Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Mol Carcinog ; 61(2): 173-199, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34559922

RESUMO

Cancer and the immune system share an intimate relationship. Chronic inflammation increases the risk of cancer occurrence and can also drive inflammatory mediators into the tumor microenvironment enhancing tumor growth and survival. The p38 MAPK pathway is activated both acutely and chronically by stress, inflammatory chemokines, chronic inflammatory conditions, and cancer. These properties have led to extensive efforts to find effective drugs targeting p38, which have been unsuccessful. The immediate downstream serine/threonine kinase and substrate of p38 MAPK, mitogen-activated-protein-kinase-activated-protein-kinase-2 (MK2) protects cells against stressors by regulating the DNA damage response, transcription, protein and messenger RNA stability, and motility. The phosphorylation of downstream substrates by MK2 increases inflammatory cytokine production, drives an immune response, and contributes to wound healing. By binding directly to p38 MAPK, MK2 is responsible for the export of p38 MAPK from the nucleus which gives MK2 properties that make it unique among the large number of p38 MAPK substrates. Many of the substrates of both p38 MAPK and MK2 are separated between the cytosol and nucleus and interfering with MK2 and altering this intracellular translocation has implications for the actions of both p38 MAPK and MK2. The inhibition of MK2 has shown promise in combination with both chemotherapy and radiotherapy as a method for controlling cancer growth and metastasis in a variety of cancers. Whereas the current data are encouraging the field requires the development of selective and well tolerated drugs to target MK2 and a better understanding of its effects for effective clinical use.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias , Proteínas Serina-Treonina Quinases/metabolismo , Sobrevivência Celular , Humanos , Sistema de Sinalização das MAP Quinases , Microambiente Tumoral , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
2.
Nat Immunol ; 11(3): 265-72, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20139987

RESUMO

Voltage-gated proton currents regulate generation of reactive oxygen species (ROS) in phagocytic cells. In B cells, stimulation of the B cell antigen receptor (BCR) results in the production of ROS that participate in B cell activation, but the involvement of proton channels is unknown. We report here that the voltage-gated proton channel HVCN1 associated with the BCR complex and was internalized together with the BCR after activation. BCR-induced generation of ROS was lower in HVCN1-deficient B cells, which resulted in attenuated BCR signaling via impaired BCR-dependent oxidation of the tyrosine phosphatase SHP-1. This resulted in less activation of the kinases Syk and Akt, impaired mitochondrial respiration and glycolysis and diminished antibody responses in vivo. Our findings identify unanticipated functions for proton channels in B cells and demonstrate the importance of ROS in BCR signaling and downstream metabolism.


Assuntos
Linfócitos B/imunologia , Canais Iônicos/imunologia , Espécies Reativas de Oxigênio/imunologia , Receptores de Antígenos de Linfócitos B/imunologia , Animais , Linfócitos B/enzimologia , Ativação Enzimática/imunologia , Immunoblotting , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Camundongos , Camundongos Knockout , Microscopia Confocal , Mitocôndrias/imunologia , Proteína Oncogênica v-akt/imunologia , Proteínas Tirosina Quinases/imunologia , Transdução de Sinais , Quinase Syk
3.
Proc Natl Acad Sci U S A ; 116(38): 18951-18961, 2019 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-31462498

RESUMO

The hydrophobic gasket (HG), a ring of hydrophobic amino acids in the voltage-sensing domain of most voltage-gated ion channels, forms a constriction between internal and external aqueous vestibules. Cationic Arg or Lys side chains lining the S4 helix move through this "gating pore" when the channel opens. S4 movement may occur during gating of the human voltage-gated proton channel, hHV1, but proton current flows through the same pore in open channels. Here, we replaced putative HG residues with less hydrophobic residues or acidic Asp. Substitution of individuals, pairs, or all 3 HG positions did not impair proton selectivity. Evidently, the HG does not act as a secondary selectivity filter. However, 2 unexpected functions of the HG in HV1 were discovered. Mutating HG residues independently accelerated channel opening and compromised the closed state. Mutants exhibited open-closed gating, but strikingly, at negative voltages where "normal" gating produces a nonconducting closed state, the channel leaked protons. Closed-channel proton current was smaller than open-channel current and was inhibited by 10 µM Zn2+ Extreme hyperpolarization produced a deeper closed state through a weakly voltage-dependent transition. We functionally identify the HG as Val109, Phe150, Val177, and Val178, which play a critical and exclusive role in preventing H+ influx through closed channels. Molecular dynamics simulations revealed enhanced mobility of Arg208 in mutants exhibiting H+ leak. Mutation of HG residues produces gating pore currents reminiscent of several channelopathies.


Assuntos
Ativação do Canal Iônico , Canais Iônicos/química , Canais Iônicos/metabolismo , Prótons , Aminoácidos , Células HEK293 , Humanos , Interações Hidrofóbicas e Hidrofílicas , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/genética , Canais Iônicos/genética , Potenciais da Membrana , Simulação de Dinâmica Molecular , Mutação , Conformação Proteica , Zinco/farmacologia
4.
Nature ; 480(7376): 273-7, 2011 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-22020278

RESUMO

The ion selectivity of pumps and channels is central to their ability to perform a multitude of functions. Here we investigate the mechanism of the extraordinary selectivity of the human voltage-gated proton channel, H(V)1 (also known as HVCN1). This selectivity is essential to its ability to regulate reactive oxygen species production by leukocytes, histamine secretion by basophils, sperm capacitation, and airway pH. The most selective ion channel known, H(V)1 shows no detectable permeability to other ions. Opposing classes of selectivity mechanisms postulate that (1) a titratable amino acid residue in the permeation pathway imparts proton selectivity, or (2) water molecules 'frozen' in a narrow pore conduct protons while excluding other ions. Here we identify aspartate 112 as a crucial component of the selectivity filter of H(V)1. When a neutral amino acid replaced Asp 112, the mutant channel lost proton specificity and became anion-selective or did not conduct. Only the glutamate mutant remained proton-specific. Mutation of the nearby Asp 185 did not impair proton selectivity, indicating that Asp 112 has a unique role. Although histidine shuttles protons in other proteins, when histidine or lysine replaced Asp 112, the mutant channel was still anion-permeable. Evidently, the proton specificity of H(V)1 requires an acidic group at the selectivity filter.


Assuntos
Ácido Aspártico/metabolismo , Ativação do Canal Iônico/genética , Canais Iônicos/química , Canais Iônicos/metabolismo , Prótons , Ácido Aspártico/genética , Condutividade Elétrica , Histidina/genética , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Canais Iônicos/genética , Soluções Isotônicas/farmacologia , Lisina/genética , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação/genética , Fases de Leitura Aberta/genética , Concentração Osmolar , Permeabilidade/efeitos dos fármacos , Especificidade por Substrato/efeitos dos fármacos , Sacarose/farmacologia
5.
Proc Natl Acad Sci U S A ; 111(50): 18078-83, 2014 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-25425665

RESUMO

HVCN1 (Hydrogen voltage-gated channel 1) is the only mammalian voltage-gated proton channel. In human B lymphocytes, HVCN1 associates with the B-cell receptor (BCR) and is required for optimal BCR signaling and redox control. HVCN1 is expressed in malignant B cells that rely on BCR signaling, such as chronic lymphocytic leukemia (CLL) cells. However, little is known about its regulation in these cells. We found that HVCN1 was expressed in B cells as two protein isoforms. The shorter isoform (HVCN1S) was enriched in B cells from a cohort of 76 CLL patients. When overexpressed in a B-cell lymphoma line, HVCN1S responded more profoundly to protein kinase C-dependent phosphorylation. This more potent enhanced gating response was mediated by increased phosphorylation of the same residue responsible for enhanced gating in HVCN1L, Thr(29). Furthermore, the association of HVCN1S with the BCR was weaker, which resulted in its diminished internalization upon BCR stimulation. Finally, HVCN1S conferred a proliferative and migratory advantage as well as enhanced BCR-dependent signaling. Overall, our data show for the first time, to our knowledge, the existence of a shorter isoform of HVCN1 with enhanced gating that is specifically enriched in malignant B cells. The properties of HVCN1S suggest that it may contribute to the pathogenesis of BCR-dependent B-cell malignancies.


Assuntos
Linfócitos B/metabolismo , Neoplasias Hematológicas/imunologia , Canais Iônicos/metabolismo , Leucemia Linfocítica Crônica de Células B/imunologia , Animais , Linhagem Celular Tumoral , Células HEK293 , Humanos , Camundongos , Técnicas de Patch-Clamp , Fosforilação , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteína Quinase C/metabolismo , Espécies Reativas de Oxigênio/metabolismo
6.
Proc Natl Acad Sci U S A ; 108(44): 18162-7, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-22006335

RESUMO

Fogel and Hastings first hypothesized the existence of voltage-gated proton channels in 1972 in bioluminescent dinoflagellates, where they were thought to trigger the flash by activating luciferase. Proton channel genes were subsequently identified in human, mouse, and Ciona intestinalis, but their existence in dinoflagellates remained unconfirmed. We identified a candidate proton channel gene from a Karlodinium veneficum cDNA library based on homology with known proton channel genes. K. veneficum is a predatory, nonbioluminescent dinoflagellate that produces toxins responsible for fish kills worldwide. Patch clamp studies on the heterologously expressed gene confirm that it codes for a genuine voltage-gated proton channel, kH(V)1: it is proton-specific and activated by depolarization, its g(H)-V relationship shifts with changes in external or internal pH, and mutation of the selectivity filter (which we identify as Asp(51)) results in loss of proton-specific conduction. Indirect evidence suggests that kH(V)1 is monomeric, unlike other proton channels. Furthermore, kH(V)1 differs from all known proton channels in activating well negative to the Nernst potential for protons, E(H). This unique voltage dependence makes the dinoflagellate proton channel ideally suited to mediate the proton influx postulated to trigger bioluminescence. In contrast to vertebrate proton channels, whose main function is acid extrusion, we propose that proton channels in dinoflagellates have fundamentally different functions of signaling and excitability.


Assuntos
Dinoflagellida/fisiologia , Ativação do Canal Iônico , Animais , Dinoflagellida/genética , Mutação , Prótons
7.
Explor Target Antitumor Ther ; 5(1): 108-119, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38468824

RESUMO

Head and neck cancer (HNC) is a challenging disease that lacks effective treatment, particularly in the cases that spread locoregionally and metastasize distantly, dramatically reducing patient survival rates. Expanding the understanding of the mechanisms of the metastatic cascade is critical for creating more effective therapeutics that improve outcomes for HNC patients. A true grasp of cancer metastasis requires the consideration of all cell types that contribute to the inflammatory HNC microenvironment as drivers of this process. More emphasis now is being placed on exploring the roles of the different immune cells in cancer control, tumorigenesis and metastasis. Myeloid cells are the most numerous immune cell types in the body, and they are actively recruited and reprogrammed by tumor cells to behave in a variety of ways. These cells are remarkably diverse in phenotype and function, and the part they play in tumor spread greatly differs based on the cell type. This review will focus on summarizing the roles of macrophages, neutrophils, myeloid derived suppressor cells (MDSCs), and dendritic cells (DCs) in driving HNC metastasis by examining the current knowledge base and offering potential new routes through which to target and treat this deadly process.

8.
FEBS J ; 290(4): 1008-1026, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36062330

RESUMO

Voltage-gated ion channels, whose first identified function was to generate action potentials, are divided into subfamilies with numerous members. The family of voltage-gated proton channels (HV ) is tiny. To date, all species found to express HV have exclusively one gene that codes for this unique ion channel. Here we report the discovery and characterization of three proton channel genes in the classical model system of neural plasticity, Aplysia californica. The three channels (AcHV 1, AcHV 2, and AcHV 3) are distributed throughout the whole animal. Patch-clamp analysis confirmed proton selectivity of these channels but they all differed markedly in gating. AcHV 1 gating resembled HV in mammalian cells where it is responsible for proton extrusion and charge compensation. AcHV 2 activates more negatively and conducts extensive inward proton current, properties likely to acidify the cytosol. AcHV 3, which differs from AcHV 1 and AcHV 2 in lacking the first arginine in the S4 helix, exhibits proton selective leak currents and weak voltage dependence. We report the expansion of the proton channel family, demonstrating for the first time the expression of three functionally distinct proton channels in a single species.


Assuntos
Ativação do Canal Iônico , Prótons , Animais , Ativação do Canal Iônico/fisiologia , Canais Iônicos/metabolismo , Arginina , Citosol/metabolismo , Mamíferos/metabolismo
9.
Proc Natl Acad Sci U S A ; 106(42): 18022-7, 2009 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-19805063

RESUMO

Phagocytosis of microbial invaders represents a fundamental defense mechanism of the innate immune system. The subsequent killing of microbes is initiated by the respiratory burst, in which nicotinamide adenine dinucleotide phosphate (NADPH) oxidase generates vast amounts of superoxide anion, precursor to bactericidal reactive oxygen species. Cytoplasmic pH regulation is crucial because NADPH oxidase functions optimally at neutral pH, yet produces enormous quantities of protons. We monitored pH(i) in individual human neutrophils during phagocytosis of opsonized zymosan, using confocal imaging of the pH sensing dye SNARF-1, enhanced by shifted excitation and emission ratioing, or SEER. Despite long-standing dogma that Na(+)/H(+) antiport regulates pH during the phagocyte respiratory burst, we show here that voltage-gated proton channels are the first transporter to respond. During the initial phagocytotic event, pH(i) decreased sharply, and recovery required both Na(+)/H(+) antiport and proton current. Inhibiting myeloperoxidase attenuated the acidification, suggesting that diffusion of HOCl into the cytosol comprises a substantial acid load. Inhibiting proton channels with Zn(2+) resulted in profound acidification to levels that inhibit NADPH oxidase. The pH changes accompanying phagocytosis in bone marrow phagocytes from HVCN1-deficient mice mirrored those in control mouse cells treated with Zn(2+). Both the rate and extent of acidification in HVCN1-deficient cells were twice larger than in control cells. In summary, acid extrusion by proton channels is essential to the production of reactive oxygen species during phagocytosis.


Assuntos
Canais Iônicos/metabolismo , Neutrófilos/fisiologia , Fagocitose/fisiologia , Adulto , Animais , Benzopiranos , Corantes Fluorescentes , Humanos , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Canais Iônicos/deficiência , Canais Iônicos/genética , Camundongos , Camundongos Knockout , Microscopia Confocal , NADPH Oxidases/metabolismo , Naftóis , Prótons , Rodaminas , Trocadores de Sódio-Hidrogênio/metabolismo
10.
J Biol Chem ; 285(8): 5117-21, 2010 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-20037153

RESUMO

Voltage-gated proton channels and NADPH oxidase function cooperatively in phagocytes during the respiratory burst, when reactive oxygen species are produced to kill microbial invaders. Agents that activate NADPH oxidase also enhance proton channel gating profoundly, facilitating its roles in charge compensation and pH(i) regulation. The "enhanced gating mode" appears to reflect protein kinase C (PKC) phosphorylation. Here we examine two candidates for PKC-delta phosphorylation sites in the human voltage-gated proton channel, H(V)1 (Hvcn1), Thr(29) and Ser(97), both in the intracellular N terminus. Channel phosphorylation was reduced in single mutants S97A or T29A, and further in the double mutant T29A/S97A, by an in vitro kinase assay with PKC-delta. Enhanced gating was evaluated by expressing wild-type (WT) or mutant H(V)1 channels in LK35.2 cells, a B cell hybridoma. Stimulation by phorbol myristate acetate enhanced WT channel gating, and this effect was reversed by treatment with the PKC inhibitor GF109203X. The single mutant T29A or double mutant T29A/S97A failed to respond to phorbol myristate acetate or GF109203X. In contrast, the S97A mutant responded like cells transfected with WT H(V)1. We conclude that under these conditions, direct phosphorylation of the proton channel molecule at Thr(29) is primarily responsible for the enhancement of proton channel gating. This phosphorylation is crucial to activation of the proton conductance during the respiratory burst in phagocytes.


Assuntos
Ativação do Canal Iônico/fisiologia , Canais Iônicos/metabolismo , Leucócitos/metabolismo , Explosão Respiratória/fisiologia , Treonina/metabolismo , Substituição de Aminoácidos , Carcinógenos/farmacologia , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Humanos , Indóis/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Canais Iônicos/genética , Maleimidas/farmacologia , Mutação de Sentido Incorreto , NADPH Oxidases/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Proteína Quinase C-delta/antagonistas & inibidores , Proteína Quinase C-delta/metabolismo , Explosão Respiratória/efeitos dos fármacos , Acetato de Tetradecanoilforbol/farmacologia , Treonina/genética
11.
Proc Natl Acad Sci U S A ; 105(31): 11020-5, 2008 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-18664579

RESUMO

Eosinophils and other phagocytes use NADPH oxidase to kill bacteria. Proton channels in human eosinophils and neutrophils are thought to sustain NADPH oxidase activity, and their opening is greatly enhanced by a variety of NADPH oxidase activators, including phorbol myristate acetate (PMA). In nonphagocytic cells that lack NADPH oxidase, no clear effect of PMA on proton channels has been reported. The basophil is a granulocyte that is developmentally closely related to the eosinophil but nevertheless does not express NADPH oxidase. Thus, one might expect that stimulating basophils with PMA would not affect proton currents. However, stimulation of human basophils in perforated-patch configuration with PMA, N-formyl-methionyl-leucyl-phenylalanine, or anti-IgE greatly enhanced proton currents, the latter suggesting involvement of proton channels during activation of basophils by allergens through their highly expressed IgE receptor (Fc epsilonRI). The anti-IgE-stimulated response occurred in a fraction of cells that varied among donors and was less profound than that to PMA. PKC inhibition reversed the activation of proton channels, and the proton channel response to anti-IgE or PMA persisted in Ca(2+)-free solutions. Zn(2+) at concentrations that inhibit proton current inhibited histamine release elicited by PMA or anti-IgE. Studied with confocal microscopy by using SNARF-AM and the shifted excitation and emission ratioing of fluorescence approach, anti-IgE produced acidification that was exacerbated in the presence of 100 microM Zn(2+). Evidently, proton channels are active in basophils during IgE-mediated responses and prevent excessive acidification, which may account for their role in histamine release.


Assuntos
Basófilos/imunologia , Hipersensibilidade/imunologia , Imunoglobulina E/imunologia , Bombas de Próton/metabolismo , Anticorpos Anti-Idiotípicos/metabolismo , Basófilos/metabolismo , Benzopiranos , Eletrofisiologia , Histamina/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Microscopia Confocal , N-Formilmetionina Leucil-Fenilalanina/metabolismo , Inibidores da Bomba de Prótons , Acetato de Tetradecanoilforbol/metabolismo , Zinco/farmacologia
12.
J Physiol ; 588(Pt 9): 1435-49, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20231140

RESUMO

Voltage-gated proton channels are strongly inhibited by Zn(2+), which binds to His residues. However, in a molecular model, the two externally accessible His are too far apart to coordinate Zn(2+). We hypothesize that high-affinity Zn(2+) binding occurs at the dimer interface between pairs of His residues from both monomers. Consistent with this idea, Zn(2+) effects were weaker in monomeric channels. Mutation of His(193) and His(140) in various combinations and in tandem dimers revealed that channel opening was slowed by Zn(2+) only when at least one His was present in each monomer, suggesting that in wild-type (WT) H(V)1, Zn(2+) binding between His of both monomers inhibits channel opening. In addition, monomeric channels opened exponentially, and dimeric channels opened sigmoidally. Monomeric channel gating had weaker temperature dependence than dimeric channels. Finally, monomeric channels opened 6.6 times faster than dimeric channels. Together, these observations suggest that in the proton channel dimer, the two monomers are closely apposed and interact during a cooperative gating process. Zn(2+) appears to slow opening by preventing movement of the monomers relative to each other that is prerequisite to opening. These data also suggest that the association of the monomers is tenuous and allows substantial freedom of movement. The data support the idea that native proton channels are dimeric. Finally, the idea that monomer-dimer interconversion occurs during activation of phagocytes appears to be ruled out.


Assuntos
Ativação do Canal Iônico/efeitos dos fármacos , Canais Iônicos/antagonistas & inibidores , Zinco/farmacologia , Linhagem Celular , Dimerização , Eletrofisiologia , Proteínas de Fluorescência Verde/metabolismo , Histidina/química , Humanos , Concentração de Íons de Hidrogênio , Ativação do Canal Iônico/genética , Canais Iônicos/química , Canais Iônicos/genética , Cinética , Modelos Moleculares , Mutação/fisiologia , Técnicas de Patch-Clamp , Temperatura , Transfecção
13.
PLoS One ; 15(5): e0227522, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32374759

RESUMO

Expression of the voltage gated proton channel (Hv1) as identified by immunocytochemistry has been reported previously in breast cancer tissue. Increased expression of HV1 was correlated with poor prognosis and decreased overall and disease-free survival but the mechanism of its involvement in the disease is unknown. Here we present electrophysiological recordings of HV1 channel activity, confirming its presence and function in the plasma membrane of a breast cancer cell line, MDA-MB-231. With western blotting we identify significant levels of HV1 expression in 3 out of 8 "triple negative" breast cancer cell lines (estrogen, progesterone, and HER2 receptor expression negative). We examine the function of HV1 in breast cancer using MDA-MB-231 cells as a model by suppressing the expression of HV1 using shRNA (knock-down; KD) and by eliminating HV1 using CRISPR/Cas9 gene editing (knock-out; KO). Surprisingly, these two approaches produced incongruous effects. Knock-down of HV1 using shRNA resulted in slower cell migration in a scratch assay and a significant reduction in H2O2 release. In contrast, HV1 Knock-out cells did not show reduced migration or H2O2 release. HV1 KO but not KD cells showed an increased glycolytic rate accompanied by an increase in p-AKT (phospho-AKT, Ser473) activity. The expression of CD171/LCAM-1, an adhesion molecule and prognostic indicator for breast cancer, was reduced in HV1 KO cells. When we compared MDA-MB-231 xenograft growth rates in immunocompromised mice, tumors from HV1 KO cells grew less than WT in mass, with lower staining for the Ki-67 marker for cell proliferation rate. Therefore, deletion of HV1 expression in MDA-MB-231 cells limits tumor growth rate. The limited growth thus appears to be independent of oxidant production by NADPH oxidase molecules and to be mediated by cell adhesion molecules. Although HV1 KO and KD affect certain cellular mechanisms differently, both implicate HV1-mediated pathways for control of tumor growth in the MDA-MB-231 cell line.


Assuntos
Proliferação de Células/genética , Canais Iônicos/genética , Proteínas de Membrana/genética , Neoplasias de Mama Triplo Negativas/genética , Animais , Sistemas CRISPR-Cas/genética , Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Técnicas de Inativação de Genes , Xenoenxertos , Humanos , Peróxido de Hidrogênio/farmacologia , Imuno-Histoquímica , Camundongos , NADPH Oxidases/genética , RNA Interferente Pequeno/genética , Neoplasias de Mama Triplo Negativas/patologia
14.
PLoS One ; 15(6): e0234146, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32525891

RESUMO

Approximately 20% of breast cancers are HER2-positive. Trastuzumab has improved patient outcomes significantly for these cancers. However, acquired resistance remains a major hurdle in the clinical management of these patients. Therefore, identifying molecular changes that cause trastuzumab resistance is worthwhile. STAT6 is a transcription factor that regulates a variety of genes involved in cell cycle regulation, growth inhibition, and apoptosis. STAT6 expression is lost in approximately 3% of breast cancers, but little work has been done in the context of trastuzumab resistance in breast cancer. In isogenic cell line pairs, we observed that trastuzumab-resistant cells expressed significantly lower levels of STAT6 compared to trastuzumab-sensitive cells. Therefore, in order to study the consequences of STAT6 loss in HER2+ breast cancer, we knocked out both alleles of the STAT6 gene using somatic cell gene targeting. Interestingly, loss of STAT6 resulted in anchorage-independent growth and changes in several genes involved in epithelial to mesenchymal transition. This study suggests that STAT6 may play a role in the pathophysiology of HER2+ human breast cancer.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Receptor ErbB-2/metabolismo , Fator de Transcrição STAT6/genética , Trastuzumab/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Transição Epitelial-Mesenquimal , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , RNA Guia de Cinetoplastídeos/metabolismo , Receptor ErbB-2/genética , Fator de Transcrição STAT6/deficiência
15.
J Gen Physiol ; 152(10)2020 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-32902579

RESUMO

The voltage-gated proton channel (HV1) is a voltage sensor that also conducts protons. The singular ability of protons to penetrate proteins complicates distinguishing closed and open channels. When we replaced valine with histidine at position 116 in the external vestibule of hHV1, current was potently inhibited by externally applied Zn2+ in a construct lacking the two His that bind Zn2+ in WT channels. High-affinity binding with profound effects at 10 nM Zn2+ at pHo 7 suggests additional groups contribute. We hypothesized that Asp185, which faces position 116 in our closed-state model, contributes to Zn2+ chelation. Confirming this prediction, V116H/D185N abolished Zn2+ binding. Studied in a C-terminal truncated monomeric construct, V116H channels activated rapidly. Anomalously, Zn2+ slowed activation, producing a time constant independent of both voltage and Zn2+ concentration. We hypothesized that slow turn-on of H+ current in the presence of Zn2+ reflects the rate of Zn2+ unbinding from the channel, analogous to drug-receptor dissociation reactions. This behavior in turn suggests that the affinity for Zn2+ is greater in the closed state of hHV1. Supporting this hypothesis, pulse pairs revealed a rapid component of activation whose amplitude decreased after longer intervals at negative voltages as closed channels bound Zn2+. The lower affinity of Zn2+ in open channels is consistent with the idea that structural rearrangements within the transmembrane region bring Arg205 near position 116, electrostatically expelling Zn2+. This phenomenon provides direct evidence that Asp185 opposes position 116 in closed channels and that Arg205 moves between them when the channel opens.


Assuntos
Canais Iônicos , Prótons , Zinco , Sítios de Ligação , Humanos , Ativação do Canal Iônico , Canais Iônicos/metabolismo , Zinco/metabolismo
16.
J Gen Physiol ; 150(6): 835-850, 2018 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-29743301

RESUMO

Voltage-gated proton channels, HV1, were first reported in Helix aspersa snail neurons. These H+ channels open very rapidly, two to three orders of magnitude faster than mammalian HV1. Here we identify an HV1 gene in the snail Helisoma trivolvis and verify protein level expression by Western blotting of H. trivolvis brain lysate. Expressed in mammalian cells, HtHV1 currents in most respects resemble those described in other snails, including rapid activation, 476 times faster than hHV1 (human) at pHo 7, between 50 and 90 mV. In contrast to most HV1, activation of HtHV1 is exponential, suggesting first-order kinetics. However, the large gating charge of ∼5.5 e0 suggests that HtHV1 functions as a dimer, evidently with highly cooperative gating. HtHV1 opening is exquisitely sensitive to pHo, whereas closing is nearly independent of pHo Zn2+ and Cd2+ inhibit HtHV1 currents in the micromolar range, slowing activation, shifting the proton conductance-voltage (gH-V) relationship to more positive potentials, and lowering the maximum conductance. This is consistent with HtHV1 possessing three of the four amino acids that coordinate Zn2+ in mammalian HV1. All known HV1 exhibit ΔpH-dependent gating that results in a 40-mV shift of the gH-V relationship for a unit change in either pHo or pHi This property is crucial for all the functions of HV1 in many species and numerous human cells. The HtHV1 channel exhibits normal or supernormal pHo dependence, but weak pHi dependence. Under favorable conditions, this might result in the HtHV1 channel conducting inward currents and perhaps mediating a proton action potential. The anomalous ΔpH-dependent gating of HtHV1 channels suggests a structural basis for this important property, which is further explored in this issue (Cherny et al. 2018. J. Gen. Physiol. https://doi.org/10.1085/jgp.201711968).


Assuntos
Ativação do Canal Iônico , Canais Iônicos/metabolismo , Potenciais da Membrana , Prótons , Animais , Cádmio/metabolismo , Células HEK293 , Humanos , Canais Iônicos/química , Caramujos , Zinco/metabolismo
17.
J Gen Physiol ; 150(6): 851-862, 2018 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-29743300

RESUMO

We recently identified a voltage-gated proton channel gene in the snail Helisoma trivolvis, HtHV1, and determined its electrophysiological properties. Consistent with early studies of proton currents in snail neurons, HtHV1 opens rapidly, but it unexpectedly exhibits uniquely defective sensitivity to intracellular pH (pHi). The H+ conductance (gH)-V relationship in the voltage-gated proton channel (HV1) from other species shifts 40 mV when either pHi or pHo (extracellular pH) is changed by 1 unit. This property, called ΔpH-dependent gating, is crucial to the functions of HV1 in many species and in numerous human tissues. The HtHV1 channel exhibits normal pHo dependence but anomalously weak pHi dependence. In this study, we show that a single point mutation in human hHV1-changing His168 to Gln168, the corresponding residue in HtHV1-compromises the pHi dependence of gating in the human channel so that it recapitulates the HtHV1 response. This location was previously identified as a contributor to the rapid gating kinetics of HV1 in Strongylocentrotus purpuratus His168 mutation in human HV1 accelerates activation but accounts for only a fraction of the species difference. H168Q, H168S, or H168T mutants exhibit normal pHo dependence, but changing pHi shifts the gH-V relationship on average by <20 mV/unit. Thus, His168 is critical to pHi sensing in hHV1. His168, located at the inner end of the pore on the S3 transmembrane helix, is the first residue identified in HV1 that significantly impairs pH sensing when mutated. Because pHo dependence remains intact, the selective erosion of pHi dependence supports the idea that there are distinct internal and external pH sensors. Although His168 may itself be a pHi sensor, the converse mutation, Q229H, does not normalize the pHi sensitivity of the HtHV1 channel. We hypothesize that the imidazole group of His168 interacts with nearby Phe165 or other parts of hHV1 to transduce pHi into shifts of voltage-dependent gating.


Assuntos
Ativação do Canal Iônico , Canais Iônicos/metabolismo , Mutação Puntual , Prótons , Animais , Cricetinae , Células HEK293 , Histidina/química , Histidina/genética , Humanos , Concentração de Íons de Hidrogênio , Canais Iônicos/química , Canais Iônicos/genética , Potenciais da Membrana , Camundongos , Domínios Proteicos , Ratos , Homologia de Sequência , Caramujos
18.
J Gen Physiol ; 127(6): 659-72, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16702353

RESUMO

Electrophysiological events are of central importance during the phagocyte respiratory burst, because NADPH oxidase is electrogenic and voltage sensitive. We investigated the recent suggestion that large-conductance, calcium-activated K(+) (BK) channels, rather than proton channels, play an essential role in innate immunity (Ahluwalia, J., A. Tinker, L.H. Clapp, M.R. Duchen, A.Y. Abramov, S. Page, M. Nobles, and A.W. Segal. 2004. Nature. 427:853-858). In PMA-stimulated human neutrophils or eosinophils, we did not detect BK currents, and neither of the BK channel inhibitors iberiotoxin or paxilline nor DPI inhibited any component of outward current. BK inhibitors did not inhibit the killing of bacteria, nor did they affect NADPH oxidase-dependent degradation of bacterial phospholipids by extracellular gIIA-PLA(2) or the production of superoxide anion (O(2*)(-)). Moreover, an antibody against the BK channel did not detect immunoreactive protein in human neutrophils. A required role for voltage-gated proton channels is demonstrated by Zn(2+) inhibition of NADPH oxidase activity assessed by H(2)O(2) production, thus validating previous studies showing that Zn(2+) inhibited O(2*)(-) production when assessed by cytochrome c reduction. In conclusion, BK channels were not detected in human neutrophils or eosinophils, and BK inhibitors did not impair antimicrobial activity. In contrast, we present additional evidence that voltage-gated proton channels serve the essential role of charge compensation during the respiratory burst.


Assuntos
Atividade Bactericida do Sangue , Eosinófilos/microbiologia , Eosinófilos/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/fisiologia , Neutrófilos/microbiologia , Neutrófilos/fisiologia , Prótons , Animais , Células COS , Linhagem Celular Tumoral , Cloretos/farmacologia , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Humanos , Canais Iônicos/antagonistas & inibidores , Canais Iônicos/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/antagonistas & inibidores , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/fisiologia , Compostos de Zinco/farmacologia
19.
Methods Mol Biol ; 412: 139-75, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18453111

RESUMO

The past decade has seen increasing use of the patch clamp technique on neutrophils and eosinophils. The main goal of these electrophysiological studies has been to elucidate the mechanisms underlying the phagocyte respiratory burst. NADPH oxidase activity, which defines the respiratory burst in granulocytes, is electrogenic because electrons from NADPH are transported across the cell membrane, where they reduce oxygen to form superoxide anion (O2-). This passage of electrons comprises an electrical current that would rapidly depolarize the membrane if the charge movement were not balanced by proton efflux. The patch clamp technique enables simultaneous recording of NADPH oxidase-generated electron current and H+ flux through the closely related H+ channel. Increasing evidence suggests that other ion channels may play crucial roles in degranulation, phagocytosis, and chemotaxis, highlighting the importance of electrophysiological studies to advance knowledge of granulocyte function. Several configurations of the patch clamp technique exist. Each has advantages and limitations that are discussed here. Meaningful measurements of ion channels cannot be achieved without an understanding of their fundamental properties. We describe the types of measurements that are necessary to characterize a particular ion channel.


Assuntos
Eletrofisiologia/métodos , Neutrófilos/fisiologia , Animais , Adesão Celular/fisiologia , Eletrodos , Elétrons , Eletrofisiologia/instrumentação , Humanos , Canais Iônicos/fisiologia , Modelos Biológicos , Técnicas de Patch-Clamp/instrumentação , Bombas de Próton/fisiologia
20.
PLoS One ; 12(2): e0171594, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28178296

RESUMO

In 1972, J. Woodland Hastings and colleagues predicted the existence of a proton selective channel (HV1) that opens in response to depolarizing voltage across the vacuole membrane of bioluminescent dinoflagellates and conducts protons into specialized luminescence compartments (scintillons), thereby causing a pH drop that triggers light emission. HV1 channels were subsequently identified and demonstrated to have important functions in a multitude of eukaryotic cells. Here we report a predicted protein from Lingulodinium polyedrum that displays hallmark properties of bona fide HV1, including time-dependent opening with depolarization, perfect proton selectivity, and characteristic ΔpH dependent gating. Western blotting and fluorescence confocal microscopy of isolated L. polyedrum scintillons immunostained with antibody to LpHV1 confirm LpHV1's predicted organellar location. Proteomics analysis demonstrates that isolated scintillon preparations contain peptides that map to LpHV1. Finally, Zn2+ inhibits both LpHV1 proton current and the acid-induced flash in isolated scintillons. These results implicate LpHV1 as the voltage gated proton channel that triggers bioluminescence in L. polyedrum, confirming Hastings' hypothesis. The same channel likely mediates the action potential that communicates the signal along the tonoplast to the scintillon.


Assuntos
Dinoflagellida/metabolismo , Ativação do Canal Iônico , Canais Iônicos/metabolismo , Prótons , Vacúolos/metabolismo , Membrana Celular/metabolismo , Concentração de Íons de Hidrogênio , Espectrometria de Massas , Zinco/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa