (TLR4) contributes to pro-apoptotic signaling under hypoxic conditions. Activation of intracellular AKT and ERK pathways opposes this signal and improves cell survival. It is unknown whether ablation of TLR4 affects these pathways after hypoxic injury in MSCs. We hypothesized that: 1) TLR4 knockout (TLR4KO) in MSCs improves survival after hypoxic injury; and 2) this survival difference is due to improved signaling in the AKT and ERK pathways. MATERIALS AND METHODS: Murine wild-type (WT) and TLR4KO MSCs were harvested from bone marrow and grown in vitro. A total of 0.1 × 10(6) cells/well were incubated in hypoxic conditions versus normoxic controls. After 24 h, these groups were examined for cell survival via counting and compared using a t-test with P < 0.05 = statistical significance. AKT and ERK concentrations were measured in lysate using Western blot analysis. RESULTS: The morphology of WT and TLR4KO MSCs was similar. In line with our previous findings, hypoxia did significantly increase cell death in WT cells (1.79 × 10(5) living cells/mL control versus 0.88 × 10(5) hypoxia, P < 0.05). Hypoxic injury did not increase cell death in the TLR4KO group (1.68 × 10(5) control versus 1.82 × 10(5) hypoxia, P < 0.05). Increased AKT activation was observed in all TLR4KO groups. TLR4 did not affect phosphorylated ERK levels. CONCLUSION: TLR4-knockout MSCs show improved survival after hypoxic injury because of increased AKT pathway signal. Use of TLR4-knockout MSCs in ischemia/reperfusion studies results in enhanced cardioprotection; improved stem cell survival was likely a contributing factor.


Assuntos
Hipóxia/fisiopatologia , Sistema de Sinalização das MAP Quinases , Células-Tronco Mesenquimais/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor 4 Toll-Like/fisiologia , Animais , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
4.
J Surg Res ; 173(1): 113-26, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22225756

RESUMO

BACKGROUND: Sepsis remains a source of morbidity and mortality in the postoperative patient despite appropriate resuscitative and antimicrobial approaches. Recent research has focused upon additional interventions such as exogenous cell-based therapy. Mesenchymal stem cells (MSCs) exhibit multiple beneficial properties through their capacity for homing, attenuating the inflammatory response, modulating immune cells, and promoting tissue healing. Recent animal trials have provided evidence that MSCs may be useful therapeutic adjuncts. MATERIALS AND METHODS: A directed search of recent medical literature was performed utilizing PubMed to examine the pathophysiology of sepsis, mechanisms of mesenchymal stem cell interaction with host cells, sepsis animal models, and recent trials utilizing stem cells in sepsis. RESULTS: MSCs continue to show promise in the treatment of sepsis by their intrinsic ability to home to injured tissue, secrete paracrine signals to limit systemic and local inflammation, decrease apoptosis in threatened tissues, stimulate neoangiogenesis, activate resident stem cells, beneficially modulate immune cells, and exhibit direct antimicrobial activity. These effects are associated with reduced organ dysfunction and improved survival in animal models. CONCLUSION: Research utilizing animal models of sepsis has provided a greater understanding of the beneficial properties of MSCs. Their capacity to home to sites of injury and use paracrine mechanisms to change the local environment to ultimately improve organ function and survival make MSCs attractive in the treatment of sepsis. Future studies are needed to further evaluate the complex interactions between MSCs and host tissues.


Assuntos
Pesquisa Biomédica/tendências , Terapia Baseada em Transplante de Células e Tecidos/métodos , Transplante de Células-Tronco Mesenquimais/métodos , Sepse/terapia , Animais , Apoptose/fisiologia , Modelos Animais de Doenças , Neovascularização Fisiológica/fisiologia , Comunicação Parácrina/fisiologia , Sepse/fisiopatologia
5.
J Surg Res ; 176(2): 386-94, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22316665

RESUMO

BACKGROUND: Neonatal mesenchymal stem cells exhibit less cardioprotective potential than their adult counterparts. Transforming growth factor-α (TGF-α) has been shown to stimulate adult stem cell VEGF production, however, it remains unknown whether it may augment neonatal stem cell paracrine function. We hypothesized that TGF-α would equalize adult and neonatal stem cell paracrine function and cardioprotection during acute ischemia/reperfusion. MATERIALS AND METHODS: Bone marrow mesenchymal stem cells isolated from adult and 2.5 wk-old mice were treated with TGF-α (250 ng/mL) for 24 h. VEGF, HGF, IGF-1, IL-1ß, and IL-6 production were measure in vitro, and cells were infused via an intracoronary route using a model of isolated heart perfusion. RESULTS: TGF-α equalized adult and neonatal stem cell VEGF production but did not affect production of HGF, IGF-1, IL-1ß, or IL-6. ERK, p38 MAPK, and JNK phosphorylation were greater in adult cells in response to TGF-α. Whereas infusion of adult but not neonatal stem cells was associated with improved myocardial functional recovery during reperfusion, infusions of either TGF-α-pretreated cell group were associated with the greatest functional recovery. TGF-α equalizes adult and neonatal mesenchymal stem cell VEGF production and cardioprotection in association with differential regulation of ERK, p38 MAPK, and JNK phosphorylation.


Assuntos
Células-Tronco Adultas/efeitos dos fármacos , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/efeitos dos fármacos , Traumatismo por Reperfusão Miocárdica/terapia , Fator de Crescimento Transformador alfa/farmacologia , Doença Aguda , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Fatores Etários , Animais , Animais Recém-Nascidos , Caspase 3/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Traumatismo por Reperfusão Miocárdica/metabolismo , Miocárdio/citologia , Miocárdio/metabolismo , Comunicação Parácrina/efeitos dos fármacos , Comunicação Parácrina/fisiologia , Fator de Crescimento Transformador alfa/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
Proc Natl Acad Sci U S A ; 106(41): 17499-504, 2009 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-19805173

RESUMO

IL-18 is a proinflammatory cytokine known to cause tissue injury by inducing inflammation and cell death. Increased levels of IL-18 are associated with myocardial injury after ischemia or infarction. IL-18-binding protein (IL-18BP), the naturally occurring inhibitor of IL-18 activity, decreases the severity of inflammation in response to injury. In the present study, mesenchymal stem cells (MSCs) derived from mice transgenic for over expression of human IL-18BP were tested in rat models of global myocardial ischemia and acute myocardial infarction. Improved myocardial function is associated with production of VEGF, and in vitro, IL-18BP MSCs secreted higher levels of constitutive VEGF compared to wild-type MSCs. Whereas IL-18 increased cell death and reduced VEGF in wild-type MSCs, IL-18BP MSCs were protected. In an isolated heart model, intracoronary infusion of IL-18BP MSCs before ischemia increased postischemic left ventricular (LV) developed pressure to 79.5 + or - 9.47 mmHg compared to 59.3 + or - 7.8 mmHg in wild-type MSCs and 37.8 + or - 5 mmHg in the vehicle group. Similarly, using a coronary artery ligation model, intramyocardial injection of IL-18BP MSCs improved LV ejection fraction to 67.8 + or - 1.76% versus wild-type MSCs (57.4 + or - 1.33%) and vehicle (39.2 + or - 2.07%), increased LV fractional shortening 1.25-fold over wild-type MSCs and 1.95-fold over vehicle, decreased infarct size to 38.8 + or - 2.16% compared to 46.4 + or - 1.92% in wild-type MSCs and 60.7 + or - 2.2% in vehicle, reduced adverse ventricular remodeling, increased myocardial VEGF production, and decreased IL-6 levels. This study provides the concept that IL-18BP genetically modified stem cells improve cardioprotection over that observed with unmodified stem cells.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/uso terapêutico , Interleucina-18/uso terapêutico , Células-Tronco Mesenquimais/fisiologia , Infarto do Miocárdio/prevenção & controle , Isquemia Miocárdica/prevenção & controle , Animais , Divisão Celular , Células Cultivadas , Regulação para Baixo , Feminino , Coração/efeitos dos fármacos , Coração/fisiopatologia , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/patologia , Camundongos , Imagem de Perfusão do Miocárdio , Ratos , Fator de Necrose Tumoral alfa/farmacologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/fisiologia
7.
Am J Physiol Regul Integr Comp Physiol ; 300(6): R1506-14, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21451141

RESUMO

Mesenchymal stem cells (MSCs) may offer therapeutic benefit in the setting of sepsis and endotoxemia. Previous studies suggest that MSCs from female donors may possess better protective capabilities than their male counterparts. The present study examined whether female MSCs may offer a greater protective advantage in the setting of endotoxemic cardiac dysfunction compared with male MSCs. Adult male Sprague-Dawley rats were injected intraperitoneally with LPS and then treated with intraperitoneal injections of either saline, female MSCs, or male MSCs. Hearts and serum were then collected for analysis of myocardial function, myocardial protein, and myocardial and serum cytokines. Compared with male MSC or vehicle-treated animals, female MSC treatment resulted in greater preservation of myocardial function (P < 0.001). Serum and myocardial levels of all measured cytokines were comparable between rats given MSCs from male or female donors but substantially improved over rats given vehicle (P < 0.05). Reduced myocardial inflammation correlated with reduced levels of phosphorylated p38 MAPK expression in the myocardium of animals injected with MSCs of either sex (P < 0.05). The Bcl-xL/Bax ratio was increased to a greater extent following treatment with female MSCs vs. male MSCs (P < 0.05). Intraperitoneal administration of MSCs is effective in limiting myocardial inflammation and dysfunction in the rat endotoxemia model. Compared with treatment with their male counterparts, MSC treatment from female donors is associated with greater cardiac protection against acute endotoxemic injury.


Assuntos
Cardiomiopatias/etiologia , Cardiomiopatias/prevenção & controle , Terapia Baseada em Transplante de Células e Tecidos/métodos , Endotoxemia/complicações , Células-Tronco Mesenquimais/fisiologia , Animais , Cardiomiopatias/fisiopatologia , Modelos Animais de Doenças , Endotoxemia/induzido quimicamente , Endotoxemia/fisiopatologia , Feminino , Lipopolissacarídeos/efeitos adversos , Masculino , Transplante de Células-Tronco Mesenquimais/métodos , Camundongos , Camundongos Endogâmicos C57BL , Miocárdio/metabolismo , Ratos , Ratos Sprague-Dawley , Proteína X Associada a bcl-2/metabolismo , Proteína bcl-X/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
J Surg Res ; 168(1): e51-9, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20850784

RESUMO

BACKGROUND: Mesenchymal stem cells (MSCs) hold promise for the treatment of renal disease. While MSCs have been shown to accelerate recovery and prevent acute renal failure in multiple disease models, the effect of MSC therapy on chronic obstruction-induced renal fibrosis has not previously been evaluated. MATERIALS AND METHODS: Male Sprague-Dawley rats underwent renal artery injection of vehicle or fluorescent-labeled human bone marrow-derived MSCs immediately prior to sham operation or induction of left ureteral obstruction (UUO). One or 4 wk later, the kidneys were harvested and the renal cortex analyzed for evidence of stem cell infiltration, epithelial-mesenchymal transition (EMT) as evidenced by E-cadherin/α-smooth muscle actin (α-SMA) expression and fibroblast specific protein (FSP+) staining, renal fibrosis (collagen content, Masson's trichrome staining), and cytokine and growth factor activity (ELISA and real time RT-PCR). RESULTS: Fluorescent-labeled MSCs were detected in the interstitium of the kidney up to 4 wk post-obstruction. Arterially delivered MSCs significantly reduced obstruction-induced α-SMA expression, FSP+ cell accumulation, total collagen content, and tubulointerstitial fibrosis, while simultaneously preserving E-cadherin expression, suggesting that MSCs prevent obstruction-induced EMT and renal fibrosis. Exogenous MSCs reduced obstruction-induced tumor necrosis factor-α (TNF-α) levels, but did not alter transforming growth factor-ß1 (TGF-ß1), vascular endothelial growth factor (VEGF), interleukin-10 (IL-10), fibroblast growth factor (FGF), or hepatocyte growth factor (HGF) expression. CONCLUSIONS: Human bone marrow-derived MSCs remain viable several weeks after delivery into the kidney and provide protection against obstruction-induced EMT and chronic renal fibrosis. While the mechanism of MSCs-induced renal protection during obstruction remains unclear, our results demonstrate that alterations in TNF-α production may be involved.


Assuntos
Nefropatias/prevenção & controle , Rim/patologia , Transplante de Células-Tronco Mesenquimais/métodos , Transplante Heterólogo/métodos , Obstrução Ureteral/prevenção & controle , Animais , Células Cultivadas , Colágeno/metabolismo , Citocinas/metabolismo , Fibrose , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Nefropatias/metabolismo , Nefropatias/patologia , Masculino , Modelos Animais , Ratos , Ratos Sprague-Dawley , Obstrução Ureteral/metabolismo , Obstrução Ureteral/patologia
9.
J Surg Res ; 167(1): 78-86, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20869073

RESUMO

Mesenchymal stem cells (MSCs) have been used experimentally and clinically in the treatment of a wide variety of pathologies. It is now clear that a number of different mechanisms contribute to the therapeutic effects exerted by these cells. The ability of MSCs to interact with and modulate the functions of a wide variety of immune cells has been recognized as one such mechanism. The implications that the immunomodulatory properties of MSCs may have for the treatment of solid organ rejection, the Systemic Inflammatory Response Syndrome, cancer, and Crohn's disease are reviewed herein.


Assuntos
Imunomodulação/fisiologia , Células-Tronco Mesenquimais/fisiologia , Doença de Crohn/fisiopatologia , Doença de Crohn/cirurgia , Rejeição de Enxerto/fisiopatologia , Rejeição de Enxerto/cirurgia , Humanos , Transplante de Células-Tronco Mesenquimais , Neoplasias/fisiopatologia , Neoplasias/cirurgia , Sepse/fisiopatologia , Sepse/cirurgia
10.
J Surg Res ; 166(1): 138-45, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20828719

RESUMO

BACKGROUND: Stem cell-based therapies for myocardial ischemia have demonstrated promising early clinical results, but their benefits have been limited in duration due to impaired donor cell engraftment and function. Several strategies have emerged for enhancing stem cell function prior to their therapeutic use particularly with regard to stem cell homing, paracrine function, and survival. This review discusses current understandings of stem cell-mediated cardioprotection as well as methods of enhancing post-transplantation stem cell function and survival through hypoxic preconditioning, genetic manipulation, and pharmacologic pretreatment. MATERIALS AND METHODS: A literature search was performed using the MEDLINE and PubMed databases using the keywords "stem cell therapy," "myocardial ischemia," "hypoxic preconditioning," "paracrine function," and "stem cell pretreatment." Studies published in English since January 1990 were selected. In addition, studies were identified from references cited in publications found using the search terms. RESULTS: All included studies utilized animal studies and/or in vitro techniques. Stem cell modifications generally targeted stem cell homing (SDF-1, CXCR4), paracrine function (VEGF, angiogenin, Ang-1, HGF, IL-18 binding protein, TNFR1/2), or survival (Akt, Bcl-2, Hsp20, HO-1, FGF-2). However, individual modifications commonly exhibited pleiotropic effects involving some or all of these general categories. CONCLUSION: These strategies for optimizing stem cell-mediated cardioprotection present unique potential sets of advantages and disadvantages for clinical application. Additional questions remain including those that are most efficacious in terms of magnitude and duration of benefit as well as whether combinations may yield greater benefits in both the preclinical and clinical settings.


Assuntos
Sobrevivência de Enxerto , Precondicionamento Isquêmico , Isquemia Miocárdica/terapia , Transplante de Células-Tronco/métodos , Transplante de Células-Tronco/normas , Humanos , Comunicação Parácrina
11.
Am J Physiol Heart Circ Physiol ; 298(5): H1529-36, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20173040

RESUMO

Toll-like receptor 2 (TLR2), a key component of the innate immune system, is linked to inflammation and myocardial dysfunction after ischemia-reperfusion injury (I/R). Treatment of the heart with mesenchymal stem cells (MSCs) is known to improve myocardial recovery after I/R in part by paracrine factors such as VEGF. However, it is unknown whether TLR2 activation on the MSCs affects MSC-mediated myocardial recovery and VEGF production. We hypothesized that the knockout of TLR2 on the MSCs (TLR2KO MSCs) would 1) improve MSC-mediated myocardial recovery and 2) increase myocardial and MSC VEGF release. With the isolated heart perfusion system, Sprague-Dawley rat hearts were subjected to I/R and received one of three intracoronary treatments: vehicle, male wild-type MSCs (MWT MSCs), or TL2KO MSCs. All treatments were performed immediately before ischemia, and heart function was measured continuously. Postreperfusion, heart homogenates were analyzed for myocardial VEGF production. Contrary to our hypothesis, only MWT MSC treatment significantly improved the recovery of left ventricular developed pressure and the maximal positive and negative values of the first derivative of pressure. In addition, VEGF production was greatest in hearts treated with MWT MSCs. To investigate MSC production of VEGF, MSCs were activated with TNF in vitro and the supernatants collected for ELISA. In vitro basal levels of MSC VEGF production were similar. However, with TNF activation, MWT MSCs produced significantly more VEGF, whereas activated TLR2KO MSC production of VEGF was unchanged. Finally, we observed that MWT MSCs proliferated more rapidly than TLR2KO MSCs. These data indicate that TLR2 may be essential to MSC-mediated myocardial recovery and VEGF production.


Assuntos
Coração/fisiologia , Células-Tronco Mesenquimais/fisiologia , Traumatismo por Reperfusão Miocárdica/patologia , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/fisiologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Adipócitos/fisiologia , Animais , Diferenciação Celular/fisiologia , Proliferação de Células , Técnicas In Vitro , Proteínas de Membrana/metabolismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Traumatismo por Reperfusão Miocárdica/metabolismo , Osteogênese/fisiologia , Recuperação de Função Fisiológica
12.
J Urol ; 184(1): 26-33, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20478602

RESUMO

PURPOSE: The potential use of stem cells for acute and chronic renal injury is under intensive investigation. We summarized the current literature on the potential therapeutic role of mesenchymal stem cells for kidney injury. MATERIALS AND METHODS: We reviewed the pertinent literature on mesenchymal stem cell therapy for acute and chronic renal injury. RESULTS: Experimental evidence suggests that administering exogenous mesenchymal stem cells during acute and chronic kidney injury may improve functional and structural recovery of the tubular, glomerular and interstitial kidney compartments. Several studies point to a paracrine and/or endocrine mechanism of action rather than to direct repopulation of cells in the injured nephron. Multiple questions remain unanswered regarding the protective action of mesenchymal stem cells during renal injury, including signals that regulate stem cell homing to injured tissue, factors regulating paracrine and/or endocrine activity of exogenous mesenchymal stem cells and particularly the long-term behavior of administered stem cells in vivo. CONCLUSIONS: Many questions remain unanswered but mesenchymal stem cell based therapy is a promising new strategy for acute and chronic kidney disease.


Assuntos
Nefropatias/cirurgia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/fisiologia , Animais , Diferenciação Celular , Humanos , Nefropatias/fisiopatologia , Comunicação Parácrina/fisiologia , Regeneração , Transdução de Sinais
13.
Am J Physiol Regul Integr Comp Physiol ; 299(1): R371-8, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20484699

RESUMO

Stem cell-based therapies offer promising potential for myocardial infarction (MI), but endogenous molecules released in response to injury likely impair posttransplantation stem cell function. Stem cell-mediated cardioprotection occurs in part via paracrine effects, and transforming growth factor-alpha (TGF-alpha) has been shown to enhance paracrine function. However, it is unknown whether pretreating stem cells with TGF-alpha increases stem cell-mediated cardioprotection after acute MI. Mesenchymal stem cells (MSCs) were treated with TGF-alpha (250 ng/ml) for 24 h. Adult male Sprague-Dawley rat hearts were isolated and perfused using the Langendorff method. MI was induced by ligating the left anterior descending coronary artery. Postligation (30 min), vehicle or 1 x 10(6) MSCs with or without pretreatment were injected in the infarct border zones, and the hearts were perfused for an additional 60 min. Left ventricular function was continuously measured, and infarct size was assessed with Evans blue dye and 2,3,5-triphenyltetrazolium chloride staining. Myocardial production of interleukin (IL)-1beta and IL-6 and caspase 3 activation was also measured. Left ventricular function decreased significantly following coronary artery ligation but improved following injection of untreated MSCs and to a greater extent after injection of pretreated MSCs. In addition, the infarct area, myocardial caspase 3 activation, and IL-6 production were lowest in hearts injected with pretreated cells. Intramyocardial injection of TGF-alpha-pretreated MSCs after acute MI is associated with increased myocardial function and decreased myocardial injury. This strategy may be useful for optimizing the therapeutic efficacy of stem cells for the treatment of acute MI.


Assuntos
Células-Tronco Mesenquimais , Infarto do Miocárdio/terapia , Fator de Crescimento Transformador alfa/farmacologia , Animais , Coração/efeitos dos fármacos , Coração/fisiologia , Coração/fisiopatologia , Interleucina-6/farmacologia , Interleucina-6/fisiologia , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/fisiologia , Infarto do Miocárdio/fisiopatologia , Miocárdio , Ratos , Ratos Sprague-Dawley , Transplante de Células-Tronco , Sais de Tetrazólio , Fator de Crescimento Transformador alfa/fisiologia , Fator A de Crescimento do Endotélio Vascular/farmacologia , Fator A de Crescimento do Endotélio Vascular/fisiologia , Função Ventricular Esquerda/fisiologia
14.
J Surg Res ; 162(2): 239-49, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20053409

RESUMO

Over the past century, numerous animal models have been developed in an attempt to understand myocardial and vascular injury. However, the successful translation of results observed in animals to human therapy remains low. To understand this problem, we present several animal models of cardiac and vascular injury that are of particular relevance to the cardiac or vascular surgeon. We also explore the potential clinical implications and limitations of each model with respect to the human disease state. Our results underscore the concept that animal research requires an in-depth understanding of the model, animal physiology, and the potential confounding factors. Future outcome analyses with standardized animal models may improve translation of animal research from the bench to the bedside.


Assuntos
Doença das Coronárias/fisiopatologia , Endotélio Vascular/fisiopatologia , Insuficiência Cardíaca/fisiopatologia , Coração/fisiologia , Infarto do Miocárdio/fisiopatologia , Isquemia Miocárdica/fisiopatologia , Animais , Animais Geneticamente Modificados , Doença das Coronárias/epidemiologia , Doença das Coronárias/mortalidade , Modelos Animais de Doenças , Extremidades/irrigação sanguínea , Coração/fisiopatologia , Humanos , Incidência , Isquemia/fisiopatologia , Camundongos , Modelos Animais , Modelos Biológicos , Infarto do Miocárdio/epidemiologia , Estados Unidos/epidemiologia
15.
Am J Physiol Cell Physiol ; 297(5): C1284-93, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19692652

RESUMO

Human bone marrow mesenchymal stem cells (MSCs) are a potent source of growth factors, which are partly responsible for their beneficial paracrine effects. We reported previously that transforming growth factor-alpha (TGF-alpha), a putative mediator of wound healing and the injury response, increases the release of vascular endothelial growth factor (VEGF), augments tumor necrosis factor-alpha (TNF-alpha)-stimulated VEGF production, and activates mitogen-activated protein kinases and phosphatidylinositol 3-kinase (PI-3K) pathway in human MSCs. The experiments described in this report indicate that TGF-alpha increases MSC-derived hepatocyte growth factor (HGF) production. TGF-alpha-stimulated HGF production was abolished by inhibition of MEK, p38, PI-3K, or by small interfering RNA (siRNA) targeting TNF receptor 2 (TNFR2), but was not attenuated by siRNA targeting TNF receptor 1 (TNFR1). Ablation of TNFR1 significantly increased basal and stimulated HGF. A potent synergy between TGF-alpha and TNF-alpha was noted in MSC HGF production. This synergistic effect was abolished by MEK, P38, PI-3K inhibition, or by ablation of both TNF receptors using siRNA. We conclude that 1) novel cross talk occurs between tumor necrosis factor receptor and TGF-alpha/epidermal growth factor receptor in stimulating MSC HGF production; 2) this cross talk is mediated, at least partially, via activation of MEK, p38, and PI-3K; 3) TGF-alpha stimulates MSCs to produce HGF by MEK, p38, PI-3K, and TNFR2-dependent mechanisms; and 4) TNFR1 acts to decrease basal TGF-alpha and TNF-alpha-stimulated HGF.


Assuntos
Receptores ErbB/metabolismo , Fator de Crescimento de Hepatócito/biossíntese , Células-Tronco Mesenquimais/metabolismo , Receptor Cross-Talk/fisiologia , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais/fisiologia , Western Blotting , Ensaio de Imunoadsorção Enzimática , Humanos , MAP Quinase Quinase Quinases/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , RNA Interferente Pequeno , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Fator de Crescimento Transformador alfa/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
Circulation ; 118(14 Suppl): S38-45, 2008 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-18824767

RESUMO

BACKGROUND: TNFR1/TNFR2 signaling may mediate different cellular and molecular responses (injury versus protection) and the balance may be affected by sex hormones. Previous studies have shown that females have improved myocardial functional recovery, TNFR1 signaling resistance, and increased SOCS3 expression after acute ischemia/reperfusion when compared with males. However, it is unknown whether the TNFR2 pathway protects the myocardium from ischemia/reperfusion injury, and if so, whether sex differences exist in TNFR2-mediated cardioprotection. Therefore, we hypothesized that (1) TNFR2 mediates myocardial protection from ischemia/reperfusion through STAT3, SOCS3, and vascular endothelial growth factor in both sexes; and (2) TNFR2 elicits greater protective signaling in females compared with males. METHODS AND RESULTS: Isolated male and female mouse hearts from TNFR2 knockout, TNFR1/2 knockout, and wild-type (C57BL/6J or B6129SF2/J; n=5 to 6/group) were subjected to 20 minutes ischemia followed by 60 minutes reperfusion. TNFR2 deficiency decreased postischemic myocardial recovery in both sexes but had a greater effect on females. The deleterious effects of TNFR2 ablation were associated with a decrease in mRNA and protein levels of SOCS3, STAT3, and vascular endothelial growth factor as well as an increase in myocardial interleukin-1-beta production in female hearts. However, a significant increase in JNK activation and interleukin-1-beta protein levels was noted in male TNFR2KO hearts after ischemia/reperfusion. Additionally, TNFR1/2 knockout decreased myocardial function in female hearts but not males. This observation was associated with a decrease in mRNA levels of SOCS3, STAT3, and vascular endothelial growth factor and an increase in myocardial p38 mitogen-activated protein kinase activation in females. CONCLUSIONS: Sex differences in the mechanisms of TNFR2-mediated cardioprotection occur by increasing STAT3, SOCS3, and vascular endothelial growth factor in females and by decreasing JNK in males.


Assuntos
Traumatismo por Reperfusão Miocárdica/prevenção & controle , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Fator de Transcrição STAT3/metabolismo , Caracteres Sexuais , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Apoptose , Regulação para Baixo , Ativação Enzimática , Feminino , Coração/fisiopatologia , Técnicas In Vitro , Interleucina-1beta/biossíntese , Masculino , Camundongos , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miocárdio/metabolismo , RNA Mensageiro/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/deficiência , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/deficiência , Recuperação de Função Fisiológica , Fator de Transcrição STAT3/genética , Transdução de Sinais , Proteínas Supressoras da Sinalização de Citocina/genética , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/genética
17.
Kidney Int ; 76(5): 500-11, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19536084

RESUMO

Ureteral obstruction results in renal fibrosis in part due to inflammatory injury. The role of interleukin-18 (IL-18), an important mediator of inflammation, in the genesis of renal fibrosis was studied using transgenic mice overexpressing human IL-18-binding protein. In addition, HK-2 cells were analyzed following direct exposure to IL-18 compared to control media. Two weeks after ureteral obstruction, the kidneys of wild-type mice had a significant increase in IL-18 production, collagen deposition, alpha-smooth muscle actin and RhoA expression, fibroblast and macrophage accumulation, chemokine expression, and transforming growth factor-beta1 (TGF-beta1) and tumor necrosis factor-alpha (TNF-alpha) production, whereas E-cadherin expression was simultaneously decreased. The transgenic mice with neutralized IL-18 activity exhibited significant reductions in these indicators of obstruction-induced renal fibrosis and epithelial- mesenchymal transition, without demonstrating alterations in TGF-beta1 or TNF-alpha activity. Similarly, the HK-2 cells exhibited increased alpha-smooth muscle actin expression and collagen production, and decreased E-cadherin expression in response to IL-18 stimulation without alterations in TNF-alpha or TGF-beta1 activity. Our study demonstrates that IL-18 is a significant mediator of obstruction-induced renal fibrosis and epithelial- mesenchymal transition independent of downstream TGF-beta1 or TNF-alpha production.


Assuntos
Células Epiteliais/patologia , Interleucina-18/fisiologia , Rim/patologia , Mesoderma/patologia , Actinas/análise , Animais , Linhagem Celular , Quimiocinas/genética , Colágeno/genética , Colágeno/metabolismo , Fibrose , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/análise , Fator de Crescimento Transformador beta1/biossíntese , Fator de Necrose Tumoral alfa/biossíntese
18.
Ann Surg ; 250(1): 19-27, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19561461

RESUMO

OBJECTIVE: To review the characteristics of stem cells that may qualify them to be useful as therapeutic agents in sepsis. SUMMARY BACKGROUND DATA: Sepsis is a devastating syndrome and is the leading cause of death among critically ill surgical patients in the United States. Despite decades of research and numerous clinical trials, little progress has been made in the development of new treatments and mortality rates are much the same as they have been for the last 20 to 30 years. As such, sepsis remains a formidable adversary for surgeons and their patients and new therapeutic modalities must continue to be explored. METHODS: Recent literature regarding sepsis and the use of stem cells in the treatment of various inflammatory conditions including sepsis was reviewed. Our experience with the use of stem cells in our own laboratory was included. RESULTS: Stem cells have recently emerged as a promising therapy for a variety of commonly encountered surgical pathologies including cardiovascular disease, neurodegenerative disease, peripheral vascular disease, renal disease, and several others. Their beneficial effects are owed chiefly to their abilities to home to sites of injury and inflammation, to attenuate the inflammatory response, and to accelerate tissue healing and neoangiogenesis in the face of noxious stimuli. CONCLUSIONS: Experimental evidence indicates that stem cells are immunologically responsive cells that home to sites of inflammation and tissue injury. Stem cells also secrete growth factors in response to lipopolysaccharide and tumor necrosis factor that may limit apoptosis and organ injury. Stem cells represent an endogenous therapeutic strategy that may be enhanced for maximum clinical benefit.


Assuntos
Sepse/imunologia , Células-Tronco/fisiologia , Animais , Humanos , Sepse/fisiopatologia , Sepse/terapia , Transplante de Células-Tronco , Células-Tronco/imunologia
19.
J Surg Res ; 152(2): 325-30, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18805555

RESUMO

BACKGROUND: Bone marrow stem cells (BMSCs) may be a novel treatment modality for organ ischemia, possibly through beneficial paracrine mechanisms. However, stem cells from older hosts exhibit decreased function during stress. We therefore hypothesized that (1) BMSCs derived from neonatal hosts would provide protection to ischemic myocardium, and (2) neonatal stem cells would enhance postischemic myocardial recovery above that seen with adult stem cell therapy. MATERIALS AND METHODS: Female adult Sprague Dawley rat hearts were subjected to an ischemia/reperfusion protocol via Langendorff isolated heart preparation (15 min equilibration, 25 min ischemia, and 60 min reperfusion). BMSCs were harvested from adult and neonatal mice and cultured through several passages under normal conditions (37 degrees C, 5% CO(2)/air). Immediately prior to ischemia, 1 million adult or neonatal BMSCs were infused into the coronary circulation. Cardiac functional parameters were continuously recorded. RESULTS: Pretreatment with adult BMSCs significantly increased postischemic myocardial recovery as noted by improved left ventricular developed pressure, end diastolic pressure, contractility, and rate of relaxation. Neonatal stem cells, however, did not cause any noticeable improvement in myocardial functional parameters following ischemia. CONCLUSION: Neonatal and adult BMSCs are distinctly different in the degree of beneficial tissue protection that they can provide. The data herein suggests that a critical age exists as to when stem cells become fully activated to provide their beneficial protective properties. Defining the genes that initiate these protective properties may allow for genetic amplification of beneficial signals, and the generation of "super stem cells" that provide maximum protection to ischemic tissues.


Assuntos
Transplante de Medula Óssea/métodos , Doença das Coronárias/cirurgia , Coração/fisiologia , Traumatismo por Reperfusão/prevenção & controle , Transplante de Células-Tronco/métodos , Envelhecimento/fisiologia , Animais , Animais Recém-Nascidos , Débito Cardíaco , Feminino , Fêmur , Técnicas In Vitro , Transplante de Células-Tronco Mesenquimais/métodos , Camundongos , Camundongos Endogâmicos C57BL , Isquemia Miocárdica/cirurgia , Ratos , Ratos Sprague-Dawley , Células Estromais/transplante , Tíbia , Coleta de Tecidos e Órgãos/métodos
20.
J Surg Res ; 152(2): 319-24, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18511080

RESUMO

BACKGROUND: Stem cell therapy is a promising treatment modality for injured cardiac tissue. A novel mechanism for this cardioprotection may include paracrine actions. Our lab has recently shown that gender differences exist in mesenchymal stem cell (MSC) paracrine function. Estrogen is implicated in the cardioprotection found in females. It remains unknown whether 17beta-estradiol (E2) affects MSC paracrine function and whether E2-treated MSCs may better protect injured cardiac tissue. We hypothesize that E2-exposed MSCs infused into hearts prior to ischemia may demonstrate increased vascular endothelial growth factor (VEGF) production and greater protection of myocardial function compared to untreated MSCs. MATERIALS AND METHODS: Untreated and E2-treated MSCs were isolated, cultured, and plated and supernatants were harvested for VEGF assay (enzyme-linked immunosorbent assay). Adult male Sprague-Dawley rat hearts (n = 13) were isolated and perfused via Langendorff model and subjected to 15 min equilibration, 25 min warm global ischemia, and 40 min reperfusion. Hearts were randomly assigned to perfusate vehicle, untreated male MSC, or E2-treated male MSC. Transcoronary delivery of 1 million MSCs was performed immediately prior to ischemia in experimental hearts. RESULTS: E2-treated MSCs provoked significantly more VEGF production than untreated MSCs (933.2 +/- 64.9 versus 595.8 +/- 10.7 pg/mL). Postischemic recovery of left ventricular developed pressure was significantly greater in hearts infused with E2-treated MSCs (66.9 +/- 3.3%) than untreated MSCs (48.7 +/- 3.7%) and vehicle (28.9 +/- 4.6%) at end reperfusion. There was also greater recovery of the end diastolic pressure with E2-treated MSCs than untreated MSCs and vehicle. CONCLUSIONS: Preischemic infusion of MSCs protects myocardial function and viability. E2-treated MSCs may enhance this paracrine protection, which suggests that ex vivo modification of MSCs may improve therapeutic outcome.


Assuntos
Estradiol/farmacologia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/fisiologia , Isquemia Miocárdica/fisiopatologia , Animais , Técnicas de Cultura de Células/métodos , Diástole/efeitos dos fármacos , Diástole/fisiologia , Coração/efeitos dos fármacos , Coração/fisiologia , Coração/fisiopatologia , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Isquemia Miocárdica/cirurgia , Reperfusão Miocárdica/métodos , Ratos , Ratos Sprague-Dawley , Fator A de Crescimento do Endotélio Vascular/metabolismo , Função Ventricular Esquerda/efeitos dos fármacos , Função Ventricular Esquerda/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA