Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Antioxidants (Basel) ; 12(6)2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37372017

ABSTRACT

OBJECTIVES: Homozygous familial hypercholesteremia (HoFH) is a rare, life-threatening metabolic disease, mainly caused by a mutation in the LDL receptor. If untreated, HoFH causes premature death from acute coronary syndrome. Lomitapide is approved by the FDA as a therapy to lower lipid levels in adult patients with HoFH. Nevertheless, the beneficial effect of lomitapide in HoFH models remains to be defined. In this study, we investigated the effect of lomitapide on cardiovascular function using LDL receptor-knockout mice (LDLr-/-). METHODS: Six-week-old LDLr-/- mice were fed a standard diet (SD) or a high-fat diet (HFD) for 12 weeks. Lomitapide (1 mg/Kg/Day) was given by oral gavage for the last 2 weeks in the HFD group. Body weight and composition, lipid profile, blood glucose, and atherosclerotic plaques were measured. Vascular reactivity and markers for endothelial function were determined in conductance arteries (thoracic aorta) and resistance arteries (mesenteric resistance arteries (MRA)). Cytokine levels were measured by using the Mesoscale discovery V-Plex assays. RESULTS: Body weight (47.5 ± 1.5 vs. 40.3 ± 1.8 g), % of fat mass (41.6 ± 1.9% vs. 31.8 ± 1.7%), blood glucose (215.5 ± 21.9 vs. 142.3 ± 7.7 mg/dL), and lipid levels (cholesterol: 600.9 ± 23.6 vs. 451.7 ± 33.4 mg/dL; LDL/VLDL: 250.6 ± 28.9 vs. 161.1 ± 12.24 mg/dL; TG: 299.5 ± 24.1 vs. 194.1 ± 28.1 mg/dL) were significantly decreased, and the % of lean mass (56.5 ± 1.8% vs. 65.2 ± 2.1%) was significantly increased in the HFD group after lomitapide treatment. The atherosclerotic plaque area also decreased in the thoracic aorta (7.9 ± 0.5% vs. 5.7 ± 0.1%). After treatment with lomitapide, the endothelium function of the thoracic aorta (47.7 ± 6.3% vs. 80.7 ± 3.1%) and mesenteric resistance artery (66.4 ± 4.3% vs. 79.5 ± 4.6%) was improved in the group of LDLr-/- mice on HFD. This was correlated with diminished vascular endoplasmic (ER) reticulum stress, oxidative stress, and inflammation. CONCLUSIONS: Treatment with lomitapide improves cardiovascular function and lipid profile and reduces body weight and inflammatory markers in LDLr-/- mice on HFD.

2.
Antioxidants (Basel) ; 12(1)2023 Jan 10.
Article in English | MEDLINE | ID: mdl-36671022

ABSTRACT

OBJECTIVES: Short-chain fatty acids (SCFAs), the main metabolites released from the gut microbiota, are altered during hypertension and obesity. SCFAs play a beneficial role in the cardiovascular system. However, the effect of SCFAs on cerebrovascular endothelial cells is yet to be uncovered. In this study, we use brain endothelial cells to investigate the in vitro effect of SCFAs on heme oxygenase 2 (HO-2) and mitochondrial function after angiotensin II (Ang-II) treatment. METHODS: Brain human microvascular endothelial cells were treated with Ang-II (500 nM for 24 h) in the presence and absence of an SCFAs cocktail (1 µM; acetate, propionate, and butyrate) and/or HO-2 inhibitor (SnPP 5 µM). At the end of the treatment, HO-2, endothelial markers (p-eNOS and NO production), inflammatory markers (TNFα, NFκB-p50, and -p65), calcium homeostasis, mitochondrial membrane potential, mitochondrial ROS and H2O2, and mitochondrial respiration were determined in all groups of treated cells. KEY RESULTS: Our data showed that SCFAs rescued HO-2 after Ang-II treatment. Additionally, SCFAs rescued Ang-II-induced eNOS reduction and mitochondrial membrane potential impairment and mitochondrial respiration damage. On the other hand, SCFAs reduced Ang-II-induced inflammation, calcium dysregulation, mitochondrial ROS, and H2O2. All of the beneficial effects of SCFAs on endothelial cells and mitochondrial function occurred through HO-2. CONCLUSIONS: SCFAs treatment restored endothelial cells and mitochondrial function following Ang-II-induced oxidative stress. SCFAs exert these beneficial effects by acting on HO-2. Our results are opening the door for more studies to investigate the effect the of SCFAs/HO-2 axis on hypertension and obesity-induced cerebrovascular diseases.

4.
Obesity (Silver Spring) ; 30(4): 893-901, 2022 04.
Article in English | MEDLINE | ID: mdl-35253407

ABSTRACT

OBJECTIVE: In this study, the effect of lomitapide, a microsomal triglyceride transfer protein inhibitor, on the cardiovascular function in obesity was investigated. METHODS: Eight-week-old C57BL/6 mice were fed with high-fat diet for 12 weeks in the presence and absence of lomitapide. Lomitapide was administered by gavage (1 mg/kg/d) during the last 2 weeks of high-fat feeding. Body weight, blood glucose, body composition, and lipid profile were determined. Vascular function and endothelial function markers were studied in the aorta and mesenteric resistance arteries. RESULTS: Lomitapide treatment reduced body weight in mice with obesity. Blood glucose, percentage of fat mass, total cholesterol, and low-density lipoprotein levels were significantly reduced, and the percentage of lean mass was significantly increased after lomitapide treatment. The vascular response to sodium nitroprusside in the aorta and mesenteric arteries was similar among groups. However, the vascular response to acetylcholine was improved in the treated group. This was associated with decreased levels of vascular endoplasmic reticulum stress, inflammation, and oxidative stress. CONCLUSIONS: Treatment with lomitapide attenuated the increase in body weight in mice with obesity and restored the lipid profile and vascular function. These effects were accompanied by a decrease in inflammation and oxidative stress.


Subject(s)
Anticholesteremic Agents , Hyperlipoproteinemia Type II , Animals , Anticholesteremic Agents/pharmacology , Benzimidazoles , Blood Glucose , Carrier Proteins , Diet, High-Fat , Hyperlipoproteinemia Type II/metabolism , Hyperlipoproteinemia Type II/therapy , Inflammation , Lipids , Mice , Mice, Inbred C57BL , Obesity/drug therapy
5.
Cureus ; 14(2): e21913, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35155042

ABSTRACT

The prevalence of obesity is increasing worldwide, and novel therapeutic strategies such as enhancement of thermogenic pathways in white adipose tissue (WAT) are gaining more attention. The gut/brain axis plays an essential role in promoting the browning of WAT. However, the mechanism by which this axis regulates WAT function is not fully understood. On the other hand, the role of microRNAs (miRNAs) in the control of WAT browning has already been established. Therefore, understanding the communication pathways linking the gut/brain axis and miRNAs might establish a promising intervention for obesity. Our published data showed that microRNA-204 (miR-204), a microRNA that plays an important role in the control of the central nervous system (CNS) and the pathogenesis of obesity, is affected by gut dysbiosis. Therefore, miR-204 could be a key element that controls the browning of WAT by acting as a potential link between the gut microbiota and the brain. In this review, we summarized the current knowledge about communication pathways between the brain, gut, and miR-204 and examined the literature to discuss potential research directions that might lead to a better understanding of the mechanisms underlying the browning of WAT in obesity.

6.
Cureus ; 13(10): e18783, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34692262

ABSTRACT

There is abundant evidence demonstrating the association between gut dysbiosis and neurogenic diseases such as hypertension. A common characteristic of resistant hypertension is the chronic elevation in sympathetic nervous system (SNS) activity accompanied by increased release of norepinephrine (NE), indicating a neurogenic component that contributes to the development of hypertension. Factors that modulate the sympathetic tone to the cardiovascular system in hypertensive patients are still poorly understood. Research has identified an interaction between the brain and the gut, and this interaction plays a possible role in the mechanism of heart damage-induced hypertension. Data, however, remain scarce, and further study is required to define the role of microbiota in sympathetic neural function and its relationship with heart damage and blood pressure (BP) control. Experimental evidence has pointed toward a bidirectional relationship between alterations in the types of bacteria present in the gut and neurogenic diseases, such as hypertension. Our published data showed that miR-204, a microRNA that plays an important role in the CNS function, is affected by gut dysbiosis. Therefore, miR-204 could be a key element that regulates normal sinus rhythm and neuronal hypertension. In this review, we will shed light on the potential mechanism by which microbiota affects hypothalamic miR-204, which in turn, could hinder the sympathetic nerve drive to the cardiovascular system leading to arrhythmia and hypertension.

7.
Cureus ; 13(9): e18194, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34589374

ABSTRACT

Anticancer drugs play an important role in reducing mortality rates and increasing life expectancy in cancer patients. Treatments include monotherapy and/or a combination of radiation therapy, chemotherapy, hormone therapy, or immunotherapy. Despite great advances in drug development, some of these treatments have been shown to induce cardiotoxicity directly affecting heart function and structure, as well as accelerating the development of cardiovascular disease. Such side effects restrict treatment options and can negatively affect disease management. Consequently, when managing cancer patients, it is vital to understand the mechanisms causing cardiotoxicity to better monitor heart function, develop preventative measures against cardiotoxicity, and treat heart failure when it occurs in this patient population. This review discusses the role and mechanism of major chemotherapy agents with principal cardiovascular complications in cancer patients.

8.
Cardiovasc Diabetol ; 19(1): 136, 2020 09 09.
Article in English | MEDLINE | ID: mdl-32907629

ABSTRACT

The endothelium plays a pivotal role in maintaining vascular health. Obesity is a global epidemic that has seen dramatic increases in both adult and pediatric populations. Obesity perturbs the integrity of normal endothelium, leading to endothelial dysfunction which predisposes the patient to cardiovascular diseases. MicroRNAs (miRNAs) are short, single-stranded, non-coding RNA molecules that play important roles in a variety of cellular processes such as differentiation, proliferation, apoptosis, and stress response; their alteration contributes to the development of many pathologies including obesity. Mediators of obesity-induced endothelial dysfunction include altered endothelial nitric oxide synthase (eNOS), Sirtuin 1 (SIRT1), oxidative stress, autophagy machinery and endoplasmic reticulum (ER) stress. All of these factors have been shown to be either directly or indirectly caused by gene regulatory mechanisms of miRNAs. In this review, we aim to provide a comprehensive description of the therapeutic potential of miRNAs to treat obesity-induced endothelial dysfunction. This may lead to the identification of new targets for interventions that may prevent or delay the development of obesity-related cardiovascular disease.


Subject(s)
Endothelium/physiopathology , MicroRNAs/genetics , Obesity/physiopathology , Antagomirs , Autophagy/genetics , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/therapeutic use , Molecular Mimicry , Molecular Targeted Therapy , Nitric Oxide Synthase Type III/genetics , Obesity/genetics , Oxidative Stress/genetics , RNAi Therapeutics , Sirtuin 1/genetics
9.
Obesity (Silver Spring) ; 27(7): 1050-1058, 2019 07.
Article in English | MEDLINE | ID: mdl-30938942

ABSTRACT

Over the past three decades, the increasing rates of obesity have led to an alarming obesity epidemic worldwide. Obesity is associated with an increased risk of cardiovascular diseases; thus, it is essential to define the molecular mechanisms by which obesity affects heart function. Individuals with obesity and overweight have shown changes in cardiac structure and function, leading to cardiomyopathy, hypertrophy, atrial fibrillation, and arrhythmia. Autophagy is a highly conserved recycling mechanism that delivers proteins and damaged organelles to lysosomes for degradation. In the hearts of patients and mouse models with obesity, this process is impaired. Furthermore, it has been shown that autophagy flux restoration in obesity models improves cardiac function. Therefore, autophagy may play an important role in mitigating the adverse effects of obesity on the heart. Throughout this review, we will discuss the benefits of autophagy on the heart in obesity and how regulating autophagy might be a therapeutic tool to reduce the risk of obesity-associated cardiovascular diseases.


Subject(s)
Autophagy , Cardiovascular Diseases/physiopathology , Heart Diseases/physiopathology , Obesity/complications , Humans , Obesity/pathology
10.
Hypertens Res ; 42(7): 960-969, 2019 07.
Article in English | MEDLINE | ID: mdl-30664704

ABSTRACT

Metformin is an antidiabetic drug. However, the pleiotropic beneficial effects of metformin in nondiabetic models still need to be defined. The objective of this study is to investigate the effect of metformin on angiotensin II (Ang II)-induced hypertension and cardiovascular diseases. Mice were infused with Ang II (400 ng/kg per min) with or without metformin for 2 weeks. Mice infused with angiotensin II displayed an increase in blood pressure associated with enhanced vascular endoplasmic reticulum (ER) stress markers, which were blunted after metformin treatment. Moreover, hypertension-induced reduction in phosphorylated AMPK, endothelial nitric oxide synthase (eNOs) phosphorylation, and endothelium-dependent relaxation (EDR) in mesenteric resistance arteries (MRA) were rescued after metformin treatment. Infusion of ER stress inducer (tunicamycin, Tun) in control mice induced ER stress in MRA and reduced phosphorylation of AMPK, eNOS synthase phosphorylation, and EDR in MRA without affecting systolic blood pressure (SBP). All these factors were reversed subsequently with metformin treatment. ER stress inhibition by metformin improves vascular function in hypertension. Therefore, metformin could be a potential therapy for cardiovascular diseases in hypertension independent of its effects on diabetes.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Endothelium, Vascular/drug effects , Hypertension/metabolism , Metformin/therapeutic use , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , Angiotensin II , Animals , Endothelium, Vascular/metabolism , Hypertension/chemically induced , Male , Mesenteric Arteries/metabolism , Metformin/pharmacology , Mice , Nitric Oxide Synthase Type III/metabolism , Phosphorylation/drug effects
11.
FASEB J ; 33(1): 1209-1225, 2019 01.
Article in English | MEDLINE | ID: mdl-30169110

ABSTRACT

Statins, which reduce LDL-cholesterol by inhibition of 3-hydroxy-3-methylglutaryl-coenzyme A reductase, are among the most widely prescribed drugs. Skeletal myopathy is a known statin-induced adverse effect associated with mitochondrial changes. We hypothesized that similar effects would occur in cardiac myocytes in a lipophilicity-dependent manner between 2 common statins: atorvastatin (lipophilic) and pravastatin (hydrophilic). Neonatal cardiac ventricular myocytes were treated with atorvastatin and pravastatin for 48 h. Both statins induced endoplasmic reticular (ER) stress, but only atorvastatin inhibited ERK1/2T202/Y204, AktSer473, and mammalian target of rapamycin signaling; reduced protein abundance of caveolin-1, dystrophin, epidermal growth factor receptor, and insulin receptor-ß; decreased Ras homolog gene family member A activation; and induced apoptosis. In cardiomyocyte-equivalent HL-1 cells, atorvastatin, but not pravastatin, reduced mitochondrial oxygen consumption. When male mice underwent atorvastatin and pravastatin administration per os for up to 7 mo, only long-term atorvastatin, but not pravastatin, induced elevated serum creatine kinase; swollen, misaligned, size-variable, and disconnected cardiac mitochondria; alteration of ER structure; repression of mitochondria- and endoplasmic reticulum-related genes; and a 21% increase in mortality in cardiac-specific vinculin-knockout mice during the first 2 months of administration. To our knowledge, we are the first to demonstrate in vivo that long-term atorvastatin administration alters cardiac ultrastructure, a finding with important clinical implications.-Godoy, J. C., Niesman, I. R., Busija, A. R., Kassan, A., Schilling, J. M., Schwarz, A., Alvarez, E. A., Dalton, N. D., Drummond, J. C., Roth, D. M., Kararigas, G., Patel, H. H., Zemljic-Harpf, A. E. Atorvastatin, but not pravastatin, inhibits cardiac Akt/mTOR signaling and disturbs mitochondrial ultrastructure in cardiac myocytes.


Subject(s)
Atorvastatin/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mitochondria, Heart/drug effects , Myocytes, Cardiac/drug effects , Pravastatin/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line , Cell Survival , Cholesterol, LDL/blood , Creatine Kinase/blood , Male , Mice , Mitochondria, Heart/metabolism , Mitochondria, Heart/ultrastructure , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/metabolism , Transcriptome , Vinculin/genetics , rhoA GTP-Binding Protein/metabolism
12.
J Sex Med ; 14(10): 1177-1186, 2017 10.
Article in English | MEDLINE | ID: mdl-28923309

ABSTRACT

BACKGROUND: The pathophysiology of increased severity of erectile dysfunction in men with diabetes and their poor response to oral pharmacotherapy are unclear. Defective vascular endothelium and consequent impairment in the formation and action of nitric oxide (NO) are implicated as potential mechanisms. Endothelial NO synthase, critical for NO generation, is localized to caveolae, plasma membrane lipid rafts enriched in structural proteins, and caveolins. Type 2 diabetes mellitus (T2DM)-induced changes in caveolin expression are recognized to play a role in cardiovascular dysfunction. AIMS: To evaluate DM-related changes to male erectile tissue in a mouse model that closely resembles human T2DM and study the specific role of caveolins in penile blood flow and microvascular perfusion using mice lacking caveolin (Cav)-1 or Cav-3. METHODS: We used wild-type C57BL6 (control) and Cav-1 and Cav-3 knockout (KO) male mice. T2DM was induced by streptozotocin followed by a high-fat diet for 4 months. Penile expressions of Cav-1, Cav-3, and endothelial NO synthase were determined by western blot, and phosphodiesterase type 5 activity was measured using [3H] cyclic guanosine monophosphate as a substrate. For hemodynamic studies, Cav-1 and Cav-3 KO mice were anesthetized, and penile blood flow (peak systolic velocity and end-diastolic velocity; millimeters per second) was determined using a high-frequency and high-resolution digital imaging color Doppler system. Penile tissue microcirculatory blood perfusion (arbitrary perfusion units) was measured using a novel PeriCam PSI system. OUTCOMES: Penile erectile tissues were harvested for histologic studies to assess Cav-1, Cav-3, and endothelial NO synthase expression, phosphodiesterase type 5 activity, and blood flow, and perfusion measurements were assessed for hemodynamic studies before and after an intracavernosal injection of prostaglandin E1 (50 ng). RESULTS: In T2DM mice, decreased Cav-1 and Cav-3 penile protein expression and increased phosphodiesterase type 5 activity were observed. Decreased response to prostaglandin E1 in peak systolic velocity (33 ± 4 mm/s in Cav-1 KO mice vs 62 ± 5 mm/s in control mice) and perfusion (146 ± 12 AU in Cav-1 KO mice vs 256 ± 12 AU in control mice) was observed. Hemodynamic changes in Cav-3 KO mice were insignificant. CLINICAL TRANSLATION: Our findings provide novel mechanistic insights into erectile dysfunction severity and poor pharmacotherapy that could have potential application to patients with T2DM. STRENGTHS AND LIMITATIONS: Use of KO mice and novel hemodynamic techniques are the strengths. A limitation is the lack of direct evaluation of penile hemodynamics in T2DM mice. CONCLUSION: Altered penile Cav-1 expression in T2DM mice and impaired penile hemodynamics in Cav-1 KO mice suggests a regulatory role for Cav-1 in DM-related erectile dysfunction. Parikh J, Zemljic-Harpf A, Fu J, et al. Altered Penile Caveolin Expression in Diabetes: Potential Role in Erectile Dysfunction. J Sex Med 2017;14:1177-1186.


Subject(s)
Caveolin 1/metabolism , Diabetes Mellitus, Type 2/complications , Erectile Dysfunction/metabolism , Nitric Oxide Synthase Type III/metabolism , Animals , Cyclic GMP/metabolism , Diabetes Mellitus, Type 2/metabolism , Endothelium, Vascular/metabolism , Male , Mice , Mice, Knockout , Microcirculation , Penile Erection/physiology , Penis/blood supply
14.
Sci Rep ; 7: 42265, 2017 02 09.
Article in English | MEDLINE | ID: mdl-28181559

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.


Subject(s)
Caveolin 1/metabolism , Endoplasmic Reticulum Stress , Endothelial Cells/metabolism , MicroRNAs/metabolism , Sirtuin 1/metabolism , Animals , Down-Regulation , Male , Mice, Inbred C57BL , MicroRNAs/genetics , Models, Biological , Vasodilation
15.
J Neurophysiol ; 117(1): 436-444, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27832597

ABSTRACT

Schizophrenia is a debilitating psychiatric disorder manifested in early adulthood. Disrupted-in-schizophrenia-1 (DISC1) is a susceptible gene for schizophrenia (Hodgkinson et al. 2004; Millar et al. 2000; St Clair et al. 1990) implicated in neuronal development, brain maturation, and neuroplasticity (Brandon and Sawa 2011; Chubb et al. 2008). Therefore, DISC1 is a promising candidate gene for schizophrenia, but the molecular mechanisms underlying its role in the pathogenesis of the disease are still poorly understood. Interestingly, caveolin-1 (Cav-1), a cholesterol binding and scaffolding protein, regulates neuronal signal transduction and promotes neuroplasticity. In this study we examined the role of Cav-1 in mediating DISC1 expression in neurons in vitro and the hippocampus in vivo. Overexpressing Cav-1 specifically in neurons using a neuron-specific synapsin promoter (SynCav1) increased expression of DISC1 and proteins involved in synaptic plasticity (PSD95, synaptobrevin, synaptophysin, neurexin, and syntaxin 1). Similarly, SynCav1-transfected differentiated human neurons derived from induced pluripotent stem cells (hiPSCs) exhibited increased expression of DISC1 and markers of synaptic plasticity. Conversely, hippocampi from Cav-1 knockout (KO) exhibited decreased expression of DISC1 and proteins involved in synaptic plasticity. Finally, SynCav1 delivery to the hippocampus of Cav-1 KO mice and Cav-1 KO neurons in culture restored expression of DISC1 and markers of synaptic plasticity. Furthermore, we found that Cav-1 coimmunoprecipitated with DISC1 in brain tissue. These findings suggest an important role by which neuron-targeted Cav-1 regulates DISC1 neurobiology with implications for synaptic plasticity. Therefore, SynCav1 might be a potential therapeutic target for restoring neuronal function in schizophrenia. NEW & NOTEWORTHY: The present study is the first to demonstrate that caveolin-1 can regulate DISC1 expression in neuronal models. Furthermore, the findings are consistent across three separate neuronal models that include rodent neurons (in vitro and in vivo) and human differentiated neurons derived from induced pluripotent stem cells. These findings justify further investigation regarding the modulatory role by caveolin on synaptic function and as a potential therapeutic target for the treatment of schizophrenia.


Subject(s)
Caveolin 1/metabolism , Gene Expression Regulation/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Animals , Caveolin 1/genetics , Cells, Cultured , Hippocampus/cytology , Humans , Immunoprecipitation , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Rats , Synapses/metabolism , Synapsins/genetics , Synapsins/metabolism , Transduction, Genetic , Red Fluorescent Protein
16.
Am J Physiol Cell Physiol ; 311(6): C854-C865, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27707689

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.


Subject(s)
Autophagy/physiology , Caveolin 3/metabolism , Ischemia/metabolism , Ischemia/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Animals , Apoptosis Regulatory Proteins/metabolism , Caspase 3/metabolism , Caveolae/metabolism , Cytochromes c/metabolism , Heart/physiology , Mice , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Reperfusion/methods
17.
Am J Physiol Cell Physiol ; 310(3): C181-92, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26771520

ABSTRACT

When considering which components of the cell are the most critical to function and physiology, we naturally focus on the nucleus, the mitochondria that regulate energy and apoptotic signaling, or other organelles such as the endoplasmic reticulum, Golgi, ribosomes, etc. Few people will suggest that the membrane is the most critical element of a cell in terms of function and physiology. Those that consider the membrane critical will point to its obvious barrier function regulated by the lipid bilayer and numerous ion channels that regulate homeostatic gradients. What becomes evident upon closer inspection is that not all membranes are created equal and that there are lipid-rich microdomains that serve as platforms of signaling and a means of communication with the intracellular environment. In this review, we explore the evolution of membranes, focus on lipid-rich microdomains, and advance the novel concept that membranes serve as "capacitors for energy and metabolism." Within this framework, the membrane then is the primary and critical regulator of stress and disease adaptation of the cell.


Subject(s)
Cell Membrane/metabolism , Electric Capacitance , Energy Metabolism , Animals , Biological Evolution , Elasticity , Humans , Membrane Fluidity , Membrane Lipids/metabolism , Membrane Microdomains/metabolism , Membrane Proteins/metabolism , Models, Biological , Oxidation-Reduction , Oxygen/metabolism , Signal Transduction
18.
J Endocrinol ; 224(1): 75-84, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25453115

ABSTRACT

Results from previous investigations have indicated that glucose-stimulated insulin secretion (GSIS) is affected by changes in cholesterol and its intermediates, but the precise link between secretion and cholesterol has not been thoroughly investigated. In this study, we show the contribution of both protein isoprenylation and cholesterol-dependent plasma membrane structural integrity to insulin secretion in INS-1E cells and mouse islets. Acute (2 h) inhibition of hydroxyl-methylglutaryl-CoA reductase by simvastatin (SIM) resulted in inhibition of GSIS without reduction in total cellular cholesterol content. This effect was prevented by cell loading with the isoprenyl molecule geranylgeranyl pyrophosphate. Chronic (24 h) inhibition of cholesterol biosynthesis resulted in inhibition of GSIS with a significant reduction in total cellular cholesterol content, which was also observed after the inhibition of cholesterol biosynthesis downstream of isoprenoid formation. Electron paramagnetic resonance analyses of INS-1E cells showed that the SIM-induced reduction in cholesterol increased plasma membrane fluidity. Thus, the blockade of cholesterol biosynthesis resulted in the reduction of availability of isoprenoids, followed by a reduction in the total cholesterol content associated with an increase in plasma membrane fluidity. Herein, we show the different contributions of cholesterol biosynthesis to GSIS, and propose that isoprenoid molecules and cholesterol-dependent signaling are dual regulators of proper ß-cell function.


Subject(s)
Cholesterol/biosynthesis , Insulin/metabolism , Animals , Anticholesteremic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cells, Cultured , Glucose/pharmacology , Insulin Secretion , Membrane Fluidity/drug effects , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/physiology , Mice , Polyisoprenyl Phosphates/pharmacology , Simvastatin/pharmacology , Tricarboxylic Acids/pharmacology
19.
J Cell Biol ; 203(6): 985-1001, 2013 Dec 23.
Article in English | MEDLINE | ID: mdl-24368806

ABSTRACT

Control of lipid droplet (LD) nucleation and copy number are critical, yet poorly understood, processes. We use model peptides that shift from the endoplasmic reticulum (ER) to LDs in response to fatty acids to characterize the initial steps of LD formation occurring in lipid-starved cells. Initially, arriving lipids are rapidly packed in LDs that are resistant to starvation (pre-LDs). Pre-LDs are restricted ER microdomains with a stable core of neutral lipids. Subsequently, a first round of "emerging" LDs is nucleated, providing additional lipid storage capacity. Finally, in proportion to lipid concentration, new rounds of LDs progressively assemble. Confocal microscopy and electron tomography suggest that emerging LDs are nucleated in a limited number of ER microdomains after a synchronized stepwise process of protein gathering, lipid packaging, and recognition by Plin3 and Plin2. A comparative analysis demonstrates that the acyl-CoA synthetase 3 is recruited early to the assembly sites, where it is required for efficient LD nucleation and lipid storage.


Subject(s)
Coenzyme A Ligases/physiology , Endoplasmic Reticulum/metabolism , Lipid Metabolism , Animals , COS Cells , Caveolin 1/chemistry , Chlorocebus aethiops , Coenzyme A Ligases/analysis , Coenzyme A Ligases/metabolism , Endoplasmic Reticulum/ultrastructure , Green Fluorescent Proteins/analysis , Intracellular Space , Methyltransferases/chemistry , Protein Engineering , Protein Sorting Signals
20.
Elife ; 1: e00003, 2012 Nov 13.
Article in English | MEDLINE | ID: mdl-23150794

ABSTRACT

We previously discovered histones bound to cytosolic lipid droplets (LDs); here we show that this forms a cellular antibacterial defense system. Sequestered on droplets under normal conditions, in the presence of bacterial lipopolysaccharide (LPS) or lipoteichoic acid (LTA), histones are released from the droplets and kill bacteria efficiently in vitro. Droplet-bound histones also function in vivo: when injected into Drosophila embryos lacking droplet-bound histones, bacteria grow rapidly. In contrast, bacteria injected into embryos with droplet-bound histones die. Embryos with droplet-bound histones displayed more than a fourfold survival advantage when challenged with four different bacterial species. Our data suggests that this intracellular antibacterial defense system may function in adult flies, and also potentially in mice.DOI:http://dx.doi.org/10.7554/eLife.00003.001.


Subject(s)
Drosophila melanogaster/immunology , Embryo, Nonmammalian/immunology , Histones/immunology , Lipid Droplets/immunology , Liver/immunology , Animals , Bacillus subtilis/drug effects , Bacillus subtilis/growth & development , Drosophila melanogaster/metabolism , Drosophila melanogaster/microbiology , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/microbiology , Escherichia coli/drug effects , Escherichia coli/growth & development , Histones/metabolism , Histones/pharmacology , Lipid Droplets/chemistry , Lipid Droplets/metabolism , Lipopolysaccharides/pharmacology , Listeria monocytogenes/drug effects , Listeria monocytogenes/growth & development , Liver/drug effects , Liver/metabolism , Liver/microbiology , Mice , Mice, Inbred C57BL , Staphylococcus epidermidis/drug effects , Staphylococcus epidermidis/growth & development , Teichoic Acids/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...