Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Sci Rep ; 10(1): 4561, 2020 03 12.
Article in English | MEDLINE | ID: mdl-32165710

ABSTRACT

Insulin resistance is associated with aging in mice and humans. We have previously shown that administration of recombinant GDF11 (rGDF11) to aged mice alters aging phenotypes in the brain, skeletal muscle, and heart. While the closely related protein GDF8 has a role in metabolism, limited data are available on the potential metabolic effects of GDF11 or GDF8 in aging. To determine the metabolic effects of these two ligands, we administered rGDF11 or rGDF8 protein to young or aged mice fed a standard chow diet, short-term high-fat diet (HFD), or long-term HFD. Under nearly all of these diet conditions, administration of exogenous rGDF11 reduced body weight by 3-17% and significantly improved glucose tolerance in aged mice fed a chow (~30% vs. saline) or HF (~50% vs. saline) diet and young mice fed a HFD (~30%). On the other hand, exogenous rGDF8 showed signifcantly lesser effect or no effect at all on glucose tolerance compared to rGDF11, consistent with data demonstrating that GFD11 is a more potent signaling ligand than GDF8. Collectively, our results show that administration of exogenous rGDF11, but not rGDF8, can reduce diet-induced weight gain and improve metabolic homeostasis.


Subject(s)
Aging/metabolism , Body Weight/drug effects , Bone Morphogenetic Proteins/administration & dosage , Diet, High-Fat/adverse effects , Insulin Resistance , Myostatin/administration & dosage , Aging/blood , Aging/drug effects , Animals , Bone Morphogenetic Proteins/pharmacology , Energy Metabolism/drug effects , Growth Differentiation Factors/administration & dosage , Growth Differentiation Factors/pharmacology , Male , Mice , Mice, Inbred C57BL , Myostatin/pharmacology , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Signal Transduction/drug effects
2.
Proc Natl Acad Sci U S A ; 113(51): 14852-14857, 2016 12 20.
Article in English | MEDLINE | ID: mdl-27930302

ABSTRACT

Electrophysiological studies of excitable organs usually focus on action potential (AP)-generating cells, whereas nonexcitable cells are generally considered as barriers to electrical conduction. Whether nonexcitable cells may modulate excitable cell function or even contribute to AP conduction via direct electrotonic coupling to AP-generating cells is unresolved in the heart: such coupling is present in vitro, but conclusive evidence in situ is lacking. We used genetically encoded voltage-sensitive fluorescent protein 2.3 (VSFP2.3) to monitor transmembrane potential in either myocytes or nonmyocytes of murine hearts. We confirm that VSFP2.3 allows measurement of cell type-specific electrical activity. We show that VSFP2.3, expressed solely in nonmyocytes, can report cardiomyocyte AP-like signals at the border of healed cryoinjuries. Using EM-based tomographic reconstruction, we further discovered tunneling nanotube connections between myocytes and nonmyocytes in cardiac scar border tissue. Our results provide direct electrophysiological evidence of heterocellular electrotonic coupling in native myocardium and identify tunneling nanotubes as a possible substrate for electrical cell coupling that may be in addition to previously discovered connexins at sites of myocyte-nonmyocyte contact in the heart. These findings call for reevaluation of cardiac nonmyocyte roles in electrical connectivity of the heterocellular heart.


Subject(s)
Connexins/metabolism , Gap Junctions/metabolism , Heart Conduction System/metabolism , Myocardium/cytology , Myocytes, Cardiac/metabolism , Optogenetics , Action Potentials , Animals , Bacterial Proteins/metabolism , Cell Communication , Cell Count , Cell Membrane/metabolism , Electric Conductivity , Female , Fibroblasts/metabolism , Heart/physiology , Luminescent Proteins/metabolism , Male , Membrane Potentials , Mice , Mice, Transgenic , Muscle Cells/metabolism
3.
Circ Res ; 118(7): 1125-41; discussion 1142, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27034275

ABSTRACT

Growth differentiation factor 11 (GDF11) and myostatin (or GDF8) are closely related members of the transforming growth factor ß superfamily and are often perceived to serve similar or overlapping roles. Yet, despite commonalities in protein sequence, receptor utilization and signaling, accumulating evidence suggests that these 2 ligands can have distinct functions in many situations. GDF11 is essential for mammalian development and has been suggested to regulate aging of multiple tissues, whereas myostatin is a well-described negative regulator of postnatal skeletal and cardiac muscle mass and modulates metabolic processes. In this review, we discuss the biochemical regulation of GDF11 and myostatin and their functions in the heart, skeletal muscle, and brain. We also highlight recent clinical findings with respect to a potential role for GDF11 and/or myostatin in humans with heart disease. Finally, we address key outstanding questions related to GDF11 and myostatin dynamics and signaling during development, growth, and aging.


Subject(s)
Bone Morphogenetic Proteins/physiology , Growth Differentiation Factors/physiology , Myostatin/physiology , Adult , Aging/physiology , Amino Acid Sequence , Animals , Bone Morphogenetic Proteins/chemistry , Bone Morphogenetic Proteins/deficiency , Brain/growth & development , Brain/physiology , Dimerization , Female , Follistatin/metabolism , Follistatin-Related Proteins/metabolism , Growth Differentiation Factors/chemistry , Growth Differentiation Factors/deficiency , Growth Differentiation Factors/therapeutic use , Heart/physiology , Heart Diseases/metabolism , Humans , Male , Mice , Models, Molecular , Molecular Sequence Data , Muscles/physiology , Myocardium/metabolism , Myostatin/chemistry , Myostatin/deficiency , Organ Specificity , Protein Conformation , Protein Structure, Tertiary , Rats , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction , Structure-Activity Relationship
4.
Circ Res ; 118(1): 29-37, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26489925

ABSTRACT

RATIONALE: Growth differentiation factor 11 (GDF11) and GDF8 are members of the transforming growth factor-ß superfamily sharing 89% protein sequence homology. We have previously shown that circulating GDF11 levels decrease with age in mice. However, a recent study by Egerman et al reported that GDF11/8 levels increase with age in mouse serum. OBJECTIVE: Here, we clarify the direction of change of circulating GDF11/8 levels with age and investigate the effects of GDF11 administration on the murine heart. METHODS AND RESULTS: We validated our previous finding that circulating levels of GDF11/8 decline with age in mice, rats, horses, and sheep. Furthermore, we showed by Western analysis that the apparent age-dependent increase in GDF11 levels, as reported by Egerman et al, is attributable to cross-reactivity of the anti-GDF11 antibody with immunoglobulin, which is known to increase with age. GDF11 administration in mice rapidly activated SMAD2 and SMAD3 signaling in myocardium in vivo and decreased cardiac mass in both young (2-month-old) and old (22-month-old) mice in a dose-dependent manner after only 9 days. CONCLUSIONS: Our study confirms an age-dependent decline in serum GDF11/8 levels in multiple mammalian species and that exogenous GDF11 rapidly activates SMAD signaling and reduces cardiomyocyte size. Unraveling the molecular basis for the age-dependent decline in GDF11/8 could yield insight into age-dependent cardiac pathologies.


Subject(s)
Aging/blood , Bone Morphogenetic Proteins/blood , Growth Differentiation Factors/blood , Myostatin/blood , Animals , Biomarkers/blood , Horses , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Sheep
5.
J Vis Exp ; (83): e51064, 2014 Jan 24.
Article in English | MEDLINE | ID: mdl-24513973

ABSTRACT

Previous studies showed that cell delivery promotes cardiac function amelioration by release of cytokines and factors that increase cardiac tissue revascularization and cell survival. In addition, further observations revealed that specific stem cells, such as cardiac stem cells, mesenchymal stem cells and cardiospheres have the ability to integrate within the surrounding myocardium by differentiating into cardiomyocytes, smooth muscle cells and endothelial cells. Here, we present the materials and methods to reliably deliver noncontractile cells into the left ventricular wall of immunodepleted mice. The salient steps of this microsurgical procedure involve anesthesia and analgesia injection, intratracheal intubation, incision to open the chest and expose the heart and delivery of cells by a sterile 30-gauge needle and a precision microliter syringe. Tissue processing consisting of heart harvesting, embedding, sectioning and histological staining showed that intramyocardial cell injection produced a small damage in the epicardial area, as well as in the ventricular wall. Noncontractile cells were retained into the myocardial wall of immunocompromised mice and were surrounded by a layer of fibrotic tissue, likely to protect from cardiac pressure and mechanical load.


Subject(s)
Heart/physiology , Myocardium/cytology , Stem Cell Transplantation/methods , Animals , HEK293 Cells , Humans , Immunocompromised Host , Mice , Mice, Inbred NOD , Models, Animal
6.
PLoS One ; 8(11): e80268, 2013.
Article in English | MEDLINE | ID: mdl-24265802

ABSTRACT

Serum and glucocorticoid inducible kinase 1 (SGK1) plays a pivotal role in early angiogenesis during embryonic development. In this study, we sought to define the SGK1 downstream signalling pathways in the adult heart and to elucidate their role in cardiac neo-angiogenesis and wound healing after myocardial ischemia. To this end, we employed a viable SGK1 knockout mouse model generated in a 129/SvJ background. Ablation of SGK1 in these mice caused a significant decrease in phosphorylation of SGK1 target protein NDRG1, which correlated with alterations in NF-κB signalling and expression of its downstream target protein, VEGF-A. Disruption of these signalling pathways was accompanied by smaller heart and body size. Moreover, the lack of SGK1 led to defective endothelial cell (ECs) migration and tube formation in vitro, and increased scarring with decreased angiogenesis in vivo after myocardial infarct. This study underscores the importance of SGK1 signalling in cardiac neo-angiogenesis and wound healing after an ischemic insult in vivo.


Subject(s)
Endothelial Cells/metabolism , Immediate-Early Proteins/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Neovascularization, Pathologic/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Movement/genetics , Cell Size , Disease Models, Animal , Fibrosis , Immediate-Early Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Ischemia/genetics , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , NF-kappa B/metabolism , Neovascularization, Pathologic/genetics , Phenotype , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proteomics , Signal Transduction
7.
Dis Model Mech ; 6(6): 1470-6, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23929940

ABSTRACT

Tamoxifen-inducible Cre-mediated manipulation of animal genomes has achieved wide acceptance over the last decade, with numerous important studies heavily relying on this technique. Recently, a number of groups have reported transient complications of using this protocol in the heart. In the present study we observed a previously unreported focal fibrosis and depressed left-ventricular function in tamoxifen-treated αMHC-MerCreMer-positive animals in a Tß4shRNAflox × αMHC-MerCreMer cross at 6-7 weeks following standard tamoxifen treatment, regardless of the presence of the floxed transgene. The phenotype was reproduced by treating mice from the original αMHC-MerCreMer strain with tamoxifen. In the acute phase after tamoxifen treatment, cell infiltration into the myocardium was accompanied by increased expression of pro-inflammatory cytokines (IL-1ß, IL-6, TNFα, IFNγ, Ccl2) and markers of hypertrophy (ANF, BNP, Col3a1). These observations highlight the requirement for including tamoxifen-treated MerCreMer littermate controls to avert misinterpretation of conditional mutant phenotypes. A survey of the field as well as the protocols presented here suggests that controlling the parameters of tamoxifen delivery is important in avoiding the chronic MerCreMer-mediated cardiac phenotype reported here.


Subject(s)
Integrases/metabolism , Myocardium/enzymology , Animals , Mice
8.
Cell Mol Neurobiol ; 33(5): 673-80, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23552977

ABSTRACT

Parkinson's disease (PD) is one of the most common neurodegenerative disorders whose etiology is multifactorial including both hereditary and environmental factors. Currently, pathogenic mutations in at least five genes have been implicated in familial PD generally accounting for less than 10 % of all PD cases in most populations. It has been suggested that polymorphisms in other genes such as those encoding enzymes involved in oxidative metabolism and detoxification could be involved in predisposition to PD since oxidative stress in dopaminergic neurons is thought to be of central importance in the pathogenesis of the disease. The aim of our work was to study the association of genetic polymorphisms in genes involved in oxidative metabolism and detoxification mechanism, namely GSTM1, GSTT1, GSTP1, and those involved in DNA damage repair, OGG1 and XRCC1, in an Italian cohort of sporadic PD patients. We did not detect any association between GSTT1 and GTTM1 null polymorphisms and PD, whereas the 104GSTP1 polymorphism was associated with PD in male patients but not in females. Furthermore, we detected a protective effect of wild type genotype of XRCC1 in women.


Subject(s)
DNA Repair/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Glutathione Transferase/genetics , Parkinson Disease/epidemiology , Parkinson Disease/genetics , Polymorphism, Genetic , Aged , Aged, 80 and over , Case-Control Studies , DNA Glycosylases/genetics , DNA-Binding Proteins/genetics , Demography , Female , Gene Frequency/genetics , Glutathione S-Transferase pi/genetics , Humans , Italy/epidemiology , Male , Middle Aged , Odds Ratio , Parkinson Disease/enzymology , X-ray Repair Cross Complementing Protein 1
9.
Biochem Biophys Res Commun ; 410(2): 201-7, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21621517

ABSTRACT

The aim of this study was to investigate whether supplemental IGF-1Ea transgene expression induces activation of local cardiac and bone marrow stem cell population to mediate mammalian heart repair. In physiologic conditions, cardiac overexpression of the IGF-1Ea propeptide is associated with an enrichment of c-Kit/Sca-1 positive side population cells in the bone marrow and the occurrence of an endothelial-primed CD34 positive side population in the heart. This cellular profile is shown here to correlate with the expression of cytokines involved in stem cell mobilization and vessel formation. This molecular and cellular interplay favored IGF-1Ea-mediated vessel formation in injured hearts. The physiologic and pathologic connection between cytokines and stem cells in response to IGF-1Ea may represent an important model to understand how to elicit endogenous reparative signaling.


Subject(s)
Bone Marrow Cells/physiology , Cytokines/metabolism , Heart/physiology , Insulin-Like Growth Factor I/physiology , Myocardial Infarction/physiopathology , Neovascularization, Physiologic , Regeneration , Animals , Antigens, Ly/metabolism , Bone Marrow Cells/metabolism , Coronary Vessels/physiology , Insulin-Like Growth Factor I/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Proto-Oncogene Proteins c-kit/metabolism , Stem Cells/metabolism , Stem Cells/physiology
10.
J Radiat Res ; 51(5): 615-9, 2010.
Article in English | MEDLINE | ID: mdl-20921829

ABSTRACT

Breast cancer (BC) is the most common type of malignancy in female patients and radio-treatment is the conventional therapy even if a great number of studies reported that enhanced sensitivity to ionizing radiation as measured as chromosome effects is present in a significant proportion of cancer patients, including breast cancer ones. In this study we analysed whether peripheral blood lymphocytes from sporadic BC patients and healthy subjects showed a different sensitivity to ionizing radiation and whether cytogenetic radiosensitivity may serve as a breast cancer risk biomarker. To test this hypothesis, the in vitro radiation sensitivity was measured by using both G(0) and G(2) chromosome radiosensitivity assays, on 46 subjects (23 BC patients and 23 healthy subjects). Results show that cancer patients are more radiosensitive than healthy controls and that G(2) assay could be more appropriate to define the individual radiosensitivity if compared to G(0) assay.


Subject(s)
Breast Neoplasms/radiotherapy , Cytogenetic Analysis , G2 Phase , Radiation Tolerance , Resting Phase, Cell Cycle , Adult , Aged , Biomarkers, Tumor/radiation effects , Female , G2 Phase/radiation effects , Humans , Italy/epidemiology , Middle Aged , Resting Phase, Cell Cycle/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...