Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
J Endod ; 48(7): 872-879, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35447294

RESUMEN

INTRODUCTION: Regeneration of the pulp-dentin complex hinges on functionally diverse growth factors, cytokines, chemokines, signaling molecules, and other secreted factors collectively referred to as trophic factors. The delivery of exogenous factors and the induced release of endogenous dentin-bound factors by conditioning agents have been explored toward these goals. The aim of this study was to investigate a promising regeneration strategy based on the conditioning of dental pulp cells (DPCs) with polyinosinic-polycytidylic acid (poly[I:C]) for the amplification of endogenous trophic factors. METHODS: DPCs were isolated from human dental pulps, propagated in culture, and treated with an optimized dose of poly(I:C). The 3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyl tetrazolium bromide assay and metabolite analysis were conducted to monitor the cytotoxicity of poly(I:C). Enzyme-linked immunosorbent assays and quantitative polymerase chain reaction assays were performed to quantify the induction of trophic factors in response to DPC conditioning. Statistical significance was P < .05. RESULTS: The analysis of 32 trophic factors involved in Wnt signaling, cell migration and chemotaxis, cell proliferation and differentiation, extracellular matrix remodeling and angiogenesis, and immunoregulation revealed that DPCs abundantly express many trophic factors including AMF, BDNF, BMP2, FGF1, FGF2, FGF5, HGF, MCP1, NGF, SDF1, TGFß1, TIMP1, TIMP2, TIMP3, and VEGFA, many of which were further induced by DPC conditioning; induction was significant for BDNF, EGF, HGF, LIF, MCP1, SDF1, IL6, IL11, MMP9, and TIMP1. Both DPC proliferation and lactate production (P < .05) were inhibited by 8 µg/mL poly(I:C) relative to the control. CONCLUSIONS: In vitro DPC conditioning through poly(I:C) activation of toll-like receptor 3 led to the amplification of trophic factors involved in tissue repair. The strategy offers promise for endodontic regeneration and tooth repair and warrants further investigation.


Asunto(s)
Pulpa Dental , Poli I-C , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Diferenciación Celular , Células Cultivadas , Humanos , Poli I-C/metabolismo , Poli I-C/farmacología , Receptor Toll-Like 3/metabolismo
3.
Biochem Biophys Res Commun ; 529(4): 1101-1105, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32819571

RESUMEN

The current gold standard for diagnosis of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) is through a liver biopsy, and there is an urgent need to develop non-invasive methods for early detection. We previously demonstrated metabolic remodeling in the mouse fatty liver, which is marked by increased hepatic expression and activities of phosphoglucose isomerase (PGI) and several other glycolytic enzymes. Since PGI is actively transported out of the cell, acting as a multifunctional cytokine referred to as autocrine motility factor (AMF), we explored the possibility that PGI secreted from the fatty liver may be targeted for early detection of the silent disease. We report here that mice with NASH exhibited significantly elevated serum PGI enzyme activities compared to normal control (P < 0.005). We further confirmed the finding using serum/plasma samples (n = 73) collected from a cohort of NASH patients who were diagnosed according to Kleiner's criteria, showing a normal mean PGI of 19.5 ± 8.8 IU/L and patient mean PGI of 105.6 ± 79.9 IU/L (P < 0.005). In addition, elevated blood PGI in NASH patients coincided with increased blood L-lactate. Cell culture experiments were then conducted to delineate the PGI-lactate axis, which revealed that treatment of HepG2 cells with recombinant PGI protein stimulated glycolysis and lactate output, suggesting that the disease-induced PGI likely contributed to the increased lactate in NASH patients. Taken together, the preclinical and clinical data validate secreted PGI as a useful biomarker of the fatty liver that can be easily screened at the point of care.


Asunto(s)
Glucosa-6-Fosfato Isomerasa/metabolismo , Enfermedad del Hígado Graso no Alcohólico/enzimología , Adolescente , Animales , Biomarcadores/metabolismo , Niño , Preescolar , Glucosa-6-Fosfato Isomerasa/sangre , Células Hep G2 , Humanos , Ácido Láctico/metabolismo , Modelos Lineales , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/sangre
4.
Trends Cell Mol Biol ; 13: 47-56, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30853754

RESUMEN

Wnt ligands regulate metabolic pathways, and dysregulation of Wnt signaling contributes to chronic inflammatory disease. A knowledge gap exists concerning the role of aberrant Wnt signaling in non-alcoholic steatohepatitis (NASH), which exhibits metabolic syndrome and inflammation. Using a mouse model of methionine-choline deficient diet (MCDD)-induced NASH, we investigated the Wnt signaling pathways in relation to hepatic glucose oxidation. Mice fed the MCD diet for 6 weeks developed prominent NASH marked by macrovesicular steatosis, inflammation and lipid peroxidation. qPCR analysis reveals differential hepatic expression of canonical and non-canonical Wnt ligands. While expression of Wnt3a was decreased in NASH vs chow diet control, expression of Wnt5a and Wnt11 were increased 3 fold and 15 fold, respectively. Consistent with activation of non-canonical Wnt signaling, expression of the alternative Wnt receptor ROR2 was increased 5 fold with no change in LRP6 expression. Activities of the metabolic enzymes glucokinase, phosphoglucoisomerase, glyceraldehyde-3-phosphate dehydrogenase, pyruvate kinase, and pyruvate dehydrogenase were all elevated by MCDD. NASH-driven glucose oxidation was accompanied by a 6-fold increase in lactate dehydrogenase (LDH)-B with no change in LDH-A. In addition, glucose-6-phosphate dehydrogenase, the regulatory and NADPH-producing enzyme of the pentose phosphate pathway, was elevated in NASH. These data support a role of accelerated glucose oxidation in the development of NASH, which may be driven by non-canonical Wnt signaling.

5.
Am J Physiol Cell Physiol ; 311(5): C710-C719, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27605451

RESUMEN

Secreted Frizzled-related protein 2 (sFRP2) plays a key role in chronic fibrosis after myocardial infarction and in heart failure. The aim of this study was to elucidate the mechanisms through which sFRP2 may regulate the growth and extracellular matrix (ECM) remodeling of adult mouse cardiac fibroblasts (CFs). We found that sFRP2 activates CFs in part through canonical Wnt/ß-catenin signaling, as evidenced by increased expression of Axin2 and Wnt3a, but not Wnt5a, as well as accumulation of nuclear ß-catenin. In response to sFRP2, CFs exhibited robust cell proliferation associated with increased glucose consumption and lactate production, a phenomenon termed "the Warburg effect" in oncology. The coupling between CF expansion and anaerobic glycolysis is marked by upregulation of glyceraldehyde-3-phosphate dehydrogenase and tissue-nonspecific alkaline phosphatase. In conjunction with these phenotypic changes, CFs accelerated ECM remodeling through upregulation of expression of the matrix metalloproteinase (MMP) 1 and MMP13 genes, two members of the collagenase subfamily, and enzyme activities of MMP2 and MMP9, two members of the gelatinase subfamily. Consistent with the induction of multiple MMPs possessing collagenolytic activities, the steady-state level of collagen type 1 in CF-spent medium was reduced by sFRP2. Analysis of non-CF cell types revealed that the multifaceted effects of sFRP2 on growth control, glucose metabolism, and ECM regulation are largely restricted to CFs and highly sensitive to Wnt signaling perturbation. The study provides a molecular framework on which the functional versatility and signaling complexity of sFRP2 in cardiac fibrosis may be better defined.


Asunto(s)
Metabolismo Energético/fisiología , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Corazón/fisiología , Proteínas de la Membrana/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Proliferación Celular/fisiología , Células Cultivadas , Colágeno Tipo I/metabolismo , Colagenasas/metabolismo , Fibrosis/metabolismo , Metaloendopeptidasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/metabolismo , Miocardio/metabolismo
6.
Am J Physiol Cell Physiol ; 309(3): C139-47, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25972450

RESUMEN

Recent studies of myocardial infarction in secreted Frizzled-related protein 2 (sFRP2) knockout mice and our hamster heart failure therapy based on sFRP2 blockade have established sFRP2 as a key profibrotic cytokine in the heart. The failing hamster heart is marked by prominent fibrosis and calcification with elevated expression of sFRP2. Noting the involvement of tissue-nonspecific alkaline phosphatase (TNAP) in bone mineralization and vascular calcification, we determined whether sFRP2 might be an upstream regulator of TNAP. Biochemical assays revealed an approximately twofold increase in the activity of TNAP and elevated levels of inorganic phosphate (Pi) in the failing heart compared with the normal heart. Neither was this change detected in the liver or hamstring muscle nor was it associated with systemic hyperphosphatemia. TNAP was readily cloned from the hamster heart and upon overexpression increased the level of extracellular but not intracellular Pi, which is consistent with the cell surface location of the ectoenzyme. In line with the previous demonstration that sFRP2 blockade attenuated fibrosis, we show here that the therapy downregulated TNAP. This in vivo finding is corroborated by the in vitro study showing that cultured cardiac fibroblasts treated with recombinant sFRP2 protein exhibited progressive increase in the expression and activity of TNAP, which was completely abrogated by cycloheximide or tunicamycin. Induction of TNAP by sFRP2 is restricted to cardiac fibroblasts among the multiple cell types examined, and was not observed with sFRP4. The current work indicates that sFRP2 may promote cardiac fibrocalcification through coordinate activation of tolloid-like metalloproteinases and TNAP.


Asunto(s)
Fosfatasa Alcalina/biosíntesis , Fibroblastos/metabolismo , Proteínas de la Membrana/biosíntesis , Miocitos Cardíacos/metabolismo , Animales , Animales Modificados Genéticamente , Células Cultivadas , Cricetinae , Fibroblastos/patología , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Miocitos Cardíacos/patología
7.
World J Stem Cells ; 6(2): 82-93, 2014 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-24772236

RESUMEN

Mesenchymal stem cell (MSC) therapy is entering a challenging phase after completion of many preclinical and clinical trials. Among the major hurdles encountered in MSC therapy are inconsistent stem cell potency, poor cell engraftment and survival, and age/disease-related host tissue impairment. The recognition that MSCs primarily mediate therapeutic benefits through paracrine mechanisms independent of cell differentiation provides a promising framework for enhancing stem cell potency and therapeutic benefits. Several MSC priming approaches are highlighted, which will likely allow us to harness the full potential of adult stem cells for their future routine clinical use.

8.
Am J Physiol Cell Physiol ; 306(6): C531-9, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24336656

RESUMEN

Progressive fibrosis is a pathological hallmark of many chronic diseases responsible for organ failure. Although there is currently no therapy on the market that specifically targets fibrosis, the dynamic fibrogenic process is known to be regulated by multiple soluble mediators that may be therapeutically intervened. The failing hamster heart exhibits marked fibrosis and increased expression of secreted Frizzled-related protein 2 (sFRP2) amenable to reversal by mesenchymal stem cell (MSC) therapy. Given the previous demonstration that sFRP2-null mice subjected to myocardial infarction exhibited reduced fibrosis and improved function, we tested whether antibody-based sFRP2 blockade might counteract the fibrogenic pathway and repair cardiac injury. Cardiomyopathic hamsters were injected intraperitoneally twice a week each with 20 µg of sFRP2 antibody. Echocardiography, histology, and biochemical analyses were performed after 1 mo. sFRP2 antibody increased left ventricular ejection fraction from 40 ± 1.2 to 49 ± 6.5%, whereas saline and IgG control exhibited a further decline to 37 ± 0.9 and 31 ± 3.2%, respectively. Functional improvement is associated with a ∼ 50% reduction in myocardial fibrosis, ∼ 65% decrease in apoptosis, and ∼ 75% increase in wall thickness. Consistent with attenuated fibrosis, both MSC therapy and sFRP2 antibody administration significantly increased the activity of myocardial matrix metalloproteinase-2. Gene expression analysis of the hamster heart and cultured fibroblasts identified Axin2 as a downstream target, the expression of which was activated by sFRP2 but inhibited by therapeutic intervention. sFRP2 blockade also increased myocardial levels of VEGF and hepatocyte growth factor (HGF) along with increased angiogenesis. These findings highlight the pathogenic effect of dysregulated sFRP2, which may be specifically targeted for antifibrotic therapy.


Asunto(s)
Anticuerpos/uso terapéutico , Proteína Axina/metabolismo , Insuficiencia Cardíaca/terapia , Proteínas de la Membrana/metabolismo , Miocardio/patología , Animales , Anticuerpos/inmunología , Apoptosis/inmunología , Células Cultivadas , Cricetinae , Fibrosis , Corazón , Insuficiencia Cardíaca/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Infarto del Miocardio/patología , Miocardio/metabolismo , Neovascularización Fisiológica , Transducción de Señal , Volumen Sistólico/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Función Ventricular Izquierda/fisiología
9.
Biomaterials ; 34(33): 8213-22, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23891082

RESUMEN

Mesenchymal stem/stromal cells (MSCs) are an important candidate for cell-based therapy since they can be easily isolated and expanded, secrete beneficial paracrine factors, and differentiate into multiple lineages. Since the endothelium at sites of injury and inflammation often express adhesion molecules belonging to the selectin family, methods to endow MSCs with selectin-ligands can enhance the efficacy of cell delivery and tissue engraftment. Here, we describe a construct 19Fc[FUT7(+)], where the first 19 amino acids of the pan-selectin ligand PSGL-1 (P-selectin glycoprotein ligand-1) was fused to a human IgG1. When expressed in HEK293T cells over-expressing the α(1,3)fucosyltransferase FUT7, 19Fc[FUT7(+)] is decorated by a core-2 sialyl Lewis-X sialofucosylated O-glycan. The non-covalent coupling of this protein onto MSC surface using palmitated protein G (PPG) enhanced cell binding to E- and P-selectin under hydrodynamic shear, without altering MSC multipotency. MSCs functionalized with 19Fc[FUT7(+)] were captured/tethered onto stimulated endothelial cell monolayers at wall shear stresses up to 4 dyn/cm(2). Once captured, the cells rolled robustly up to the highest shear stress tested, 10 dyn/cm(2). Unlike previous work where MSCs could only be captured onto selectin-bearing substrates at low or no-flow conditions, the current work presents a 'glycan engineering' strategy to enable leukocyte-like capture and rolling.


Asunto(s)
Glicoproteínas de Membrana/farmacología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Fosfatasa Alcalina/metabolismo , Adhesión Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Selectina E/metabolismo , Humanos , Microscopía Confocal , Selectina-P/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
10.
Am J Physiol Cell Physiol ; 303(10): C1021-33, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22843797

RESUMEN

Clinical trials of bone marrow mesenchymal stem cell (MSC) therapy have thus far demonstrated moderate and inconsistent benefits, indicating an urgent need to improve therapeutic efficacy. Although administration of sufficient cells is necessary to achieve maximal therapeutic benefits, documented MSC clinical trials have largely relied on injections of ∼1 × 10(6) cells/kg, which appears too low to elicit a robust therapeutic response according to published preclinical studies. However, repeated cell passaging necessary for large-scale expansion of MSC causes cellular senescence and reduces stem cell potency. Using the RNA mimetic polyinosinic-polycytidylic acid [poly(I:C)] to engage MSC Toll-like receptor 3 (TLR3), we found that poly(I:C), signaling through multiple mitogen-activated protein kinase pathways, induced therapeutically relevant trophic factors such as interleukin-6-type cytokines, stromal-derived factor 1, hepatocyte growth factor, and vascular endothelial growth factor while slightly inhibiting the proliferation and migration potentials of MSC. At the suboptimal injection dose of 1 × 10(6) cells/kg, poly(I:C)-treated MSC, but not untreated MSC, effectively stimulated regeneration of the failing hamster heart 1 mo after cell administration. The regenerating heart exhibited increased CD34(+)/Ki67(+) and CD34(+)/GATA4(+) progenitor cells in the presence of decreased inflammatory cells and cytokines. Cardiac functional improvement was associated with a ∼50% reduction in fibrosis, a ∼40% reduction in apoptosis, and a ∼55% increase in angiogenesis, culminating in prominent cardiomyogenesis evidenced by abundant distribution of small myocytes and a ∼90% increase in wall thickening. These functional, histological, and molecular characterizations thus establish the utility of TLR3 engagement for enabling the low-dose MSC therapy that may be translated to more efficacious clinical applications.


Asunto(s)
Cardiomiopatías/terapia , Células Madre Mesenquimatosas/fisiología , Trasplante de Células Madre , Receptor Toll-Like 3/metabolismo , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/fisiología , Proliferación Celular , Cricetinae , Regulación de la Expresión Génica/fisiología , Péptidos y Proteínas de Señalización Intercelular , Inductores de Interferón/farmacología , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Miocardio/patología , Poli I-C/farmacología , Porcinos , Receptor Toll-Like 3/genética
11.
World J Stem Cells ; 4(12): 120-4, 2012 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-23516128

RESUMEN

Mesenchymal stem cell (MSC) therapy is entering a new era shifting the focus from initial feasibility study to optimization of therapeutic efficacy. However, how MSC therapy facilitates tissue regeneration remains incompletely characterized. Consistent with the emerging notion that secretion of multiple growth factors/cytokines (trophic factors) by MSC provides the underlying tissue regenerative mechanism, the recent study by Bai et al demonstrated a critical therapeutic role of MSC-derived hepatocyte growth factor (HGF) in two animal models of multiple sclerosis (MS), which is a progressive autoimmune disorder caused by damage to the myelin sheath and loss of oligodendrocytes. Although current MS therapies are directed toward attenuation of the immune response, robust repair of myelin sheath likely requires a regenerative approach focusing on long-term replacement of the lost oligodendrocytes. This approach appears feasible because adult organs contain various populations of multipotent resident stem/progenitor cells that may be activated by MSC trophic factors as demonstrated by Bai et al This commentary highlights and discusses the major findings of their studies, emphasizing the anti-inflammatory function and trophic cross-talk mechanisms mediated by HGF and other MSC-derived trophic factors in sustaining the treatment benefits. Identification of multiple functionally synergistic trophic factors, such as HGF and vascular endothelial growth factor, can eventually lead to the development of efficacious cell-free therapeutic regimens targeting a broad spectrum of degenerative conditions.

12.
Am J Physiol Heart Circ Physiol ; 301(6): H2422-32, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21963833

RESUMEN

The skeletal muscle is endowed with an impressive ability to regenerate after injury, and this ability is coupled to paracrine production of many trophic factors possessing cardiovascular benefits. Taking advantage of this humoral capacity of the muscle, we recently demonstrated an extracardiac therapeutic regimen based on intramuscular delivery of VEGF-A(165) for repair of the failing hamster heart. This distal organ repair mechanism activates production from the injected hamstring of many trophic factors, among which stromal-derived factor-1 (SDF1) prominently mobilized multi-lineage progenitor cells expressing CXCR4 and their recruitment to the heart. The mobilized bone marrow progenitor cells express the cardiac transcription factors myocyte enhancer factor 2c and GATA4 and several major trophic factors, most notably IGF1 and VEGF. SDF1 blockade abrogated myocardial recruitment of CXCR4(+) and c-kit(+) progenitor cells with an insignificant effect on the hematopoietic progenitor lineage. The knockdown of cardiac progenitor cells led to deprivation of myocardial trophic factors, resulting in compromised cardiomyogenesis and angiogenesis. However, the VEGF-injected hamstring continued to synthesize cardioprotective factors, contributing to moderate myocardial tissue viability and function even in the presence of SDF1 blockade. These findings thus uncover two distinct but synergistic cardiac therapeutic mechanisms activated by intramuscular VEGF. Whereas the SDF1/CXCR4 axis activates the progenitor cell cascade and its trophic support of cardiomyogenesis intramuscularly, VEGF amplifies the skeletal muscle paracrine cascade capable of directly promoting myocardial survival independent of SDF1. Given that recent clinical trials of cardiac repair based on the use of marrow-mobilizing agents have been disappointing, the proposed dual therapeutic modality warrants further investigation.


Asunto(s)
Cardiomiopatías/tratamiento farmacológico , Quimiocina CXCL12/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Miocardio/metabolismo , Comunicación Paracrina/efectos de los fármacos , Regeneración/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/administración & dosificación , Animales , Anticuerpos/administración & dosificación , Apoptosis/efectos de los fármacos , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Cardiomiopatías/fisiopatología , Células Cultivadas , Quimiocina CXCL12/inmunología , Quimiotaxis , Cricetinae , Modelos Animales de Enfermedad , Inyecciones Intramusculares , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Miocardio/patología , Neovascularización Fisiológica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptores CXCR4/metabolismo , Proteínas Recombinantes/administración & dosificación , Factores de Tiempo , Factores de Transcripción/metabolismo , Función Ventricular Izquierda/efectos de los fármacos
13.
Circ Res ; 109(9): 1044-54, 2011 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-21885831

RESUMEN

RATIONALE: Mesenchymal stem cells (MSCs) improve function after infarction, but their mechanism of action remains unclear, and the importance of reduced scar volume, cardiomyocyte proliferation, and perfusion is uncertain. OBJECTIVE: The present study was conducted to test the hypothesis that MSCs mobilize bone marrow progenitor cells and improve function by stimulating myocyte proliferation in collateral-dependent hibe rnating myocardium. METHODS AND RESULTS: Swine with chronic hibernating myocardium received autologous intracoronary MSCs (icMSCs; ≈44 ×10(6) cells, n = 10) 4 months after instrumentation and were studied up to 6 weeks later. Physiological and immunohistochemical findings were compared with untreated hibernating animals (n = 7), sham-normal animals (n = 5), and icMSC-treated sham-normal animals (n = 6). In hibernating myocardium, icMSCs increased function (percent wall thickening of the left anterior descending coronary artery 24 ± 4% to 43 ± 5%, P < 0.05), although left anterior descending coronary artery flow reserve (adenosine/rest) remained critically impaired (1.2 ± 0.1 versus 1.2 ± 0.1). Circulating cKit+ and CD133+ bone marrow progenitor cells increased transiently after icMSC administration, with a corresponding increase in myocardial cKit+/CD133+ and cKit+/CD133- bone marrow progenitor cells (total cKit+ from 223 ± 49 to 4415 ± 866/10(6) cardiomyocytes, P < 0.05). In hibernating hearts, icMSCs increased Ki67+ cardiomyocytes (from 410 ± 83 to 2460 ± 610/10(6) nuclei, P < 0.05) and phospho-histone H3-positive cardiomyocytes (from 9 ± 5 to 116 ± 12/10(6) nuclei, P < 0.05). Myocyte nuclear number (from 75 336 ± 5037 to 114 424 ± 9564 nuclei/mm3, P < 0.01) and left ventricular mass (from 2.5 ± 0.1 to 2.8 ± 0.1 g/kg, P < 0.05) increased, yet myocytes were smaller (14.5 ± 0.4 versus 16.5 ± 0.4 µm, P < 0.05), which supports endogenous cardiomyocyte proliferation. In sham-normal animals, icMSCs increased myocardial bone marrow progenitor cells with no effect on myocyte proliferation or regional function. CONCLUSIONS: Our results indicate that icMSCs improve function in hibernating myocardium independent of coronary flow or reduced scar volume. This arises from stimulation of myocyte proliferation with increases in cKit+/CD133+ bone marrow progenitor cells and cKit+/CD133- resident stem cells, which increase myocyte number and reduce cellular hypertrophy.


Asunto(s)
Antígenos CD/metabolismo , Células de la Médula Ósea/metabolismo , Glicoproteínas/metabolismo , Corazón/fisiología , Células Madre Mesenquimatosas/fisiología , Aturdimiento Miocárdico/fisiopatología , Péptidos/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Células Madre/metabolismo , Antígeno AC133 , Animales , Células de la Médula Ósea/citología , Ciclo Celular/fisiología , Movimiento Celular/fisiología , Proliferación Celular , Quimiocinas/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Modelos Animales , Aturdimiento Miocárdico/terapia , Miocitos Cardíacos/patología , Neovascularización Fisiológica/fisiología , Células Madre/citología , Porcinos
14.
World J Stem Cells ; 2(4): 61-6, 2010 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-21031156

RESUMEN

Preclinical and clinical trials of stem cell therapy have been carried out for treating a broad spectrum of diseases using several types of adult stem cells. While encouraging therapeutic results have been obtained, much remains to be investigated regarding the best cell type to use, cell dosage, delivery route, long-term safety, clinical feasibility, and ultimately treatment cost. Logistic aspects of stem cell therapeutics remain an area that requires urgent attention from the medical community. Recent cardiovascular trial studies have demonstrated that growth factors and cytokines derived from the injected stem cells and host tissue appear to contribute largely to the observed therapeutic benefits, indicating that trophic actions rather than the multilineage potential (or stemness) of the administered stem cells may provide the underlying tissue healing power. However, the capacity for trophic factor production can be aberrantly downregulated as seen in human heart disease. Skeletal muscle is a dynamic tissue with an impressive ability to continuously respond to environmental stimuli. Indeed, a relation exists between active skeletal muscle and low cardiovascular risk, highlighting the critical link between the skeletal muscle and cardiovascular systems. Adding to this notion are recent studies showing that stem cells injected into skeletal muscle can rescue the failing rodent heart through activation of the muscle trophic factor network and mobilization of bone marrow multilineage progenitor cells. However, aging and disease can adversely affect the host tissue into which stem cells are injected. A better understanding of the host tissue response in stem cell therapy is necessary to advance the field and bridge the gap between preclinical and clinical findings.

15.
Am J Physiol Heart Circ Physiol ; 299(5): H1428-38, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20852053

RESUMEN

We recently demonstrated a cardiac therapeutic regimen based on injection of bone marrow mesenchymal stem cells (MSCs) into the skeletal muscle. Although the injected MSCs were trapped in the local musculature, the extracardiac cell delivery approach repaired the failing hamster heart. This finding uncovers a tissue repair mechanism mediated by trophic factors derived from the injected MSCs and local musculature that can be explored for minimally invasive stem cell therapy. However, the trophic factors involved in cardiac repair and their actions remain largely undefined. We demonstrate here a role of MSC-derived IL-6-type cytokines in cardiac repair through engagement of the skeletal muscle JAK-STAT3 axis. The MSC IL-6-type cytokines activated JAK-STAT3 signaling in cultured C2C12 skeletal myocytes and caused increased expression of the STAT3 target genes hepatocyte growth factor (HGF) and VEGF, which was inhibited by glycoprotein 130 (gp130) blockade. These in vitro findings were corroborated by in vivo studies, showing that the MSC-injected hamstrings exhibited activated JAK-STAT3 signaling and increased growth factor/cytokine production. Elevated host tissue growth factor levels were also detected in quadriceps, liver, and brain, suggesting a possible global trophic effect. Paracrine actions of these host tissue-derived factors activated the endogenous cardiac repair mechanisms in the diseased heart mediated by Akt, ERK, and JAK-STAT3. Administration of the cell-permeable JAK-STAT inhibitor WP1066 abrogated MSC-mediated host tissue growth factor expression and functional improvement. The study illustrates that the host tissue trophic factor network can be activated by MSC-mediated JAK-STAT3 signaling for tissue repair.


Asunto(s)
Factor de Crecimiento de Hepatocito/metabolismo , Quinasas Janus/metabolismo , Células Madre Mesenquimatosas/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Células Cultivadas , Cricetinae , Citocinas/metabolismo , Quinasas Janus/antagonistas & inhibidores , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Modelos Animales , Fibras Musculares Esqueléticas/citología , Miocardio/metabolismo , Piridinas/farmacología , Factor de Transcripción STAT3/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Tirfostinos/farmacología
16.
Biochem Biophys Res Commun ; 390(3): 834-8, 2009 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-19836359

RESUMEN

We recently demonstrated a novel effective therapeutic regimen for treating hamster heart failure based on injection of bone marrow mesenchymal stem cells (MSCs) or MSC-conditioned medium into the skeletal muscle. The work highlights an important cardiac repair mechanism mediated by the myriad of trophic factors derived from the injected MSCs and local musculature that can be explored for non-invasive stem cell therapy. While this therapeutic regimen provides the ultimate proof that MSC-based cardiac repair is mediated by the trophic actions independent of MSC differentiation or stemness, the trophic factors responsible for cardiac regeneration after MSC therapy remain largely undefined. Toward this aim, we took advantage of the finding that human and porcine MSCs exhibit species-related differences in expression of trophic factors. We demonstrate that human MSCs when compared to porcine MSCs express and secrete 5-fold less vascular endothelial growth factor (VEGF) in conditioned medium (40+/-5 and 225+/-17 pg/ml VEGF, respectively). This deficit in VEGF output was associated with compromised cardiac therapeutic efficacy of human MSC-conditioned medium. Over-expression of VEGF in human MSCs however completely restored the therapeutic potency of the conditioned medium. This finding indicates VEGF as a key therapeutic trophic factor in MSC-mediated myocardial regeneration, and demonstrates the feasibility of human MSC therapy using trophic factor-based cell-free strategies, which can eliminate the concern of potential stem cell transformation.


Asunto(s)
Células de la Médula Ósea/metabolismo , Insuficiencia Cardíaca/cirugía , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Cricetinae , Medios de Cultivo Condicionados/metabolismo , Medios de Cultivo Condicionados/farmacología , Corazón/efectos de los fármacos , Corazón/fisiología , Humanos , Masculino , Regeneración/efectos de los fármacos , Porcinos
17.
Am J Physiol Regul Integr Comp Physiol ; 297(5): R1503-15, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19759338

RESUMEN

Skeletal muscle produces a myriad of mitogenic factors possessing cardiovascular regulatory effects that can be explored for cardiac repair. Given the reported findings that VEGF may modulate muscle regeneration, we investigated the therapeutic effects of chronic injections of low doses of human recombinant VEGF-A(165) (0.1-1 microg/kg) into the dystrophic hamstring muscle in a hereditary hamster model of heart failure and muscular dystrophy. In vitro, VEGF stimulated proliferation, migration, and growth factor production of cultured C2C12 skeletal myocytes. VEGF also induced production of HGF, IGF2, and VEGF by skeletal muscle. Analysis of skeletal muscle revealed an increase in myocyte nuclear [531 +/- 12 VEGF 1 microg/kg vs. 364 +/- 19 for saline (number/mm(2)) saline] and capillary [591 +/- 80 VEGF 1 microg/kg vs. 342 +/- 21 for saline (number/mm(2))] densities. Skeletal muscle analysis revealed an increase in Ki67(+) nuclei in the VEGF 1 microg/kg group compared with saline. In addition, VEGF mobilized c-kit(+), CD31(+), and CXCR4(+) progenitor cells. Mobilization of progenitor cells was consistent with higher SDF-1 concentrations found in hamstring, plasma, and heart in the VEGF group. Echocardiogram analysis demonstrated improvement in left ventricular ejection fraction (0.60 +/- 0.02 VEGF 1 microg/kg vs. 0.45 +/- 0.01 mm for saline) and an attenuation in ventricular dilation [5.59 +/- 0.12 VEGF 1 microg/kg vs. 6.03 +/- 0.09 for saline (mm)] 5 wk after initiating therapy. Hearts exhibited higher cardiomyocyte nuclear [845 +/- 22 VEGF 1 microg/kg vs. 519 +/- 40 for saline (number/mm(2))] and capillary [2,159 +/- 119 VEGF 1 microg/kg vs. 1,590 +/- 66 for saline (number/mm(2))] densities. Myocardial analysis revealed approximately 2.5 fold increase in Ki67+ cells and approximately 2.8-fold increase in c-kit(+) cells in the VEGF group, which provides evidence for cardiomyocyte regeneration and progenitor cell expansion. This study provides novel evidence of a salutary effect of VEGF in the cardiomyopathic hamster via induction of myogenic growth factor production by skeletal muscle and mobilization of progenitor cells, which resulted in attenuation of cardiomyopathy and repair of the heart.


Asunto(s)
Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/patología , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Fibras Musculares Esqueléticas/patología , Miocitos Cardíacos/patología , Células Madre/patología , Factor A de Crecimiento Endotelial Vascular/uso terapéutico , Angiopoyetina 1/metabolismo , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CXCL12/metabolismo , Cricetinae , Modelos Animales de Enfermedad , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Corazón/fisiología , Insuficiencia Cardíaca/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Inyecciones Intramusculares , Masculino , Ratones , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/fisiología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Regeneración/efectos de los fármacos , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Volumen Sistólico/efectos de los fármacos , Volumen Sistólico/fisiología , Factor A de Crecimiento Endotelial Vascular/administración & dosificación , Factor A de Crecimiento Endotelial Vascular/farmacología
18.
Transplantation ; 87(9): 1275-82, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19424025

RESUMEN

BACKGROUND: The use of nonautologous stem cells isolated from healthy donors for stem-cell therapy is an attractive approach, because the stem cells can be culture expanded in advance, thoroughly tested, and formulated into off-the-shelf medicine. However, human leukocyte antigen compatibility and related immunosuppressive protocols can compromise therapeutic efficacy and cause unwanted side effects. METHODS: Mesenchymal stem cells (MSCs) have been postulated to possess unique immune regulatory function. We explored the immunomodulatory property of human and porcine MSCs for the treatment of delta-sarcoglycan-deficient dystrophic hamster muscle without immunosuppression. Circulating and tissue markers of inflammation were analyzed. Muscle regeneration and stem-cell fate were characterized. RESULTS: Total white blood cell counts and leukocyte-distribution profiles were similar among the saline- and MSC-injected dystrophic hamsters 1 month posttreatment. Circulating levels of immunoglobulin A, vascular cell adhesion molecule-1, myeloperoxidase, and major cytokines involved in inflammatory response were not elevated by MSCs, nor were expression of the leukocyte common antigen CD45 and the cytokine transcriptional activator NF-kappaB in the injected muscle. Treated muscles exhibited increased cell-cycle activity and attenuated oxidative stress. Injected MSCs were found to be trapped in the musculature, contribute to both preexisting and new muscle fibers, and mediates capillary formation. CONCLUSIONS: Intramuscular injection of nonautologous MSCs can be safely used for the treatment of dystrophic muscle in immunocompetent hosts without inflaming the host immune system.


Asunto(s)
Células de la Médula Ósea/citología , Trasplante de Células Madre Mesenquimatosas/métodos , Músculo Esquelético/fisiopatología , Distrofia Muscular Animal/cirugía , Trasplante Heterólogo/métodos , Animales , Trasplante de Médula Ósea , Cricetinae , Humanos , Inmunosupresores/uso terapéutico , Inyecciones Intramusculares , Peroxidación de Lípido , Masculino , Distrofia Muscular Animal/inmunología , Regeneración , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sarcoglicanos/deficiencia , Donantes de Tejidos
19.
Am J Physiol Heart Circ Physiol ; 296(6): H1888-97, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19395555

RESUMEN

Heart failure carries a poor prognosis with few treatment options. While myocardial stem cell therapeutic trials have traditionally relied on intracoronary infusion or intramyocardial injection routes, these cell delivery methods are invasive and can introduce harmful scar tissue, arrhythmia, calcification, or microinfarction in the heart. Given that patients with heart failure are at an increased surgical risk, the development of a noninvasive stem cell therapeutic approach is logistically appealing. Taking advantage of the trophic effects of bone marrow mesenchymal stem cells (MSCs) and using a hamster heart failure model, the present study demonstrates a novel noninvasive therapeutic regimen via the direct delivery of MSCs into the skeletal muscle bed. Intramuscularly injected MSCs and MSC-conditioned medium each significantly improved ventricular function 1 mo after MSC administration. MSCs at 4 million cells/animal increased fractional shortening by approximately 40%, enhanced capillary and myocyte nuclear density by approximately 30% and approximately 80%, attenuated apoptosis by approximately 60%, and reduced fibrosis by approximately 50%. Myocyte regeneration was evidenced by an approximately twofold increase in the expression of cell cycle markers (Ki67 and phosphohistone H(3)) and an approximately 13% reduction in mean myocyte diameter. Increased circulating levels of hepatocyte growth factor (HGF), leukemia inhibitory factor, and macrophage colony-stimulating factor were associated with the mobilization of c-Kit-positive, CD31-positive, and CD133-positive progenitor cells and a subsequent increase in myocardial c-Kit-positive cells. Trophic effects of MSCs further activated the expression of HGF, IGF-II, and VEGF in the myocardium. The work highlights a cardiac repair mechanism mediated by trophic cross-talks among the injected MSCs, bone marrow, and heart that can be explored for noninvasive stem cell therapy.


Asunto(s)
Cardiomiopatías/terapia , Insuficiencia Cardíaca/terapia , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Animales , Apoptosis/fisiología , Cardiomiopatías/patología , Cricetinae , Modelos Animales de Enfermedad , Corazón/fisiología , Insuficiencia Cardíaca/patología , Movilización de Célula Madre Hematopoyética , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/metabolismo , Inyecciones Intramusculares , Factor II del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Mutación , Miocardio/metabolismo , Miocardio/patología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Regeneración/fisiología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
Mol Cell Biochem ; 321(1-2): 45-52, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18726675

RESUMEN

Dilated cardiomyopathy (DCM) is a common cause of heart failure, and identification of early pathogenic events occurring prior to the onset of cardiac dysfunction is of mechanistic, diagnostic, and therapeutic importance. The work characterized early biochemical pathogenesis in TO2 strain hamsters lacking delta-sarcoglycan. Although the TO2 hamster heart exhibits normal function at 1 month of age (presymptomatic stage), elevated levels of myeloperoxidase, monocyte chemotactic protein-1, malondialdehyde, osteopontin, and alkaline phosphatase were evident, indicating the presence of inflammation, oxidative stress, and osteogenic phenotype. These changes were localized primarily to the myocardium. Derangement in energy metabolism was identified at the symptomatic stage (4 month), and is marked by attenuated activity and expression of pyruvate dehydrogenase E1 subunit, which catalyzes the rate-limiting step in aerobic glucose metabolism. Thus, this study illustrates differential involvement of oxidative stress, osteogenic phenotype, and glucose metabolism in the initiation and early progression of delta-sarcoglycan-null DCM.


Asunto(s)
Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/fisiopatología , Metabolismo Energético , Miocardio/metabolismo , Osteogénesis/fisiología , Estrés Oxidativo , Sarcoglicanos , Animales , Biomarcadores/metabolismo , Cardiomiopatía Dilatada/patología , Cricetinae , Cricetulus , Glucosa/metabolismo , Insulina/sangre , Leptina/sangre , Masculino , Miocardio/patología , Fenotipo , Subunidades de Proteína/metabolismo , Complejo Piruvato Deshidrogenasa/metabolismo , Sarcoglicanos/genética , Sarcoglicanos/metabolismo , Extractos de Tejidos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...