Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Xenotransplantation ; 30(2): e12793, 2023 03.
Article in English | MEDLINE | ID: mdl-36748727

ABSTRACT

Neonatal porcine islets (NPIs) are a source of islets for xenotransplantation. In the pig, the pancreatic lobes remain separate, thus, when optimizing NPI isolation, the pancreatic lobes included in the pancreatic digest should be specified. These lobes are the duodenal (DL), splenic (SL) and connecting (CL) lobe that correspond to the head, body-tail, and uncinate process of the human pancreas. In this study we are the first to evaluate all three neonatal porcine pancreatic lobes and NPIs isolated from these lobes. We report, a significant difference in endocrine and progenitor cell composition between lobes, and observed pancreatic duct glands (PDG) within the mesenchyme surrounding exocrine ducts in the DL and CL. Following in vitro differentiation, NPIs isolated from each lobe differed significantly in the percent increase of endocrine cells and final cell composition. Compared to other recipients, diabetic immunodeficient mice transplanted with NPIs isolated from the SL demonstrated euglycemic control as early as 4 weeks (p < 0.05) and achieved normoglycemia by 6 weeks post-transplant (p < 0.01). For the first time we report significant differences between the neonatal porcine pancreatic lobes and demonstrate that NPIs from these lobes differ in xenograft function.


Subject(s)
Islets of Langerhans Transplantation , Islets of Langerhans , Animals , Swine , Humans , Mice , Transplantation, Heterologous , Pancreas , Stem Cells
2.
Cardiovasc Res ; 115(8): 1343-1356, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30496354

ABSTRACT

AIMS: Cardiac remodelling in the ischaemic heart determines prognosis in patients with ischaemic heart disease (IHD), while enhancement of angiogenesis and cell survival has shown great potential for IHD despite translational challenges. Phosphoinositide 3-kinase (PI3K)/Akt signalling pathways play a critical role in promoting angiogenesis and cell survival. However, the effect of PI3Kß in the ischaemic heart is poorly understood. This study investigates the role of endothelial and cardiomyocyte (CM) PI3Kß in post-infarct cardiac remodelling. METHODS AND RESULTS: PI3Kß catalytic subunit-p110ß level was increased in infarcted murine and human hearts. Using cell type-specific loss-of-function approaches, we reported novel and distinct actions of p110ß in endothelial cells (ECs) vs. CMs in response to myocardial ischaemic injury. Inactivation of endothelial p110ß resulted in marked resistance to infarction and adverse cardiac remodelling with decreased mortality, improved systolic function, preserved microvasculature, and enhanced Akt activation. Cultured ECs with p110ß knockout or inhibition displayed preferential PI3Kα/Akt/endothelial nitric oxide synthase signalling that consequently promoted protective signalling and angiogenesis. In contrast, mice with CM p110ß-deficiency exhibited adverse post-infarct ventricular remodelling with larger infarct size and deteriorated cardiac function, which was due to enhanced susceptibility of CMs to ischaemia-mediated cell death. Disruption of CM p110ß signalling compromised nuclear p110ß and phospho-Akt levels leading to perturbed gene expression and elevated pro-cell death protein levels, increasing the susceptibility to CM death. A similar divergent response of PI3Kß endothelial and CM mutant mice was seen using a model of myocardial ischaemia-reperfusion injury. CONCLUSION: These data demonstrate novel, differential, and cell-specific functions of PI3Kß in the ischaemic heart. While the loss of endothelial PI3Kß activity produces cardioprotective effects, CM PI3Kß is protective against myocardial ischaemic injury.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/metabolism , Endothelial Cells/enzymology , Myocardial Infarction/enzymology , Myocardial Reperfusion Injury/enzymology , Myocytes, Cardiac/enzymology , Ventricular Remodeling , Animals , Cells, Cultured , Class I Phosphatidylinositol 3-Kinases/deficiency , Class I Phosphatidylinositol 3-Kinases/genetics , Disease Models, Animal , Endothelial Cells/pathology , Human Umbilical Vein Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/pathology , Humans , Male , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/pathology , Neovascularization, Physiologic , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
3.
J Am Heart Assoc ; 6(1)2017 01 23.
Article in English | MEDLINE | ID: mdl-28115312

ABSTRACT

BACKGROUND: Sex-related differences in cardiac function and iron metabolism exist in humans and experimental animals. Male patients and preclinical animal models are more susceptible to cardiomyopathies and heart failure. However, whether similar differences are seen in iron-overload cardiomyopathy is poorly understood. METHODS AND RESULTS: Male and female wild-type and hemojuvelin-null mice were injected and fed with a high-iron diet, respectively, to develop secondary iron overload and genetic hemochromatosis. Female mice were completely protected from iron-overload cardiomyopathy, whereas iron overload resulted in marked diastolic dysfunction in male iron-overloaded mice based on echocardiographic and invasive pressure-volume analyses. Female mice demonstrated a marked suppression of iron-mediated oxidative stress and a lack of myocardial fibrosis despite an equivalent degree of myocardial iron deposition. Ovariectomized female mice with iron overload exhibited essential pathophysiological features of iron-overload cardiomyopathy showing distinct diastolic and systolic dysfunction, severe myocardial fibrosis, increased myocardial oxidative stress, and increased expression of cardiac disease markers. Ovariectomy prevented iron-induced upregulation of ferritin, decreased myocardial SERCA2a levels, and increased NCX1 levels. 17ß-Estradiol therapy rescued the iron-overload cardiomyopathy in male wild-type mice. The responses in wild-type and hemojuvelin-null female mice were remarkably similar, highlighting a conserved mechanism of sex-dependent protection from iron-overload-mediated cardiac injury. CONCLUSIONS: Male and female mice respond differently to iron-overload-mediated effects on heart structure and function, and females are markedly protected from iron-overload cardiomyopathy. Ovariectomy in female mice exacerbated iron-induced myocardial injury and precipitated severe cardiac dysfunction during iron-overload conditions, whereas 17ß-estradiol therapy was protective in male iron-overloaded mice.


Subject(s)
Cardiomyopathies/metabolism , Heart Failure, Diastolic/metabolism , Hemochromatosis/metabolism , Iron Overload/metabolism , Oxidative Stress , Animals , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/etiology , Cardiomyopathies/pathology , Diet , Echocardiography , Estradiol/pharmacology , Female , Ferritins/metabolism , Fibrosis , GPI-Linked Proteins , Heart/drug effects , Heart Failure, Diastolic/diagnostic imaging , Heart Failure, Diastolic/etiology , Heart Failure, Diastolic/pathology , Hemochromatosis/complications , Hemochromatosis/pathology , Hemochromatosis Protein , Iron Overload/complications , Iron Overload/pathology , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Myocardium/pathology , Ovariectomy , Oxidative Stress/drug effects , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Sex Factors , Sodium-Calcium Exchanger/metabolism
4.
Liver Int ; 36(2): 246-57, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26077449

ABSTRACT

BACKGROUND & AIMS: Abnormal iron metabolism and hepatic iron-overload is a major cause of liver injury and in the development of chronic liver diseases. Iron-overload-mediated liver disease leads to end-stage cirrhosis and/or hepatocellular carcinoma. METHODS: Using a genetic hemochromatosis (hemojuvelin knockout mice) and non-genetic (secondary iron-overload) murine models of hepatic iron-overload, we elucidated the mechanism of hepatic iron injury and the therapeutic effects of resveratrol. RESULTS: Hepatic iron-overload was associated with hepatosplenomegaly, increased oxidative stress, hepatic fibrosis, and inflammation, and a pro-apoptotic state which was markedly corrected by resveratrol therapy. Importantly our aging studies with the hemojuvelin knockout mice showed advanced liver disease in association with steatosis in the absence of a diabetic state which recapitulates the essential pathological features seen in clinical iron-overload. Chronic hepatic iron-overload showed increased nuclear localization of acetylated Forkhead fox-O-1 (FoxO1) transcription factor whereas resveratrol dietary intervention reversed the acetylation of FoxO1 in association with increased SIRT1 levels which together with its pleotropic antioxidant properties are likely key mechanisms of its therapeutic action. Importantly, resveratrol treatment did not affect the degree of hepatic iron-overload but rather direct protects the liver from iron-mediated injury. CONCLUSIONS: Our findings illustrate a novel and definitive therapeutic action of resveratrol and represent an economically feasible therapeutic intervention to treat hepatic iron-overload and liver disease.


Subject(s)
Acetylation/drug effects , Apoptosis/drug effects , Iron Overload , Iron/metabolism , Liver Diseases , Oxidative Stress/drug effects , Stilbenes/pharmacology , Animals , Antioxidants/pharmacology , Chronic Disease , Disease Models, Animal , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Hemochromatosis/complications , Hemochromatosis/metabolism , Iron Overload/drug therapy , Iron Overload/etiology , Iron Overload/metabolism , Liver Diseases/complications , Liver Diseases/drug therapy , Liver Diseases/metabolism , Mice , Models, Genetic , Resveratrol , Sirtuin 1/metabolism , Treatment Outcome
5.
Sci Rep ; 5: 18132, 2015 Dec 07.
Article in English | MEDLINE | ID: mdl-26638758

ABSTRACT

Iron-overload cardiomyopathy is a prevalent cause of heart failure on a world-wide basis and is a major cause of mortality and morbidity in patients with secondary iron-overload and genetic hemochromatosis. We investigated the therapeutic effects of resveratrol in acquired and genetic models of iron-overload cardiomyopathy. Murine iron-overload models showed cardiac iron-overload, increased oxidative stress, altered Ca(2+) homeostasis and myocardial fibrosis resulting in heart disease. Iron-overload increased nuclear and acetylated levels of FOXO1 with corresponding inverse changes in SIRT1 levels in the heart corrected by resveratrol therapy. Resveratrol, reduced the pathological remodeling and improved cardiac function in murine models of acquired and genetic iron-overload at varying stages of iron-overload. Echocardiography and hemodynamic analysis revealed a complete normalization of iron-overload mediated diastolic and systolic dysfunction in response to resveratrol therapy. Myocardial SERCA2a levels were reduced in iron-overloaded hearts and resveratrol therapy restored SERCA2a levels and corrected altered Ca(2+) homeostasis. Iron-mediated pro-oxidant and pro-fibrotic effects in human and murine cardiomyocytes and cardiofibroblasts were suppressed by resveratrol which correlated with reduction in iron-induced myocardial oxidative stress and myocardial fibrosis. Resveratrol represents a clinically and economically feasible therapeutic intervention to reduce the global burden from iron-overload cardiomyopathy at early and chronic stages of iron-overload.


Subject(s)
Cardiomyopathies/complications , Cardiomyopathies/drug therapy , Iron Overload/complications , Iron Overload/drug therapy , Models, Genetic , Stilbenes/therapeutic use , Animals , Cardiomyopathies/physiopathology , Disease Models, Animal , Down-Regulation/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , GPI-Linked Proteins , Genetic Therapy , Hemochromatosis Protein , Humans , Iron Overload/physiopathology , Male , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Oxidants/toxicity , Oxidative Stress/drug effects , Resveratrol , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Signal Transduction/drug effects , Sirtuin 1/metabolism , Stilbenes/pharmacology
6.
Cardiovasc Res ; 105(3): 292-303, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25618408

ABSTRACT

AIMS: Genetic mouse models have yielded conflicting conclusions about the role of PI3Kα in heart physiology: specifically, the question of whether PI3Kα has a direct role in regulating myocardial contractility. This has led to concerns that PI3K inhibitors currently in clinical trials for cancer may potentiate cardiotoxicity. Here we seek to clarify the role of PI3Kα in normal heart physiology and investigate changes in related signalling pathways. METHODS AND RESULTS: Targeted deletion of PI3Kα and PI3Kß in the heart with a tamoxifen-dependent Cre recombinase transgene caused transient heart dysfunction in all genotypes, but only PI3Kα deletion prevented functional recovery. Reduction in tamoxifen dosing allowed for maintained gene deletion without any cardiomyopathy, possibly through activation of survival signalling through the related ERK pathway. Similarly, mice with PI3Kα deletion induced by constitutively active Cre recombinase had normal heart function. Insulin-mediated activation of Akt, a marker of PI3Kα activity, was impaired with increased ERK1/2 activation in PI3Kα mutant hearts. Pharmacological inhibition of PI3Kα with BYL-719 also caused impaired insulin signalling in murine and human cardiomyocytes as well as in vivo in mice, with increased fasting blood glucose levels, but did not affect myocardial contractility as determined by echocardiography and invasive pressure-volume loop analysis. CONCLUSION: Our results show that PI3Kα does not directly regulate myocardial contractility, but is required for recovery from tamoxifen/Cre toxicity. The important role for PI3Kα in insulin signalling and recovery from tamoxifen/Cre toxicity justifies caution when using PI3Kα inhibitors in combination with other cardiovascular comorbidities and cardiotoxic compounds in cancer patients.


Subject(s)
Heart Diseases/enzymology , Insulin/metabolism , Integrases/metabolism , Myocardial Contraction , Myocardium/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Tamoxifen/pharmacology , Animals , Class I Phosphatidylinositol 3-Kinases , Disease Models, Animal , Genotype , Heart Diseases/genetics , Heart Diseases/physiopathology , Integrases/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myocardial Contraction/drug effects , Phenotype , Phosphatidylinositol 3-Kinases/deficiency , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Time Factors
7.
Am J Physiol Renal Physiol ; 306(8): F812-21, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24553436

ABSTRACT

The renin-angiotensin system, especially angiotensin II (ANG II), plays a key role in the development and progression of diabetic nephropathy. ANG 1-7 has counteracting effects on ANG II and is known to exert beneficial effects on diabetic nephropathy. We studied the mechanism of ANG 1-7-induced beneficial effects on diabetic nephropathy in db/db mice. We administered ANG 1-7 (0.5 mg·kg(-1)·day(-1)) or saline to 5-mo-old db/db mice for 28 days via implanted micro-osmotic pumps. ANG 1-7 treatment reduced kidney weight and ameliorated mesangial expansion and increased urinary albumin excretion, characteristic features of diabetic nephropathy, in db/db mice. ANG 1-7 decreased renal fibrosis in db/db mice, which correlated with dephosphorylation of the signal transducer and activator of transcription 3 (STAT3) pathway. ANG 1-7 treatment also suppressed the production of reactive oxygen species via attenuation of NADPH oxidase activity and reduced inflammation in perirenal adipose tissue. Furthermore, ANG 1-7 treatment decreased lipid accumulation in db/db kidneys, accompanied by increased expressions of renal adipose triglyceride lipase (ATGL). Alterations in ATGL expression correlated with increased SIRT1 expression and deacetylation of FOXO1. The upregulation of angiotensin-converting enzyme 2 levels in diabetic nephropathy was normalized by ANG 1-7. ANG 1-7 treatment exerts renoprotective effects on diabetic nephropathy, associated with reduction of oxidative stress, inflammation, fibrosis, and lipotoxicity. ANG 1-7 can represent a promising therapy for diabetic nephropathy.


Subject(s)
Angiotensin I/therapeutic use , Diabetic Nephropathies/prevention & control , Peptide Fragments/therapeutic use , Angiotensin-Converting Enzyme 2 , Animals , Diabetic Nephropathies/physiopathology , Fibrosis , Forkhead Box Protein O1 , Forkhead Transcription Factors/drug effects , Forkhead Transcription Factors/metabolism , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Lipase/biosynthesis , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Peptidyl-Dipeptidase A/biosynthesis , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/drug effects , STAT3 Transcription Factor/metabolism , Sirtuin 1/drug effects , Sirtuin 1/metabolism , Triglycerides/metabolism
8.
Circ Heart Fail ; 7(2): 327-39, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24389129

ABSTRACT

BACKGROUND: The angiotensin-converting enzyme 2 and angiotensin-(1-7) (Ang 1-7)/MasR (Mas receptor) axis are emerging as a key pathway that can modulate the development of diabetic cardiomyopathy. We studied the effects of Ang 1-7 on diabetic cardiomyopathy in db/db diabetic mice to elucidate the therapeutic effects and mechanism of action. METHODS AND RESULTS: Ang 1-7 was administered to 5-month-old male db/db mice for 28 days via implanted micro-osmotic pumps. Ang 1-7 treatment ameliorated myocardial hypertrophy and fibrosis with normalization of diastolic dysfunction assessed by pressure-volume loop analysis and echocardiography. The functional improvement by Ang 1-7 was accompanied by a reduction in myocardial lipid accumulation and systemic fat mass and inflammation and increased insulin-stimulated myocardial glucose oxidation. Increased myocardial protein kinase C levels and loss of phosphorylation of extracellular signal-regulated kinase 1/2 were prevented by Ang 1-7. Furthermore, Ang 1-7 treatment decreased cardiac triacylglycerol and ceramide levels in db/db mice, concomitantly with an increase in myocardial adipose triglyceride lipase expression. Changes in adipose triglyceride lipase expression correlated with increased SIRT1 (silent mating type information regulation 2 homolog 1) levels and deacetylation of FOXO1 (forkhead box O1). CONCLUSIONS: We identified a novel beneficial effect of Ang 1-7 on diabetic cardiomyopathy that involved a reduction in cardiac hypertrophy and lipotoxicity, adipose inflammation, and an upregulation of adipose triglyceride lipase. Ang 1-7 completely rescued the diastolic dysfunction in the db/db model. Ang 1-7 represents a promising therapy for diabetic cardiomyopathy associated with type 2 diabetes mellitus.


Subject(s)
Angiotensin I/therapeutic use , Diabetes Mellitus, Experimental , Diabetic Cardiomyopathies/drug therapy , Inflammation/drug therapy , Lipids/blood , Peptide Fragments/therapeutic use , Ventricular Dysfunction, Left/drug therapy , Ventricular Function/physiology , Animals , Blood Glucose/metabolism , Diabetic Cardiomyopathies/complications , Diabetic Cardiomyopathies/diagnosis , Diastole , Echocardiography, Doppler , Follow-Up Studies , Inflammation/metabolism , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Vasodilator Agents/therapeutic use , Ventricular Dysfunction, Left/blood , Ventricular Dysfunction, Left/physiopathology , Ventricular Pressure/drug effects
9.
Circ Res ; 112(12): 1542-56, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23553616

ABSTRACT

RATIONALE: The classic phagocyte nicotinamide adenine dinucleotide phosphate oxidase (gp91(phox) or Nox2) is expressed in the heart. Nox2 activation requires membrane translocation of the p47(phox) subunit and is linked to heart failure. We hypothesized that loss of p47(phox) subunit will result in decreased reactive oxygen species production and resistance to heart failure. OBJECTIVE: To define the role of p47(phox) in pressure overload-induced biomechanical stress. METHODS AND RESULTS: Eight-week-old male p47(phox) null (p47(phox) knockout [KO]), Nox2 null (Nox2KO), and wild-type mice were subjected to transverse aortic constriction-induced pressure overload. Contrary to our hypothesis, p47(phox)KO mice showed markedly worsened systolic dysfunction in response to pressure overload at 5 and 9 weeks after transverse aortic constriction compared with wild-type-transverse aortic constriction mice. We found that biomechanical stress upregulated N-cadherin and ß-catenin in p47(phox)KO hearts but disrupted the actin filament cytoskeleton and reduced phosphorylation of focal adhesion kinase. p47(phox) interacts with cytosolic cortactin by coimmunoprecipitation and double immunofluorescence staining in murine and human hearts and translocated to the membrane on biomechanical stress where cortactin interacted with N-cadherin, resulting in adaptive cytoskeletal remodeling. However, p47(phox)KO hearts showed impaired interaction of cortactin with N-cadherin, resulting in loss of biomechanical stress-induced actin polymerization and cytoskeletal remodeling. In contrast, Nox2 does not interact with cortactin, and Nox2-deficient hearts were protected from pressure overload-induced adverse myocardial and intracellular cytoskeletal remodeling. CONCLUSIONS: We showed a novel role of p47(phox) subunit beyond and independent of nicotinamide adenine dinucleotide phosphate oxidase activity as a regulator of cortactin and adaptive cytoskeletal remodeling, leading to a paradoxically enhanced susceptibility to biomechanical stress and heart failure.


Subject(s)
Actin Cytoskeleton/metabolism , Actins/metabolism , Cortactin/metabolism , Heart Failure/enzymology , Mechanotransduction, Cellular , Myocardium/enzymology , NADPH Oxidases/deficiency , Animals , Biomechanical Phenomena , Cadherins/metabolism , Cells, Cultured , Disease Models, Animal , Echocardiography, Doppler , Fluorescent Antibody Technique , Focal Adhesion Kinase 1/metabolism , Heart Failure/diagnostic imaging , Heart Failure/genetics , Heart Failure/physiopathology , Heart Failure/prevention & control , Humans , Immunoprecipitation , Male , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , NADPH Oxidase 2 , NADPH Oxidases/genetics , Oxidative Stress , Phosphorylation , Polymerization , Reactive Oxygen Species/metabolism , Stress, Mechanical , Time Factors , Ventricular Remodeling , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...