Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 11(12): e0168572, 2016.
Article in English | MEDLINE | ID: mdl-28030556

ABSTRACT

BACKGROUND: Obstructive sleep apnea (OSA) is associated with the progression of non-alcoholic fatty liver disease (NAFLD) to steatohepatitis and fibrosis. This progression correlates with the severity of OSA-associated hypoxia. In mice with diet induced obesity, hepatic steatosis leads to liver tissue hypoxia, which worsens with exposure to intermittent hypoxia. Emerging data has implicated hepatocyte cell signaling as an important factor in hepatic fibrogenesis. We hypothesized that hepatocyte specific knockout of the oxygen sensing α subunit of hypoxia inducible factor-1 (HIF-1), a master regulator of the global response to hypoxia, may be protective against the development of liver fibrosis. METHODS: Wild-type mice and mice with hepatocyte-specific HIF-1α knockout (Hif1a-/-hep) were fed a high trans-fat diet for six months, as a model of NAFLD. Hepatic fibrosis was evaluated by Sirius red stain and hydroxyproline assay. Liver enzymes, fasting insulin, and hepatic triglyceride content were also assessed. Hepatocytes were isolated from Hif1a-/-hep mice and wild-type controls and were exposed to sustained hypoxia (1% O2) or normoxia (16% O2) for 24 hours. The culture media was used to reconstitute type I collagen and the resulting matrices were examined for collagen cross-linking. RESULTS: Wild-type mice on a high trans-fat diet had 80% more hepatic collagen than Hif1a-/-hep mice (2.21 µg collagen/mg liver tissue, versus 1.23 µg collagen/mg liver tissue, p = 0.03), which was confirmed by Sirius red staining. Body weight, liver weight, mean hepatic triglyceride content, and fasting insulin were similar between groups. Culture media from wild-type mouse hepatocytes exposed to hypoxia allowed for avid collagen cross-linking, but very little cross-linking was seen when hepatocytes were exposed to normoxia, or when hepatocytes from Hif1a-/-hep mice were used in hypoxia or normoxia. CONCLUSIONS: Hepatocyte HIF-1 mediates an increase in liver fibrosis in a mouse model of NAFLD, perhaps due to liver tissue hypoxia in hepatic steatosis. HIF-1 is necessary for collagen cross-linking in an in vitro model of fibrosis.


Subject(s)
Disease Models, Animal , Hepatocytes/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Hypoxia/pathology , Liver Cirrhosis/pathology , Non-alcoholic Fatty Liver Disease/complications , Animals , Cells, Cultured , Hepatocytes/metabolism , Hypoxia/etiology , Hypoxia/metabolism , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Male , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/physiopathology
2.
Oncotarget ; 6(14): 11959-78, 2015 May 20.
Article in English | MEDLINE | ID: mdl-26003168

ABSTRACT

Stem cell function declines with age largely due to the biochemical imbalances in their tissue niches, and this work demonstrates that aging imposes an elevation in transforming growth factor ß (TGF-ß) signaling in the neurogenic niche of the hippocampus, analogous to the previously demonstrated changes in the myogenic niche of skeletal muscle with age. Exploring the hypothesis that youthful calibration of key signaling pathways may enhance regeneration of multiple old tissues, we found that systemically attenuating TGF-ß signaling with a single drug simultaneously enhanced neurogenesis and muscle regeneration in the same old mice, findings further substantiated via genetic perturbations. At the levels of cellular mechanism, our results establish that the age-specific increase in TGF-ß1 in the stem cell niches of aged hippocampus involves microglia and that such an increase is pro-inflammatory both in brain and muscle, as assayed by the elevated expression of ß2 microglobulin (B2M), a component of MHC class I molecules. These findings suggest that at high levels typical of aged tissues, TGF-ß1 promotes inflammation instead of its canonical role in attenuating immune responses. In agreement with this conclusion, inhibition of TGF-ß1 signaling normalized B2M to young levels in both studied tissues.


Subject(s)
Aging/physiology , Hippocampus/drug effects , Muscle Development/drug effects , Neurogenesis/drug effects , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Blotting, Western , Female , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Neural Stem Cells/drug effects , Polymerase Chain Reaction , Rats , Rats, Inbred F344 , Stem Cell Niche/drug effects , Stem Cell Niche/physiology
3.
Stem Cells ; 33(5): 1577-88, 2015 May.
Article in English | MEDLINE | ID: mdl-25538007

ABSTRACT

Hippocampal neurogenesis, the product of resident neural stem cell proliferation and differentiation, persists into adulthood but decreases with organismal aging, which may contribute to the age-related decline in cognitive function. The mechanisms that underlie this decrease in neurogenesis are not well understood, although evidence in general indicates that extrinsic changes in an aged stem cell niche can contribute to functional decline in old stem cells. Bone morphogenetic protein (BMP) family members are intercellular signaling proteins that regulate stem and progenitor cell quiescence, proliferation, and differentiation in various tissues and are likewise critical regulators of neurogenesis in young adults. Here, we establish that BMP signaling increases significantly in old murine hippocampi and inhibits neural progenitor cell proliferation. Furthermore, direct in vivo attenuation of BMP signaling via genetic and transgenic perturbations in aged mice led to elevated neural stem cell proliferation, and subsequent neurogenesis, in old hippocampi. Such advances in our understanding of mechanisms underlying decreased hippocampal neurogenesis with age may offer targets for the treatment of age-related cognitive decline.


Subject(s)
Aging/metabolism , Bone Morphogenetic Proteins/metabolism , Hippocampus/metabolism , Neurogenesis , Signal Transduction , Animals , Cell Proliferation , Endothelial Cells/metabolism , Integrases/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Phosphorylation , Protein Transport , Smad Proteins/metabolism
4.
Aging (Albany NY) ; 6(8): 602-20, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25109702

ABSTRACT

Adult stem cells grow poorly in vitro compared to embryonic stem cells, and in vivo stem cell maintenance and proliferation by tissue niches progressively deteriorates with age. We previously reported that factors produced by human embryonic stem cells (hESCs) support a robust regenerative capacity for adult and old mouse muscle stem/progenitor cells. Here we extend these findings to human muscle progenitors and investigate underlying molecular mechanisms. Our results demonstrate that hESC-conditioned medium enhanced the proliferation of mouse and human muscle progenitors. Furthermore, hESC-produced factors activated MAPK and Notch signaling in human myogenic progenitors, and Delta/Notch-1 activation was dependent on MAPK/pERK. The Wnt, TGF-ß and BMP/pSmad1,5,8 pathways were unresponsive to hESC-produced factors, but BMP signaling was dependent on intact MAPK/pERK. c-Myc, p57, and p18 were key effectors of the enhanced myogenesis promoted by the hECS factors. To define some of the active ingredients of the hESC-secretome which may have therapeutic potential, a comparative proteomic antibody array analysis was performed and identified several putative proteins, including FGF2, 6 and 19 which as ligands for MAPK signaling, were investigated in more detail. These studies emphasize that a "youthful" signaling of multiple signaling pathways is responsible for the pro-regenerative activity of the hESC factors.


Subject(s)
Embryonic Stem Cells/metabolism , Muscle Development/physiology , Signal Transduction/physiology , Animals , Culture Media, Conditioned , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mice , Receptor, Notch1/metabolism , Transforming Growth Factor beta/metabolism
5.
Aging (Albany NY) ; 5(5): 357-72, 2013 May.
Article in English | MEDLINE | ID: mdl-23793469

ABSTRACT

This work builds upon our findings that proteins secreted by hESCs exhibit pro-regenerative activity, and determines that hESC-conditioned medium robustly enhances the proliferation of both muscle and neural progenitor cells. Importantly, this work establishes that it is the proteins that bind heparin which are responsible for the pro-myogenic effects of hESC-conditioned medium, and indicates that this strategy is suitable for enriching the potentially therapeutic factors. Additionally, this work shows that hESC-secreted proteins act independently of the mitogen FGF-2, and suggests that FGF-2 is unlikely to be a pro-aging molecule in the physiological decline of old muscle repair. Moreover, hESC-secreted factors improve the viability of human cortical neurons in an Alzheimer's disease (AD) model, suggesting that these factors can enhance the maintenance and regeneration of multiple tissues in the aging body.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental/physiology , Heparin/metabolism , Aging/metabolism , Aging/physiology , Animals , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/pharmacology , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Heparin/chemistry , Humans , Mice , Mice, Inbred C57BL , Muscle Development/drug effects , Muscle, Skeletal/injuries , Muscle, Skeletal/physiology , Neurons/drug effects , Neurons/physiology , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/drug effects , Satellite Cells, Skeletal Muscle/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...