Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Physiol ; 14: 1142354, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36935756

RESUMO

An adequate supply of oxygen (O2) is essential for most life forms on earth, making the delivery of appropriate levels of O2 to tissues a fundamental physiological challenge. When O2 levels in the alveoli and/or blood are low, compensatory adaptive reflexes are produced that increase the uptake of O2 and its distribution to tissues within a few seconds. This paper analyzes the most important acute vasomotor responses to lack of O2 (hypoxia): hypoxic pulmonary vasoconstriction (HPV) and hypoxic vasodilation (HVD). HPV affects distal pulmonary (resistance) arteries, with its homeostatic role being to divert blood to well ventilated alveoli to thereby optimize the ventilation/perfusion ratio. HVD is produced in most systemic arteries, in particular in the skeletal muscle, coronary, and cerebral circulations, to increase blood supply to poorly oxygenated tissues. Although vasomotor responses to hypoxia are modulated by endothelial factors and autonomic innervation, it is well established that arterial smooth muscle cells contain an acute O2 sensing system capable of detecting changes in O2 tension and to signal membrane ion channels, which in turn regulate cytosolic Ca2+ levels and myocyte contraction. Here, we summarize current knowledge on the nature of O2 sensing and signaling systems underlying acute vasomotor responses to hypoxia. We also discuss similarities and differences existing in O2 sensors and effectors in the various arterial territories.

2.
Int J Mol Sci ; 24(6)2023 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-36982650

RESUMO

Antiparkinsonian carotid body (CB) cell therapy has been proven to be effective in rodent and nonhuman primate models of Parkinson's disease (PD), exerting trophic protection and restoration of the dopaminergic nigrostriatal pathway. These neurotrophic actions are mediated through the release of high levels of glial-cell-line-derived neurotrophic factor (GDNF) by the CB transplant. Pilot clinical trials have also shown that CB autotransplantation can improve motor symptoms in PD patients, although its effectiveness is affected by the scarcity of the grafted tissue. Here, we analyzed the antiparkinsonian efficacy of in vitro-expanded CB dopaminergic glomus cells. Intrastriatal xenografts of rat CB neurospheres were shown to protect nigral neurons from degeneration in a chronic MPTP mouse PD model. In addition, grafts performed at the end of the neurotoxic treatment resulted in the repair of striatal dopaminergic terminals through axonal sprouting. Interestingly, both neuroprotective and reparative effects induced by in vitro-expanded CB cells were similar to those previously reported by the use of CB transplants. This action could be explained because stem-cell-derived CB neurospheres produce similar amounts of GDNF compared to native CB tissue. This study provides the first evidence that in vitro-expanded CB cells could be a clinical option for cell therapy in PD.


Assuntos
Corpo Carotídeo , Doença de Parkinson , Camundongos , Ratos , Humanos , Animais , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Corpo Carotídeo/metabolismo , Doença de Parkinson/terapia , Doença de Parkinson/metabolismo , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Transplante de Células , Substância Negra/metabolismo , Modelos Animais de Doenças , Corpo Estriado/metabolismo
3.
Sci Immunol ; 7(70): eabm8161, 2022 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-35486677

RESUMO

Effective T cell-mediated immune responses require the proper allocation of metabolic resources to sustain growth, proliferation, and cytokine production. Epigenetic control of the genome also governs T cell transcriptome and T cell lineage commitment and maintenance. Cellular metabolic programs interact with epigenetic regulation by providing substrates for covalent modifications of chromatin. By using complementary genetic, epigenetic, and metabolic approaches, we revealed that tricarboxylic acid (TCA) cycle flux fueled biosynthetic processes while controlling the ratio of succinate/α-ketoglutarate (α-KG) to modulate the activities of dioxygenases that are critical for driving T cell inflammation. In contrast to cancer cells, where succinate dehydrogenase (SDH)/complex II inactivation drives cell transformation and growth, SDH/complex II deficiency in T cells caused proliferation and survival defects when the TCA cycle was truncated, blocking carbon flux to support nucleoside biosynthesis. Replenishing the intracellular nucleoside pool partially relieved the dependence of T cells on SDH/complex II for proliferation and survival. SDH deficiency induced a proinflammatory gene signature in T cells and promoted T helper 1 and T helper 17 lineage differentiation. An increasing succinate/α-KG ratio in SDH-deficient T cells promoted inflammation by changing the pattern of the transcriptional and chromatin accessibility signatures and consequentially increasing the expression of the transcription factor, PR domain zinc finger protein 1. Collectively, our studies revealed a role of SDH/complex II in allocating carbon resources for anabolic processes and epigenetic regulation in T cell proliferation and inflammation.


Assuntos
Epigênese Genética , Succinato Desidrogenase , Proliferação de Células , Cromatina , Complexo II de Transporte de Elétrons/deficiência , Humanos , Inflamação/genética , Ácidos Cetoglutáricos/química , Ácidos Cetoglutáricos/metabolismo , Ácidos Cetoglutáricos/farmacologia , Erros Inatos do Metabolismo , Doenças Mitocondriais , Nucleosídeos , Succinato Desidrogenase/genética , Succinato Desidrogenase/metabolismo , Succinatos
4.
FASEB J ; 35(2): e21227, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33247500

RESUMO

Mutations in any of the genes encoding the four subunits of succinate dehydrogenase (SDH), a mitochondrial membrane-bound enzyme complex that is involved in both the tricarboxylic acid cycle and the electron transport chain, can lead to a variety of disorders. Recognized conditions with such mutations include Leigh syndrome and hereditary tumors such as pheochromocytoma and paraganglioma (PPGL), renal cell carcinoma, and gastrointestinal stromal tumor. Tumors appear in SDH mutation carriers with dominant inheritance due to loss of heterozygosity in susceptible cells. Here, we describe a mouse model intended to reproduce hereditary PPGL through Cre-mediated loss of SDHC in cells that express tyrosine hydroxylase (TH), a compartment where PPGL is known to originate. We report that while there is modest expansion of TH+ glomus cells in the carotid body upon SDHC loss, PPGL is not observed in such mice, even in the presence of a conditional dominant negative p53 protein and chronic hypoxia. Instead, we report an unexpected phenotype of nondiabetic obesity beginning at about 20 weeks of age. We hypothesize that this obesity is caused by TH+ cell loss or altered phenotype in key compartments of the central nervous system responsible for regulating feeding behavior, coupled with metabolic changes due to loss of peripheral catecholamine production.


Assuntos
Neoplasias das Glândulas Suprarrenais/genética , Modelos Animais de Doenças , Síndromes Neoplásicas Hereditárias/genética , Obesidade/genética , Fenótipo , Feocromocitoma/genética , Succinato Desidrogenase/genética , Neoplasias das Glândulas Suprarrenais/patologia , Animais , Carcinogênese/genética , Carcinogênese/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Síndromes Neoplásicas Hereditárias/patologia , Obesidade/patologia , Feocromocitoma/patologia , Succinato Desidrogenase/deficiência
5.
Front Physiol ; 11: 614893, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33329066

RESUMO

Carotid body glomus cells are multimodal arterial chemoreceptors able to sense and integrate changes in several physical and chemical parameters in the blood. These cells are also essential for O2 homeostasis. Glomus cells are prototypical peripheral O2 sensors necessary to detect hypoxemia and to elicit rapid compensatory responses (hyperventilation and sympathetic activation). The mechanisms underlying acute O2 sensing by glomus cells have been elusive. Using a combination of mouse genetics and single-cell optical and electrophysiological techniques, it has recently been shown that activation of glomus cells by hypoxia relies on the generation of mitochondrial signals (NADH and reactive oxygen species), which modulate membrane ion channels to induce depolarization, Ca2+ influx, and transmitter release. The special sensitivity of glomus cell mitochondria to changes in O2 tension is due to Hif2α-dependent expression of several atypical mitochondrial subunits, which are responsible for an accelerated oxidative metabolism and the strict dependence of mitochondrial complex IV activity on O2 availability. A mitochondrial-to-membrane signaling model of acute O2 sensing has been proposed, which explains existing data and provides a solid foundation for future experimental tests. This model has also unraveled new molecular targets for pharmacological modulation of carotid body activity potentially relevant in the treatment of highly prevalent medical conditions.

6.
Brain Commun ; 2(2): fcaa105, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32954345

RESUMO

Administration of recombinant glial cell line-derived neurotrophic factor into the putamen has been tested in preclinical and clinical studies to evaluate its neuroprotective effects on the progressive dopaminergic neuronal degeneration that characterizes Parkinson's disease. However, intracerebral glial cell line-derived neurotrophic factor infusion is a challenging therapeutic strategy, with numerous potential technical and medical limitations. Most of these limitations could be avoided if the production of endogenous glial cell line-derived neurotrophic factor could be increased. Glial cell line-derived neurotrophic factor is naturally produced in the striatum from where it exerts a trophic action on the nigrostriatal dopaminergic pathway. Most of striatal glial cell line-derived neurotrophic factor is synthesized by a subset of GABAergic interneurons characterized by the expression of parvalbumin. We sought to identify molecular targets specific to those neurons and which are putatively associated with glial cell line-derived neurotrophic factor synthesis. To this end, the transcriptomic differences between glial cell line-derived neurotrophic factor-positive parvalbumin neurons in the striatum and parvalbumin neurons located in the nearby cortex, which do not express glial cell line-derived neurotrophic factor, were analysed. Using mouse reporter models, we have defined the genomic signature of striatal parvalbumin interneurons obtained by fluorescence-activated cell sorting followed by microarray comparison. Short-listed genes were validated by additional histological and molecular analyses. These genes code for membrane receptors (Kit, Gpr83, Tacr1, Tacr3, Mc3r), cytosolic proteins (Pde3a, Crabp1, Rarres2, Moxd1) and a transcription factor (Lhx8). We also found the proto-oncogene cKit to be highly specific of parvalbumin interneurons in the non-human primate striatum, thus highlighting a conserved expression between species and suggesting that specific genes identified in mouse parvalbumin neurons could be putative targets in the human brain. Pharmacological stimulation of four G-protein-coupled receptors enriched in the striatal parvalbumin interneurons inhibited Gdnf expression presumably by decreasing cyclic adenosine monophosphate formation. Additional experiments with pharmacological modulators of adenylyl cyclase and protein kinase A indicated that this pathway is a relevant intracellular route to induce Gdnf gene activation. This preclinical study is an important step in the ongoing development of a specific pro-endo-glial cell line-derived neurotrophic factor pharmacological strategy to treat Parkinson's disease.

7.
Mov Disord ; 35(4): 565-576, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31930748

RESUMO

BACKGROUND: The glial cell line-derived neurotrophic factor has a potent neuroprotective action on mesencephalic dopamine neurons, which are progressively lost in Parkinson's disease. Intrastriatal administration of this factor is a promising therapy for Parkinson's disease. Glial cell line-derived neurotrophic factor is naturally produced in restricted cerebral regions, such as the striatum, septum, and thalamus; however, its effects in the adult brain remain under debate. OBJECTIVES: We sought to clarify the physiologic role of endogenous glial cell line-derived neurotrophic factor in the survival of catecholaminergic neurons of the substantia nigra pars compacta and the locus coeruleus in adult mice. METHODS: We used 2 new Cre recombinase-based mouse models to delete a floxed-glial cell line-derived neurotrophic factor gene. The first model had Cre expression in the parvalbumin expressing interneurons, as these cells represent the major source of striatal glial cell line-derived neurotrophic factor. The second model was an estrogen receptor 2-based inducible Cre triggered by tamoxifen at 2 months of age. RESULTS: We found that the floxed-glial cell line-derived neurotrophic factor gene was resilient to ablation by Cre-induced recombination and that parvalbumin-driven Cre was particularly inefficient to do so. The inducible-Cre model allowed an average 70% to 80% reduction in glial cell line-derived neurotrophic factor messenger ribonucleic acid and protein in striatum and septum with moderate significant loss of catecholamine neurons in the nigrostriatal pathway and, more markedly, in the locus coeruleus. This was accompanied with mild locomotor decline. CONCLUSIONS: Our data support qualitatively the view that brain glial cell line-derived neurotrophic factor is needed for the maintenance of adult central catecholaminergic neurons. © 2020 International Parkinson and Movement Disorder Society.


Assuntos
Fármacos Neuroprotetores , Doença de Parkinson , Animais , Corpo Estriado , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Camundongos , Neostriado , Neurônios , Substância Negra
8.
Cell Tissue Res ; 372(2): 417-425, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29368257

RESUMO

The carotid body (CB) is the major arterial chemoreceptor responsible for the detection of acute decreases in O2 tension (hypoxia) in arterial blood that trigger hyperventilation and sympathetic activation. The CB contains O2-sensitive glomus (chief) cells, which respond to hypoxia with the release of transmitters to activate sensory nerve fibers impinging upon the brain respiratory and autonomic centers. During exposure to sustained hypoxia (for weeks or months), the CB grows several-fold in size, a response associated with acclimatization to high altitude or to medical conditions presenting hypoxemia. Here, I briefly present recent advances on the mechanisms underlying glomus cell sensitivity to hypoxia, in particular the role of mitochondrial complex I in acute oxygen sensing. I also summarize the properties of adult CB stem cells and of glomus cell-stem cell synapses, which contribute to CB hypertrophy in chronic hypoxia. A note on the relationship between hypoxic CB growth and tumorigenesis is included. Finally, the medical implications of CB pathophysiology are discussed.


Assuntos
Corpo Carotídeo/metabolismo , Hipóxia/metabolismo , Oxigênio/metabolismo , Células-Tronco/metabolismo , Animais , Carcinogênese/metabolismo , Carcinogênese/patologia , Humanos
9.
EMBO Rep ; 19(3)2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29335248

RESUMO

Unlike other neural peripheral organs, the adult carotid body (CB) has a remarkable structural plasticity, as it grows during acclimatization to hypoxia. The CB contains neural stem cells that can differentiate into oxygen-sensitive glomus cells. However, an extended view is that, unlike other catecholaminergic cells of the same lineage (sympathetic neurons or chromaffin cells), glomus cells can divide and thus contribute to CB hypertrophy. Here, we show that O2-sensitive mature glomus cells are post-mitotic. However, we describe an unexpected population of pre-differentiated, immature neuroblasts that express catecholaminergic markers and contain voltage-dependent ion channels, but are unresponsive to hypoxia. Neuroblasts are quiescent in normoxic conditions, but rapidly proliferate and differentiate into mature glomus cells during hypoxia. This unprecedented "fast neurogenesis" is stimulated by ATP and acetylcholine released from mature glomus cells. CB neuroblasts, which may have evolved to facilitate acclimatization to hypoxia, could contribute to the CB oversensitivity observed in highly prevalent human diseases.


Assuntos
Adaptação Fisiológica/genética , Corpo Carotídeo/crescimento & desenvolvimento , Diferenciação Celular/genética , Hipóxia , Neurogênese/genética , Trifosfato de Adenosina/metabolismo , Corpo Carotídeo/metabolismo , Proliferação de Células/genética , Humanos , Hipóxia/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Oxigênio/metabolismo
10.
Endocr Relat Cancer ; 24(11): 579-591, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28928232

RESUMO

Mutations in genes encoding enzymes in the tricarboxylic acid cycle (TCA, also known as the Krebs cycle) have been implicated as causative genetic lesions in a number of human cancers, including renal cell cancers, glioblastomas and pheochromocytomas. In recent studies, missense mutations in the succinate dehydrogenase (SDH) complex have also been proposed to cause differentiated thyroid cancer. In order to gain mechanistic insight into this process, we generated mice lacking the SDH subunit D (Sdhd) in the thyroid. We report that these mice develop enlarged thyroid glands with follicle hypercellularity and increased proliferation. In vitro, human thyroid cell lines with knockdown of SDHD exhibit an enhanced migratory capability, despite no change in proliferative capacity. Interestingly, these cells acquire stem-like features which are also observed in the mouse tumors. The stem-like characteristics are reversed by α-ketoglutarate, suggesting that SDH-associated tumorigenesis results from dedifferentiation driven by an imbalance in cellular metabolites of the TCA cycle. The results of this study reveal a metabolic vulnerability for potential future treatment of SDH-associated neoplasia.


Assuntos
Complexo II de Transporte de Elétrons/genética , Proteínas de Membrana/genética , Neoplasias da Glândula Tireoide/patologia , Animais , Carcinogênese , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Metilação de DNA , Complexo II de Transporte de Elétrons/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Camundongos Transgênicos , Fenótipo , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Succinato Desidrogenase , Cicatrização
11.
PLoS One ; 11(10): e0164391, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27741271

RESUMO

Gender difference in Parkinson's disease (PD) suggests that female sex steroids may promote dopaminergic neuron survival and protect them from degeneration. The glial cell line-derived neurotrophic factor (GDNF) is believed to be dopaminotrophic; thus it is considered as a potential therapeutic target in PD. Additionally, GDNF is endogenously synthetized in the caudate/putamen of humans and striatum in rodents. A neuroprotective role of estrogens on the nigrostriatal pathway via the stimulation of GDNF has been proposed. Since the GDNF-producing parvalbumin (Parv) interneurons express the estrogen receptor alpha in the mouse striatum, we sought to determine whether ectopic estrogenic compound modulates the GDNF synthesis in mice. Using an ovariectomized-estradiol (E2) replacement regimen, which reliably generates a rise of plasma estradiol, we assessed the effects of different levels of E2 on the activation of striatal neuronal populations, and GDNF production. A strong correlation was found between plasma E2 and the expression of the immediate early gene cFos in the striatum, as well as in other cortical regions. However, moderate and high E2 treatments failed to induce any striatal GDNF mRNA and protein synthesis. High E2 only stimulates cFos induction in a low percentage of striatal Parv neurons whereas the majority of cFos-positive cells are medium spiny neurons. Activation of these projecting neurons by E2 suggests a role of circulating sex steroids in the modulation of striatal neural pathways.


Assuntos
Estradiol/sangue , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Animais , Corpo Estriado/metabolismo , Modelos Animais de Doenças , Neurônios Dopaminérgicos/metabolismo , Receptor alfa de Estrogênio/metabolismo , Feminino , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Imunoensaio , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ovariectomia , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Mensageiro/metabolismo
12.
J Physiol ; 594(24): 7229-7248, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27570189

RESUMO

KEY POINTS: Biotin, a vitamin whose main role is as a coenzyme for carboxylases, accumulates at unusually large amounts within cells of the carotid body (CB). In biotin-deficient rats biotin rapidly disappears from the blood; however, it remains at relatively high levels in CB glomus cells. The CB contains high levels of mRNA for SLC5a6, a biotin transporter, and SLC19a3, a thiamine transporter regulated by biotin. Animals with biotin deficiency exhibit pronounced metabolic lactic acidosis. Remarkably, glomus cells from these animals have normal electrical and neurochemical properties. However, they show a marked decrease in the size of quantal dopaminergic secretory events. Inhibitors of the vesicular monoamine transporter 2 (VMAT2) mimic the effect of biotin deficiency. In biotin-deficient animals, VMAT2 protein expression decreases in parallel with biotin depletion in CB cells. These data suggest that dopamine transport and/or storage in small secretory granules in glomus cells depend on biotin. ABSTRACT: Biotin is a water-soluble vitamin required for the function of carboxylases as well as for the regulation of gene expression. Here, we report that biotin accumulates in unusually large amounts in cells of arterial chemoreceptors, carotid body (CB) and adrenal medulla (AM). We show in a biotin-deficient rat model that the vitamin rapidly disappears from the blood and other tissues (including the AM), while remaining at relatively high levels in the CB. We have also observed that, in comparison with other peripheral neural tissues, CB cells contain high levels of SLC5a6, a biotin transporter, and SLC19a3, a thiamine transporter regulated by biotin. Biotin-deficient rats show a syndrome characterized by marked weight loss, metabolic lactic acidosis, aciduria and accelerated breathing with normal responsiveness to hypoxia. Remarkably, CB cells from biotin-deficient animals have normal electrophysiological and neurochemical (ATP levels and catecholamine synthesis) properties; however, they exhibit a marked decrease in the size of quantal catecholaminergic secretory events, which is not seen in AM cells. A similar differential secretory dysfunction is observed in CB cells treated with tetrabenazine, a selective inhibitor of the vesicular monoamine transporter 2 (VMAT2). VMAT2 is highly expressed in glomus cells (in comparison with VMAT1), and in biotin-deficient animals VMAT2 protein expression decreases in parallel with the decrease of biotin accumulated in CB cells. These data suggest that biotin has an essential role in the homeostasis of dopaminergic transmission modulating the transport and/or storage of transmitters within small secretory granules in glomus cells.


Assuntos
Biotina/metabolismo , Corpo Carotídeo/metabolismo , Dopamina/metabolismo , Trifosfato de Adenosina/metabolismo , Medula Suprarrenal/metabolismo , Animais , Artérias/metabolismo , Biotina/sangue , Biotina/deficiência , Células Cromafins/metabolismo , Exocitose , Hipóxia/fisiopatologia , Ácido Láctico/sangue , Ratos Wistar , Gânglio Cervical Superior/metabolismo , Tetrabenazina/farmacologia , Proteínas Vesiculares de Transporte de Monoamina/antagonistas & inibidores , Proteínas Vesiculares de Transporte de Monoamina/metabolismo
13.
Stem Cells ; 34(6): 1637-50, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26866353

RESUMO

Neural stem cells (NSCs) are promising tools for understanding nervous system plasticity and repair, but their use is hampered by the lack of markers suitable for their prospective isolation and characterization. The carotid body (CB) contains a population of peripheral NSCs, which support organ growth during acclimatization to hypoxia. We have set up CB neurosphere (NS) cultures enriched in differentiated neuronal (glomus) cells versus undifferentiated progenitors to investigate molecular hallmarks of cell classes within the CB stem cell (CBSC) niche. Microarray gene expression analysis in NS is compatible with CBSCs being neural crest derived-multipotent progenitor cells able to sustain CB growth upon exposure to hypoxia. Moreover, we have identified CD10 as a marker suitable for isolation of a population of CB mesectoderm-committed progenitor cells. CD10 + cells are resting in normoxia, and during hypoxia they are activated to proliferate and to eventually complete maturation into mesectodermal cells, thus participating in the angiogenesis necessary for CB growth. Our results shed light into the molecular and cellular mechanisms involved in CBSC fate choice, favoring a potential use of these cells for cell therapy. Stem Cells 2016;34:1637-1650.


Assuntos
Corpo Carotídeo/citologia , Linhagem da Célula , Ectoderma/citologia , Perfilação da Expressão Gênica , Mesoderma/citologia , Neprilisina/metabolismo , Crista Neural/citologia , Células-Tronco Neurais/citologia , Animais , Biomarcadores/metabolismo , Contagem de Células , Diferenciação Celular/genética , Hipóxia Celular/genética , Endotelina-1/metabolismo , Regulação da Expressão Gênica , Camundongos Transgênicos , Células-Tronco Neurais/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Ratos Wistar , Esferoides Celulares/citologia , Nicho de Células-Tronco/genética
14.
Cell Metab ; 22(5): 825-37, 2015 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-26437605

RESUMO

O2 sensing is essential for mammalian homeostasis. Peripheral chemoreceptors such as the carotid body (CB) contain cells with O2-sensitive K(+) channels, which are inhibited by hypoxia to trigger fast adaptive cardiorespiratory reflexes. How variations of O2 tension (PO2) are detected and the mechanisms whereby these changes are conveyed to membrane ion channels have remained elusive. We have studied acute O2 sensing in conditional knockout mice lacking mitochondrial complex I (MCI) genes. We inactivated Ndufs2, which encodes a protein that participates in ubiquinone binding. Ndufs2-null mice lose the hyperventilatory response to hypoxia, although they respond to hypercapnia. Ndufs2-deficient CB cells have normal functions and ATP content but are insensitive to changes in PO2. Our data suggest that chemoreceptor cells have a specialized succinate-dependent metabolism that induces an MCI state during hypoxia, characterized by the production of reactive oxygen species and accumulation of reduced pyridine nucleotides, which signal neighboring K(+) channels.


Assuntos
Células Quimiorreceptoras/metabolismo , Complexo I de Transporte de Elétrons/metabolismo , Mitocôndrias/metabolismo , NADH Desidrogenase/genética , Oxigênio/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Corpo Carotídeo/citologia , Corpo Carotídeo/metabolismo , Hipóxia Celular , Homeostase , Camundongos , Camundongos Knockout , NADH Desidrogenase/metabolismo , Canais de Potássio/metabolismo , Transdução de Sinais
15.
EMBO Mol Med ; 6(12): 1577-92, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25385837

RESUMO

Mutations of the von Hippel-Lindau (VHL) gene are associated with pheochromocytomas and paragangliomas, but the role of VHL in sympathoadrenal homeostasis is unknown. We generated mice lacking Vhl in catecholaminergic cells. They exhibited atrophy of the carotid body (CB), adrenal medulla, and sympathetic ganglia. Vhl-null animals had an increased number of adult CB stem cells, although the survival of newly generated neuron-like glomus cells was severely compromised. The effects of Vhl deficiency were neither prevented by pharmacological inhibition of prolyl hydroxylases or selective genetic down-regulation of prolyl hydroxylase-3, nor phenocopied by hypoxia inducible factor overexpression. Vhl-deficient animals appeared normal in normoxia but survived for only a few days in hypoxia, presenting with pronounced erythrocytosis, pulmonary edema, and right cardiac hypertrophy. Therefore, in the normal sympathoadrenal setting, Vhl deletion does not give rise to tumors but impairs development and plasticity of the peripheral O2-sensing system required for survival in hypoxic conditions.


Assuntos
Neoplasias das Glândulas Suprarrenais/metabolismo , Apoptose , Células Quimiorreceptoras/metabolismo , Deleção de Genes , Hipóxia/metabolismo , Feocromocitoma/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Neoplasias das Glândulas Suprarrenais/genética , Neoplasias das Glândulas Suprarrenais/fisiopatologia , Medula Suprarrenal/citologia , Medula Suprarrenal/metabolismo , Animais , Morte Celular , Células Quimiorreceptoras/citologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Hipóxia/genética , Hipóxia/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Oxigênio/metabolismo , Feocromocitoma/genética , Feocromocitoma/fisiopatologia , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo
16.
PLoS One ; 9(1): e85528, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24465590

RESUMO

Mutations in mitochondrial complex II (MCII; succinate dehydrogenase, Sdh) genes cause familiar pheochromocytoma/paraganglioma tumors. Several mechanisms have been proposed to account for Sdh-mutation-induced tumorigenesis, the most accepted of which is based on the constitutive expression of the hypoxia-inducible factor 1α (Hif1α) at normal oxygen tension, a theory referred to as "pseudo-hypoxic drive". Other molecular processes, such as oxidative stress, apoptosis, or chromatin remodeling have been also proposed to play a causative role. Nevertheless, the actual contribution of each of these mechanisms has not been definitively established. Moreover, the biological factors that determine the tissue-specificity of these tumors have not been identified. In this work, we made use of the inducible SDHD-ESR mouse, a conditional mutant in the SdhD gene, which encodes the small subunit of MCII, and that acts as a tumor suppressor gene in humans. The analysis of the Hif1α pathway in SDHD-ESR tissues and in two newly derived cell lines after complete SdhD loss -a requirement for hereditary paraganglioma type-1 tumor formation in humans- partially recapitulated the "pseudo-hypoxic" response and rendered inconsistent results. Therefore, we performed microarray analysis of adrenal medulla and kidney in order to identify other early gene expression changes elicited by SdhD deletion. Our results revealed that each mutant tissue displayed different variations in their gene expression profiles affecting to different biological processes. However, we found that the Cdkn1a gene was up-regulated in both tissues. This gene encodes the cyclin-dependent kinase inhibitor p21(WAF1/Cip1), a factor implicated in cell cycle, senescence, and cancer. The two SDHD-ESR cell lines also showed accumulation of this protein. This new and unprecedented evidence for a link between SdhD dysfunction and p21(WAF1/Cip1) will open new avenues for the study of the mechanisms that cause tumors in Sdh mutants. Finally, we discuss the actual role of Hif1α in tumorigenesis.


Assuntos
Carcinogênese/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Complexo II de Transporte de Elétrons/genética , Proteínas de Membrana/genética , Mitocôndrias/genética , Neoplasias das Glândulas Suprarrenais/genética , Neoplasias das Glândulas Suprarrenais/metabolismo , Neoplasias das Glândulas Suprarrenais/patologia , Glândulas Suprarrenais/metabolismo , Glândulas Suprarrenais/patologia , Animais , Carcinogênese/metabolismo , Carcinogênese/patologia , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Complexo II de Transporte de Elétrons/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Rim/metabolismo , Rim/patologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Mutação , Paraganglioma/genética , Paraganglioma/metabolismo , Paraganglioma/patologia , Feocromocitoma/genética , Feocromocitoma/metabolismo , Feocromocitoma/patologia , Succinato Desidrogenase , Regulação para Cima
17.
Mol Cell Biol ; 32(16): 3347-57, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22711987

RESUMO

The SDHD gene (subunit D of succinate dehydrogenase) has been shown to be involved in the generation of paragangliomas and pheochromocytomas. Loss of heterozygosity of the normal allele is necessary for tumor transformation of the affected cells. As complete SdhD deletion is lethal, we have generated mouse models carrying a "floxed" SdhD allele and either an inducible (SDHD-ESR strain) or a catecholaminergic tissue-specific (TH-SDHD strain) CRE recombinase. Ablation of both SdhD alleles in adult SDHD-ESR mice did not result in generation of paragangliomas or pheochromocytomas. In contrast, carotid bodies from these animals showed smaller volume than controls. In accord with these observations, the TH-SDHD mice had decreased cell numbers in the adrenal medulla, carotid body, and superior cervical ganglion. They also manifested inhibited postnatal maturation of mesencephalic dopaminergic neurons and progressive cell loss during the first year of life. These alterations were particularly intense in the substantia nigra, the most affected neuronal population in Parkinson's disease. Unexpectedly, TH(+) neurons in the locus coeruleus and group A13, also lacking the SdhD gene, were unaltered. These data indicate that complete loss of SdhD is not sufficient to induce tumorigenesis in mice. They suggest that substantia nigra neurons are more susceptible to mitochondrial damage than other catecholaminergic cells, particularly during a critical postnatal maturation period.


Assuntos
Complexo II de Transporte de Elétrons/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Trifosfato de Adenosina/metabolismo , Alelos , Animais , Catecolaminas/metabolismo , Morte Celular , DNA Mitocondrial/metabolismo , Modelos Animais de Doenças , Complexo II de Transporte de Elétrons/genética , Complexo II de Transporte de Elétrons/fisiologia , Genótipo , Camundongos , Camundongos Transgênicos , Microscopia Confocal/métodos , Mitocôndrias/metabolismo , Modelos Genéticos , Neurônios/metabolismo , Oxigênio/química , RNA Mensageiro/metabolismo , Succinato Desidrogenase
18.
Adv Exp Med Biol ; 741: 206-16, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22457112

RESUMO

Parkinson's disease (PD), one of the most frequent neurodegenerative disorders, is primarily caused by the selective degeneration of specific neuronal populations, particularly dopaminergic neurons within the substantia nigra projecting to the striatum (nigrostriatal neurons). The current pharmacological treatments are efficient in the early stage of the disease but with the continuous use of the pro-dopaminergic medication may become less effective and cause motor complications. Cell therapy is an emergent alternative therapeutic strategy to PD and consists in the replacement of damaged neurons by new cells that could help to restore the nigrostriatal pathway. For this purpose, stem cells constitute a promising tool that could provide new sources of cells to be used for experimental transplantation studies in PD, as well as in other neurological disorders. Numerous studies are being made with the use of tissue specific neural progenitors obtained from either fetal or adult nervous systems. In this chapter we will summarize the numerous preclinical and clinical studies performed so far in animal models of the disease as well as in patients and how the use of neural stem cells might improve the current cell therapies.


Assuntos
Células-Tronco Neurais/fisiologia , Doença de Parkinson/cirurgia , Transplante de Células-Tronco , Adulto , Animais , Diferenciação Celular , Modelos Animais de Doenças , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/fisiologia , Humanos , Sistema Nervoso/anatomia & histologia , Células-Tronco Neurais/citologia , Doença de Parkinson/patologia , Nicho de Células-Tronco
19.
J Biol Chem ; 287(12): 9651-8, 2012 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-22308030

RESUMO

Early adaptive responses to hypoxia are essential for cell survival, but their nature and underlying mechanisms are poorly known. We have studied the post-transcriptional changes in the proteome of mammalian cells elicited by acute hypoxia and found that phosphorylation of eukaryotic elongation factor 2 (eEF2), a ribosomal translocase whose phosphorylation inhibits protein synthesis, is under the precise and reversible control of O(2) tension. Upon exposure to hypoxia, phosphorylation of eEF2 at Thr(56) occurred rapidly (<15 min) and resulted in modest translational arrest, a fundamental homeostatic response to hypoxia that spares ATP and thus facilitates cell survival. Acute inhibitory eEF2 phosphorylation occurred without ATP depletion or AMP kinase activation. Furthermore, eEF2 phosphorylation was mimicked by prolyl hydroxylase (PHD) inhibition with dimethyloxalylglycine or by selective PHD2 siRNA silencing but was independent of hypoxia-inducible factor α stabilization. Moreover, overexpression of PHD2 blocked hypoxic accumulation of phosphorylated eEF2. Therefore, our findings suggest that eEF2 phosphorylation status (and, as a consequence, translation rate) is controlled by PHD2 activity. They unravel a novel pathway for cell adaptation to hypoxia that could have pathophysiologic relevance in tissue ischemia and cancer.


Assuntos
Regulação Enzimológica da Expressão Gênica , Hipóxia/enzimologia , Hipóxia/genética , Fator 2 de Elongação de Peptídeos/genética , Fator 2 de Elongação de Peptídeos/metabolismo , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Biossíntese de Proteínas , Trifosfato de Adenosina/metabolismo , Linhagem Celular , Humanos , Hipóxia/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia , Pró-Colágeno-Prolina Dioxigenase/genética
20.
Cardiovasc Res ; 93(4): 702-10, 2012 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-22215723

RESUMO

AIMS: Alveolar hypoxia acutely elicits contraction of pulmonary arteries, leading to a rise in pulmonary arterial pressure (PAP) and shifting blood to better ventilated areas of the lung. The molecular mechanisms underlying this hypoxic pulmonary vasoconstriction (HPV) are still incompletely understood. Here, we investigated the role of succinate dehydrogenase (SDH; synonymous to mitochondrial complex II) in HPV, with particular emphasis on regional differences along the vascular bed and consequences for PAP and perfusion-to-ventilation matching, using mutant mice heterozygous for the SDHD subunit of complex II (SDHD(+/-)). METHODS AND RESULTS: Western blots revealed reduced protein content of complex II subunits SDHA, SDHB, and SDHC in lungs of SDHD(+/-) mice, despite unaffected mRNA content as determined by real-time PCR. Hypoxic pulmonary vasoconstriction of small (20-50 µm) intra-acinar and larger (51-100 µm) pre-acinar arteries was evaluated by videomorphometric analysis of precision-cut lung slices. The hypoxic response was detectable in pre-acinar arteries but absent from intra-acinar arteries of SDHD(+/-) mice. In isolated perfused lungs, basal PAP and its hypoxia-induced increase were indistinguishable between both mouse strains. Arterial oxygenation was measured after provocation of regional ventilatory failure by tracheal fluid instillation in anaesthetized mice, and it declined more in SDHD(+/-) than in wild-type mice. CONCLUSION: SDHD is required for the formation of a stable mitochondrial complex II and it is selectively important for HPV of intra-acinar vessels. This specialized vascular segment participates in perfusion-to-ventilation matching but does not significantly contribute to the acute hypoxic rise in PAP that results from more proximal vasoconstriction.


Assuntos
Hipóxia/fisiopatologia , Pulmão/irrigação sanguínea , Artéria Pulmonar/fisiopatologia , Succinato Desidrogenase/fisiologia , Vasoconstrição/fisiologia , Animais , Pressão Sanguínea/fisiologia , Complexo II de Transporte de Elétrons/genética , Complexo II de Transporte de Elétrons/fisiologia , Heterozigoto , Pulmão/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Mutantes , Modelos Animais , RNA Mensageiro/metabolismo , Succinato Desidrogenase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA