Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
FASEB J ; 36(11): e22584, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36190314

RESUMO

ARHGAP25, a RAC-specific GTPase activating protein (GAP), is an essential regulator of phagocyte effector functions such as phagocytosis, superoxide production, and transendothelial migration. Furthermore, its complex role in tumor behavior has recently been recognized. We previously demonstrated that phosphorylation of serine 363 in ARHGAP25 regulates hematopoietic stem cells and progenitor cells in mouse bone marrow. However, the significance of other potential phosphorylation sites of ARHGAP25 remained unknown. Now, we developed a novel, real-time bioluminescence resonance energy transfer (BRET) assay to monitor the GAP activity of ARHGAP25 in vitro. Using this approach, we revealed that phosphorylation of S363 and S488, but not that of S379-380, controls ARHGAP25's RACGAP activity. On the other hand, we found in granulocyte-differentiated human PLB-985 cells that superoxide production and actin depolymerization are regulated by residues S363 and S379-380. The present data demonstrate the value of our BRET-GAP assay and show that different phosphorylation patterns regulate ARHGAP25's GAP activity and its effect on superoxide production and phagocytosis.


Assuntos
Proteínas Ativadoras de GTPase , Superóxidos , Animais , Transferência de Energia , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Humanos , Fosforilação , Serina/metabolismo , Superóxidos/metabolismo
2.
J Cell Sci ; 131(10)2018 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-29661848

RESUMO

We have previously demonstrated in H295R adrenocortical cells that the Ca2+-dependent production of mitochondrial cAMP (mt-cAMP) by the matrix soluble adenylyl cyclase (sAC; encoded by ADCY10) is associated with enhanced aldosterone production. Here, we examined whether mitochondrial sAC and mt-cAMP fine tune mitochondrial Ca2+ metabolism to support steroidogenesis. Reduction of mt-cAMP formation resulted in decelerated mitochondrial Ca2+ accumulation in intact cells during K+-induced Ca2+ signalling and also in permeabilized cells exposed to elevated perimitochondrial [Ca2+]. By contrast, treatment with the membrane-permeable cAMP analogue 8-Br-cAMP, inhibition of phosphodiesterase 2 and overexpression of sAC in the mitochondrial matrix all intensified Ca2+ uptake into the organelle. Identical mt-cAMP dependence of mitochondrial Ca2+ uptake was also observed in HeLa cells. Importantly, the enhancing effect of mt-cAMP on Ca2+ uptake was independent from both the mitochondrial membrane potential and Ca2+ efflux, but was reduced by Epac1 (also known as RAPGEF3) blockade both in intact and in permeabilized cells. Finally, overexpression of sAC in the mitochondrial matrix potentiated aldosterone production implying that the observed positive feedback mechanism of mt-cAMP on mitochondrial Ca2+ accumulation may have a role in the rapid initiation of steroidogenesis.This article has an associated First Person interview with the first author of the paper.


Assuntos
Cálcio/metabolismo , AMP Cíclico/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Mitocôndrias/metabolismo , Adenilil Ciclases/genética , Adenilil Ciclases/metabolismo , Transporte Biológico , Fatores de Troca do Nucleotídeo Guanina/genética , Células HeLa , Humanos , Potencial da Membrana Mitocondrial , Mitocôndrias/genética
3.
Pflugers Arch ; 470(8): 1141-1148, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29876637

RESUMO

The biological effects of physiological stimuli of adrenocortical glomerulosa cells are predominantly mediated by the Ca2+ and the cAMP signal transduction pathways. The complex interplay between these signalling systems fine-tunes aldosterone secretion. In addition to the well-known cytosolic interactions, a novel intramitochondrial Ca2+-cAMP interplay has been recently recognised. The cytosolic Ca2+ signal is rapidly transferred into the mitochondrial matrix where it activates Ca2+-sensitive dehydrogenases, thus enhancing the formation of NADPH, a cofactor of steroid synthesis. Quite a few cell types, including H295R adrenocortical cells, express the soluble adenylyl cyclase within the mitochondria and the elevation of mitochondrial [Ca2+] activates the enzyme, thus resulting in the Ca2+-dependent formation of cAMP within the mitochondrial matrix. On the other hand, mitochondrial cAMP (mt-cAMP) potentiates the transfer of cytosolic Ca2+ into the mitochondrial matrix. This cAMP-mediated positive feedback control of mitochondrial Ca2+ uptake may facilitate the rapid hormonal response to emergency situations since knockdown of soluble adenylyl cyclase attenuates aldosterone production whereas overexpression of the enzyme facilitates steroidogenesis in vitro. Moreover, the mitochondrial Ca2+-mt-cAMP-Ca2+ uptake feedback loop is not a unique feature of adrenocortical cells; a similar signalling system has been described in HeLa cells as well.


Assuntos
Córtex Suprarrenal/metabolismo , Cálcio/metabolismo , AMP Cíclico/metabolismo , Mitocôndrias/metabolismo , Animais , Sinalização do Cálcio/fisiologia , Humanos
4.
Adv Exp Med Biol ; 993: 257-275, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28900919

RESUMO

Mitochondria extensively modify virtually all cellular Ca2+ transport processes, and store-operated Ca2+ entry (SOCE) is no exception to this rule. The interaction between SOCE and mitochondria is complex and reciprocal, substantially altering and, ultimately, fine-tuning both capacitative Ca2+ influx and mitochondrial function. Mitochondria, owing to their considerable Ca2+ accumulation ability, extensively buffer the cytosolic Ca2+ in their vicinity. In turn, the accumulated ion is released back into the neighboring cytosol during net Ca2+ efflux. Since store depletion itself and the successive SOCE are both Ca2+-regulated phenomena, mitochondrial Ca2+ handling may have wide-ranging effects on capacitative Ca2+ influx at any given time. In addition, mitochondria may also produce or consume soluble factors known to affect store-operated channels. On the other hand, Ca2+ entering the cell during SOCE is sensed by mitochondria, and the ensuing mitochondrial Ca2+ uptake boosts mitochondrial energy metabolism and, if Ca2+ overload occurs, may even lead to apoptosis or cell death. In several cell types, mitochondria seem to be sterically excluded from the confined space that forms between the plasma membrane (PM) and endoplasmic reticulum (ER) during SOCE. This implies that high-Ca2+ microdomains comparable to those observed between the ER and mitochondria do not form here. In the following chapter, the above aspects of the many-sided SOCE-mitochondrion interplay will be discussed in greater detail.


Assuntos
Canais de Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Mitocôndrias/metabolismo , Animais , Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Humanos
5.
Proc Natl Acad Sci U S A ; 111(50): E5420-8, 2014 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-25422468

RESUMO

Diabetic nephropathy is a major cause of end-stage kidney disease, and overactivity of the endocannabinoid/cannabinoid 1 receptor (CB1R) system contributes to diabetes and its complications. Zucker diabetic fatty (ZDF) rats develop type 2 diabetic nephropathy with albuminuria, reduced glomerular filtration, activation of the renin-angiotensin system (RAS), oxidative/nitrative stress, podocyte loss, and increased CB1R expression in glomeruli. Peripheral CB1R blockade initiated in the prediabetic stage prevented these changes or reversed them when animals with fully developed diabetic nephropathy were treated. Treatment of diabetic ZDF rats with losartan, an angiotensin II receptor-1 (Agtr1) antagonist, attenuated the development of nephropathy and down-regulated renal cortical CB1R expression, without affecting the marked hyperglycemia. In cultured human podocytes, CB1R and desmin gene expression were increased and podocin and nephrin content were decreased by either the CB1R agonist arachydonoyl-2'-chloroethylamide, angiotensin II, or high glucose, and the effects of all three were antagonized by CB1R blockade or siRNA-mediated knockdown of CNR1 (the cannabinoid type 1 receptor gene). We conclude that increased CB1R signaling in podocytes contributes to the development of diabetic nephropathy and represents a common pathway through which both hyperglycemia and increased RAS activity exert their deleterious effects, highlighting the therapeutic potential of peripheral CB1R blockade.


Assuntos
Diabetes Mellitus Tipo 2/complicações , Nefropatias Diabéticas/metabolismo , Modelos Biológicos , Podócitos/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Análise de Variância , Angiotensina II/farmacologia , Animais , Ácidos Araquidônicos/farmacologia , Desmina/metabolismo , Nefropatias Diabéticas/etiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Losartan/farmacologia , Pirazóis/síntese química , Pirazóis/farmacologia , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Ratos , Ratos Zucker , Receptor Tipo 1 de Angiotensina/metabolismo , Receptor CB1 de Canabinoide/agonistas , Receptor CB1 de Canabinoide/antagonistas & inibidores , Sulfonamidas/síntese química , Sulfonamidas/farmacologia
6.
Mol Pharmacol ; 88(2): 238-44, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26013543

RESUMO

6-Alkoxy-5-aryl-3-pyridincarboxamides, including the brain-penetrant compound 14G: [5-(4-chlorophenyl)-6-(cyclopropylmethoxy)-N-[(1R,2R)-2-hydroxy-cyclohexyl]-3-pyridinecarboxamide] and its peripherally restricted analog 14H: [5-(4-chlorophenyl)-N-[(1R,2R)-2-hydroxycyclohexyl]-6-(2-methoxyethoxy)-3-pyridinecarboxamide], have been recently introduced as selective, high-affinity antagonists of the human cannabinoid-1 receptor (hCB1R). Binding analyses revealed two orders of magnitude lower affinity of these compounds for mouse and rat versus human CB1R, whereas the affinity of rimonabant is comparable for all three CB1Rs. Modeling of ligand binding to CB1R and binding assays with native and mutant (Ile105Met) hCB1Rs indicate that the Ile105 to Met mutation in rodent CB1Rs accounts for the species-dependent affinity of 14G: and 14H: . Our work identifies Ile105 as a new pharmacophore component for developing better hCB1R antagonists and invalidates rodent models for assessing the antiobesity efficacy of 14G: and 14H: .


Assuntos
Encéfalo/metabolismo , Antagonistas de Receptores de Canabinoides/síntese química , Antagonistas de Receptores de Canabinoides/farmacologia , Niacinamida/análogos & derivados , Piperidinas/farmacologia , Pirazóis/farmacologia , Receptor CB1 de Canabinoide/genética , Animais , Antagonistas de Receptores de Canabinoides/química , Células HEK293 , Humanos , Isoleucina/metabolismo , Camundongos , Modelos Moleculares , Niacinamida/síntese química , Niacinamida/química , Niacinamida/farmacologia , Piperidinas/química , Pirazóis/química , Ratos , Receptor CB1 de Canabinoide/metabolismo , Rimonabanto , Especificidade da Espécie , Relação Estrutura-Atividade , Difração de Raios X
7.
Am J Physiol Endocrinol Metab ; 306(4): E457-68, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24381003

RESUMO

The adipocyte-derived hormone adiponectin promotes fatty acid oxidation and improves insulin sensitivity and thus plays a key role in the regulation of lipid and glucose metabolism and energy homeostasis. Chronic cannabinoid type 1 (CB1) receptor blockade also increases lipid oxidation and improves insulin sensitivity in obese individuals or animals, resulting in reduced cardiometabolic risk. Chronic CB1 blockade reverses the obesity-related decline in serum adiponectin levels, which has been proposed to account for the metabolic effects of CB1 antagonists. Here, we investigated the metabolic actions of the CB1 inverse agonist rimonabant in high-fat diet (HFD)-induced obese adiponectin knockout (Adipo(-/-)) mice and their wild-type littermate controls (Adipo(+/+)). HFD-induced obesity and its hormonal/metabolic consequences were indistinguishable in the two strains. Daily treatment of obese mice with rimonabant for 7 days resulted in significant and comparable reductions in body weight, serum leptin, free fatty acid, cholesterol, and triglyceride levels in the two strains. Rimonabant treatment improved glucose homeostasis and insulin sensitivity to the same extent in Adipo(+/+) and Adipo(-/-) mice, whereas it reversed the HFD-induced hepatic steatosis, fibrosis, and hepatocellular damage only in the former. The adiponectin-dependent, antisteatotic effect of rimonabant was mediated by reduced uptake and increased ß-oxidation of fatty acids in the liver. We conclude that reversal of the HFD-induced hepatic steatosis and fibrosis by chronic CB1 blockade, but not the parallel reduction in adiposity and improved glycemic control, is mediated by adiponectin.


Assuntos
Adiponectina/metabolismo , Peso Corporal/efeitos dos fármacos , Antagonistas de Receptores de Canabinoides/farmacologia , Fígado Gorduroso/metabolismo , Obesidade/metabolismo , Piperidinas/farmacologia , Pirazóis/farmacologia , Receptor CB1 de Canabinoide/antagonistas & inibidores , Adiponectina/genética , Adiposidade/efeitos dos fármacos , Animais , Dieta Hiperlipídica , Ingestão de Energia/efeitos dos fármacos , Fígado Gorduroso/genética , Leptina/metabolismo , Metabolismo dos Lipídeos , Lipídeos/sangue , Fígado/efeitos dos fármacos , Fígado/metabolismo , Camundongos , Camundongos Knockout , Obesidade/genética , Rimonabanto
8.
iScience ; 26(7): 107207, 2023 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-37534180

RESUMO

Molecular interactions between anorexigenic leptin and orexigenic endocannabinoids, although of great metabolic significance, are not well understood. We report here that hypothalamic STAT3 signaling in mice, initiated by physiological elevations of leptin, is diminished by agonists of the cannabinoid receptor 1 (CB1R). Measurement of STAT3 activation by semi-automated confocal microscopy in cultured neurons revealed that this CB1R-mediated inhibition requires both T cell protein tyrosine phosphatase (TC-PTP) and ß-arrestin1 but is independent of changes in cAMP. Moreover, ß-arrestin1 translocates to the nucleus upon CB1R activation and binds both STAT3 and TC-PTP. Consistently, CB1R activation failed to suppress leptin signaling in ß-arrestin1 knockout mice in vivo, and in neural cells deficient in CB1R, ß-arrestin1 or TC-PTP. Altogether, CB1R activation engages ß-arrestin1 to coordinate the TC-PTP-mediated inhibition of the leptin-evoked neuronal STAT3 response. This mechanism may restrict the anorexigenic effects of leptin when hypothalamic endocannabinoid levels rise, as during fasting or in diet-induced obesity.

9.
Pflugers Arch ; 464(1): 43-50, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22395411

RESUMO

Aldosterone, secreted by adrenal glomerulosa cells, allows the adaptation of the vertebrate organism to a wide range of physiological and pathological stimuli including acute haemodynamic challenges and long-term changes in dietary sodium and potassium intake. Most of the extracellular signals are mediated by cytosolic Ca²âº signal deriving from Ca²âº release, store-operated and/or voltage-gated Ca²âº influx. Mitochondria in glomerulosa cells play a fundamental role in generating and modulating the final biological response. These organelles not only house several enzymes of aldosterone biosynthesis but also-in a Ca²âº-dependent manner-provide NADPH for the function of these enzymes. Moreover, mitochondria, constituting a high portion of cytoplasmic volume and displaying a uniquely low-threshold Ca²âº sequestering ability, shape and thus modulate the decoding of the complex cytosolic Ca²âº response. The unusual features of mitochondrial Ca²âº signalling that permit such an integrative function in adrenal glomerulosa cells are hereby described.


Assuntos
Sinalização do Cálcio , Cálcio/metabolismo , Células Endócrinas/metabolismo , Mitocôndrias/metabolismo , Zona Glomerulosa/metabolismo , Aldosterona/metabolismo , Animais , Canais de Cálcio/metabolismo , Citosol/metabolismo , Humanos , Proteínas Mitocondriais/metabolismo , NADP/metabolismo
10.
Nat Commun ; 13(1): 1783, 2022 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-35379807

RESUMO

Activation of the cannabinoid-1 receptor (CB1R) and the mammalian target of rapamycin complex 1 (mTORC1) in the renal proximal tubular cells (RPTCs) contributes to the development of diabetic kidney disease (DKD). However, the CB1R/mTORC1 signaling axis in the kidney has not been described yet. We show here that hyperglycemia-induced endocannabinoid/CB1R stimulation increased mTORC1 activity, enhancing the transcription of the facilitative glucose transporter 2 (GLUT2) and leading to the development of DKD in mice; this effect was ameliorated by specific RPTCs ablation of GLUT2. Conversely, CB1R maintained the normal activity of mTORC1 by preventing the cellular excess of amino acids during normoglycemia. Our findings highlight a novel molecular mechanism by which the activation of mTORC1 in RPTCs is tightly controlled by CB1R, either by enhancing the reabsorption of glucose and inducing kidney dysfunction in diabetes or by preventing amino acid uptake and maintaining normal kidney function in healthy conditions.


Assuntos
Nefropatias Diabéticas , Receptor CB1 de Canabinoide , Animais , Nefropatias Diabéticas/patologia , Rim/metabolismo , Túbulos Renais Proximais/metabolismo , Mamíferos , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Receptor CB1 de Canabinoide/genética , Receptor CB1 de Canabinoide/metabolismo
11.
Front Endocrinol (Lausanne) ; 12: 740913, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34745007

RESUMO

The G protein-coupled type 1 cannabinoid receptor (CB1R) mediates virtually all classic cannabinoid effects, and both its agonists and antagonists hold major therapeutic potential. Heterologous expression of receptors is vital for pharmacological research, however, overexpression of these proteins may fundamentally alter their localization pattern, change the signalling partner preference and may also spark artificial clustering. Additionally, recombinant CB1Rs are prone to intense proteasomal degradation, which may necessitate substantial modifications, such as N-terminal truncation or signal sequence insertion, for acceptable cell surface expression. We report here that tuning down the expression intensity of the full-length CB1R reduces proteasomal degradation and offers receptor levels that are comparable to those of endogenous CB1 receptors. As opposed to high-efficiency expression with conventional promoters, weak promoter-driven CB1R expression provides ERK 1/2 and p38 MAPK signalling that closely resemble the activity of endogenous CB1Rs. Moreover, weakly expressed CB1R variants exhibit plasma membrane localization, preserve canonical Gi-signalling but prevent CB1R-Gs coupling observed with high-expression variants. Based on these findings, we propose that lowering the expression level of G protein-coupled receptors should always be considered in heterologous expression systems in order to reduce the pressure on the proteasomal machinery and to avoid potential signalling artefacts.


Assuntos
Receptor CB1 de Canabinoide/biossíntese , Linhagem Celular , Estresse do Retículo Endoplasmático , Humanos , Sistema de Sinalização das MAP Quinases , Proteína Quinase 3 Ativada por Mitógeno , Regiões Promotoras Genéticas , Complexo de Endopeptidases do Proteassoma , RNA Interferente Pequeno/farmacologia , Receptor CB1 de Canabinoide/genética , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
Elife ; 92020 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-33210603

RESUMO

The soluble isoform of leptin receptor (sOb-R), secreted by the liver, regulates leptin bioavailability and bioactivity. Its reduced levels in diet-induced obesity (DIO) contribute to hyperleptinemia and leptin resistance, effects that are regulated by the endocannabinoid (eCB)/CB1R system. Here we show that pharmacological activation/blockade and genetic overexpression/deletion of hepatic CB1R modulates sOb-R levels and hepatic leptin resistance. Interestingly, peripheral CB1R blockade failed to reverse DIO-induced reduction of sOb-R levels, increased fat mass and dyslipidemia, and hepatic steatosis in mice lacking C/EBP homologous protein (CHOP), whereas direct activation of CB1R in wild-type hepatocytes reduced sOb-R levels in a CHOP-dependent manner. Moreover, CHOP stimulation increased sOb-R expression and release via a direct regulation of its promoter, while CHOP deletion reduced leptin sensitivity. Our findings highlight a novel molecular aspect by which the hepatic eCB/CB1R system is involved in the development of hepatic leptin resistance and in the regulation of sOb-R levels via CHOP.


When the human body has stored enough energy from food, it releases a hormone called leptin that travels to the brain and stops feelings of hunger. This hormone moves through the bloodstream and can affect other organs, such as the liver, which also help control our body's energy levels. Most people with obesity have very high levels of leptin in their blood, but are resistant to its effects and will therefore continue to feel hungry despite having stored enough energy. One of the proteins that controls the levels of leptin is a receptor called sOb-R, which is released by the liver and binds to leptin as it travels in the blood. Individuals with high levels of this receptor often have less free leptin in their bloodstream and a lower body weight. Another protein that helps the body to regulate its energy levels is the cannabinoid-1 receptor, or CB1R for short. In people with obesity, this receptor is overactive and has been shown to contribute to leptin resistance, which is when the brain becomes less receptive to leptin. Previous work in mice showed that blocking CB1R reduced the levels of leptin and allowed mice to react to this hormone normally again, but it remained unclear whether CB1R affects how other organs, such as the liver, respond to leptin. To answer this question, Drori et al. blocked the CB1R receptor in the liver of mice eating a high-fat diet, either by using a drug or by deleting the gene that codes for this protein. This caused mice to have higher levels of sOb-R circulating in their bloodstream. Further experiments showed that this change in sOb-R was caused by the levels of a protein called CHOP increasing in the liver when CB1R was blocked. Drori et al. found that inhibiting CB1R caused these obese mice to lose weight and have healthier, less fatty livers as a result of their livers no longer being resistant to the effects of leptin. Scientists, doctors and pharmaceutical companies are trying to develop new strategies to combat obesity. The results from these experiments suggest that blocking CB1R in the liver could allow this organ to react to leptin appropriately again. Drugs blocking CB1R, including the one used in this study, will be tested in clinical trials and could provide a new approach for treating obesity.


Assuntos
Estresse do Retículo Endoplasmático , Hepatócitos/metabolismo , Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Receptores para Leptina/metabolismo , Fator de Transcrição CHOP/metabolismo , Fator 4 Ativador da Transcrição/genética , Fator 4 Ativador da Transcrição/metabolismo , Animais , Antagonistas de Receptores de Canabinoides/farmacologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Dieta Hiperlipídica , Modelos Animais de Doenças , Endocanabinoides/metabolismo , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/patologia , Obesidade/complicações , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptor CB1 de Canabinoide/genética , Receptores para Leptina/genética , Transdução de Sinais , Fator de Transcrição CHOP/genética
13.
Cell Calcium ; 44(1): 51-63, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18242694

RESUMO

The Ca(2+) coupling between endoplasmic reticulum (ER) and mitochondria is central to multiple cell survival and cell death mechanisms. Cytoplasmic [Ca(2+)] ([Ca(2+)](c)) spikes and oscillations produced by ER Ca(2+) release are effectively delivered to the mitochondria. Propagation of [Ca(2+)](c) signals to the mitochondria requires the passage of Ca(2+) across three membranes, namely the ER membrane, the outer mitochondrial membrane (OMM) and the inner mitochondrial membrane (IMM). Strategic positioning of the mitochondria by cytoskeletal transport and interorganellar tethers provides a means to promote the local transfer of Ca(2+) between the ER membrane and OMM. In this setting, even >100 microM [Ca(2+)] may be attained to activate the low affinity mitochondrial Ca(2+) uptake. However, a mitochondrial [Ca(2+)] rise has also been documented during submicromolar [Ca(2+)](c) elevations. Evidence has been emerging that Ca(2+) exerts allosteric control on the Ca(2+) transport sites at each membrane, providing mechanisms that may facilitate the Ca(2+) delivery to the mitochondria. Here we discuss the fundamental mechanisms of ER and mitochondrial Ca(2+) transport, particularly the control of their activity by Ca(2+) and evaluate both high- and low-[Ca(2+)]-activated mitochondrial calcium signals in the context of cell physiology.


Assuntos
Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Retículo Endoplasmático/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Mitocôndrias , Canal de Liberação de Cálcio do Receptor de Rianodina/metabolismo , Regulação Alostérica/genética , Animais , Citoesqueleto/fisiologia , Retículo Endoplasmático/genética , Humanos , Receptores de Inositol 1,4,5-Trifosfato/química , Receptores de Inositol 1,4,5-Trifosfato/genética , Mitocôndrias/genética , Mitocôndrias/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Membranas Mitocondriais/fisiologia , Poro de Transição de Permeabilidade Mitocondrial , Canal de Liberação de Cálcio do Receptor de Rianodina/genética
14.
Cell Calcium ; 43(3): 250-9, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17628663

RESUMO

Angiotensin II elicits cytosolic and mitochondrial Ca2+ signal in H295R adrenocortical cells. We found that Ca2+ uptake rate and peak values in small mitochondrial regions both depend on the colocalization of these mitochondrial regions with GFP-marked endoplasmic reticular (ER) vesicles. The dependence of the Ca2+ response on this colocalization is abolished by SB202190 and PD169316, inhibitors of p38 MAPK, as well as by transfection with siRNA against p38 MAPK mRNA. The same manoeuvres result in an increased ratio of global mitochondrial to global cytosolic Ca2+ response, indicating that inhibition of p38 MAPK is followed by enhanced mitochondrial Ca2+ uptake. alpha-Toxin and TNFalpha, agents which similarly to angiotensin II increase the phosphorylation of p38, failed to affect mitochondrial Ca2+ uptake, indicating that activation of p38 MAPK is necessary but not sufficient for the inhibition of Ca2+ uptake. Bisindolylmaleimide, an inhibitor of the conventional and novel-type protein kinase C isoforms also evokes enhanced mitochondrial Ca2+ uptake, whereas Gö6976 that inhibits the conventional isoforms only failed to exert any effect. These data show that angiotensin II attenuates Ca2+ uptake predominantly into mitochondria that do not colocalize with ER, by a mechanism involving p38 MAPK and a novel-type PKC.


Assuntos
Cálcio/metabolismo , Mitocôndrias/metabolismo , Proteína Quinase C-épsilon/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Angiotensina II/farmacologia , Toxinas Bacterianas/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Linhagem Celular , Citosol/efeitos dos fármacos , Citosol/enzimologia , Ativação Enzimática/efeitos dos fármacos , Humanos , Immunoblotting , Isoenzimas/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Fosforilação/efeitos dos fármacos , Proteína Quinase C-épsilon/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , RNA Interferente Pequeno/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
16.
Mol Metab ; 6(10): 1113-1125, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-29031713

RESUMO

OBJECTIVE: In visceral obesity, an overactive endocannabinoid/CB1 receptor (CB1R) system promotes increased caloric intake and decreases energy expenditure, which are mitigated by global or peripheral CB1R blockade. In mice with diet-induced obesity (DIO), inhibition of food intake by the peripherally restricted CB1R antagonist JD5037 could be attributed to endogenous leptin due to the rapid reversal of hyperleptinemia that maintains leptin resistance, but the signaling pathway engaged by leptin has remained to be determined. METHODS: We analyzed the hypothalamic circuitry targeted by leptin following chronic treatment of DIO mice with JD5037. RESULTS: Leptin treatment or an increase in endogenous leptin following fasting/refeeding induced STAT3 phosphorylation in neurons in the arcuate nucleus (ARC) in lean and JD5037-treated DIO mice, but not in vehicle-treated DIO animals. Co-localization of pSTAT3 in leptin-treated mice was significantly less common with NPY+ than with POMC+ ARC neurons. The hypophagic effect of JD5037 was absent in melanocortin-4 receptor (MC4R) deficient obese mice or DIO mice treated with a MC4R antagonist, but was maintained in NPY-/- mice kept on a high-fat diet. CONCLUSIONS: Peripheral CB1R blockade in DIO restores sensitivity to endogenous leptin, which elicits hypophagia via the re-activation of melanocortin signaling in the ARC.


Assuntos
Leptina/metabolismo , Receptor CB1 de Canabinoide/metabolismo , Receptores para Leptina/efeitos dos fármacos , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Peso Corporal/fisiologia , Canabinoides/metabolismo , Dieta Hiperlipídica , Gorduras na Dieta/metabolismo , Ingestão de Alimentos/fisiologia , Hipotálamo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Neuropeptídeo Y/metabolismo , Obesidade/metabolismo , Pró-Opiomelanocortina/metabolismo , Pirazóis/farmacologia , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptor CB1 de Canabinoide/fisiologia , Receptor Tipo 4 de Melanocortina/metabolismo , Receptores de Canabinoides/metabolismo , Receptores para Leptina/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/farmacologia
17.
Cell Rep ; 18(10): 2291-2300, 2017 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-28273446

RESUMO

Mitochondrial Ca2+ uptake through the Ca2+ uniporter supports cell functions, including oxidative metabolism, while meeting tissue-specific calcium signaling patterns and energy needs. The molecular mechanisms underlying tissue-specific control of the uniporter are unknown. Here, we investigated a possible role for tissue-specific stoichiometry between the Ca2+-sensing regulators (MICUs) and pore unit (MCU) of the uniporter. Low MICU1:MCU protein ratio lowered the [Ca2+] threshold for Ca2+ uptake and activation of oxidative metabolism but decreased the cooperativity of uniporter activation in heart and skeletal muscle compared to liver. In MICU1-overexpressing cells, MICU1 was pulled down by MCU proportionally to MICU1 overexpression, suggesting that MICU1:MCU protein ratio directly reflected their association. Overexpressing MICU1 in the heart increased MICU1:MCU ratio, leading to liver-like mitochondrial Ca2+ uptake phenotype and cardiac contractile dysfunction. Thus, the proportion of MICU1-free and MICU1-associated MCU controls these tissue-specific uniporter phenotypes and downstream Ca2+ tuning of oxidative metabolism.


Assuntos
Canais de Cálcio/metabolismo , Sinalização do Cálcio , Proteínas de Ligação ao Cálcio/metabolismo , Mitocôndrias/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Proteínas Mitocondriais/metabolismo , Especificidade de Órgãos , Feminino , Humanos , Fígado/metabolismo , Músculos/metabolismo , Miocárdio/metabolismo , Oxirredução
18.
Diabetes ; 66(4): 994-1007, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28082458

RESUMO

Islet inflammation promotes ß-cell loss and type 2 diabetes (T2D), a process replicated in Zucker Diabetic Fatty (ZDF) rats in which ß-cell loss has been linked to cannabinoid-1 receptor (CB1R)-induced proinflammatory signaling in macrophages infiltrating pancreatic islets. Here, we analyzed CB1R signaling in macrophages and its developmental role in T2D. ZDF rats with global deletion of CB1R are protected from ß-cell loss, hyperglycemia, and nephropathy that are present in ZDF littermates. Adoptive transfer of CB1R-/- bone marrow to ZDF rats also prevents ß-cell loss and hyperglycemia but not nephropathy. ZDF islets contain elevated levels of CB1R, interleukin-1ß, tumor necrosis factor-α, the chemokine CCL2, and interferon regulatory factor-5 (IRF5), a marker of inflammatory macrophage polarization. In primary cultured rodent and human macrophages, CB1R activation increased Irf5 expression, whereas knockdown of Irf5 blunted CB1R-induced secretion of inflammatory cytokines without affecting CCL2 expression, which was p38MAPKα dependent. Macrophage-specific in vivo knockdown of Irf5 protected ZDF rats from ß-cell loss and hyperglycemia. Thus, IRF5 is a crucial downstream mediator of diabetogenic CB1R signaling in macrophages and a potential therapeutic target.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Nefropatias Diabéticas/genética , Hiperglicemia/genética , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Macrófagos/metabolismo , Receptor CB1 de Canabinoide/genética , Animais , Quimiocina CCL2/metabolismo , Nefropatias Diabéticas/metabolismo , Técnicas de Inativação de Genes , Hiperglicemia/metabolismo , Fatores Reguladores de Interferon/metabolismo , Interleucina-1beta , Masculino , Ratos , Ratos Zucker , Receptor CB1 de Canabinoide/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
19.
JCI Insight ; 2(8)2017 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-28422760

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a life-threatening disease without effective treatment, highlighting the need for identifying new targets and treatment modalities. The pathogenesis of IPF is complex, and engaging multiple targets simultaneously might improve therapeutic efficacy. To assess the role of the endocannabinoid/cannabinoid receptor 1 (endocannabinoid/CB1R) system in IPF and its interaction with inducible nitric oxide synthase (iNOS) as dual therapeutic targets, we analyzed lung fibrosis and the status of the endocannabinoid/CB1R system and iNOS in mice with bleomycin-induced pulmonary fibrosis (PF) and in lung tissue and bronchoalveolar lavage fluid (BALF) from patients with IPF, as well as controls. In addition, we investigated the antifibrotic efficacy in the mouse PF model of an orally bioavailable and peripherally restricted CB1R/iNOS hybrid inhibitor. We report that increased activity of the endocannabinoid/CB1R system parallels disease progression in the lungs of patients with idiopathic PF and in mice with bleomycin-induced PF and is associated with increased tissue levels of interferon regulatory factor-5. Furthermore, we demonstrate that simultaneous engagement of the secondary target iNOS by the hybrid CB1R/iNOS inhibitor has greater antifibrotic efficacy than inhibition of CB1R alone. This hybrid antagonist also arrests the progression of established fibrosis in mice, thus making it a viable candidate for future translational studies in IPF.

20.
Cell Calcium ; 40(5-6): 527-37, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17069884

RESUMO

The mitochondrial Ca(2+) uniporter has low affinity for Ca(2+), therefore it has been assumed that submicromolar Ca(2+) signals cannot induce mitochondrial Ca(2+) uptake. The close apposition of the plasma membrane or the endoplamic reticulum (ER) to the mitochondria and the limited Ca(2+) diffusion in the cytoplasm result in the formation of perimitochondrial high-Ca(2+) microdomains (HCMDs) capable of activating mitochondrial Ca(2+) uptake. The possibility of mitochondrial Ca(2+) uptake at low submicromolar [Ca(2+)](c) has not yet been generally accepted. Earlier we found in permeabilized glomerulosa, luteal and pancreatic beta cells that [Ca(2+)](m) increased when [Ca(2+)](c) was raised from 60 nM to less than 200 nM. Here we report data obtained from H295R (adrenocortical) cells transfected with ER-targeted GFP. Cytoplasmic Ca(2+) response to angiotensin II was different in mitochondrion-rich and mitochondrion-free domains. The mitochondrial Ca(2+) response to angiotensin II correlated with GFP fluorescence indicating the vicinity of ER. When the cells were exposed to K(+) (inducing Ca(2+) influx), no correlation was found between the mitochondrial Ca(2+) signal and the vicinity of the plasma membrane or the ER. The results presented here provide evidence that mitochondrial Ca(2+) uptake may occur both with and without the formation of HCMDs within the same cell.


Assuntos
Cálcio/metabolismo , Microdomínios da Membrana/metabolismo , Mitocôndrias/metabolismo , Angiotensina II/farmacologia , Canais de Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Linhagem Celular Tumoral , Retículo Endoplasmático/metabolismo , Humanos , Receptores de Inositol 1,4,5-Trifosfato/fisiologia , Microdomínios da Membrana/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Potássio/farmacologia , Zona Glomerulosa/citologia , Zona Glomerulosa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA