123 results on '"Hemal H. Patel"'
Search Results
2. aPC/PAR1 confers endothelial anti-apoptotic activity via a discrete, β-arrestin-2–mediated SphK1-S1PR1-Akt signaling axis
- Author
-
Luisa J. Coronel, Hemal H. Patel, Neil Grimsey, Cisneros-Aguirre M, Mark A. Lawson, Olivia Molinar-Inglis, Anand Patwardhan, Gomez-Menzies Pk, JoAnn Trejo, Dequina A. Nicholas, Cierra A. Birch, Huilan Lin, and Brian H. Chen
- Subjects
Pyrrolidines ,Pyridines ,Organophosphonates ,PAR-1 ,Apoptosis ,3-Ring ,endothelial dysfunction ,chemistry.chemical_compound ,Lactones ,GPCR ,cytoprotection ,Heterocyclic Compounds ,Heterotrimeric G protein ,Caveolae ,Humans ,Receptor, PAR-1 ,Anilides ,Sulfones ,Enzyme Inhibitors ,Receptor ,Protein kinase B ,Sphingosine-1-Phosphate Receptors ,S1PR1 ,biased signaling ,Multidisciplinary ,Sphingosine ,Chemistry ,Methanol ,Endothelial Cells ,Biological Sciences ,beta-Arrestin 2 ,Cell biology ,Phosphotransferases (Alcohol Group Acceptor) ,Gene Expression Regulation ,cardiovascular system ,Phosphorylation ,Signal transduction ,Heterocyclic Compounds, 3-Ring ,Proto-Oncogene Proteins c-akt ,Platelet Aggregation Inhibitors ,Protein C - Abstract
Endothelial dysfunction is associated with vascular disease and results in disruption of endothelial barrier function and increased sensitivity to apoptosis. Currently, there are limited treatments for improving endothelial dysfunction. Activated protein C (aPC), a promising therapeutic, signals via protease-activated receptor-1 (PAR1) and mediates several cytoprotective responses, including endothelial barrier stabilization and anti-apoptotic responses. We showed that aPC-activated PAR1 signals preferentially via β-arrestin-2 (β-arr2) and dishevelled-2 (Dvl2) scaffolds rather than G proteins to promote Rac1 activation and barrier protection. However, the signaling pathways utilized by aPC/PAR1 to mediate anti-apoptotic activities are not known. aPC/PAR1 cytoprotective responses also require coreceptors; however, it is not clear how coreceptors impact different aPC/PAR1 signaling pathways to drive distinct cytoprotective responses. Here, we define a β-arr2-mediated sphingosine kinase-1 (SphK1)-sphingosine-1-phosphate receptor-1 (S1PR1)-Akt signaling axis that confers aPC/PAR1-mediated protection against cell death. Using human cultured endothelial cells, we found that endogenous PAR1 and S1PR1 coexist in caveolin-1 (Cav1)-rich microdomains and that S1PR1 coassociation with Cav1 is increased by aPC activation of PAR1. Our study further shows that aPC stimulates β-arr2-dependent SphK1 activation independent of Dvl2 and is required for transactivation of S1PR1-Akt signaling and protection against cell death. While aPC/PAR1-induced, extracellular signal-regulated kinase 1/2 (ERK1/2) activation is also dependent on β-arr2, neither SphK1 nor S1PR1 are integrated into the ERK1/2 pathway. Finally, aPC activation of PAR1-β-arr2-mediated protection against apoptosis is dependent on Cav1, the principal structural protein of endothelial caveolae. These studies reveal that different aPC/PAR1 cytoprotective responses are mediated by discrete, β-arr2-driven signaling pathways in caveolae.
- Published
- 2021
3. Synapsin-Promoted Caveolin-1 Overexpression Maintains Mitochondrial Morphology and Function in PSAPP Alzheimer’s Disease Mice
- Author
-
Brian P. Head, Yuki Terada, Hemal H. Patel, Taiga Ichinomiya, Dongsheng Wang, and Shanshan Wang
- Subjects
Scaffold protein ,Aging ,Caveolin 1 ,Mitochondrion ,Neurodegenerative ,Inbred C57BL ,Alzheimer's Disease ,Hippocampus ,Transgenic ,Amyloid beta-Protein Precursor ,Mice ,MFN1 ,2.1 Biological and endogenous factors ,oxidative stress ,Aetiology ,Phosphorylation ,Biology (General) ,Neurons ,Chemistry ,Presenilins ,General Medicine ,Synapsin ,gene therapy ,Cell biology ,Mitochondria ,mitochondria ,Neuroprotective Agents ,mitochondrial fusion ,Neurological ,Alzheimer’s disease ,QH301-705.5 ,1.1 Normal biological development and functioning ,Mice, Transgenic ,Neuroprotection ,Article ,Underpinning research ,Alzheimer Disease ,Acquired Cognitive Impairment ,Animals ,transgenic ,Animal ,Neurosciences ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Synapsins ,Brain Disorders ,Mice, Inbred C57BL ,Disease Models, Animal ,Disease Models ,Dementia ,caveolin - Abstract
Mitochondrial dysfunction plays a pivotal role in the Alzheimer’s Disease (AD) pathology. Disrupted mitochondrial dynamics (i.e., fusion/fission balance), which are essential for normal mitochondria structure and function, are documented in AD. Caveolin-1 (Cav-1), a membrane/lipid raft (MLR) scaffolding protein regulates metabolic pathways in several different cell types such as hepatocytes and cancer cells. Previously, we have shown decreased expression of Cav-1 in the hippocampus of 9-month (m) old PSAPP mice, while hippocampal overexpression of neuron-targeted Cav-1 using the synapsin promoter (i.e., SynCav1) preserved cognitive function, neuronal morphology, and synaptic ultrastructure in 9 and 12 m PSAPP mice. Considering the central role of energy production in maintaining normal neuronal and synaptic function and survival, the present study reveals that PSAPP mice exhibit disrupted mitochondrial distribution, morphometry, and respiration. In contrast, SynCav1 mitigates mitochondrial damage and loss and enhances mitochondrial respiration. Furthermore, by examining mitochondrial dynamics, we found that PSAPP mice showed a significant increase in the phosphorylation of mitochondrial dynamin-related GTPase protein (DRP1), resulting in excessive mitochondria fragmentation and dysfunction. In contrast, hippocampal delivery of SynCav1 significantly decreased p-DRP1 and augmented the level of the mitochondrial fusion protein, mitofusin1 (Mfn1) in PSAPP mice, a molecular event, which may mechanistically explain for the preserved balance of mitochondria fission/fusion and metabolic resilience in 12 m PSAPP-SynCav1 mice. Our data demonstrate the critical role for Cav-1 in maintaining normal mitochondrial morphology and function through affecting mitochondrial dynamics and explain a molecular and cellular mechanism underlying the previously reported neuroprotective and cognitive preservation induced by SynCav1 in PSAPP mouse model of AD.
- Published
- 2021
- Full Text
- View/download PDF
4. Neuron‐targeted caveolin‐1 improves neuromuscular function and extends survival in SOD1 G93A mice
- Author
-
Jesse Wackerbarth, Martin Marsala, Oleksandr Platoshyn, David M. Roth, Atsushi Sawada, Piyush M. Patel, Joseph Leem, Hemal H. Patel, Junji Egawa, Brian P. Head, Alice E. Zemljic-Harpf, Shanshan Wang, Minyu Jian, and Jan M. Schilling
- Subjects
0301 basic medicine ,biology ,Tropomyosin receptor kinase B ,Spinal cord ,medicine.disease ,Biochemistry ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Caveolin 1 ,Genetics ,medicine ,biology.protein ,Neuron ,Amyotrophic lateral sclerosis ,Receptor ,Molecular Biology ,Lipid raft ,030217 neurology & neurosurgery ,Biotechnology ,Neurotrophin - Abstract
Interventions that preserve motor neurons or restore functional motor neuroplasticity may extend longevity in amyotrophic lateral sclerosis (ALS). Delivery of neurotrophins may potentially revive degenerating motor neurons, yet this approach is dependent on the proper subcellular localization of neurotrophin receptor (NTR) to plasmalemmal signaling microdomains, termed membrane/lipid rafts (MLRs). We previously showed that overexpression of synapsin-driven caveolin-1 (Cav-1) (SynCav1) increases MLR localization of NTR [e.g., receptor tyrosine kinase B (TrkB)], promotes hippocampal synaptic and neuroplasticity, and significantly improves learning and memory in aged mice. The present study crossed a SynCav1 transgene-positive (SynCav1+) mouse with the mutant human superoxide dismutase glycine to alanine point mutation at amino acid 93 (hSOD1G93A) mouse model of ALS. When compared with hSOD1G93A, hSOD1G93A/SynCav1+ mice exhibited greater body weight and longer survival as well as better motor function. Microscopic analyses of hSOD1G93A/SynCav1+ spinal cords revealed preserved spinal cord α-motor neurons and preserved mitochondrial morphology. Moreover, hSOD1G93A/SynCav1+ spinal cords contained more MLRs (cholera toxin subunit B positive) and MLR-associated TrkB and Cav-1 protein expression. These findings demonstrate that SynCav1 delays disease progression in a mouse model of ALS, potentially by preserving or restoring NTR expression and localization to MLRs.-Sawada, A., Wang, S., Jian, M., Leem, J., Wackerbarth, J., Egawa, J., Schilling, J. M., Platoshyn, O., Zemljic-Harpf, A., Roth, D. M., Patel, H. H., Patel, P. M., Marsala, M., Head, B. P. Neuron-targeted caveolin-1 improves neuromuscular function and extends survival in SOD1G93A mice.
- Published
- 2019
5. Metformin intervention prevents cardiac dysfunction in a murine model of adult congenital heart disease
- Author
-
Nadia Rosenthal, Milena B. Furtado, Vivek M. Philip, Stuart K. Archer, Jan M. Schilling, Anjana Chandran, Raghav Pandey, Hemal H. Patel, Julia C. Wilmanns, Gael Cagnone, Qizhu Wu, James T. Pearson, Joerg Heineke, Olivia J Hon, Preeti Bais, Mauro W. Costa, Heidi Kocalis, Mirana Ramialison, Elvira Forte, and David Coleman
- Subjects
0301 basic medicine ,Male ,Heart disease ,Physiology ,Left ,Inbred C57BL ,Cardiovascular ,Ventricular Dysfunction, Left ,Mice ,Congenital ,0302 clinical medicine ,Ventricular Dysfunction ,2.1 Biological and endogenous factors ,Adult congenital heart disease ,Cardiac Output ,Aetiology ,Heart Defects ,Metabolic Syndrome ,Diabetes ,Metformin ,3. Good health ,Heart Disease ,Cardiology ,Original Article ,Energy source ,medicine.drug ,Heart Defects, Congenital ,medicine.medical_specialty ,lcsh:Internal medicine ,030209 endocrinology & metabolism ,03 medical and health sciences ,Internal medicine ,Diabetes mellitus ,Genetic model ,medicine ,Animals ,Hypoglycemic Agents ,Obesity ,lcsh:RC31-1245 ,Molecular Biology ,Metabolic and endocrine ,Nutrition ,business.industry ,Prevention ,Cell Biology ,medicine.disease ,Mice, Inbred C57BL ,030104 developmental biology ,Metabolism ,Heart failure ,Biochemistry and Cell Biology ,Metabolic syndrome ,business ,Energy Metabolism - Abstract
Objective Congenital heart disease (CHD) is the most frequent birth defect worldwide. The number of adult patients with CHD, now referred to as ACHD, is increasing with improved surgical and treatment interventions. However the mechanisms whereby ACHD predisposes patients to heart dysfunction are still unclear. ACHD is strongly associated with metabolic syndrome, but how ACHD interacts with poor modern lifestyle choices and other comorbidities, such as hypertension, obesity, and diabetes, is mostly unknown. Methods We used a newly characterized mouse genetic model of ACHD to investigate the consequences and the mechanisms associated with combined obesity and ACHD predisposition. Metformin intervention was used to further evaluate potential therapeutic amelioration of cardiac dysfunction in this model. Results ACHD mice placed under metabolic stress (high fat diet) displayed decreased left ventricular ejection fraction. Comprehensive physiological, biochemical, and molecular analysis showed that ACHD hearts exhibited early changes in energy metabolism with increased glucose dependence as main cardiac energy source. These changes preceded cardiac dysfunction mediated by exposure to high fat diet and were associated with increased disease severity. Restoration of metabolic balance by metformin administration prevented the development of heart dysfunction in ACHD predisposed mice. Conclusions This study reveals that early metabolic impairment reinforces heart dysfunction in ACHD predisposed individuals and diet or pharmacological interventions can be used to modulate heart function and attenuate heart failure. Our study suggests that interactions between genetic and metabolic disturbances ultimately lead to the clinical presentation of heart failure in patients with ACHD. Early manipulation of energy metabolism may be an important avenue for intervention in ACHD patients to prevent or delay onset of heart failure and secondary comorbidities. These interactions raise the prospect for a translational reassessment of ACHD presentation in the clinic., Highlights • Adult congenital heart disease (ACHD) incidence is rapidly increasing. • Obesity triggers cardiac dysfunction in ACHD genetic predisposition mouse model. • Early metabolic and mitochondrial dysfunction was detected in ACHD hearts. • Energy modulation by metformin prevents progression of cardiac dysfunction in ACHD.
- Published
- 2019
6. Regulation of theβ-Adrenergic Receptor Signaling Pathway in Sustained Ligand-Activated Preconditioning
- Author
-
J. P. Headrick, L. Wendt, Jason Nigel John Peart, Hemal H. Patel, Simon R. Foster, and L. E. See Hoe
- Subjects
0301 basic medicine ,Pharmacology ,Gs alpha subunit ,Cell biology ,Wortmannin ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,chemistry ,Molecular Medicine ,Phosphorylation ,Signal transduction ,Protein kinase A ,Protein kinase B ,Rho-associated protein kinase ,030217 neurology & neurosurgery ,PI3K/AKT/mTOR pathway - Abstract
Sustained ligand-activated preconditioning (SLP), induced with chronic opioid receptor (OR) agonism, enhances tolerance to ischemia/reperfusion injury in young and aged hearts. Underlying mechanisms remain ill-defined, although early data implicate phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) during the induction phase, and β2-adrenoceptor (β2-AR), Gs alpha subunit (Gαs), and protein kinase A (PKA) involvement in subsequent cardioprotection. Here, we tested for induction of a protective β2-AR/Gαs/PKA signaling axis with SLP to ascertain whether signaling changes were PI3K-dependent (by sustained cotreatment with wortmannin), and whether the downstream PKA target Rho kinase (ROCK) participates in subsequent cardioprotection (by acute treatment with fasudil). A protected phenotype was evident after 5 days of OR agonism (using morphine) in association with increased membrane versus reduced cytosolic levels of total and phosphorylated β2-ARs; increased membrane and cytosolic expression of 52 and 46 kDa Gαs isoforms, respectively; and increased phosphorylation of PKA and Akt. Nonetheless, functional sensitivities of β2-ARs and adenylyl cyclase were unchanged based on concentration-response analyses for formoterol, fenoterol, and 6-[3-(dimethylamino)propionyl]-forskolin. Protection with SLP was not modified by ROCK inhibition, and changes in β2-AR, Gαs, and PKA expression appeared insensitive to PI3K inhibition, although 5 days of wortmannin alone exerted unexpected effects on signaling (also increasing membrane β2-AR and PKA expression/phosphorylation and Gαs levels). In summary, sustained OR agonism upregulates cardiac membrane β2-AR expression and phosphorylation in association with increased Gαs subtype levels and PKA phosphorylation. While Akt phosphorylation was evident, PI3K activity appears nonessential to OR upregulation of the β2-AR signal axis. This opioidergic remodeling of β2-AR signaling may explain β2-AR, Gαs, and PKA dependence of SLP protection.
- Published
- 2019
7. Subpial Gene Delivery of synapsin‐promoted Caveolin‐1 Prolongs Survival in hSOD G93A mice Model of ALS
- Author
-
Hemal H. Patel, Takahiro Tadokoro, Shanshan Wang, Martin Marsala, Taiga Ichinomiya, and Brian P. Head
- Subjects
Caveolin 1 ,Genetics ,Synapsin ,Gene delivery ,Biology ,Molecular Biology ,Biochemistry ,Biotechnology ,Cell biology - Published
- 2021
8. Loss of Immunohistochemical Reactivity in Association With Handling-Induced Dark Neurons in Mouse Brains
- Author
-
Rachel C Chang, Benchawanna Soontornniyomkij, Hemal H. Patel, Jan M. Schilling, Virawudh Soontornniyomkij, and Dilip V. Jeste
- Subjects
Male ,Aging ,hippocampus ,Hippocampus ,Hippocampal formation ,Toxicology ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,C57BL/6 mouse ,2.1 Biological and endogenous factors ,Paraformaldehyde ,Neurons ,0303 health sciences ,Glial fibrillary acidic protein ,biology ,Microglia ,artifact ,Immunohistochemistry ,medicine.anatomical_structure ,immunohistochemistry ,Neurological ,Artifacts ,medicine.medical_specialty ,Neurite ,1.1 Normal biological development and functioning ,Clinical Sciences ,Article ,Specimen Handling ,Pathology and Forensic Medicine ,03 medical and health sciences ,Internal medicine ,medicine ,Animals ,immunofluorescence ,Molecular Biology ,mouse ,030304 developmental biology ,Nutrition ,Neurosciences ,Cell Biology ,Mice, Inbred C57BL ,C57BL ,Endocrinology ,chemistry ,nervous system ,biology.protein ,Neuron ,Biomarkers ,030217 neurology & neurosurgery ,dark neurons - Abstract
The handling-induced dark neuron is a histological artifact observed in brain samples handled before fixation with aldehydes. To explore associations between dark neurons and immunohistochemical alterations in mouse brains, we examined protein products encoded by Cav3 (neuronal perikarya/neurites), Rbbp4 (neuronal nuclei), Gfap (astroglia), and Aif1 (microglia) genes in adjacent tissue sections. Here, dark neurons were incidental findings from our prior project, studying the effects of age and high-fat diet on metabolic homeostasis in male C57BL/6N mice. Available were brains from 4 study groups: middle-aged/control diet, middle-aged/high-fat diet, old/control diet, and old/high-fat diet. Young/control diet mice were used as baseline. The hemibrains were immersion-fixed with paraformaldehyde and paraffin-embedded. In the hippocampal formation, we found negative correlations between dark neuron hyperbasophilia and immunoreactivity for CAV3, RBBP4, and glial fibrillary acidic protein (GFAP) using quantitative image analysis. There was no significant difference in dark neuron hyperbasophilia or immunoreactivity for any protein examined among all groups. In contrast, in the hippocampal fimbria, old age seemed to be associated with higher immunoreactivity for GFAP and allograft inflammatory factor-1. Our findings suggest that loss of immunohistochemical reactivity for CAV3, RBBP4, and GFAP in the hippocampal formation is an artifact associated with the occurrence of dark neurons. The unawareness of dark neurons may lead to misinterpretation of immunohistochemical reactivity alterations.
- Published
- 2020
9. Phosphorylation of protein kinase A (PKA) regulatory subunit RIα by protein kinase G (PKG) primes PKA for catalytic activity in cells
- Author
-
Kristofer J. Haushalter, Darren E. Casteel, Andrea Raffeiner, Susan S. Taylor, Hemal H. Patel, and Eduard Stefan
- Subjects
0301 basic medicine ,Gene isoform ,Biochemistry & Molecular Biology ,post-translational modification (PTM) ,Cyclic AMP-Dependent Protein Kinase RIalpha Subunit ,Protein subunit ,Mutant ,Medical and Health Sciences ,Biochemistry ,Serine ,03 medical and health sciences ,Catalytic Domain ,Cyclic AMP ,Cyclic GMP-Dependent Protein Kinases ,2.1 Biological and endogenous factors ,Humans ,PKA Regulatory Subunit RI alpha ,Aetiology ,protein kinase G (PKG) ,Protein kinase A ,Molecular Biology ,Binding Sites ,protein kinase G ,phosphorylation ,Chemistry ,protein kinase ,Cell Biology ,Biological Sciences ,In vitro ,Cell biology ,protein kinase A (PKA) ,HEK293 Cells ,030104 developmental biology ,post-translational modification ,Chemical Sciences ,Phosphorylation ,protein kinase A ,PKA Regulatory Subunit RI alpha (RI&α) ,cGMP-dependent protein kinase ,Signal Transduction ,Protein Binding - Abstract
cAMP-dependent protein kinase (PKAc) is a pivotal signaling protein in eukaryotic cells. PKAc has two well-characterized regulatory subunit proteins, RI and RII (each having α and β isoforms), which keep the PKAc catalytic subunit in a catalytically inactive state until activation by cAMP. Previous reports showed that the RIα regulatory subunit is phosphorylated by cGMP-dependent protein kinase (PKG) in vitro, whereupon phosphorylated RIα no longer inhibits PKAc at normal (1:1) stoichiometric ratios. However, the significance of this phosphorylation as a mechanism for activating type I PKA holoenzymes has not been fully explored, especially in cellular systems. In this study, we further examined the potential of RIα phosphorylation to regulate physiologically relevant "desensitization" of PKAc activity. First, the serine 101 site of RIα was validated as a target of PKGIα phosphorylation both in vitro and in cells. Analysis of a phosphomimetic substitution in RIα (S101E) showed that modification of this site increases PKAc activity in vitro and in cells, even without cAMP stimulation. Numerous techniques were used to show that although Ser101 variants of RIα can bind PKAc, the modified linker region of the S101E mutant has a significantly reduced affinity for the PKAc active site. These findings suggest that RIα phosphorylation may be a novel mechanism to circumvent the requirement of cAMP stimulus to activate type I PKA in cells. We have thus proposed a model to explain how PKG phosphorylation of RIα creates a "sensitized intermediate" state that is in effect primed to trigger PKAc activity.
- Published
- 2018
10. Caveolins and cavins in the trafficking, maturation, and degradation of caveolae: implications for cell physiology
- Author
-
Paul A. Insel, Hemal H. Patel, and Anna R. Busija
- Subjects
0301 basic medicine ,Cell physiology ,Scaffold protein ,Physiology ,Membrane Proteins ,RNA-Binding Proteins ,Review ,Cell Biology ,Biology ,Caveolae ,Caveolins ,Models, Biological ,Cell Physiological Phenomena ,Cell biology ,Protein Transport ,03 medical and health sciences ,030104 developmental biology ,Caveolin ,cardiovascular system ,Animals ,Humans ,Lipid raft ,Cavin - Abstract
Caveolins (Cavs) are ~20 kDa scaffolding proteins that assemble as oligomeric complexes in lipid raft domains to form caveolae, flask-shaped plasma membrane (PM) invaginations. Caveolae (“little caves”) require lipid-lipid, protein-lipid, and protein-protein interactions that can modulate the localization, conformational stability, ligand affinity, effector specificity, and other functions of proteins that are partners of Cavs. Cavs are assembled into small oligomers in the endoplasmic reticulum (ER), transported to the Golgi for assembly with cholesterol and other oligomers, and then exported to the PM as an intact coat complex. At the PM, cavins, ~50 kDa adapter proteins, oligomerize into an outer coat complex that remodels the membrane into caveolae. The structure of caveolae protects their contents (i.e., lipids and proteins) from degradation. Cellular changes, including signal transduction effects, can destabilize caveolae and produce cavin dissociation, restructuring of Cav oligomers, ubiquitination, internalization, and degradation. In this review, we provide a perspective of the life cycle (biogenesis, degradation), composition, and physiologic roles of Cavs and caveolae and identify unanswered questions regarding the roles of Cavs and cavins in caveolae and in regulating cell physiology.1
- Published
- 2017
11. Caveolin-3 plays a critical role in autophagy after ischemia-reperfusion
- Author
-
Adam Kassan, Piyush M. Patel, Uyen Pham, Melissa E. Reichelt, Quynhmy Nguyen, Eunbyul Cho, Brian P. Head, David M. Roth, and Hemal H. Patel
- Subjects
0301 basic medicine ,Programmed cell death ,Caveolin 3 ,Physiology ,Myocardial Reperfusion Injury ,030204 cardiovascular system & hematology ,Mitochondrion ,Biology ,Caveolae ,ATG12 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Ischemia ,Caveolin ,Autophagy ,medicine ,Animals ,Myocytes, Cardiac ,Cardiac muscle cell ,Caspase 3 ,Cytochromes c ,Heart ,Articles ,Cell Biology ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Reperfusion ,cardiovascular system ,Apoptosis Regulatory Proteins - Abstract
Autophagy is a dynamic recycling process responsible for the breakdown of misfolded proteins and damaged organelles, providing nutrients and energy for cellular renovation and homeostasis. Loss of autophagy is associated with cardiovascular diseases. Caveolin-3 (Cav-3), a muscle-specific isoform, is a structural protein within caveolae and is critical to stress adaptation in the heart. Whether Cav-3 plays a role in regulating autophagy to modulate cardiac stress responses remains unknown. In the present study, we used HL-1 cells, a cardiac muscle cell line, with stable Cav-3 knockdown (Cav-3 KD) and Cav-3 overexpression (Cav-3 OE) to study the impact of Cav-3 in regulation of autophagy. We show that traditional stimulators of autophagy (i.e., rapamycin and starvation) result in upregulation of the process in Cav-3 OE cells while Cav-3 KD cells have a blunted response. Cav-3 coimmunoprecipitated with beclin-1 and Atg12, showing an interaction of caveolin with autophagy-related proteins. In the heart, autophagy may be a major regulator of protection from ischemic stress. We found that Cav-3 KD cells have a decreased expression of autophagy markers [beclin-1, light chain (LC3-II)] after simulated ischemia and ischemia-reperfusion (I/R) compared with WT, whereas OE cells showed increased expression. Moreover, Cav-3 KD cells showed increased cell death and higher level of apoptotic proteins (cleaved caspase-3 and cytochrome c) with suppressed mitochondrial function in response to simulated ischemia and I/R, whereas Cav-3 OE cells were protected and had preserved mitochondrial function. Taken together, these results indicate that autophagy regulates adaptation to cardiac stress in a Cav-3-dependent manner.
- Published
- 2016
12. Cardiac ischemia-reperfusion injury induces ROS-dependent loss of PKA regulatory subunit RIα
- Author
-
Kristofer J. Haushalter, Jan M. Schilling, Hemal H. Patel, Guy Perkins, Mira Sastri, Young Duk Song, Stefan Strack, and Susan S. Taylor
- Subjects
Male ,Physiology ,Protein subunit ,Cyclic AMP-Dependent Protein Kinase RIalpha Subunit ,A Kinase Anchor Proteins ,Myocardial Reperfusion Injury ,Mitochondrion ,medicine.disease_cause ,Mitochondria, Heart ,Cell Line ,Mice ,Physiology (medical) ,medicine ,Animals ,Myocytes, Cardiac ,Cells, Cultured ,Chemistry ,Cardiac ischemia ,medicine.disease ,Cell biology ,Mice, Inbred C57BL ,Pka signaling ,Cardiology and Cardiovascular Medicine ,Reactive Oxygen Species ,Reperfusion injury ,Oxidative stress ,Signal Transduction ,Research Article - Abstract
Type I PKA regulatory α-subunit (RIα; encoded by the Prkar1a gene) serves as the predominant inhibitor protein of the catalytic subunit of cAMP-dependent protein kinase (PKAc). However, recent evidence suggests that PKA signaling can be initiated by cAMP-independent events, especially within the context of cellular oxidative stress such as ischemia-reperfusion (I/R) injury. We determined whether RIα is actively involved in the regulation of PKA activity via reactive oxygen species (ROS)-dependent mechanisms during I/R stress in the heart. Induction of ex vivo global I/R injury in mouse hearts selectively downregulated RIα protein expression, whereas RII subunit expression appears to remain unaltered. Cardiac myocyte cell culture models were used to determine that oxidant stimulus (i.e., H2O2) alone is sufficient to induce RIα protein downregulation. Transient increase of RIα expression (via adenoviral overexpression) negatively affects cell survival and function upon oxidative stress as measured by increased induction of apoptosis and decreased mitochondrial respiration. Furthermore, analysis of mitochondrial subcellular fractions in heart tissue showed that PKA-associated proteins are enriched in subsarcolemmal mitochondria (SSM) fractions and that loss of RIα is most pronounced at SSM upon I/R injury. These data were supported via electron microscopy in A-kinase anchoring protein 1 (AKAP1)-knockout mice, where loss of AKAP1 expression leads to aberrant mitochondrial morphology manifested in SSM but not interfibrillar mitochondria. Thus, we conclude that modification of RIα via ROS-dependent mechanisms induced by I/R injury has the potential to sensitize PKA signaling in the cell without the direct use of the canonical cAMP-dependent activation pathway.NEW & NOTEWORTHY We uncovered a previously undescribed phenomenon involving oxidation-induced activation of PKA signaling in the progression of cardiac ischemia-reperfusion injury. Type I PKA regulatory subunit RIα, but not type II PKA regulatory subunits, is dynamically regulated by oxidative stress to trigger the activation of the catalytic subunit of PKA in cardiac myocytes. This effect may play a critical role in the regulation of subsarcolemmal mitochondria function upon the induction of ischemic injury in the heart.
- Published
- 2019
13. Caveolin-1 Phosphorylation Is Essential for Axonal Growth of Human Neurons Derived From iPSCs
- Author
-
Shanshan Wang, Zheng Zhang, Angels Almenar-Queralt, Joseph Leem, Celine DerMardirossian, David M. Roth, Piyush M. Patel, Hemal H. Patel, and Brian P. Head
- Subjects
0301 basic medicine ,caveolin-1 ,Intellectual and Developmental Disabilities (IDD) ,1.1 Normal biological development and functioning ,iPSCs ,RAC1 ,Rett syndrome ,Biology ,Regenerative Medicine ,axonal growth ,lcsh:RC321-571 ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Stem Cell Research - Nonembryonic - Human ,Underpinning research ,Neuroplasticity ,medicine ,Axon ,Progenitor cell ,Stem Cell Research - Embryonic - Human ,Induced pluripotent stem cell ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Original Research ,Rac1/Cdc42 ,Pediatric ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Stem Cell Research - Induced Pluripotent Stem Cell ,phosphorylation ,Neurosciences ,medicine.disease ,Stem Cell Research ,NPCs ,Cell biology ,Brain Disorders ,030104 developmental biology ,medicine.anatomical_structure ,Mental Health ,nervous system ,Caveolin 1 ,Neurological ,Neuron differentiation ,cardiovascular system ,Stem Cell Research - Nonembryonic - Non-Human ,Biochemistry and Cell Biology ,030217 neurology & neurosurgery ,Neuroscience - Abstract
Proper axonal growth and guidance is essential for neuron differentiation and development. Abnormal neuronal development due to genetic or epigenetic influences can contribute to neurological and mental disorders such as Down syndrome, Rett syndrome, and autism. Identification of the molecular targets that promote proper neuronal growth and differentiation may restore structural and functional neuroplasticity, thus improving functional performance in neurodevelopmental disorders. Using differentiated human neuronal progenitor cells (NPCs) derived from induced pluripotent stem cells (iPSCs), the present study demonstrates that during early stage differentiation of human NPCs, neuron-targeted overexpression constitutively active Rac1 (Rac1CA) and constitutively active Cdc42 (Cdc42CA) enhance expression of P-Cav-1, T-Cav-1, and P-cofilin and increases axonal growth. Similarly, neuron-targeted over-expression of Cav-1 (termed SynCav1) increases axonal development by increasing both axon length and volume. Moreover, inhibition of Cav-1(Y14A) phosphorylation blunts Rac1/Cdc42-mediated both axonal growth and differentiation of human NPCs and SynCav1(Y14A)-treated NPCs exhibited blunted axonal growth. These results suggest that: (1) SynCav1-mediated dendritic and axonal growth in human NPCs is dependent upon P-Cav-1, (2) P-Cav-1 is necessary for proper axonal growth during early stages of neuronal differentiation, and (3) Rac1/Cdc42CA-mediated neuronal growth is in part dependent upon P-Cav-1. In conclusion, Cav-1 phosphorylation is essential for human neuronal axonal growth during early stages of neuronal differentiation.
- Published
- 2019
14. Deletion of caveolin scaffolding domain alters cancer cell migration
- Author
-
Aayush Boddu, Yousuke T. Horikawa, Jonathan Okerblom, Sunaho Okada, Sadaf Azad Raja, Fiona Murray, Hideshi Okada, Itta Kawamura, Supriyo Ray, Yoshiteru Murofushi, and Hemal H. Patel
- Subjects
0301 basic medicine ,STAT3 Transcription Factor ,Cell signaling ,Cellular differentiation ,Caveolin 1 ,Biology ,HeLa ,03 medical and health sciences ,0302 clinical medicine ,Protein Domains ,Cell Movement ,Neoplasms ,Caveolin ,Humans ,Neoplasm Metastasis ,Molecular Biology ,Cells, Cultured ,Sequence Deletion ,Cell migration ,Cell Biology ,Cell cycle ,biology.organism_classification ,HCT116 Cells ,Cell biology ,G2 Phase Cell Cycle Checkpoints ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer cell ,cardiovascular system ,Signal transduction ,HT29 Cells ,Developmental Biology ,HeLa Cells ,Research Paper - Abstract
Caveolin-1 (Cav-1) is an integral membrane protein that plays an important role in proliferative and terminally differentiated cells. As a structural component of Caveolae, Cav-1 interacts with signaling molecules via a caveolin scaffolding domain (CSD) regulating cell signaling. Recent reports have shown that Cav-1 is a negative regulator in tumor metastasis. Therefore, we hypothesize that Cav-1 inhibits cell migration through its CSD. HeLa cells were engineered to overexpress Cav-1 (Cav-1 OE), Cav-1 without a functional CSD (∆CSD), or enhanced green fluorescent protein (EGFP) as a control. HeLa cell migration was suppressed in Cav-1 OE cells while ∆CSD showed increased migration, which corresponded to a decrease in the tight junction protein, zonula occludens (ZO-1). The migration phenotype was confirmed in multiple cancer cell lines. Phosphorylated STAT-3 was decreased in Cav-1 OE cells compared to control and ∆CSD cells; reducing STAT-3 expression alone decreased cell migration. ∆CSD blunted HeLa proliferation by increasing the number of cells in the G2/M phase of the cell cycle. Overexpressing the CSD peptide alone suppressed HeLa cell migration and inhibited pSTAT3. These findings suggest that Cav-1 CSD may be critical in controlling the dynamic phenotype of cancer cells by facilitating the interaction of specific signal transduction pathways, regulating STAT3 and participating in a G2/M checkpoint. Modulating the CSD and targeting specific proteins may offer potential new therapies in the treatment of cancer metastasis.
- Published
- 2019
15. A Human Model of Whole‐Body Ischemia‐Reperfusion: Changes in Exosome Release
- Author
-
Ying Qiu (Kim) Zhou, Liem Nguyen, David M. Roth, Victor Pretorius, Antonio De Maio, Hemal H. Patel, Michael M. Madani, and Jonathan Okerblom
- Subjects
business.industry ,Genetics ,Ischemia ,medicine ,Whole body ,medicine.disease ,business ,Molecular Biology ,Biochemistry ,Exosome ,Biotechnology ,Cell biology - Published
- 2019
16. Caveolin scaffolding domain plays an important role in cancer cell migration
- Author
-
Sadaf Azad Raja, Supriyo Ray, Yoshiteru Murofushi, Hemal H. Patel, Fiona Murray, Itta Kawamura, Hideshi Okada, Sunaho Okada, and Jonathan Okerblom
- Subjects
Cell invasion ,Scaffold ,Chemistry ,Caveolin ,Genetics ,Molecular Biology ,Biochemistry ,Biotechnology ,Domain (software engineering) ,Cell biology - Published
- 2019
17. Helium-Induced Changes in Circulating Caveolin in Mice Suggest a Novel Mechanism of Cardiac Protection
- Author
-
Raphaela P. Kerindongo, Jan M. Schilling, Nina C. Weber, Hemal H. Patel, Benedikt Preckel, Markus W. Hollmann, Moritz Valerian Warmbrunn, Jamila H. Siamwala, Mehul Dhanani, McKenzie J. Fannon, David M. Roth, Alice E. Zemljic-Harpf, Young Hwan Song, Anesthesiology, ACS - Atherosclerosis & ischemic syndromes, ACS - Diabetes & metabolism, ACS - Heart failure & arrhythmias, APH - Quality of Care, and ACS - Microcirculation
- Subjects
0301 basic medicine ,Male ,noble gas ,Cardioprotection ,Inbred C57BL ,Exosomes ,Cardiovascular ,Helium ,Mitochondria, Heart ,lcsh:Chemistry ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,conditioning ,Caveolae ,Caveolin ,Noble gas ,Myocyte ,lcsh:QH301-705.5 ,Spectroscopy ,Cells, Cultured ,Cultured ,Inhalation ,Chemistry ,Heart ,General Medicine ,3. Good health ,Computer Science Applications ,Cell biology ,Mitochondria ,Heart Disease ,membranes ,cardioprotection ,cardiovascular system ,Ceramide ,Cardiotonic Agents ,Cells ,Myocardial Reperfusion Injury ,helium ,Caveolins ,Catalysis ,Article ,Inorganic Chemistry ,03 medical and health sciences ,In vivo ,Genetics ,Animals ,Physical and Theoretical Chemistry ,Molecular Biology ,Chemical Physics ,Membranes ,Cholesterol ,Organic Chemistry ,Mice, Inbred C57BL ,030104 developmental biology ,lcsh:Biology (General) ,lcsh:QD1-999 ,Other Biological Sciences ,Other Chemical Sciences ,030217 neurology & neurosurgery ,Conditioning - Abstract
The noble gas helium (He) induces cardioprotection in vivo through unknown molecular mechanisms. He can interact with and modify cellular membranes. Caveolae are cholesterol and sphingolipid-enriched invaginations of the plasma-membrane-containing caveolin (Cav) proteins that are critical in protection of the heart. Mice (C57BL/6J) inhaled either He gas or adjusted room air. Functional measurements were performed in the isolated Langendorff perfused heart at 24 h post He inhalation. Electron paramagnetic resonance spectrometry (EPR) of samples was carried out at 24 h post He inhalation. Immunoblotting was used to detect Cav-1/3 expression in whole-heart tissue, exosomes isolated from platelet free plasma (PFP) and membrane fractions. Additionally, transmission electron microscopy analysis of cardiac tissue and serum function and metabolomic analysis were performed. In contrast to cardioprotection observed in in vivo models, the isolated Langendorff perfused heart revealed no protection after He inhalation. However, levels of Cav-1/3 were reduced 24 h after He inhalation in whole-heart tissue, and Cav-3 was increased in exosomes from PFP. Addition of serum to muscle cells in culture or naï, ve ventricular tissue increased mitochondrial metabolism without increasing reactive oxygen species generation. Primary and lipid metabolites determined potential changes in ceramide by He exposure. In addition to direct effects on myocardium, He likely induces the release of secreted membrane factors enriched in caveolae. Our results suggest a critical role for such circulating factors in He-induced organ protection.
- Published
- 2019
18. Genetically Encoded Biosensors Reveal PKA Hyperphosphorylation on the Myofilaments in Rabbit Heart Failure
- Author
-
Federica Barbagallo, Qian Shi, Julie Bossuyt, Kenneth S. Ginsburg, Qingtong Wang, Bing Xu, Toni M. West, Andrea M. Isidori, Minghui Li, Fabio Naro, Qin Fu, G. R. Reddy, Yang Kevin Xiang, William T. Ferrier, Hemal H. Patel, and Donald M. Bers
- Subjects
0301 basic medicine ,Myofilament ,medicine.medical_specialty ,Physiology ,Cells ,Clinical Sciences ,heart failure ,Hyperphosphorylation ,Biosensing Techniques ,Cardiorespiratory Medicine and Haematology ,Biology ,Cardiovascular ,Article ,03 medical and health sciences ,Myofibrils ,Internal medicine ,medicine ,Animals ,2.1 Biological and endogenous factors ,Myocyte ,Myocytes, Cardiac ,Protein phosphorylation ,Aetiology ,Protein kinase A ,myofibrils ,Cells, Cultured ,adrenergic receptor ,Heart Failure ,phosphorylation ,protein kinase A ,Cardiology and Cardiovascular Medicine ,Myocytes ,Cultured ,Sarcolemma ,Prevention ,Cyclic AMP-Dependent Protein Kinases ,Cell biology ,Heart Disease ,030104 developmental biology ,Endocrinology ,Cardiovascular System & Hematology ,Phosphorylation ,Rabbits ,Myofibril ,Cardiac - Abstract
Rationale: In heart failure, myofilament proteins display abnormal phosphorylation, which contributes to contractile dysfunction. The mechanisms underlying the dysregulation of protein phosphorylation on myofilaments is not clear. Objective: This study aims to understand the mechanisms underlying altered phosphorylation of myofilament proteins in heart failure. Methods and Results: We generate a novel genetically encoded protein kinase A (PKA) biosensor anchored onto the myofilaments in rabbit cardiac myocytes to examine PKA activity at the myofilaments in responses to adrenergic stimulation. We show that PKA activity is shifted from the sarcolemma to the myofilaments in hypertrophic failing rabbit myocytes. In particular, the increased PKA activity on the myofilaments is because of an enhanced β 2 adrenergic receptor signal selectively directed to the myofilaments together with a reduced phosphodiesterase activity associated with the myofibrils. Mechanistically, the enhanced PKA activity on the myofilaments is associated with downregulation of caveolin-3 in the hypertrophic failing rabbit myocytes. Reintroduction of caveolin-3 in the failing myocytes is able to normalize the distribution of β 2 adrenergic receptor signal by preventing PKA signal access to the myofilaments and to restore contractile response to adrenergic stimulation. Conclusions: In hypertrophic rabbit myocytes, selectively enhanced β 2 adrenergic receptor signaling toward the myofilaments contributes to elevated PKA activity and PKA phosphorylation of myofilament proteins. Reintroduction of caveolin-3 is able to confine β 2 adrenergic receptor signaling and restore myocyte contractility in response to β adrenergic stimulation.
- Published
- 2016
19. Caveolin‐1 frame‐shift mutation induce changes in cellular morphology and function
- Author
-
Juan Pablo Zuniga Hertz and Hemal H. Patel
- Subjects
Chemistry ,Caveolin 1 ,Genetics ,Cellular Morphology ,Molecular Biology ,Biochemistry ,Function (biology) ,Biotechnology ,Cell biology ,Frameshift mutation - Published
- 2020
20. Loss of Caveolin‐3 (Cav‐3) Promotes G‐protein‐regulated Matrix Metalloprotease 14 (MMP14) Activation in the Aged Heart
- Author
-
Kristofer J. Haushalter, Hemal H. Patel, Paul A. Insel, and Aaron C. Overland
- Subjects
Caveolin 3 ,G protein ,Chemistry ,Genetics ,MMP14 ,Matrix metalloproteinase ,Molecular Biology ,Biochemistry ,Biotechnology ,Cell biology - Published
- 2018
21. Caveolin‐1 Phosphorylation is Essential for iPSC‐derived Human Neuronal Axonal Growth during Early Stage of Differentiation
- Author
-
Hemal H. Patel, Zheng Zhang, Shanshan Wang, Joseph Leem, and Brian P. Head
- Subjects
Caveolin 1 ,Genetics ,Phosphorylation ,Biology ,Stage (cooking) ,Molecular Biology ,Biochemistry ,Biotechnology ,Cell biology - Published
- 2018
22. Novel Marine Compounds Modulate Mitochondrial Function in H9c2 Cells: Potential New Pharmaceutical Targets to Control Cardiac Metabolism
- Author
-
Lena Gerwick, Jan M. Schilling, Hemal H. Patel, Moritz Valerian Warmbrunn, Evgenia Glukhov, William Gerwick, and Mehul Dhanani
- Subjects
Chemistry ,Genetics ,Cardiac metabolism ,Molecular Biology ,Biochemistry ,Function (biology) ,Biotechnology ,Cell biology - Published
- 2018
23. Non-canonical roles for caveolin in regulation of membrane repair and mitochondria: implications for stress adaptation with age
- Author
-
Jan M. Schilling and Hemal H. Patel
- Subjects
0301 basic medicine ,Physiology ,Regulator ,Mitochondrion ,Biology ,Bioinformatics ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,Signalling ,Ageing ,Caveolin ,Signal transduction ,Barrier function ,Function (biology) - Abstract
Many different theories of ageing have been proposed but none has served the unifying purpose of defining a molecular target that can limit the structural and functional decline associated with age that ultimately leads to the demise of the organism. We propose that the search for a molecule with these unique properties must account for regulation of the signalling efficiency of multiple cellular functions that degrade with age due to a loss of a particular protein. We suggest caveolin as one such molecule that serves as a regulator of key processes in signal transduction. We define a particular distinction between cellular senescence and ageing and propose that caveolin plays a distinct role in each of these processes. Caveolin is traditionally thought of as a membrane-localized protein regulating signal transduction via membrane enrichment of specific signalling molecules. Ultimately we focus on two non-canonical roles for caveolin - membrane repair and regulation of mitochondrial function - which may be novel features of stress adaptation, especially in the setting of ageing. The end result of preserving membrane structure and mitochondrial function is maintenance of homeostatic signalling, preserving barrier function, and regulating energy production for cell survival and resilient ageing.
- Published
- 2015
24. Role of caveolin-3 in lymphocyte activation
- Author
-
Brandon Bentley, Hemal H. Patel, Creed M. Stary, Jan M. Schilling, David M. Roth, and Chinh Tran
- Subjects
Caveolin 3 ,Knockout ,1.1 Normal biological development and functioning ,medicine.medical_treatment ,Lymphocyte ,Inflammation ,Biology ,Inbred C57BL ,Lymphocyte Activation ,Article ,General Biochemistry, Genetics and Molecular Biology ,Flow cytometry ,Mice ,Immune system ,Underpinning research ,medicine ,Animals ,Humans ,2.1 Biological and endogenous factors ,Lymphocyte Count ,Pharmacology & Pharmacy ,Aetiology ,General Pharmacology, Toxicology and Pharmaceutics ,Cytokine ,Mice, Knockout ,medicine.diagnostic_test ,Inflammatory and immune system ,Pharmacology and Pharmaceutical Sciences ,General Medicine ,Leukocyte ,Flow Cytometry ,Signaling ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Immune System ,cardiovascular system ,Cytokines ,Tumor necrosis factor alpha ,Biochemistry and Cell Biology ,medicine.symptom ,Signal transduction ,Signal Transduction - Abstract
Aims Caveolins are structural proteins clustered in lipid-rich regions of plasma membrane involved in coordinating signal transduction in various organ systems. While caveolin-1 (Cav-1) has been shown to regulate lymphocyte activation, the role of caveolin-3 (Cav-3) in immune system signaling has not been investigated. We tested the hypothesis that Cav-3 modulates lymphocyte activation. Main methods Lymphocyte/leukocyte subpopulations from WT and Cav-3 mice were profiled with flow cytometry. Cytokine production in quiescent and activated splenocytes from WT and Cav-3 mice was assessed with ELISA. Key findings Levels of T-cells, monocytes, and natural killer cells were not different between WT and KO mice, however KO mice had lower B-cell population-percentage. Functionally, activated lymphocytes from Cav-3 KO mice demonstrated significantly reduced expression of IL-2 compared to WT, while expression of TNFα, IL-6, and IL-10 was not different. Finally, expression of IL-17 was significantly reduced in T-helper cells from KO mice, while IFNγ was not, suggesting that Cav-3 is a determinant in the development of the Th-17 subpopulation. Significance This study is the first to demonstrate that Cav-3 may be a novel participant in B-cell expression, T-cell cytokine production and activation of inflammation.
- Published
- 2015
25. Modulation of caveolins, integrins and plasma membrane repair proteins in anthracycline-induced heart failure in rabbits
- Author
-
Hemal H. Patel, David M. Roth, Ana Maria Manso, H. Kirk Hammond, Zheng Zhang, Yasuhiro Ichikawa, M. Dan McKirnan, Robert S. Ross, Alice E. Zemljic-Harpf, and Kukreja, Rakesh
- Subjects
0301 basic medicine ,Integrins ,Cell Membranes ,030204 cardiovascular system & hematology ,Pharmacology ,Cardiovascular ,Biochemistry ,0302 clinical medicine ,Medicine and Health Sciences ,Anthracyclines ,Receptor ,Energy-Producing Organelles ,Mammals ,Microscopy ,Multidisciplinary ,biology ,Blotting ,Heart ,Animal Models ,Immunohistochemistry ,3. Good health ,Extracellular Matrix ,Mitochondria ,Heart Disease ,Cholesterol ,Experimental Organism Systems ,Echocardiography ,Vertebrates ,Medicine ,Rabbits ,Anatomy ,Cellular Structures and Organelles ,Western ,medicine.drug ,Research Article ,Anthracycline ,General Science & Technology ,Daunorubicin ,1.1 Normal biological development and functioning ,Science ,Integrin ,Blotting, Western ,Cardiology ,Bioenergetics ,Research and Analysis Methods ,Electron ,Caveolins ,03 medical and health sciences ,Underpinning research ,medicine ,Cell Adhesion ,Animals ,Heart Failure ,Cardiotoxicity ,Myocardium ,Organisms ,Plasma membrane repair ,Biology and Life Sciences ,Membrane Proteins ,Cell Biology ,medicine.disease ,Microscopy, Electron ,030104 developmental biology ,Membrane protein ,Coated Pits ,Heart failure ,Immunology ,Amniotes ,biology.protein ,Cardiovascular Anatomy - Abstract
Anthracyclines are chemotherapeutic drugs known to induce heart failure in a dose-dependent manner. Mechanisms involved in anthracycline cardiotoxicity are an area of relevant investigation. Caveolins bind, organize and regulate receptors and signaling molecules within cell membranes. Caveolin-3 (Cav-3), integrins and related membrane repair proteins can function as cardioprotective proteins. Expression of these proteins in anthracycline-induced heart failure has not been evaluated. We tested the hypothesis that daunorubicin alters cardioprotective protein expression in the heart. Rabbits were administered daunorubicin (3 mg/kg, IV) weekly, for three weeks or nine weeks. Nine weeks but not three weeks of daunorubicin resulted in progressive reduced left ventricular function. Cav-3 expression in the heart was unchanged at three weeks of daunorubicin and increased in nine week treated rabbits when compared to control hearts. Electron microscopy showed caveolae in the heart were increased and mitochondrial number and size were decreased after nine weeks of daunorubicin. Activated beta-1 (β1) integrin and the membrane repair protein MG53 were increased after nine weeks of daunorubicin vs. controls with no change at the three week time point. The results suggest a potential pathophysiological role for Cav3, integrins and membrane repair in daunorubicin-induced heart failure.
- Published
- 2017
26. Caveolin modulates integrin function and mechanical activation in the cardiomyocyte
- Author
-
Ingrid R. Niesman, David M. Roth, Daniel N. Deussen, Robert S. Ross, Hemal H. Patel, Sharon Israeli-Rosenberg, Chao Chen, Hideshi Okada, and Ruixia Li
- Subjects
Integrins ,Caveolin 3 ,Integrin ,Mice, Transgenic ,Mechanotransduction, Cellular ,Biochemistry ,Collagen receptor ,Focal adhesion ,Research Communication ,Mice ,Sarcolemma ,Caveolae ,Caveolin ,Genetics ,Animals ,Myocyte ,Myocytes, Cardiac ,Mechanotransduction ,Microscopy, Immunoelectron ,Molecular Biology ,Aorta ,Mice, Knockout ,biology ,Integrin beta1 ,Cell Membrane ,Heart ,Protein Structure, Tertiary ,Cell biology ,biology.protein ,Integrin, beta 6 ,Signal Transduction ,Biotechnology - Abstract
β1 integrins (β1) transduce mechanical signals in many cells, including cardiac myocytes (CM). Given their close localization, as well as their role in mechanotransduction and signaling, we hypothesized that caveolin (Cav) proteins might regulate integrins in the CM. β1 localization, complex formation, activation state, and signaling were analyzed using wild-type, Cav3 knockout, and Cav3 CM-specific transgenic heart and myocyte samples. Studies were performed under basal and mechanically loaded conditions. We found that: 1) β1 and Cav3 colocalize in CM and coimmunoprecipitate from CM protein lysates; 2) β1 is detected in a subset of caveolae; 3) loss of Cav3 caused reduction of β1D integrin isoform and active β1 integrin from the buoyant domains in the heart; 4) increased expression of myocyte Cav3 correlates with increased active β1 integrin in adult CM; 5) in vivo pressure overload of the wild-type heart results in increased activated integrin in buoyant membrane domains along with increased association between active integrin and Cav3; and 6) Cav3-deficient myocytes have perturbed basal and stretch mediated signaling responses. Thus, Cav3 protein can modify integrin function and mechanotransduction in the CM and intact heart.—Israeli-Rosenberg, S., Chen, C., Li, R., Deussen, D. N., Niesman, I. R., Okada, H., Patel, H. H., Roth, D. M., Ross, R. S. Caveolin modulates integrin function and mechanical activation in the cardiomyocyte.
- Published
- 2014
27. Signaling Epicenters: The Role of Caveolae and Caveolins in Volatile Anesthetic Induced Cardiac Protection
- Author
-
Yousuke T. Horikawa, Yasuo M. Tsutsumi, Hemal H. Patel, and David M. Roth
- Subjects
cardiac protection ,Cell signaling ,Heart Diseases ,Medicinal & Biomolecular Chemistry ,Biology ,Caveolae ,Cardiovascular ,Caveolins ,Article ,Drug Discovery ,Animals ,Humans ,volatile anesthetics ,Anesthetics ,Pharmacology ,Extramural ,Volatile anesthetic ,Pharmacology and Pharmaceutical Sciences ,lipid raft ,Cell biology ,Inhalation ,Anesthetics, Inhalation ,caveolin ,Signal transduction ,Signal Transduction - Abstract
Caveolae are flask-like invaginations of the cell surface that have been identified as signaling epicenters. Within these microdomains, caveolins are structural proteins of caveolae, which are able to interact with numerous signaling molecules affecting temporal and spatial dimensions required in cardiac protection. This complex moiety is essential to the mechanisms involved in volatile anesthetics. In this review, we will outline a general overview of caveolae and caveolins and their role in protective signaling, with a focus on the effects of volatile anesthetics. These recent developments have allowed us to better understand the mechanistic effect of volatile anesthetics and their potential in cardiac protection.
- Published
- 2014
28. Regulation of intracellular signaling and function by caveolin
- Author
-
Heidi N. Fridolfsson, David M. Roth, Paul A. Insel, and Hemal H. Patel
- Subjects
Scaffold protein ,Cell physiology ,Endoplasmic reticulum ,Reviews ,Intracellular Membranes ,Biology ,Caveolins ,Biochemistry ,Cell biology ,Membrane Microdomains ,Caveolae ,Caveolin ,Genetics ,Extracellular ,Animals ,Humans ,Signal transduction ,Molecular Biology ,Intracellular ,Signal Transduction ,Biotechnology - Abstract
Caveolae, flask-like invaginations of the plasma membrane, were discovered nearly 60 years ago. Originally regarded as fixation artifacts of electron microscopy, the functional role for these structures has taken decades to unravel. The discovery of the caveolin protein in 1992 (by the late Richard G.W. Anderson) accelerated progress in defining the contribution of caveolae to cellular physiology and pathophysiology. The three isoforms of caveolin (caveolin-1, -2, and -3) are caveolae-resident structural and scaffolding proteins that are critical for the formation of caveolae and their localization of signaling entities. A PubMed search for “caveolae” reveals ∼280 publications from their discovery in the 1950s to the early 1990s, whereas a search for “caveolae or caveolin” after 1990, identifies ∼7000 entries. Most work on the regulation of biological responses by caveolae and caveolin since 1990 has focused on caveolae as plasma membrane microdomains and the function of caveolin proteins at the plasma membrane. By contrast, our recent work and that of others has explored the localization of caveolins in multiple cellular membrane compartments and in the regulation of intracellular signaling. Cellular organelles that contain caveolin include mitochondria, nuclei and the endoplasmic reticulum. Such intracellular localization allows for a complexity of responses to extracellular stimuli by caveolin and the possibility of novel organelle-targeted therapeutics. This review focuses on the impact of intracellular localization of caveolin on signal transduction and cell regulation.—Fridolfsson, H. N., Roth, D. M., Insel, P. A., Patel, H. H. Regulation of intracellular signaling and function by caveolin.
- Published
- 2014
29. Interaction of membrane/lipid rafts with the cytoskeleton: Impact on signaling and function
- Author
-
Paul A. Insel, Hemal H. Patel, and Brian P. Head
- Subjects
0303 health sciences ,Cell signaling ,Cellular polarity ,Biophysics ,Cell Biology ,Biology ,Biochemistry ,Cell biology ,Cell membrane ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Caveolae ,Caveolin ,medicine ,Signal transduction ,Cytoskeleton ,Lipid raft ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
The plasma membrane in eukaryotic cells contains microdomains that are enriched in certain glycosphingolipids, gangliosides, and sterols (such as cholesterol) to form membrane/lipid rafts (MLR). These regions exist as caveolae, morphologically observable flask-like invaginations, or as a less easily detectable planar form. MLR are scaffolds for many molecular entities, including signaling receptors and ion channels that communicate extracellular stimuli to the intracellular milieu. Much evidence indicates that this organization and/or the clustering of MLR into more active signaling platforms depends upon interactions with and dynamic rearrangement of the cytoskeleton. Several cytoskeletal components and binding partners, as well as enzymes that regulate the cytoskeleton, localize to MLR and help regulate lateral diffusion of membrane proteins and lipids in response to extracellular events (e.g., receptor activation, shear stress, electrical conductance, and nutrient demand). MLR regulate cellular polarity, adherence to the extracellular matrix, signaling events (including ones that affect growth and migration), and are sites of cellular entry of certain pathogens, toxins and nanoparticles. The dynamic interaction between MLR and the underlying cytoskeleton thus regulates many facets of the function of eukaryotic cells and their adaptation to changing environments. Here, we review general features of MLR and caveolae and their role in several aspects of cellular function, including polarity of endothelial and epithelial cells, cell migration, mechanotransduction, lymphocyte activation, neuronal growth and signaling, and a variety of disease settings. This article is part of a Special Issue entitled: Reciprocal influences between cell cytoskeleton and membrane channels, receptors and transporters. Guest Editor: Jean Claude Herve.
- Published
- 2014
- Full Text
- View/download PDF
30. Sirtuin1 protects endothelial Caveolin-1 expression and preserves endothelial function via suppressing miR-204 and endoplasmic reticulum stress
- Author
-
Kaikobad Irani, Modar Kassan, Qiuxia Li, Julia S. Jacobs, Hemal H. Patel, Adam Kassan, Jing Liu, Mohanad Gabani, Ajit Vikram, Santosh Kumar, and Young-Rae Kim
- Subjects
0301 basic medicine ,Male ,Caveolin 1 ,Down-Regulation ,030204 cardiovascular system & hematology ,Biology ,Inbred C57BL ,Cardiovascular ,Vascular endothelial growth inhibitor ,Models, Biological ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Sirtuin 1 ,Models ,Animals ,Multidisciplinary ,Endoplasmic reticulum ,Endothelial Cells ,Biological ,Endoplasmic Reticulum Stress ,Corrigenda ,Cell biology ,Vascular endothelial growth factor B ,Mice, Inbred C57BL ,Vasodilation ,Vascular endothelial growth factor A ,MicroRNAs ,030104 developmental biology ,Vascular endothelial growth factor C ,Unfolded protein response - Abstract
Sirtuin1 (Sirt1) is a class III histone deacetylase that regulates a variety of physiological processes, including endothelial function. Caveolin1 (Cav1) is also an important determinant of endothelial function. We asked if Sirt1 governs endothelial Cav1 and endothelial function by regulating miR-204 expression and endoplasmic reticulum (ER) stress. Knockdown of Sirt1 in endothelial cells, and in vivo deletion of endothelial Sirt1, induced endothelial ER stress and miR-204 expression, reduced Cav1, and impaired endothelium-dependent vasorelaxation. All of these effects were reversed by a miR-204 inhibitor (miR-204 I) or with overexpression of Cav1. A miR-204 mimic (miR-204 M) decreased Cav1 in endothelial cells. In addition, high-fat diet (HFD) feeding induced vascular miR-204 and reduced endothelial Cav1. MiR-204-I protected against HFD-induced downregulation of endothelial Cav1. Moreover, pharmacologic induction of ER stress with tunicamycin downregulated endothelial Cav1 and impaired endothelium-dependent vasorelaxation that was rescued by overexpressing Cav1. In conclusion, Sirt1 preserves Cav1-dependent endothelial function by mitigating miR-204-mediated vascular ER stress.
- Published
- 2016
31. Increase in Cellular Cyclic AMP Concentrations Reverses the Profibrogenic Phenotype of Cardiac Myofibroblasts: A Novel Therapeutic Approach for Cardiac Fibrosis
- Author
-
Hemal H. Patel, David S.K. Lu, Nakon Aroonsakool, Paul A. Insel, and Utako Yokoyama
- Subjects
Male ,Accelerated Communication ,Cardiac fibrosis ,Cardiovascular ,Receptors, G-Protein-Coupled ,Rats, Sprague-Dawley ,Adenylyl cyclase ,Extracellular matrix ,chemistry.chemical_compound ,Fibrosis ,Receptors ,Cyclic AMP ,2.1 Biological and endogenous factors ,Pharmacology & Pharmacy ,Aetiology ,Myofibroblasts ,Cells, Cultured ,Cultured ,Forskolin ,Pharmacology and Pharmaceutical Sciences ,Cell biology ,Isoenzymes ,Heart Disease ,Plasminogen activator inhibitor-1 ,Second messenger system ,Molecular Medicine ,Collagen ,5'-Cyclic-AMP Phosphodiesterases ,Myofibroblast ,Adenylyl Cyclases ,medicine.medical_specialty ,Cells ,Biology ,G-Protein-Coupled ,Internal medicine ,Plasminogen Activator Inhibitor 1 ,medicine ,3' ,Animals ,Pharmacology ,Myocardium ,Colforsin ,Neurosciences ,Fibroblasts ,medicine.disease ,Actins ,Rats ,Endocrinology ,chemistry ,3',5'-Cyclic-AMP Phosphodiesterases ,Sprague-Dawley ,Biochemistry and Cell Biology - Abstract
Tissue fibrosis is characterized by excessive production, deposition, and contraction of the extracellular matrix (ECM). The second messenger cAMP has antifibrotic effects in fibroblasts from several tissues, including cardiac fibroblasts (CFs). Increased cellular cAMP levels can prevent the transformation of CFs into profibrogenic myofibroblasts, a critical step that precedes increased ECM deposition and tissue fibrosis. Here we tested two hypotheses: 1) myofibroblasts have a decreased ability to accumulate cAMP in response to G protein-coupled receptor (GPCR) agonists, and 2) increasing cAMP will not only prevent, but also reverse, the myofibroblast phenotype. We found that myofibroblasts produce less cAMP in response to GPCR agonists or forskolin and have decreased expression of several adenylyl cyclase (AC) isoforms and increased expression of multiple cyclic nucleotide phosphodiesterases (PDEs). Furthermore, we found that forskolin-promoted increases in cAMP or N(6)-phenyladenosine-cAMP, a protein kinase A-selective analog, reverse the myofibroblast phenotype, as assessed by the expression of collagen Iα1, α-smooth muscle actin, plasminogen activator inhibitor-1, and cellular contractile abilities, all hallmarks of a fibrogenic state. These results indicate that: 1) altered expression of AC and PDE isoforms yield a decrease in cAMP concentrations of cardiac myofibroblasts (relative to CFs) that likely contributes to their profibrotic state, and 2) approaches to increase cAMP concentrations not only prevent fibroblast-to-myofibroblast transformation but also can reverse the profibrotic myofibroblastic phenotype. We conclude that therapeutic strategies designed to enhance cellular cAMP concentrations in CFs may provide a means to reverse excessive scar formation following injury and to treat cardiac fibrosis.
- Published
- 2013
32. Impairment of TRPC1–STIM1 channel assembly and AQP5 translocation compromise agonist-stimulated fluid secretion in mice lacking caveolin1
- Author
-
Xibao Liu, Kwong Tai Cheng, Biswaranjan Pani, Ingrid R. Niesman, Brij B. Singh, Sunitha Bollimuntha, Changyu Zheng, Hemal H. Patel, Indu S. Ambudkar, and Virginia R. Achen
- Subjects
Caveolin 1 ,Saliva secretion ,Biology ,Transfection ,TRPC1 ,Mice ,Transient receptor potential channel ,Microscopy, Electron, Transmission ,Acinar cell ,Animals ,Humans ,Secretion ,Stromal Interaction Molecule 1 ,Cells, Cultured ,Cellular localization ,TRPC Cation Channels ,Mice, Knockout ,Membrane Glycoproteins ,Voltage-dependent calcium channel ,ORAI1 ,Membrane Proteins ,Cell Biology ,Immunohistochemistry ,Aquaporin 5 ,Neoplasm Proteins ,Cell biology ,Calcium Channels ,Research Article - Abstract
Summary Neurotransmitter regulation of salivary fluid secretion is mediated by activation of Ca2+ influx. The Ca2+-permeable transient receptor potential canonical 1 (TRPC1) channel is crucial for fluid secretion. However, the mechanism(s) involved in channel assembly and regulation are not completely understood. We report that Caveolin1 (Cav1) is essential for the assembly of functional TRPC1 channels in salivary glands (SG) in vivo and thus regulates fluid secretion. In Cav1−/− mouse SG, agonist-stimulated Ca2+ entry and fluid secretion are significantly reduced. Microdomain localization of TRPC1 and interaction with its regulatory protein, STIM1, are disrupted in Cav1−/− SG acinar cells, whereas Orai1–STIM1 interaction is not affected. Furthermore, localization of aquaporin 5 (AQP5), but not that of inositol (1,4,5)-trisphosphate receptor 3 or Ca2+-activated K+ channel (IK) in the apical region of acinar cell was altered in Cav1−/− SG. In addition, agonist-stimulated increase in surface expression of AQP5 required Ca2+ influx via TRPC1 channels and was inhibited in Cav1−/− SG. Importantly, adenovirus-mediated expression of Cav1 in Cav1−/− SG restored interaction of STIM1 with TRPC1 and channel activation, apical targeting and regulated trafficking of AQP5, and neurotransmitter stimulated fluid-secretion. Together these findings demonstrate that, by directing cellular localization of TRPC1 and AQP5 channels and by selectively regulating the functional assembly TRPC1–STIM1 channels, Cav1 is a crucial determinant of SG fluid secretion.
- Published
- 2013
33. Role of decoy molecules in neuronal ischemic preconditioning
- Author
-
Daniel P. Davis, Hemal H. Patel, Michael W. Kidd, Ingrid R. Niesman, Piyush M. Patel, Brian P. Head, David M. Roth, Blake Chin-Lee, Satoki Inoue, and Mathivadhani Panneerselvam
- Subjects
Receptor expression ,Ischemia ,Apoptosis ,Biology ,environment and public health ,Article ,General Biochemistry, Genetics and Molecular Biology ,TNF-Related Apoptosis-Inducing Ligand ,Caffeic Acids ,parasitic diseases ,medicine ,Animals ,cardiovascular diseases ,General Pharmacology, Toxicology and Pharmaceutics ,Decoy receptors ,Ischemic Preconditioning ,skin and connective tissue diseases ,Receptor ,Cerebral Cortex ,Neurons ,NF-kappa B ,Receptors, Death Domain ,General Medicine ,Phenylethyl Alcohol ,medicine.disease ,biological factors ,Rats ,Cell biology ,Tumor Necrosis Factor Decoy Receptors ,Animals, Newborn ,Gene Expression Regulation ,Immunology ,Ischemic preconditioning ,Decoy - Abstract
Aims Decoy receptors bind with TNF related apoptosis inducing ligands (TRAIL) but do not contain the cytoplasmic domains necessary to transduce apoptotic signals. We hypothesized that decoy receptors may confer neuronal protection against lethal ischemia after ischemic preconditioning (IPC). Main method Mixed cortical neurons were exposed to IPC one day prior to TRAIL treatment or lethal ischemia. Key findings IPC increased decoy receptor but reduced death receptor expression compared to lethal ischemia. IPC-induced increase in decoy receptor expression was reduced by prior treatment with CAPE, a nuclear factor-kappa B inhibitor (NFκB). Significance Expression of decoy molecules, dependent on NFκB, may mediate neuronal survival induced by IPC.
- Published
- 2011
34. Role of Caveolae in Cardiac Protection
- Author
-
Hemal H. Patel and David M. Roth
- Subjects
Gene isoform ,medicine.medical_specialty ,Cell signaling ,Myocardial Ischemia ,Ischemia ,Myocardial Reperfusion Injury ,Ischemia/reperfusion injury ,030204 cardiovascular system & hematology ,Caveolae ,Caveolins ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Caveolin ,Humans ,Medicine ,Myocyte ,Pediatrics, Perinatology, and Child Health ,Riley Symposium ,030304 developmental biology ,0303 health sciences ,business.industry ,medicine.disease ,Cardiac protection ,Cell biology ,Pediatrics, Perinatology and Child Health ,Cardiology ,Signal transduction ,Cardiology and Cardiovascular Medicine ,business ,Reperfusion injury ,Signal Transduction - Abstract
Myocardial ischemia/reperfusion injury is a major cause of morbidity and mortality. The molecular signaling pathways involved in cardiac protection from myocardial ischemia/reperfusion injury are complex. An emerging idea in signal transduction suggests the existence of spatially organized complexes of signaling molecules in lipid-rich microdomains of the plasma membrane known as caveolae. Caveolins—proteins abundant in caveolae—provide a scaffold to organize, traffic, and regulate signaling molecules. Numerous signaling molecules involved in cardiac protection are known to exist within caveolae or interact directly with caveolins. Over the last 4 years, our laboratories have explored the hypothesis that caveolae are vitally important to cardiac protection from myocardial ischemia/reperfusion injury. We have provided evidence that (1) caveolae and the caveolin isoforms 1 and 3 are essential for cardiac protection from myocardial ischemia/reperfusion injury, (2) stimuli that produce preconditioning of cardiac myocytes, including brief periods of ischemia/reperfusion and exposure to volatile anesthetics, alter the number of membrane caveolae, and (3) cardiac myocyte-specific overexpression of caveolin-3 can produce innate cardiac protection from myocardial ischemia/reperfusion injury. The work demonstrates that caveolae and caveolins are critical elements of signaling pathways involved in cardiac protection and suggests that caveolins are unique targets for therapy in patients at risk of myocardial ischemia.
- Published
- 2011
35. Comparison of Cardiac Cell Function in Caveolin-3 Knock-Out and TAC Mice
- Author
-
Simon M. Bryant, Hemal H. Patel, David M. Roth, Judy J. Watson, Hanne C. Gadeberg, Cherrie H. Kong, Andrew F. James, Clive H. Orchard, and Mark B. Cannell
- Subjects
Caveolin 3 ,Chemistry ,Biophysics ,Cardiac cell ,Function (biology) ,Cell biology - Published
- 2018
36. Membrane rafts and caveolae in cardiovascular signaling
- Author
-
Hemal H. Patel and Paul A. Insel
- Subjects
Kidney ,Epithelial Cells ,Raft ,Biology ,Caveolae ,Article ,Cell biology ,Cardiovascular Physiological Phenomena ,Membrane Microdomains ,Membrane ,medicine.anatomical_structure ,Nephrology ,Internal Medicine ,medicine ,Animals ,Humans ,Endothelium, Vascular ,Signal transduction ,Receptor ,Signal Transduction ,Hormone - Abstract
Purpose of review—Substantial evidence documents the key role of lipid (membrane) rafts and caveolae as microdomains that concentrate a wide variety of receptors and post-receptor components regulated by hormones, neurotransmitters and growth factors. Recent findings—Recent data document that those microdomains are important in regulating vascular endothelial and smooth muscle cells and renal epithelial cells, and in particular in signal transduction across the plasma membrane. Summary—Raft/caveolae domains are cellular regions, including in cardiovascular and renal epithelial cells, that organize a large number of signal transduction components, thereby providing spatially and temporally efficient regulation of cell function.
- Published
- 2009
37. Cardiac-Specific Overexpression of Caveolin-3 Induces Endogenous Cardiac Protection by Mimicking Ischemic Preconditioning
- Author
-
Hemal H. Patel, Yasuo M. Tsutsumi, Yousuke T. Horikawa, Paul A. Insel, Atsushi Miyanohara, Brian P. Head, Yasuko Hagiwara, Yoshihiro Ishikawa, Piyush M. Patel, Utako Yokoyama, David M. Roth, Michael W. Kidd, Michelle Jennings, and Ingrid R. Niesman
- Subjects
Gene isoform ,Cell signaling ,Caveolin 3 ,Gene Expression ,Apoptosis ,Myocardial Reperfusion Injury ,Caveolae ,Article ,Adenoviridae ,Glycogen Synthase Kinase 3 ,Mice ,Sarcolemma ,GSK-3 ,Physiology (medical) ,Animals ,Medicine ,Myocyte ,Myocytes, Cardiac ,Phosphorylation ,Mice, Knockout ,Glycogen Synthase Kinase 3 beta ,business.industry ,Cell biology ,Mice, Inbred C57BL ,Microscopy, Electron ,Cholesterol ,Ischemic Preconditioning, Myocardial ,Immunology ,cardiovascular system ,Ischemic preconditioning ,Nitric Oxide Synthase ,Cardiology and Cardiovascular Medicine ,business ,Proto-Oncogene Proteins c-akt - Abstract
Background— Caveolae, lipid-rich microdomains of the sarcolemma, localize and enrich cardiac-protective signaling molecules. Caveolin-3 (Cav-3), the dominant isoform in cardiac myocytes, is a determinant of caveolar formation. We hypothesized that cardiac myocyte–specific overexpression of Cav-3 would enhance the formation of caveolae and augment cardiac protection in vivo. Methods and Results— Ischemic preconditioning in vivo increased the formation of caveolae. Adenovirus for Cav-3 increased caveolar formation and phosphorylation of survival kinases in cardiac myocytes. A transgenic mouse with cardiac myocyte–specific overexpression of Cav-3 (Cav-3 OE) showed enhanced formation of caveolae on the sarcolemma. Cav-3 OE mice subjected to ischemia/reperfusion injury had a significantly reduced infarct size relative to transgene-negative mice. Endogenous cardiac protection in Cav-3 OE mice was similar to wild-type mice undergoing ischemic preconditioning; no increased protection was observed in preconditioned Cav-3 OE mice. Cav-3 knockout mice did not show endogenous protection and showed no protection in response to ischemic preconditioning. Cav-3 OE mouse hearts had increased basal Akt and glycogen synthase kinase-3β phosphorylation comparable to wild-type mice exposed to ischemic preconditioning. Wortmannin, a phosphoinositide 3-kinase inhibitor, attenuated basal phosphorylation of Akt and glycogen synthase kinase-3β and blocked cardiac protection in Cav-3 OE mice. Cav-3 OE mice had improved functional recovery and reduced apoptosis at 24 hours of reperfusion. Conclusions— Expression of caveolin-3 is both necessary and sufficient for cardiac protection, a conclusion that unites long-standing ultrastructural and molecular observations in the ischemic heart. The present results indicate that increased expression of caveolins, apparently via actions that depend on phosphoinositide 3-kinase, has the potential to protect hearts exposed to ischemia/reperfusion injury.
- Published
- 2008
38. The cyclic AMP effector Epac integrates pro- and anti-fibrotic signals
- Author
-
Nakon Aroonsakool, David M. Roth, Utako Yokoyama, Paul A. Insel, Hemal H. Patel, and N. Chin Lai
- Subjects
Male ,medicine.medical_specialty ,Myocardial Infarction ,Biology ,Collagen Type I ,Rats, Sprague-Dawley ,Transforming Growth Factor beta1 ,Mice ,Enzyme activator ,Cell Movement ,Internal medicine ,Cyclic AMP ,medicine ,Animals ,Guanine Nucleotide Exchange Factors ,Humans ,Fibroblast ,Protein kinase A ,Multidisciplinary ,Myocardium ,rap1 GTP-Binding Proteins ,DNA ,Biological Sciences ,Fibroblasts ,Cyclic AMP-Dependent Protein Kinases ,Fibrosis ,Rats ,Cell biology ,Enzyme Activation ,Collagen Type III ,Phenotype ,medicine.anatomical_structure ,Endocrinology ,Hepatic stellate cell ,Rap1 ,Guanine nucleotide exchange factor ,Signal transduction ,Signal Transduction ,Transforming growth factor - Abstract
Scar formation occurs during the late stages of the inflammatory response but, when excessive, produces fibrosis that can lead to functional and structural damage of tissues. Here, we show that the profibrogenic agonist, transforming growth factor β1, transcriptionally decreases expression of Exchange protein activated by cAMP 1 (Epac1) in fibroblasts/fibroblast-like cells from multiple tissues (i.e., cardiac, lung, and skin fibroblasts and hepatic stellate cells). Overexpression of Epac1 inhibits transforming growth factor β1-induced collagen synthesis, indicating that a decrease of Epac1 expression appears to be necessary for the fibrogenic phenotype, an idea supported by evidence that Epac1 expression in cardiac fibroblasts is inhibited after myocardial infarction. Epac and protein kinase A, a second mediator of cAMP action, have opposite effects on migration but both inhibit synthesis of collagen and DNA by fibroblasts. Epac is preferentially activated by low concentrations of cAMP and stimulates migration via the small G protein Rap1 but inhibits collagen synthesis in a Rap1-independent manner. The regulation of Epac expression and activation thus appear to be critical for the integration of pro- and anti-fibrotic signals and for the regulation of fibroblast function.
- Published
- 2008
39. Caveolae as Organizers of Pharmacologically Relevant Signal Transduction Molecules
- Author
-
Hemal H. Patel, Fiona Murray, and Paul A. Insel
- Subjects
Lung Diseases ,medicine.medical_specialty ,Cell signaling ,Heart Diseases ,G protein ,Caveolae ,Toxicology ,Caveolins ,Article ,Receptor tyrosine kinase ,Mice ,Drug Delivery Systems ,Internal medicine ,Heterotrimeric G protein ,Caveolin ,medicine ,Animals ,Humans ,G protein-coupled receptor ,Pharmacology ,biology ,Cell biology ,Endocrinology ,Gene Expression Regulation ,biology.protein ,Signal transduction ,Signal Transduction - Abstract
Caveolae, a subset of membrane (lipid) rafts, are flask-like invaginations of the plasma membrane that contain caveolin proteins, which serve as organizing centers for cellular signal transduction. Caveolins (-1, -2, and -3) have cytoplasmic N and C termini, palmitolylation sites, and a scaffolding domain that facilitates interaction and organization of signaling molecules so as to help provide coordinated and efficient signal transduction. Such signaling components include upstream entities (e.g., G protein–coupled receptors (GPCRs), receptor tyrosine kinases, and steroid hormone receptors) and downstream components (e.g., heterotrimeric and low-molecular-weight G proteins, effector enzymes, and ion channels). Diseases associated with aberrant signaling may result in altered localization or expression of signaling proteins in caveolae. Caveolin-knockout mice have numerous abnormalities, some of which may reflect the impact of total body knockout throughout the life span. This review provides a general overview of caveolins and caveolae, signaling molecules that localize to caveolae, the role of caveolae/caveolin in cardiac and pulmonary pathophysiology, pharmacologic implications of caveolar localization of signaling molecules, and the possibility that caveolae might serve as a therapeutic target.
- Published
- 2008
40. Pathway and gene ontology based analysis of gene expression in a rat model of cerebral ischemic tolerance
- Author
-
Zheng Feng, Daniel P. Davis, John C. Drummond, Roman Sasik, Piyush M. Patel, and Hemal H. Patel
- Subjects
Male ,Candidate gene ,Bone Morphogenetic Protein 7 ,Blotting, Western ,Ischemia ,Gene Expression ,Biology ,Bioinformatics ,Hippocampus ,Brain Ischemia ,Transforming Growth Factor beta ,Databases, Genetic ,Gene expression ,medicine ,Animals ,Cluster Analysis ,Rats, Wistar ,KEGG ,Ischemic Preconditioning ,Receptor ,Molecular Biology ,Oligonucleotide Array Sequence Analysis ,Mitogen-Activated Protein Kinase Kinases ,Reverse Transcriptase Polymerase Chain Reaction ,Microarray analysis techniques ,General Neuroscience ,Toll-Like Receptors ,Computational Biology ,medicine.disease ,Rats ,Cell biology ,Cyclooxygenase 2 ,Fluorescent Antibody Technique, Direct ,Bone Morphogenetic Proteins ,Ischemic preconditioning ,Neurology (clinical) ,Signal transduction ,Ribosomes ,Signal Transduction ,Developmental Biology - Abstract
Ischemic tolerance is a phenomenon whereby a sublethal ischemic insult [ischemic preconditioning (IPC)] provides robust protection against subsequent lethal ischemia. Activation of N-methyl-D-aspartate (NMDA) receptors and subsequent new gene transcription are required for tolerance. We utilized the NMDA antagonist, MK801, prior to the IPC stimulus to separate candidate genes from epiphenomenona. Rats were divided into four groups: vehicle/IPC (preconditioned), MK801/IPC (attenuated preconditioning), vehicle/sham (non-preconditioned), and MK801/sham (non-preconditioned). Hippocampi (5/group/time point) were harvested immediately after ischemia as well as 1, 4, and 24 h post-ischemia to profile gene expression patterns using microarray analyses. Extracted mRNAs were pooled and subsequently hybridized to Affymetrix arrays. In addition, groups of rats were sacrificed for Western blot analysis and histological studies. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and gene ontology (GO) analyses were used to identify functionally related groups of genes whose modulation was statistically significant, while hierarchical cluster analysis was used to visualize the fold expression within these groups. Significantly modulated pathways included: MAP kinase signaling pathway, Toll receptor pathway, TGF-beta signaling pathways, and pathways associated with ribosome function and oxidative phosphorylation. Our data suggest that the tolerant brain responds to subsequent ischemic stress by partially downregulating inflammatory and upregulating protein synthesis and energy metabolism pathways.
- Published
- 2007
41. Caveolin‐1 expression is essential forN‐methyl‐<scp>D</scp>‐aspartate receptor‐mediated Src and extracellular signal‐regulated kinase 1/2 activation and protection of primary neurons from ischemic cell death
- Author
-
Brian P. Head, Hemal H. Patel, Piyush M. Patel, Rosalia Mora, John C. Drummond, Yasuo M. Tsutsumi, Yue Hu, Paul A. Insel, David M. Roth, and Trisha Mejia
- Subjects
MAPK/ERK pathway ,Caveolin 1 ,Receptors, N-Methyl-D-Aspartate ,Biochemistry ,Receptor tyrosine kinase ,Brain Ischemia ,MAP2K7 ,Mice ,Genetics ,Animals ,Humans ,ASK1 ,Phosphorylation ,Molecular Biology ,Mitogen-Activated Protein Kinase 1 ,Neurons ,Mitogen-Activated Protein Kinase 3 ,Tyrosine-protein kinase CSK ,Cell Death ,biology ,Chemistry ,Akt/PKB signaling pathway ,Membrane raft ,Molecular biology ,Rats ,Up-Regulation ,Cell biology ,Enzyme Activation ,src-Family Kinases ,nervous system ,biology.protein ,Signal Transduction ,Biotechnology ,Proto-oncogene tyrosine-protein kinase Src - Abstract
N-Methyl-D-aspartate (NMDA) receptor (NMDAR) activation and downstream signaling are important for neuronal function. Activation of prosurvival Src family kinases and extracellular signal-regulated kinase (ERK) 1/2 is initiated by NMDAR activation, but the cellular organization of these kinases in relation to NMDARs is not entirely clear. We hypothesized that caveolin-1 scaffolds and coordinates protein complexes involved in NMDAR signaling and that this organization is necessary for neuronal preconditioning, whereby NMDAR activation protects neurons from subsequent ischemic cell death. We found that sublethal ischemia (SLI) or preconditioning via NMDA treatment of primary cortical neurons from neonatal rats or mice increases expression of phosphorylated (P) caveolin-1, P-Src, and P-ERK1/2. The NMDAR antagonist, MK801, or the Src inhibitor, PP2, attenuated SLI-induced preconditioning. NMDAR2B distributed to buoyant fractions and heavy fractions, partially colocalized with caveolin-1 and the membrane raft marker, cholera toxin B. Cultures of primary neurons treated with caveolin-1 small interfering RNA or from caveolin-1(-/-) mice lacked the NMDA-mediated increase in P-Src and P-ERK, as well as SLI- and NMDA-induced preconditioning. Adenovirally mediated expression of caveolin-1 in neurons from caveolin-1(-/-) mice restored NMDA-mediated enhancement of P-Src and P-ERK1/2, redistributed NMDAR2B to buoyant fractions, and enhanced NMDAR2B localization to membrane rafts. We conclude that caveolin-1, perhaps via its ability to scaffold key signaling components, is essential for NMDAR localization to neuronal membrane rafts, NMDAR/Src tyrosine kinase family/ERK signaling, and protection of neurons from ischemic injury and cell death.
- Published
- 2007
42. Expression and activity of cAMP phosphodiesterase isoforms in pulmonary artery smooth muscle cells from patients with pulmonary hypertension: role for PDE1
- Author
-
Jason X.-J. Yuan, Shen Zhang, Ryan Y. S. Suda, Fiona Murray, Paul A. Insel, Hemal H. Patel, and Patricia A. Thistlethwaite
- Subjects
Adult ,Male ,Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Physiology ,Hypertension, Pulmonary ,Phosphodiesterase 3 ,Gene Expression ,In Vitro Techniques ,PDE1 ,Cyclic nucleotide ,chemistry.chemical_compound ,Physiology (medical) ,Internal medicine ,medicine.artery ,Humans ,Medicine ,RNA, Messenger ,Aged ,DNA Primers ,Base Sequence ,Phosphoric Diester Hydrolases ,business.industry ,Phosphodiesterase ,Cell Biology ,Middle Aged ,Cyclic Nucleotide Phosphodiesterases, Type 1 ,medicine.disease ,Pulmonary hypertension ,Cyclic Nucleotide Phosphodiesterases, Type 3 ,Isoenzymes ,medicine.anatomical_structure ,Endocrinology ,chemistry ,3',5'-Cyclic-AMP Phosphodiesterases ,Case-Control Studies ,Circulatory system ,Pulmonary artery ,Female ,business ,Blood vessel - Abstract
Pulmonary hypertension (PHT) is associated with increased vascular resistance due to sustained contraction and enhanced proliferation of pulmonary arterial smooth muscle cells (PASMC); the abnormal tone and remodeling in the pulmonary vasculature may relate, at least in part, to decreased cyclic nucleotide levels. Cyclic nucleotide phosphodiesterases (PDEs), of which 11 families have been identified, catalyze the hydrolysis of cAMP and cGMP. We tested the hypothesis that PASMC isolated from patients with PHT, either idiopathic pulmonary arterial hypertension (IPAH) or secondary pulmonary hypertension (SPH), have increased expression and activity of PDE isoforms that reduce the responsiveness of agents that raise cellular cAMP. Real-time PCR and immunoblotting demonstrated that the expression of PDE1A, PDE1C, PDE3B, and PDE5A was enhanced in PASMC from both IPAH and SPH patients compared with control PASMC. Consistent with this enhanced expression of PDEs, agonist-stimulated cAMP levels were significantly reduced in IPAH and SPH PASMC unless a PDE inhibitor was present. The use of specific PDE inhibitors revealed that an increase in PDE1 and PDE3 activity largely accounted for reduced agonist-induced cAMP levels and increased proliferation in IPAH and SPH PASMC. Treatment with PDE1C-targeted small interference RNA enhanced cAMP accumulation and inhibited cellular proliferation to a greater extent in PHT PASMC than controls. The results imply that an increase in PDE isoforms, in particular PDE1C, contributes to decreased cAMP and increased proliferation of PASMC in patients with PHT. PDE1 isoforms may provide novel targets for the treatment of both primary and secondary forms of the disease.
- Published
- 2007
43. Thy-1 interaction with Fas in lipid rafts regulates fibroblast apoptosis and lung injury resolution
- Author
-
Simon S. Wong, Jeeyeon Kim, J. Cameron Finley, John E. Bradley, Xiaoqiu Liu, Hemal H. Patel, Brian P. Head, Carmen A. Taype, Tzu-Pin Shentu, Emma J. Mah, Celia R. Espinoza, and James S. Hagood
- Subjects
0301 basic medicine ,Pulmonary Fibrosis ,Apoptosis ,Pharmacology ,0302 clinical medicine ,Fibrosis ,Staurosporine ,Myofibroblasts ,Lipid raft ,Thy-1 ,Mice, Knockout ,Microscopy, Confocal ,Chemistry ,Lung Injury ,Caspase 9 ,FasL ,3. Good health ,Cell biology ,medicine.anatomical_structure ,Proto-Oncogene Proteins c-bcl-2 ,caspases ,030220 oncology & carcinogenesis ,Myofibroblast ,medicine.drug ,Protein Binding ,Signal Transduction ,Fas Ligand Protein ,Immunoblotting ,bcl-X Protein ,Lung injury ,Article ,Pathology and Forensic Medicine ,Cell Line ,03 medical and health sciences ,Bleomycin ,Membrane Microdomains ,Downregulation and upregulation ,fibroblasts ,medicine ,Animals ,fas Receptor ,Fibroblast ,Molecular Biology ,fibrosis ,Cell Biology ,Fas ,medicine.disease ,Embryo, Mammalian ,Rats ,lipid raft ,Mice, Inbred C57BL ,030104 developmental biology ,Thy-1 Antigens - Abstract
Thy-1-negative lung fibroblasts are resistant to apoptosis. The mechanisms governing this process and its relevance to fibrotic remodeling remain poorly understood. By using either sorted or transfected lung fibroblasts, we found that Thy-1 expression is associated with downregulation of anti-apoptotic molecules Bcl-2 and Bcl-xL, as well as increased levels of cleaved caspase-9. Addition of rhFasL and staurosporine, well-known apoptosis inducers, caused significantly increased cleaved caspase-3, -8, and PARP in Thy-1-transfected cells. Furthermore, rhFasL induced Fas translocation into lipid rafts and its colocalization with Thy-1. These in vitro results indicate that Thy-1, in a manner dependent upon its glycophosphatidylinositol anchor and lipid raft localization, regulates apoptosis in lung fibroblasts via Fas-, Bcl-, and caspase-dependent pathways. In vivo, Thy-1 deficient (Thy1-/-) mice displayed persistence of myofibroblasts in the resolution phase of bleomycin-induced fibrosis, associated with accumulation of collagen and failure of lung fibrosis resolution. Apoptosis of myofibroblasts is decreased in Thy1-/- mice in the resolution phase. Collectively, these findings provide new evidence regarding the role and mechanisms of Thy-1 in initiating myofibroblast apoptosis that heralds the termination of the reparative response to bleomycin-induced lung injury. Understanding the mechanisms regulating fibroblast survival/apoptosis should lead to novel therapeutic interventions for lung fibrosis.
- Published
- 2015
44. Caveolin-3 Overexpression Attenuates Cardiac Hypertrophy via Inhibition of T-type Ca2+ Current Modulated by Protein Kinase Cα in Cardiomyocytes*
- Author
-
Courtney R. Reynolds, Ravi C. Balijepalli, Alexis M Keefe, Benjamin K. August, Timothy A. Hacker, David M. Roth, Marites T. Woon, Hemal H. Patel, Laura J. Phelan, and Yogananda S. Markandeya
- Subjects
Male ,Patch-Clamp Techniques ,Caveolin 3 ,Gene Expression ,cardiomyocyte ,angiotensin II ,Inbred C57BL ,Cardiovascular ,Medical and Health Sciences ,Biochemistry ,Muscle hypertrophy ,Membrane Potentials ,Mice ,Calcium Channels, T-Type ,2.1 Biological and endogenous factors ,Myocyte ,Myocytes, Cardiac ,Aetiology ,Cells, Cultured ,Microscopy ,Cultured ,Blotting ,Reverse Transcriptase Polymerase Chain Reaction ,cardiac hypertrophy ,Angiotensin II ,NFAT ,Molecular Bases of Disease ,Biological Sciences ,T-Type ,Heart Disease ,medicine.anatomical_structure ,Hypertension ,cardiovascular system ,RNA Interference ,Cardiac ,Western ,Biochemistry & Molecular Biology ,medicine.medical_specialty ,Protein Kinase C-alpha ,Cells ,T cell ,Blotting, Western ,Cardiomegaly ,Biology ,Caveolae ,Electron ,Microscopy, Electron, Transmission ,Internal medicine ,medicine ,Transmission ,Animals ,Protein kinase A ,Molecular Biology ,Protein kinase C ,Myocytes ,Cell Biology ,Newborn ,Calcineurin ,Mice, Inbred C57BL ,Endocrinology ,Animals, Newborn ,Chemical Sciences ,calcium channel ,caveolin ,Calcium Channels ,protein kinase C - Abstract
Pathological cardiac hypertrophy is characterized by subcellular remodeling of the ventricular myocyte with a reduction in the scaffolding protein caveolin-3 (Cav-3), altered Ca(2+) cycling, increased protein kinase C expression, and hyperactivation of calcineurin/nuclear factor of activated T cell (NFAT) signaling. However, the precise role of Cav-3 in the regulation of local Ca(2+) signaling in pathological cardiac hypertrophy is unclear. We used cardiac-specific Cav-3-overexpressing mice and in vivo and in vitro cardiac hypertrophy models to determine the essential requirement for Cav-3 expression in protection against pharmacologically and pressure overload-induced cardiac hypertrophy. Transverse aortic constriction and angiotensin-II (Ang-II) infusion in wild type (WT) mice resulted in cardiac hypertrophy characterized by significant reduction in fractional shortening, ejection fraction, and a reduced expression of Cav-3. In addition, association of PKCα and angiotensin-II receptor, type 1, with Cav-3 was disrupted in the hypertrophic ventricular myocytes. Whole cell patch clamp analysis demonstrated increased expression of T-type Ca(2+) current (ICa, T) in hypertrophic ventricular myocytes. In contrast, the Cav-3-overexpressing mice demonstrated protection from transverse aortic constriction or Ang-II-induced pathological hypertrophy with inhibition of ICa, T and intact Cav-3-associated macromolecular signaling complexes. siRNA-mediated knockdown of Cav-3 in the neonatal cardiomyocytes resulted in enhanced Ang-II stimulation of ICa, T mediated by PKCα, which caused nuclear translocation of NFAT. Overexpression of Cav-3 in neonatal myocytes prevented a PKCα-mediated increase in ICa, T and nuclear translocation of NFAT. In conclusion, we show that stable Cav-3 expression is essential for protecting the signaling mechanisms in pharmacologically and pressure overload-induced cardiac hypertrophy.
- Published
- 2015
45. Ischaemic preconditioning preferentially increases protein S-nitrosylation in subsarcolemmal mitochondria
- Author
-
Mark J. Kohr, Sara Menazza, David M. Roth, Charles Steenbergen, Hemal H. Patel, Angel Aponte, Elizabeth Murphy, Tiffany Nguyen, and Junhui Sun
- Subjects
Male ,Nitric Oxide Synthase Type III ,Physiology ,Caveolin 3 ,Subsarcolemmal and interfibrillar mitochondria ,Ischaemic preconditioning ,Biology ,Cardiorespiratory Medicine and Haematology ,Inbred C57BL ,Caveolae ,Cardiovascular ,Mitochondria, Heart ,Nitric oxide ,chemistry.chemical_compound ,Mice ,Sarcolemma ,Enos ,Physiology (medical) ,parasitic diseases ,Caveolin ,Animals ,Myocardial ,2.1 Biological and endogenous factors ,cardiovascular diseases ,Protein S-nitrosylation ,Aetiology ,Ischemic Preconditioning ,Protein Processing ,Cardioprotection ,Myocardium ,Post-Translational ,Heart ,biology.organism_classification ,Cell biology ,Mitochondria ,Mice, Inbred C57BL ,Nitric oxide synthase ,chemistry ,Biochemistry ,Cardiovascular System & Hematology ,Ischemic Preconditioning, Myocardial ,biology.protein ,Ischemic preconditioning ,Cardiology and Cardiovascular Medicine ,Protein Processing, Post-Translational - Abstract
Nitric oxide (NO) and protein S-nitrosylation (SNO) have been shown to play important roles in ischaemic preconditioning (IPC)-induced acute cardioprotection. The majority of proteins that show increased SNO following IPC are localized to the mitochondria, and our recent studies suggest that caveolae transduce acute NO/SNO cardioprotective signalling in IPC hearts. Due to the close association between subsarcolemmal mitochondria (SSM) and the sarcolemma/caveolae, we tested the hypothesis that SSM, rather than the interfibrillar mitochondria (IFM), are major targets for NO/SNO signalling derived from caveolae-associated eNOS. Following either control perfusion or IPC, SSM and IFM were isolated from Langendorff perfused mouse hearts, and SNO was analysed using a modified biotin switch method with fluorescent maleimide fluors. In perfusion control hearts, the SNO content was higher in SSM compared with IFM (1.33 ± 0.19, ratio of SNO content Perf-SSM vs. Perf-IFM), and following IPC SNO content significantly increased preferentially in SSM, but not in IFM (1.72 ± 0.17 and 1.07 ± 0.04, ratio of SNO content IPC-SSM vs. Perf-IFM, and IPC-IFM vs. Perf-IFM, respectively). Consistent with these findings, eNOS, caveolin-3, and connexin-43 were detected in SSM, but not in IFM, and IPC resulted in a further significant increase in eNOS/caveolin-3 levels in SSM. Interestingly, we did not observe an IPC-induced increase in SNO or eNOS/caveolin-3 in SSM isolated from caveolin-3(-/-) mouse hearts, which could not be protected with IPC. In conclusion, these results suggest that SSM may be the preferential target of sarcolemmal signalling-derived post-translational protein modification (caveolae-derived eNOS/NO/SNO), thus providing an important role in IPC-induced cardioprotection.
- Published
- 2015
46. Cardiac Myocyte‐Specific Overexpression of Caveolin‐3 Enhances Physiological Response to β‐Adrenergic Receptors
- Author
-
Jan M. Schilling, David M. Roth, Anna R. Busija, Paul A. Insel, and Hemal H. Patel
- Subjects
Caveolin 3 ,Chemistry ,Cardiac myocyte ,Genetics ,β adrenergic receptor ,Molecular Biology ,Biochemistry ,Biotechnology ,Cell biology - Published
- 2015
47. Helium Postconditioning Regulates Caveolin‐1/‐3 Translocation and Gene Expression
- Author
-
Moritz Flick, Benedikt Preckel, Nina C. Weber, Markus W. Hollmann, Hemal H. Patel, Martin Albrecht, and Gezina T. M. L. Oei
- Subjects
Chemistry ,Gene expression ,Caveolin 1 ,Genetics ,Chromosomal translocation ,Molecular Biology ,Biochemistry ,Biotechnology ,Cell biology - Published
- 2015
48. Differential Effects of Hydrophilic versus Lipophilic Statins on RhoA Kinase Inhibition and Membrane Stability in Cardiac Myocytes
- Author
-
Alice E. Zemljic-Harpf, Elizabeth Asfaw, Joseph C. Godoy, Jan M. Schilling, Erika A. Alvarez, Anna Schwarz, Hemal H. Patel, and Nancy D. Dalton
- Subjects
RHOA ,biology ,Chemistry ,nutritional and metabolic diseases ,Kinase inhibition ,Biochemistry ,Differential effects ,Cell biology ,Membrane ,Genetics ,biology.protein ,Protein prenylation ,Myocyte ,lipids (amino acids, peptides, and proteins) ,cardiovascular diseases ,Kinase activity ,Prescribed drugs ,Molecular Biology ,Biotechnology - Abstract
Statins are among the most prescribed drugs worldwide. By inhibiting mevalonate synthesis statins inhibit protein prenylation and thereby may reduce RhoA kinase activity. Cardiac overexpression of ...
- Published
- 2015
49. Microtubules and Actin Microfilaments Regulate Lipid Raft/Caveolae Localization of Adenylyl Cyclase Signaling Components
- Author
-
Hemal H. Patel, Ingrid R. Niesman, David M. Roth, Marilyn G. Farquhar, Fiona Murray, Paul A. Insel, James S. Swaney, and Brian P. Head
- Subjects
Male ,Cytochalasin D ,Gs alpha subunit ,Filamins ,Linker for Activation of T cells ,macromolecular substances ,Biology ,Caveolae ,Cell Fractionation ,Filamin ,Caveolins ,Microtubules ,Models, Biological ,Biochemistry ,Receptors, G-Protein-Coupled ,Rats, Sprague-Dawley ,Adenylyl cyclase ,Mice ,chemistry.chemical_compound ,Contractile Proteins ,Membrane Microdomains ,Sarcolemma ,Receptors, Adrenergic, beta ,Cyclic AMP ,Animals ,Protein Isoforms ,Myocytes, Cardiac ,Molecular Biology ,Lipid raft ,Cells, Cultured ,Cytoskeleton ,Actin ,Nucleic Acid Synthesis Inhibitors ,Mice, Knockout ,Microfilament Proteins ,Isoproterenol ,Cell Biology ,Adrenergic beta-Agonists ,Actins ,Tubulin Modulators ,Rats ,Cell biology ,Actin Cytoskeleton ,chemistry ,Colchicine ,Adenylyl Cyclases ,Signal Transduction - Abstract
Microtubules and actin filaments regulate plasma membrane topography, but their role in compartmentation of caveolae-resident signaling components, in particular G protein-coupled receptors (GPCR) and their stimulation of cAMP production, has not been defined. We hypothesized that the microtubular and actin cytoskeletons influence the expression and function of lipid rafts/caveolae, thereby regulating the distribution of GPCR signaling components that promote cAMP formation. Depolymerization of microtubules with colchicine (Colch) or actin microfilaments with cytochalasin D (CD) dramatically reduced the amount of caveolin-3 in buoyant (sucrose density) fractions of adult rat cardiac myocytes. Colch or CD treatment led to the exclusion of caveolin-1, caveolin-2, beta1-adrenergic receptors (beta1-AR), beta2-AR, Galpha(s), and adenylyl cyclase (AC)5/6 from buoyant fractions, decreasing AC5/6 and tyrosine-phosphorylated caveolin-1 in caveolin-1 immunoprecipitates but in parallel increased isoproterenol (beta-AR agonist)-stimulated cAMP production. Incubation with Colch decreased co-localization (by immunofluorescence microscopy) of caveolin-3 and alpha-tubulin; both Colch and CD decreased co-localization of caveolin-3 and filamin (an F-actin cross-linking protein), decreased phosphorylation of caveolin-1, Src, and p38 MAPK, and reduced the number of caveolae/mum of sarcolemma (determined by electron microscopy). Treatment of S49 T-lymphoma cells (which possess lipid rafts but lack caveolae) with CD or Colch redistributed a lipid raft marker (linker for activation of T cells (LAT)) and Galpha(s) from lipid raft domains. We conclude that microtubules and actin filaments restrict cAMP formation by regulating the localization and interaction of GPCR-G(s)-AC in lipid rafts/caveolae.
- Published
- 2006
50. Protection of adult rat cardiac myocytes from ischemic cell death: role of caveolar microdomains and δ-opioid receptors
- Author
-
Heidi N. Petersen, Ingrid R. Niesman, Garrett J. Gross, Brian P. Head, Paul A. Insel, Diane Huang, Hemal H. Patel, and David M. Roth
- Subjects
Programmed cell death ,Cell signaling ,Physiology ,G protein ,Myocardial Ischemia ,Ischemia ,Biology ,Caveolae ,Rats, Sprague-Dawley ,Receptors, Opioid, delta ,Physiology (medical) ,medicine ,Animals ,Myocyte ,Myocytes, Cardiac ,Receptor ,Cells, Cultured ,medicine.disease ,Rats ,Cell biology ,Opioid ,Immunology ,cardiovascular system ,Cardiology and Cardiovascular Medicine ,medicine.drug - Abstract
The role of caveolae, membrane microenvironments enriched in signaling molecules, in myocardial ischemia is poorly defined. In the current study, we used cardiac myocytes prepared from adult rats to test the hypothesis that opioid receptors (OR), which are capable of producing cardiac protection in vivo, promote cardiac protection in cardiac myocytes in a caveolae-dependent manner. We determined protein expression and localization of delta-OR (DOR) using coimmunohistochemistry, caveolar fractionation, and immunoprecipitations. DOR colocalized in fractions with caveolin-3 (Cav-3), a structural component of caveolae in muscle cells, and could be immunoprecipitated by a Cav-3 antibody. Immunohistochemistry confirmed plasma membrane colocalization of DOR with Cav-3. Cardiac myocytes were subjected to simulated ischemia (2 h) or an ischemic preconditioning (IPC) protocol (10 min ischemia, 30 min recovery, 2 h ischemia) in the presence and absence of methyl-beta-cyclodextrin (MbetaCD, 2 mM), which binds cholesterol and disrupts caveolae. We also assessed the cardiac protective effects of SNC-121 (SNC), a selective DOR agonist, on cardiac myocytes with or without MbetaCD and MbetaCD preloaded with cholesterol. Ischemia, simulated by mineral oil layering to inhibit gas exchange, promoted cardiac myocyte cell death (trypan blue staining), a response blunted by SNC (37 +/- 3 vs. 59 +/- 3% dead cells in the presence and absence of 1 muM SNC, respectively, P0.01) or by use of the IPC protocol (35 +/- 4 vs. 62 +/- 3% dead cells, P0.01). MbetaCD treatment, which disrupted caveolae (as detected by electron microscopy), fully attenuated the protective effects of IPC or SNC, resulting in cell death comparable to that of the ischemic group. By contrast, SNC-induced protection was not abrogated in cells incubated with cholesterol-saturated MbetaCD, which maintained caveolae structure and function. These findings suggest a key role for caveolae, perhaps through enrichment of signaling molecules, in contributing to protection of cardiac myocytes from ischemic damage.
- Published
- 2006
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.