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










Publication year range
1.
J Am Heart Assoc ; 8(22): e012792, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31718444

ABSTRACT

Background The SNRK (sucrose-nonfermenting-related kinase) enzyme is critical for cardiac function. However, the underlying cause for heart failure observed in Snrk cardiac conditional knockout mouse is unknown. Methods and Results Previously, 6-month adult mice knocked out for Snrk in cardiomyocytes (CMs) displayed left ventricular dysfunction. Here, 4-month adult mice, on angiotensin II (Ang II) infusion, show rapid decline in cardiac systolic function, which leads to heart failure and death in 2 weeks. These mice showed increased expression of nuclear factor κ light chain enhancer of activated B cells (NF-κB), inflammatory signaling proteins, proinflammatory proteins in the heart, and fibrosis. Interestingly, under Ang II infusion, mice knocked out for Snrk in endothelial cells did not show significant systolic or diastolic dysfunction. Although an NF-κB inflammation signaling pathway was increased in Snrk knockout endothelial cells, this did not lead to fibrosis or mortality. In hearts of adult mice knocked out for Snrk in CMs, we also observed NF-κB pathway activation in CMs, and an increased presence of Mac2+ macrophages was observed in basal and Ang II-infused states. In vitro analysis of Snrk knockdown HL-1 CMs revealed similar upregulation of the NF-κB signaling proteins and proinflammatory proteins that was exacerbated on Ang II treatment. The Ang II-induced NF-κB pathway-mediated proinflammatory effects were mediated in part through protein kinase B or AKT, wherein AKT inhibition restored the proinflammatory signaling protein levels to baseline in Snrk knockdown HL-1 CMs. Conclusions During heart failure, SNRK acts as a cardiomyocyte-specific repressor of cardiac inflammation and fibrosis.


Subject(s)
Endothelial Cells/metabolism , Heart Failure/genetics , Inflammation/genetics , Myocardium/metabolism , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/genetics , Angiotensin II/pharmacology , Animals , Cell Line , Fibrosis/genetics , Fibrosis/metabolism , Fibrosis/pathology , Heart/drug effects , Heart Failure/metabolism , Heart Failure/pathology , In Vitro Techniques , Inflammation/metabolism , Inflammation/pathology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , Myocardium/pathology , Vasoconstrictor Agents/pharmacology , Ventricular Dysfunction, Left
2.
Arterioscler Thromb Vasc Biol ; 38(2): 373-385, 2018 02.
Article in English | MEDLINE | ID: mdl-29242271

ABSTRACT

OBJECTIVE: SNRK (sucrose nonfermenting 1-related kinase) is a novel member of the AMPK (adenosine monophosphate-activated protein kinase)-related superfamily that is activated in the process of angiogenesis. Currently, little is known about the function of SNRK in angiogenesis in the physiological and pathological conditions. APPROACH AND RESULTS: In this study, in Snrk global heterozygous knockout mice, retina angiogenesis and neovessel formation after hindlimb ischemia were suppressed. Consistently, mice with endothelial cell (EC)-specific Snrk deletion exhibited impaired retina angiogenesis, and delayed perfusion recovery and exacerbated muscle apoptosis in ischemic hindlimbs, compared with those of littermate wide-type mice. Endothelial SNRK expression was increased in the extremity vessel samples from nonischemic human. In ECs cultured in hypoxic conditions, HIF1α (hypoxia inducible factor 1α) bound to the SNRK promoter to upregulate SNRK expression. In the nuclei of hypoxic ECs, SNRK complexed with SP1 (specificity protein 1), and together, they bound to an SP1-binding motif in the ITGB1 (ß1 integrin) promoter, resulting in enhanced ITGB1 expression and promoted EC migration. Furthermore, SNRK or SP1 deficiency in ECs ameliorated hypoxia-induced ITGB1 expression and, consequently, inhibited EC migration and angiogenesis. CONCLUSIONS: Taken together, our data have revealed that SNRK/SP1-ITGB1 signaling axis promotes angiogenesis in vivo.


Subject(s)
Endothelial Cells/enzymology , Ischemia/enzymology , Lung/blood supply , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Protein Serine-Threonine Kinases/metabolism , Retinal Vessels/enzymology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Apoptosis , Blood Flow Velocity , Cadherins/genetics , Cadherins/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Endothelial Cells/pathology , Gene Expression Regulation, Enzymologic , Hindlimb , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Integrin beta1/genetics , Integrin beta1/metabolism , Ischemia/genetics , Ischemia/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Regional Blood Flow , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism
3.
Cancer Invest ; 35(7): 456-462, 2017 Aug 09.
Article in English | MEDLINE | ID: mdl-28722495

ABSTRACT

Sucrose non-fermenting related kinase (SNRK) is a serine/threonine kinase known to regulate cellular metabolism and adipocyte inflammation. Since alterations in adipocyte metabolism play a role in ovarian cancer metastasis, we investigated the expression of SNRK in benign and malignant human ovarian tissue using immunohistochemistry and qPCR. The number of SNRK positive (+) nuclei is increased in malignant tissue compared to benign tissue (21.03% versus 14.90%, p < .0431). The most strongly stained malignant SNRK+ nuclei were stage 1 compared to stage 2-4 disease. Differential expression of SNRK in early versus late stage disease suggests specific roles for SNRK in ovarian cancer metastasis.


Subject(s)
Biomarkers, Tumor/analysis , Neoplasms, Glandular and Epithelial/enzymology , Ovarian Neoplasms/enzymology , Protein Serine-Threonine Kinases/analysis , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Carcinoma, Ovarian Epithelial , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Middle Aged , Neoplasm Grading , Neoplasm Staging , Neoplasms, Glandular and Epithelial/genetics , Neoplasms, Glandular and Epithelial/secondary , Neoplasms, Glandular and Epithelial/therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
4.
PLoS One ; 11(12): e0167246, 2016.
Article in English | MEDLINE | ID: mdl-27936095

ABSTRACT

The mitogen-activated protein kinase (MAPK) pathway regulates many key cellular processes such as differentiation, apoptosis, and survival. The final proteins in this pathway, ERK1/2, are regulated by dual specificity phosphatase 5 (DUSP5). DUSP5 is a nuclear, inducible phosphatase with high affinity and fidelity for ERK1/2. By regulating the final step in the MAPK signaling cascade, DUSP5 exerts strong regulatory control over a central cellular pathway. Like other DUSPs, DUSP5 plays an important role in immune function. In this study, we have utilized new knockout mouse reagents to explore its function further. We demonstrate that global loss of DUSP5 does not result in any gross phenotypic changes. However, loss of DUSP5 affects memory/effector CD8+ T cell populations in response to acute viral infection. Specifically, Dusp5-/- mice have decreased proportions of short-lived effector cells (SLECs) and increased proportions of memory precursor effector cells (MPECs) in response to infection. Further, we show that this phenotype is T cell intrinsic; a bone marrow chimera model restricting loss of DUSP5 to the CD8+ T cell compartment displays a similar phenotype. Dusp5-/- T cells also display increased proliferation, increased apoptosis, and altered metabolic profiles, suggesting that DUSP5 is a pro-survival protein in T cells.


Subject(s)
Apoptosis/genetics , Cell Proliferation/genetics , Dual-Specificity Phosphatases/genetics , T-Lymphocytes/metabolism , Animals , Blotting, Western , CD8-Positive T-Lymphocytes/enzymology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Survival/genetics , Cells, Cultured , Dual-Specificity Phosphatases/metabolism , Gene Expression Regulation, Enzymologic , Interferon-gamma/metabolism , Lymphocytic choriomeningitis virus/physiology , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/enzymology , T-Lymphocytes/virology , Tumor Necrosis Factor-alpha/metabolism
5.
Circ Cardiovasc Genet ; 9(6): 474-486, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27780848

ABSTRACT

BACKGROUND: Cardiac metabolism is critical for the functioning of the heart, and disturbance in this homeostasis is likely to influence cardiac disorders or cardiomyopathy. Our laboratory has previously shown that SNRK (sucrose nonfermenting related kinase) enzyme, which belongs to the AMPK (adenosine monophosphate-activated kinase) family, was essential for cardiac metabolism in mammals. Snrk global homozygous knockout (KO) mice die at postnatal day 0, and conditional deletion of Snrk in cardiomyocytes (Snrk cmcKO) leads to cardiac failure and death by 8 to 10 months. METHODS AND RESULTS: We performed additional cardiac functional studies using echocardiography and identified further cardiac functional deficits in Snrk cmcKO mice. Nuclear magnetic resonance-based metabolomics analysis identified key metabolic pathway deficits in SNRK knockdown cardiomyocytes in vitro. Specifically, metabolites involved in lipid metabolism and oxidative phosphorylation are altered, and perturbations in these pathways can result in cardiac function deficits and heart failure. A phosphopeptide-based proteomic screen identified ROCK (Rho-associated kinase) as a putative substrate for SNRK, and mass spec-based fragment analysis confirmed key amino acid residues on ROCK that are phosphorylated by SNRK. Western blot analysis on heart lysates from Snrk cmcKO adult mice and SNRK knockdown cardiomyocytes showed increased ROCK activity. In addition, in vivo inhibition of ROCK partially rescued the in vivo Snrk cmcKO cardiac function deficits. CONCLUSIONS: Collectively, our data suggest that SNRK in cardiomyocytes is responsible for maintaining cardiac metabolic homeostasis, which is mediated in part by ROCK, and alteration of this homeostasis influences cardiac function in the adult heart.


Subject(s)
Embryonic Stem Cells/enzymology , Energy Metabolism , Heart Failure/enzymology , Myocytes, Cardiac/enzymology , Protein Serine-Threonine Kinases/metabolism , rho-Associated Kinases/metabolism , Animals , Cells, Cultured , Echocardiography , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/pathology , Energy Metabolism/drug effects , Fibrosis , Genetic Predisposition to Disease , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Homozygote , Human Umbilical Vein Endothelial Cells/enzymology , Lipid Metabolism , Magnetic Resonance Spectroscopy , Metabolomics/methods , Mice, Knockout , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Oxidative Phosphorylation , Phenotype , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , RNA Interference , Signal Transduction , Transfection , rho-Associated Kinases/antagonists & inhibitors
7.
Biol Open ; 4(1): 48-61, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25505152

ABSTRACT

In this study, we have identified a novel member of the AMPK family, namely Sucrose non-fermenting related kinase (Snrk), that is responsible for maintaining cardiac metabolism in mammals. SNRK is expressed in the heart, and brain, and in cell types such as endothelial cells, smooth muscle cells and cardiomyocytes (CMs). Snrk knockout (KO) mice display enlarged hearts, and die at postnatal day 0. Microarray analysis of embryonic day 17.5 Snrk hearts, and blood profile of neonates display defect in lipid metabolic pathways. SNRK knockdown CMs showed altered phospho-acetyl-coA carboxylase and phospho-AMPK levels similar to global and endothelial conditional KO mouse. Finally, adult cardiac conditional KO mouse displays severe cardiac functional defects and lethality. Our results suggest that Snrk is essential for maintaining cardiac metabolic homeostasis, and shows an autonomous role for SNRK during mammalian development.

8.
Dev Biol ; 386(1): 204-15, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24380800

ABSTRACT

During early development, GATA factors have been shown to be important for key events of coronary vasculogenesis, including formation of the epicardium. Myocardial GATA factors are required for coronary vascular (CV) formation; however, the role of epicardial localized GATAs in this process has not been addressed. The current study was conducted to investigate the molecular mechanisms by which the epicardium controls coronary vasculogenesis, focusing on the role of epicardial GATAs in establishing the endothelial plexus during early coronary vasculogenesis. To address the role of epicardial GATAs, we ablated GATA4 and GATA6 transcription factors specifically from the mouse epicardium and found that the number of endothelial cells in the sub-epicardium was drastically reduced, and concomitant coronary vascular plexus formation was significantly compromised. Here we present evidence for a novel role for epicardial GATA factors in controlling plexus formation by recruiting endothelial cells to the sub-epicardium.


Subject(s)
Coronary Vessels/metabolism , Endothelium, Vascular/metabolism , GATA4 Transcription Factor/physiology , GATA6 Transcription Factor/physiology , Gene Expression Regulation, Developmental , Pericardium/metabolism , Animals , Cell Differentiation , Cell Proliferation , Crosses, Genetic , GATA4 Transcription Factor/genetics , GATA6 Transcription Factor/genetics , Genotype , Heart/embryology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardium/metabolism , Signal Transduction , Time Factors
9.
PLoS One ; 8(12): e80933, 2013.
Article in English | MEDLINE | ID: mdl-24324647

ABSTRACT

Brain metastasis is a defining component of tumor pathophysiology, and the underlying mechanisms responsible for this phenomenon are not well understood. Current dogma is that tumor cells stimulate and activate astrocytes, and this mutual relationship is critical for tumor cell sustenance in the brain. Here, we provide evidence that primary rat neonatal and adult astrocytes secrete factors that proactively induced human lung and breast tumor cell invasion and metastasis capabilities. Among which, tumor invasion factors namely matrix metalloprotease-2 (MMP-2) and MMP-9 were partly responsible for the astrocyte media-induced tumor cell invasion. Inhibiting MMPs reduced the ability of tumor cell to migrate and invade in vitro. Further, injection of astrocyte media-conditioned breast cancer cells in mice showed increased invasive activity to the brain and other distant sites. More importantly, blocking the preconditioned tumor cells with broad spectrum MMP inhibitor decreased the invasion and metastasis of the tumor cells, in particular to the brain in vivo. Collectively, our data implicate astrocyte-derived MMP-2 and MMP-9 as critical players that facilitate tumor cell migration and invasion leading to brain metastasis.


Subject(s)
Astrocytes/pathology , Biological Factors/pharmacology , Breast Neoplasms/pathology , Lung Neoplasms/pathology , Matrix Metalloproteinase 2/pharmacology , Matrix Metalloproteinase 9/pharmacology , Animals , Animals, Newborn , Astrocytes/metabolism , Biological Factors/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Culture Media, Conditioned/chemistry , Female , Hippocampus/metabolism , Hippocampus/pathology , Humans , Lung Neoplasms/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Neoplasm Transplantation , Primary Cell Culture , Protease Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Tumor Cells, Cultured
10.
Differentiation ; 72(5): 239-49, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15270780

ABSTRACT

The role of thyroid hormone in Xenopus metamorphosis is particularly well understood as it plays an essential role in that process. However, recent evidence suggests that thyroid hormone may play an earlier role in amphibian embryogenesis. We demonstrate that Xenopus thyroid hormone receptor beta (XTR beta) is expressed shortly after neural fold closure, and that its expression is localized to the developing retina. Retinoid X receptor gamma (RXR gamma), a potential dimerization partner for XTR beta, was also found to be expressed in the retina at early stages, and at later stages RXR gamma was also expressed in the liver diverticulum. Addition of either thyroid hormone or 9-cis retinoic acid, the ligands for XTR beta and RXR gamma, respectively, did not alter the expression of their receptors. However, the addition of thyroid hormone and 9-cis retinoic acid did alter rhodopsin mRNA expression. Addition of thyroid hormone generates a small expansion of the rhodopsin expression domain. When 9-cis retinoic acid or a combination of thyroid hormone and 9-cis retinoic acid was administered, there was a decrease in the expression domain of rhodopsin in the developing retina. These results provide evidence for an early role for XTR beta and RXR gamma in the developing Xenopus retina.


Subject(s)
Cell Differentiation/physiology , Embryo, Nonmammalian/metabolism , Eye/metabolism , Retinoid X Receptor gamma/genetics , Thyroid Hormone Receptors beta/genetics , Animals , Eye/cytology , Eye/embryology , Gene Expression Profiling , Ligands , Retinoid X Receptor gamma/metabolism , Rhodopsin/metabolism , Thyroid Hormone Receptors beta/metabolism , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...