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











Base de dados
Intervalo de ano de publicação
1.
Sci Adv ; 10(36): eadn9361, 2024 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-39231216

RESUMO

Kv1.3 is a multifunctional potassium channel implicated in multiple pathologies, including cancer. However, how it is involved in disease progression is not fully clear. We interrogated the interactome of Kv1.3 in intact cells using BioID proximity labeling, revealing that Kv1.3 interacts with STAT3- and p53-linked pathways. To prove the relevance of Kv1.3 and of its interactome in the context of tumorigenesis, we generated stable melanoma clones, in which ablation of Kv1.3 remodeled gene expression, reduced proliferation and colony formation, yielded fourfold smaller tumors, and decreased metastasis in vivo in comparison to WT cells. Kv1.3 deletion or pharmacological inhibition of mitochondrial Kv1.3 increased mitochondrial Reactive Oxygen Species release, decreased STAT3 phosphorylation, stabilized the p53 tumor suppressor, promoted metabolic switch, and altered the expression of several BioID-identified Kv1.3-networking proteins in tumor tissues. Collectively, our work revealed the tumor-promoting Kv1.3-interactome landscape, thus opening the way to target Kv1.3 not only as an ion-conducting entity but also as a signaling hub.


Assuntos
Canal de Potássio Kv1.3 , Fator de Transcrição STAT3 , Transdução de Sinais , Proteína Supressora de Tumor p53 , Canal de Potássio Kv1.3/metabolismo , Canal de Potássio Kv1.3/genética , Proteína Supressora de Tumor p53/metabolismo , Fator de Transcrição STAT3/metabolismo , Humanos , Animais , Camundongos , Linhagem Celular Tumoral , Melanoma/metabolismo , Melanoma/patologia , Melanoma/genética , Mitocôndrias/metabolismo , Proliferação de Células , Espécies Reativas de Oxigênio/metabolismo
2.
Cell Death Discov ; 10(1): 299, 2024 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-38909013

RESUMO

Ion channels are critical in enabling ion movement into and within cells and are important targets for pharmacological interventions in different human diseases. In addition to their ion transport abilities, ion channels interact with signalling and scaffolding proteins, which affects their function, cellular positioning, and links to intracellular signalling pathways. The study of "channelosomes" within cells has the potential to uncover their involvement in human diseases, although this field of research is still emerging. LRRC8A is the gene that encodes a crucial protein involved in the formation of volume-regulated anion channels (VRACs). Some studies suggest that LRRC8A could be a valuable prognostic tool in different types of cancer, serving as a biomarker for predicting patients' outcomes. LRRC8A expression levels might be linked to tumour progression, metastasis, and treatment response, although its implications in different cancer types can be varied. Here, publicly accessible databases of cancer patients were systematically analysed to determine if a correlation between VRAC channel expression and survival rate exists across distinct cancer types. Moreover, we re-evaluated the impact of LRRC8A on cellular proliferation and migration in colon cancer via HCT116 LRRC8A-KO cells, which is a current topic of debate in the literature. In addition, to investigate the role of LRRC8A in cellular signalling, we conducted biotin proximity-dependent identification (BioID) analysis, revealing a correlation between VRAC channels and cell-cell junctions, mechanisms that govern cellular calcium homeostasis, kinases, and GTPase signalling. Overall, this dataset improves our understanding of LRRC8A/VRAC and explores new research avenues while identifying promising therapeutic targets and promoting inventive methods for disease treatment.

3.
Cell Death Dis ; 14(10): 704, 2023 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-37898636

RESUMO

Skin cutaneous melanoma (SKCM) is the deadliest form of skin cancer due to its high heterogeneity that drives tumor aggressiveness. Melanoma plasticity consists of two distinct phenotypic states that co-exist in the tumor niche, the proliferative and the invasive, respectively associated with a high and low expression of MITF, the master regulator of melanocyte lineage. However, despite efforts, melanoma research is still far from exhaustively dissecting this phenomenon. Here, we discovered a key function of Transglutaminase Type-2 (TG2) in regulating melanogenesis by modulating MITF transcription factor expression and its transcriptional activity. Importantly, we demonstrated that TG2 expression affects melanoma invasiveness, highlighting its positive value in SKCM. These results suggest that TG2 may have implications in the regulation of the phenotype switching by promoting melanoma differentiation and impairing its metastatic potential. Our findings offer potential perspectives to unravel melanoma vulnerabilities via tuning intra-tumor heterogeneity.


Assuntos
Melanoma , Neoplasias Cutâneas , Humanos , Melanoma/patologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Transglutaminases/genética , Transglutaminases/metabolismo , Regulação Neoplásica da Expressão Gênica , Melanócitos/metabolismo , Fenótipo , Fator de Transcrição Associado à Microftalmia/genética , Linhagem Celular Tumoral , Melanoma Maligno Cutâneo
4.
Cell Death Dis ; 13(12): 1055, 2022 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-36539400

RESUMO

Ion channels are non-conventional, druggable oncological targets. The intermediate-conductance calcium-dependent potassium channel (KCa3.1) is highly expressed in the plasma membrane and in the inner mitochondrial membrane (mitoKCa3.1) of various cancer cell lines. The role mitoKCa3.1 plays in cancer cells is still undefined. Here we report the synthesis and characterization of two mitochondria-targeted novel derivatives of a high-affinity KCa3.1 antagonist, TRAM-34, which retain the ability to block channel activity. The effects of these drugs were tested in melanoma, pancreatic ductal adenocarcinoma and breast cancer lines, as well as in vivo in two orthotopic models. We show that the mitochondria-targeted TRAM-34 derivatives induce release of mitochondrial reactive oxygen species, rapid depolarization of the mitochondrial membrane, fragmentation of the mitochondrial network. They trigger cancer cell death with an EC50 in the µM range, depending on channel expression. In contrast, inhibition of the plasma membrane KCa3.1 by membrane-impermeant Maurotoxin is without effect, indicating a specific role of mitoKCa3.1 in determining cell fate. At sub-lethal concentrations, pharmacological targeting of mitoKCa3.1 significantly reduced cancer cell migration by enhancing production of mitochondrial reactive oxygen species and nuclear factor-κB (NF-κB) activation, and by downregulating expression of Bcl-2 Nineteen kD-Interacting Protein (BNIP-3) and of Rho GTPase CDC-42. This signaling cascade finally leads to cytoskeletal reorganization and impaired migration. Overexpression of BNIP-3 or pharmacological modulation of NF-κB and CDC-42 prevented the migration-reducing effect of mitoTRAM-34. In orthotopic models of melanoma and pancreatic ductal adenocarcinoma, the tumors at sacrifice were 60% smaller in treated versus untreated animals. Metastasis of melanoma cells to lymph nodes was also drastically reduced. No signs of toxicity were observed. In summary, our results identify mitochondrial KCa3.1 as an unexpected player in cancer cell migration and show that its pharmacological targeting is efficient against both tumor growth and metastatic spread in vivo.


Assuntos
Carcinoma Ductal Pancreático , Melanoma , Neoplasias Pancreáticas , Canais de Potássio Cálcio-Ativados , Animais , NF-kappa B/metabolismo , Cálcio/metabolismo , Canais de Cálcio , Canais de Potássio , Espécies Reativas de Oxigênio/metabolismo , Morte Celular , Mitocôndrias/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/genética , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Neoplasias Pancreáticas
5.
Biomolecules ; 11(8)2021 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-34439838

RESUMO

Potassium ions can cross both the outer and inner mitochondrial membranes by means of multiple routes. A few potassium-permeable ion channels exist in the outer membrane, while in the inner membrane, a multitude of different potassium-selective and potassium-permeable channels mediate K+ uptake into energized mitochondria. In contrast, potassium is exported from the matrix thanks to an H+/K+ exchanger whose molecular identity is still debated. Among the K+ channels of the inner mitochondrial membrane, the most widely studied is the ATP-dependent potassium channel, whose pharmacological activation protects cells against ischemic damage and neuronal injury. In this review, we briefly summarize and compare the different hypotheses regarding the molecular identity of this patho-physiologically relevant channel, taking into account the electrophysiological characteristics of the proposed components. In addition, we discuss the characteristics of the other channels sharing localization to both the plasma membrane and mitochondria.


Assuntos
Trifosfato de Adenosina/metabolismo , Mitocôndrias/metabolismo , Membranas Mitocondriais/metabolismo , Canais de Potássio/metabolismo , Potássio/metabolismo , Animais , Humanos
6.
Pharmacol Ther ; 227: 107874, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33930454

RESUMO

The field of mitochondrial ion channels underwent a rapid development during the last decade, thanks to the molecular identification of some of the nuclear-encoded organelle channels and to advances in strategies allowing specific pharmacological targeting of these proteins. Thereby, genetic tools and specific drugs aided definition of the relevance of several mitochondrial channels both in physiological as well as pathological conditions. Unfortunately, in the case of mitochondrial K+ channels, efforts of genetic manipulation provided only limited results, due to their dual localization to mitochondria and to plasma membrane in most cases. Although the impact of mitochondrial K+ channels on human diseases is still far from being genuinely understood, pre-clinical data strongly argue for their substantial role in the context of several pathologies, including cardiovascular and neurodegenerative diseases as well as cancer. Importantly, these channels are druggable targets, and their in-depth investigation could thus pave the way to the development of innovative small molecules with huge therapeutic potential. In the present review we summarize the available experimental evidence that mechanistically link mitochondrial potassium channels to the above pathologies and underline the possibility of exploiting them for therapy.


Assuntos
Mitocôndrias , Canais de Potássio , Tratamento Farmacológico , Humanos , Mitocôndrias/metabolismo , Canais de Potássio/farmacologia
7.
Int J Mol Sci ; 22(9)2021 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-33924772

RESUMO

Wnt signaling is an important pathway mainly active during embryonic development and controlling cell proliferation. This regulatory pathway is aberrantly activated in several human diseases. Ion channels are known modulators of several important cellular functions ranging from the tuning of the membrane potential to modulation of intracellular pathways, in particular the influence of ion channels in Wnt signaling regulation has been widely investigated. This review will discuss the known links between ion channels and canonical Wnt signaling, focusing on their possible roles in human metabolic diseases, neurological disorders, and cancer.


Assuntos
Doença/etiologia , Canais Iônicos/metabolismo , Via de Sinalização Wnt , Animais , Humanos
8.
Pharmaceuticals (Basel) ; 14(2)2021 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-33562146

RESUMO

A developing family of chemotherapeutics-derived from 5-(4-phenoxybutoxy)psoralen (PAP-1)-target mitochondrial potassium channel mtKv1.3 to selectively induce oxidative stress and death of diseased cells. The key to their effectiveness is the presence of a positively charged triphenylphosphonium group which drives their accumulation in the organelles. These compounds have proven their preclinical worth in murine models of cancers such as melanoma and pancreatic adenocarcinoma. In in vitro experiments they also efficiently killed glioblastoma cells, but in vivo they were powerless against orthotopic glioma because they were completely unable to overcome the blood-brain barrier. In an effort to improve brain delivery we have now coupled one of these promising compounds, PAPTP, to well-known cell-penetrating and brain-targeting peptides TAT48-61 and Angiopep-2. Coupling has been obtained by linking one of the phenyl groups of the triphenylphosphonium to the first amino acid of the peptide via a reversible carbamate ester bond. Both TAT48-61 and Angiopep-2 allowed the delivery of 0.3-0.4 nmoles of construct per gram of brain tissue upon intravenous (i.v.) injection of 5 µmoles/kg bw to mice. This is the first evidence of PAPTP delivery to the brain; the chemical strategy described here opens the possibility to conjugate PAPTP to small peptides in order to fine-tune tissue distribution of this interesting compound.

9.
Front Physiol ; 12: 784867, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35082690

RESUMO

Voltage-dependent anion-selective channels (VDAC) are pore-forming proteins located in the outer mitochondrial membrane. Three isoforms are encoded by separate genes in mammals (VDAC1-3). These proteins play a crucial role in the cell, forming the primary interface between mitochondrial and cellular metabolisms. Research on the role of VDACs in the cell is a rapidly growing field, but the function of VDAC3 remains elusive. The high-sequence similarity between isoforms suggests a similar pore-forming structure. Electrophysiological analyzes revealed that VDAC3 works as a channel; however, its gating and regulation remain debated. A comparison between VDAC3 and VDAC1-2 underlines the presence of a higher number of cysteines in both isoforms 2 and 3. Recent mass spectrometry data demonstrated that the redox state of VDAC3 cysteines is evolutionarily conserved. Accordingly, these residues were always detected as totally reduced or partially oxidized, thus susceptible to disulfide exchange. The deletion of selected cysteines significantly influences the function of the channel. Some cysteine mutants of VDAC3 exhibited distinct kinetic behavior, conductance values and voltage dependence, suggesting that channel activity can be modulated by cysteine reduction/oxidation. These properties point to VDAC3 as a possible marker of redox signaling in the mitochondrial intermembrane space. Here, we summarize our current knowledge about VDAC3 predicted structure, physiological role and regulation, and possible future directions in this research field.

10.
Front Cell Dev Biol ; 8: 620081, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33585458

RESUMO

Mitochondria are bioenergetic organelles with a plethora of fundamental functions ranging from metabolism and ATP production to modulation of signaling events leading to cell survival or cell death. Ion channels located in the outer and inner mitochondrial membranes critically control mitochondrial function and, as a consequence, also cell fate. Opening or closure of mitochondrial ion channels allow the fine-tuning of mitochondrial membrane potential, ROS production, and function of the respiratory chain complexes. In this review, we critically discuss the intracellular regulatory factors that affect channel activity in the inner membrane of mitochondria and, indirectly, contribute to cell death. These factors include various ligands, kinases, second messengers, and lipids. Comprehension of mitochondrial ion channels regulation in cell death pathways might reveal new therapeutic targets in mitochondria-linked pathologies like cancer, ischemia, reperfusion injury, and neurological disorders.

11.
Cell Physiol Biochem ; 53(S1): 52-62, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31854954

RESUMO

Kv1.3 is a voltage gated potassium channel located in the plasma membrane, as well as at intracellular levels, such as mitochondria (mitoKv1.3), nucleus and Golgi apparatus. The plasma membrane channel has been shown to be important for cell proliferation, while the mitochondrial counterpart has been related to modulation of cell death. Moreover, altered expression of Kv1.3 was observed in various tumors and Kv1.3 seems to be involved in development and progression of various cancerous forms. Recent experimental evidences have proved that pharmacological inhibition of the mitoKv1.3 succeeded in reducing up to 90% of tumor volume in vivo in orthotopic mouse model. Furthermore, mitoKv1.3 modulation could impact on cell proliferation as well as on regulation of intracellular signaling pathways. Indeed, the treatment with sub-lethal doses of mitoKv1.3 inhibitors can downregulate Wnt-ß catenin signaling by reducing mitochondrial ATP production and triggering ER-stress. In this review, we describe the role of the mitoKv1.3 in cell death, cancer and intracellular signaling. We will discuss how pharmacological modulation of mitochondrial potassium fluxes impact on mitochondrial membrane potential, reactive oxygen species production and ATP synthesis. All these changes in mitochondrial fitness are related to cell proliferation as well as to cell death and finally on cancer development and progression, so Kv1.3 (and mitoKv1.3) could be now considered a new oncological target.


Assuntos
Canal de Potássio Kv1.3/metabolismo , Mitocôndrias/metabolismo , Neoplasias/metabolismo , Animais , Apoptose , Proliferação de Células , Estresse do Retículo Endoplasmático , Canal de Potássio Kv1.3/antagonistas & inibidores , Canal de Potássio Kv1.3/genética , Neoplasias/patologia , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
12.
SLAS Discov ; 24(9): 882-892, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31373829

RESUMO

Cancer is the consequence of aberrations in cell growth or cell death. In this scenario, mitochondria and ion channels play a critical role in regard to cell proliferation, malignant angiogenesis, migration, and metastasis. In this review, we focus on Kv1.3 and specifically on mitoKv1.3, which showed an aberrant expression in cancer cells compared with healthy tissues and which is involved in the apoptotic pathway. In recent years, mitoKv1.3 has become an oncological target since its pharmacological modulation has been demonstrated to reduce tumor growth and progression both in vitro and in vivo using preclinical mouse models of different types of tumors.


Assuntos
Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Canal de Potássio Kv1.3/metabolismo , Mitocôndrias/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Humanos , Mitocôndrias/metabolismo , Neoplasias/metabolismo , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo
13.
Nature ; 572(7771): 609-613, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31435016

RESUMO

Mitochondria provide chemical energy for endoergonic reactions in the form of ATP, and their activity must meet cellular energy requirements, but the mechanisms that link organelle performance to ATP levels are poorly understood. Here we confirm the existence of a protein complex localized in mitochondria that mediates ATP-dependent potassium currents (that is, mitoKATP). We show that-similar to their plasma membrane counterparts-mitoKATP channels are composed of pore-forming and ATP-binding subunits, which we term MITOK and MITOSUR, respectively. In vitro reconstitution of MITOK together with MITOSUR recapitulates the main properties of mitoKATP. Overexpression of MITOK triggers marked organelle swelling, whereas the genetic ablation of this subunit causes instability in the mitochondrial membrane potential, widening of the intracristal space and decreased oxidative phosphorylation. In a mouse model, the loss of MITOK suppresses the cardioprotection that is elicited by pharmacological preconditioning induced by diazoxide. Our results indicate that mitoKATP channels respond to the cellular energetic status by regulating organelle volume and function, and thereby have a key role in mitochondrial physiology and potential effects on several pathological processes.


Assuntos
Trifosfato de Adenosina/metabolismo , Mitocôndrias Cardíacas/metabolismo , Canais de Potássio/metabolismo , Animais , Cardiotônicos/farmacologia , Diazóxido/farmacologia , Fenômenos Eletrofisiológicos , Coração/efeitos dos fármacos , Coração/fisiologia , Precondicionamento Isquêmico Miocárdico , Masculino , Potencial da Membrana Mitocondrial , Camundongos , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/patologia , Mitocôndrias Cardíacas/fisiologia , Tamanho do Órgão/efeitos dos fármacos , Fosforilação Oxidativa , Potássio/metabolismo , Canais de Potássio/química , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo
14.
Br J Pharmacol ; 176(22): 4258-4283, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30440086

RESUMO

The field of mitochondrial ion channels has undergone a rapid development during the last three decades, due to the molecular identification of some of the channels residing in the outer and inner membranes. Relevant information about the function of these channels in physiological and pathological settings was gained thanks to genetic models for a few, mitochondria-specific channels. However, many ion channels have multiple localizations within the cell, hampering a clear-cut determination of their function by pharmacological means. The present review summarizes our current knowledge about the ins and outs of mitochondrial ion channels, with special focus on the channels that have received much attention in recent years, namely, the voltage-dependent anion channels, the permeability transition pore (also called mitochondrial megachannel), the mitochondrial calcium uniporter and some of the inner membrane-located potassium channels. In addition, possible strategies to overcome the difficulties of specifically targeting mitochondrial channels versus their counterparts active in other membranes are discussed, as well as the possibilities of modulating channel function by small peptides that compete for binding with protein interacting partners. Altogether, these promising tools along with large-scale chemical screenings set up to identify new, specific channel modulators will hopefully allow us to pinpoint the actual function of most mitochondrial ion channels in the near future and to pharmacologically affect important pathologies in which they are involved, such as neurodegeneration, ischaemic damage and cancer. LINKED ARTICLES: This article is part of a themed section on Mitochondrial Pharmacology: Featured Mechanisms and Approaches for Therapy Translation. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.22/issuetoc.


Assuntos
Canais Iônicos/fisiologia , Moduladores de Transporte de Membrana/farmacologia , Mitocôndrias/efeitos dos fármacos , Animais , Humanos , Mitocôndrias/fisiologia
15.
Int J Mol Sci ; 19(7)2018 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-30011966

RESUMO

In recent years, several experimental evidences have underlined a new role of ion channels in cancer development and progression. In particular, mitochondrial ion channels are arising as new oncological targets, since it has been proved that most of them show an altered expression during tumor development and the pharmacological targeting of some of them have been demonstrated to be able to modulate cancer growth and progression, both in vitro as well as in vivo in pre-clinical mouse models. In this scenario, pharmacology of mitochondrial ion channels would be in the near future a new frontier for the treatment of tumors. In this review, we discuss the new advances in the field, by focusing our attention on the improvements in new drug developments to target mitochondrial ion channels.


Assuntos
Antineoplásicos/uso terapêutico , Canais Iônicos/antagonistas & inibidores , Mitocôndrias/metabolismo , Proteínas Mitocondriais/antagonistas & inibidores , Terapia de Alvo Molecular/métodos , Neoplasias/tratamento farmacológico , Animais , Progressão da Doença , Humanos , Canais Iônicos/metabolismo , Proteínas Mitocondriais/metabolismo , Modelos Biológicos , Terapia de Alvo Molecular/tendências , Neoplasias/metabolismo , Neoplasias/patologia
16.
Biochem Biophys Res Commun ; 500(1): 51-58, 2018 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-28642134

RESUMO

Mitochondria are intracellular organelles involved in several processes from bioenergetics to cell death. In the latest years, ion channels are arising as new possible targets in controlling several cellular functions. The discovery that several plasma membrane located ion channels have intracellular counterparts, has now implemented this consideration and the number of studies enforcing the understanding of their role in different metabolic pathways. In this review, we will discuss the recent updates in the field, focusing our attention on the involvement of potassium channels during mitochondrial mediated apoptotic cell death. Since mitochondria are one of the key organelles involved in this process, it is not surprising that potassium channels located in their inner membrane could be involved in modulating mitochondrial membrane potential, ROS production, and respiratory chain complexes functions. Eventually, these events lead to changes in the mitochondrial fitness that prelude to the cytochrome c release and apoptosis. In this scenario, both the inhibition and the activation of mitochondrial potassium channels could cause cell death, and their targeting could be a novel pharmacological way to treat different human diseases.


Assuntos
Apoptose/genética , Mitocôndrias/metabolismo , Membranas Mitocondriais/metabolismo , Neoplasias/metabolismo , Canais de Potássio/metabolismo , Animais , Citocromos c/metabolismo , Metabolismo Energético/genética , Regulação da Expressão Gênica , Humanos , Transporte de Íons , Potencial da Membrana Mitocondrial/fisiologia , Mitocôndrias/ultraestrutura , Membranas Mitocondriais/ultraestrutura , Neoplasias/genética , Neoplasias/patologia , Canais de Potássio/classificação , Canais de Potássio/genética , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
17.
Oncotarget ; 7(3): 2249-68, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26760765

RESUMO

Voltage-Dependent Anion selective Channels (VDAC) are pore-forming mitochondrial outer membrane proteins. In mammals VDAC3, the least characterized isoform, presents a set of cysteines predicted to be exposed toward the intermembrane space. We find that cysteines in VDAC3 can stay in different oxidation states. This was preliminary observed when, in our experimental conditions, completely lacking any reducing agent, VDAC3 presented a pattern of slightly different electrophoretic mobilities. This observation holds true both for rat liver mitochondrial VDAC3 and for recombinant and refolded human VDAC3. Mass spectroscopy revealed that cysteines 2 and 8 can form a disulfide bridge in native VDAC3. Single or combined site-directed mutagenesis of cysteines 2, 8 and 122 showed that the protein mobility in SDS-PAGE is influenced by the presence of cysteine and by the redox status. In addition, cysteines 2, 8 and 122 are involved in the stability control of the pore as shown by electrophysiology, complementation assays and chemico-physical characterization. Furthermore, a positive correlation between the pore conductance of the mutants and their ability to complement the growth of porin-less yeast mutant cells was found. Our work provides evidence for a complex oxidation pattern of a mitochondrial protein not directly involved in electron transport. The most likely biological meaning of this behavior is to buffer the ROS load and keep track of the redox level in the inter-membrane space, eventually signaling it through conformational changes.


Assuntos
Cisteína/química , Fígado/metabolismo , Mitocôndrias/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Canais de Ânion Dependentes de Voltagem/metabolismo , Sequência de Aminoácidos , Animais , Transporte de Elétrons/fisiologia , Eletroforese em Gel de Poliacrilamida , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Fígado/citologia , Fígado/enzimologia , Espectrometria de Massas , Proteínas de Transporte da Membrana Mitocondrial/genética , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Oxirredução , Isoformas de Proteínas/metabolismo , Ratos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Canais de Ânion Dependentes de Voltagem/genética
18.
J Biol Chem ; 290(8): 4537-4544, 2015 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-25550160

RESUMO

Mitochondria of Drosophila melanogaster undergo Ca(2+)-induced Ca(2+) release through a putative channel (mCrC) that has several regulatory features of the permeability transition pore (PTP). The PTP is an inner membrane channel that forms from F-ATPase, possessing a conductance of 500 picosiemens (pS) in mammals and of 300 pS in yeast. In contrast to the PTP, the mCrC of Drosophila is not permeable to sucrose and appears to be selective for Ca(2+) and H(+). We show (i) that like the PTP, the mCrC is affected by the sense of rotation of F-ATPase, by Bz-423, and by Mg(2+)/ADP; (ii) that expression of human cyclophilin D in mitochondria of Drosophila S2R(+) cells sensitizes the mCrC to Ca(2+) but does not increase its apparent size; and (iii) that purified dimers of D. melanogaster F-ATPase reconstituted into lipid bilayers form 53-pS channels activated by Ca(2+) and thiol oxidants and inhibited by Mg(2+)/γ-imino ATP. These findings indicate that the mCrC is the PTP of D. melanogaster and that the signature conductance of F-ATPase channels depends on unique structural features that may underscore specific roles in different species.


Assuntos
Adenosina Trifosfatases/metabolismo , Canais de Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Adenosina Trifosfatases/genética , Animais , Canais de Cálcio/genética , Linhagem Celular , Proteínas de Drosophila/genética , Drosophila melanogaster , Humanos , Proteínas de Transporte da Membrana Mitocondrial/genética , Poro de Transição de Permeabilidade Mitocondrial
19.
Biophys Chem ; 182: 51-7, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23891570

RESUMO

Photosynthesis converts light energy into chemical energy, and supplies ATP and NADPH for CO2 fixation into carbohydrates and for the synthesis of several compounds which are essential for autotrophic growth. Oxygenic photosynthesis takes place in thylakoid membranes of chloroplasts and photosynthetic prokaryote cyanobacteria. An ancestral photoautotrophic prokaryote related to cyanobacteria has been proposed to give rise to chloroplasts of plants and algae through an endosymbiotic event. Indeed, photosynthetic complexes involved in the electron transport coupled to H(+) translocation and ATP synthesis are similar in higher plants and cyanobacteria. Furthermore, some of the protein and solute/ion conducting machineries also share common structure and function. Electrophysiological and biochemical evidence support the existence of ion channels in the thylakoid membrane in both types of organisms. By allowing specific ion fluxes across thylakoid membranes, ion channels have been hypothesized to either directly or indirectly regulate photosynthesis, by modulating the proton motive force. Recent molecular identification of some of the thylakoid-located channels allowed to obtain genetic proof in favor of such hypothesis. Furthermore, some ion channels of the envelope membrane in chloroplasts have also been shown to impact on this light-driven process. Here we give an overview of thylakoid/chloroplast located ion channels of higher plants and of cyanobacterium Synechocystis sp. PCC 6803. We focus on channels shown to be implicated in the regulation of photosynthesis and discuss the possible mechanisms of action.


Assuntos
Cianobactérias/metabolismo , Canais Iônicos/metabolismo , Fotossíntese , Plantas/metabolismo , Complexos de ATP Sintetase/metabolismo , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Cloroplastos/metabolismo , Transporte de Elétrons , Canais Iônicos/química
20.
Cell Physiol Biochem ; 26(6): 975-82, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21220928

RESUMO

Indirect evidence points to the presence of K(+) channels in plant mitochondria. In the present study, we report the results of the first patch clamp experiments on plant mitochondria. Single-channel recordings in 150 mM potassium gluconate have allowed the biophysical characterization of a channel with a conductance of 150 pS in the inner mitochondrial membrane of mitoplasts obtained from wheat (Triticum durum Desf.). The channel displayed sharp voltage sensitivity, permeability to potassium and cation selectivity. ATP in the mM concentration range completely abolished the activity. We discuss the possible molecular identity of the channel and its possible role in the defence mechanisms against oxidative stress in plants.


Assuntos
Trifosfato de Adenosina/farmacologia , Mitocôndrias/metabolismo , Canais de Potássio/fisiologia , Triticum/metabolismo , Estresse Oxidativo , Técnicas de Patch-Clamp , Canais de Potássio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA