Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Elife ; 122023 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-37449820

RESUMO

Cystathionine-ß-synthase (CBS)-pair domain divalent metal cation transport mediators (CNNMs) are an evolutionarily conserved family of magnesium transporters. They promote efflux of Mg2+ ions on their own and influx of divalent cations when expressed with the transient receptor potential ion channel subfamily M member 7 (TRPM7). Recently, ADP-ribosylation factor-like GTPase 15 (ARL15) has been identified as CNNM-binding partner and an inhibitor of divalent cation influx by TRPM7. Here, we characterize ARL15 as a GTP and CNNM-binding protein and demonstrate that ARL15 also inhibits CNNM2 Mg2+ efflux. The crystal structure of a complex between ARL15 and CNNM2 CBS-pair domain reveals the molecular basis for binding and allowed the identification of mutations that specifically block binding. A binding deficient ARL15 mutant, R95A, failed to inhibit CNNM and TRPM7 transport of Mg2+ and Zn2+ ions. Structural analysis and binding experiments with phosphatase of regenerating liver 2 (PRL2 or PTP4A2) showed that ARL15 and PRLs compete for binding CNNM to coordinate regulation of ion transport by CNNM and TRPM7.


Assuntos
Proteínas Monoméricas de Ligação ao GTP , Canais de Cátion TRPM , Cátions Bivalentes , Canais de Cátion TRPM/genética , Ligação Proteica , Transporte Biológico
2.
bioRxiv ; 2023 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-36711628

RESUMO

Cystathionine-ß-synthase (CBS)-pair domain divalent metal cation transport mediators (CNNMs) are an evolutionarily conserved family of magnesium transporters. They promote efflux of Mg 2+ ions on their own or uptake of divalent cations when coupled to the transient receptor potential ion channel subfamily M member 7 (TRPM7). Recently, ADP-ribosylation factor-like GTPase 15 (ARL15) has been identified as CNNM binding partner and an inhibitor of divalent cation influx by TRPM7. Here, we characterize ARL15 as a GTP-binding protein and demonstrate that it binds the CNNM CBS-pair domain with low micromolar affinity. The crystal structure of the complex between ARL15 GTPase domain and CNNM2 CBS-pair domain reveals the molecular determinants of the interaction and allowed the identification of mutations in ARL15 and CNNM2 mutations that abrogate binding. Loss of CNNM binding prevented ARL15 suppression of TRPM7 channel activity in support of previous reports that the proteins function as a ternary complex. Binding experiments with phosphatase of regenerating liver 2 (PRL2 or PTP4A2) revealed that ARL15 and PRLs compete for binding CNNM, suggesting antagonistic regulation of divalent cation transport by the two proteins.

3.
Nutrients ; 14(12)2022 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-35745255

RESUMO

The trace element zinc (Zn) binds to over ten percent of proteins in eukaryotic cells. Zn flexible chemistry allows it to regulate the activity of hundreds of enzymes and influence scores of metabolic processes in cells throughout the body. Deficiency of Zn in humans has a profound effect on development and in adults later in life, particularly in the brain, where Zn deficiency is linked to several neurological disorders. In this review, we will summarize the importance of Zn during development through a description of the outcomes of both genetic and early dietary Zn deficiency, focusing on the pathological consequences on the whole body and brain. The epidemiology and the symptomology of Zn deficiency in humans will be described, including the most studied inherited Zn deficiency disease, Acrodermatitis enteropathica. In addition, we will give an overview of the different forms and animal models of Zn deficiency, as well as the 24 Zn transporters, distributed into two families: the ZIPs and the ZnTs, which control the balance of Zn throughout the body. Lastly, we will describe the TRPM7 ion channel, which was recently shown to contribute to intestinal Zn absorption and has its own significant impact on early embryonic development.


Assuntos
Acrodermatite , Proteínas de Transporte de Cátions , Acrodermatite/metabolismo , Animais , Encéfalo/metabolismo , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Feminino , Gravidez , Zinco/deficiência
4.
PLoS Biol ; 19(12): e3001496, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34928937

RESUMO

Magnesium is essential for cellular life, but how it is homeostatically controlled still remains poorly understood. Here, we report that members of CNNM family, which have been controversially implicated in both cellular Mg2+ influx and efflux, selectively bind to the TRPM7 channel to stimulate divalent cation entry into cells. Coexpression of CNNMs with the channel markedly increased uptake of divalent cations, which is prevented by an inactivating mutation to the channel's pore. Knockout (KO) of TRPM7 in cells or application of the TRPM7 channel inhibitor NS8593 also interfered with CNNM-stimulated divalent cation uptake. Conversely, KO of CNNM3 and CNNM4 in HEK-293 cells significantly reduced TRPM7-mediated divalent cation entry, without affecting TRPM7 protein expression or its cell surface levels. Furthermore, we found that cellular overexpression of phosphatases of regenerating liver (PRLs), known CNNMs binding partners, stimulated TRPM7-dependent divalent cation entry and that CNNMs were required for this activity. Whole-cell electrophysiological recordings demonstrated that deletion of CNNM3 and CNNM4 from HEK-293 cells interfered with heterologously expressed and native TRPM7 channel function. We conclude that CNNMs employ the TRPM7 channel to mediate divalent cation influx and that CNNMs also possess separate TRPM7-independent Mg2+ efflux activities that contribute to CNNMs' control of cellular Mg2+ homeostasis.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Ciclinas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Canais de Cátion TRPM/metabolismo , Proteínas de Transporte de Cátions/fisiologia , Cátions Bivalentes/metabolismo , Linhagem Celular Tumoral , Ciclinas/fisiologia , Células HEK293 , Humanos , Magnésio/metabolismo , Técnicas de Patch-Clamp , Proteínas Serina-Treonina Quinases/fisiologia , Canais de Cátion TRPM/genética , Canais de Cátion TRPM/fisiologia
5.
Dev Dyn ; 249(8): 912-923, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32315468

RESUMO

A common theme in organogenesis is how the final structure of organs emerge from epithelial tube structures, with the formation of the neural tube being one of the best examples. Two types of cell movements co-occur during neural tube closure involving the migration of cells toward the midline of the embryo (mediolateral intercalation or convergent extension) as well as the deep movement of cells from inside the embryo to the outside of the lateral side of the neural plate (radial intercalation). Failure of either type of cell movement will prevent neural tube closure, which can produce a range of neural tube defects (NTDs), a common congenital disease in humans. Numerous studies have identified signaling pathways that regulate mediolateral intercalation during neural tube closure. Less understood are the pathways that govern radial intercalation. Using the Xenopus laevis system, our group reported the identification of transient receptor potential (TRP) channels, TRPM6 and TRPM7, and the Mg2+ ion they conduct, as novel and key factors regulating both mediolateral and radial intercalation during neural tube closure. Here we broadly discuss tubulogenesis and cell intercalation from the perspective of neural tube closure and the respective roles of TRPM7 and TRPM6 in this critical embryonic process.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Magnésio/química , Proteínas Serina-Treonina Quinases/fisiologia , Canais de Cátion TRPM/fisiologia , Proteínas de Xenopus/fisiologia , Xenopus laevis/metabolismo , Células 3T3 , Animais , Movimento Celular , Desenvolvimento Embrionário , Humanos , Íons , Magnésio/metabolismo , Camundongos , Placa Neural/metabolismo , Tubo Neural/metabolismo , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo , Neurulação , Domínios Proteicos , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais , Canais de Cátion TRPM/genética , Canais de Cátion TRPM/metabolismo , Proteínas de Xenopus/genética , Peixe-Zebra
6.
J Cardiovasc Dev Dis ; 6(4)2019 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-31547577

RESUMO

Cardiac fibrosis is the excessive deposition of extracellular matrix proteins by cardiac fibroblasts and myofibroblasts, and is a hallmark feature of most heart diseases, including arrhythmia, hypertrophy, and heart failure. This maladaptive process occurs in response to a variety of stimuli, including myocardial injury, inflammation, and mechanical overload. There are multiple signaling pathways and various cell types that influence the fibrogenesis cascade. Fibroblasts and myofibroblasts are central effectors. Although it is clear that Ca2+ signaling plays a vital role in this pathological process, what contributes to Ca2+ signaling in fibroblasts and myofibroblasts is still not wholly understood, chiefly because of the large and diverse number of receptors, transporters, and ion channels that influence intracellular Ca2+ signaling. Intracellular Ca2+ signals are generated by Ca2+ release from intracellular Ca2+ stores and by Ca2+ entry through a multitude of Ca2+-permeable ion channels in the plasma membrane. Over the past decade, the transient receptor potential (TRP) channels have emerged as one of the most important families of ion channels mediating Ca2+ signaling in cardiac fibroblasts. TRP channels are a superfamily of non-voltage-gated, Ca2+-permeable non-selective cation channels. Their ability to respond to various stimulating cues makes TRP channels effective sensors of the many different pathophysiological events that stimulate cardiac fibrogenesis. This review focuses on the mechanisms of Ca2+ signaling in fibroblast differentiation and fibrosis-associated heart diseases and will highlight recent advances in the understanding of the roles that TRP and other Ca2+-permeable channels play in cardiac fibrosis.

7.
J Biol Chem ; 293(29): 11491-11504, 2018 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-29866880

RESUMO

The channel-kinase transient receptor potential melastatin 7 (TRPM7) is a bifunctional protein with ion channel and kinase domains. The kinase activity of TRPM7 has been linked to the regulation of a broad range of cellular activities, but little is understood as to how the channel itself is regulated by its own kinase activity. Here, using several mammalian cell lines expressing WT TRPM7 or kinase-inactive variants, we discovered that compared with the cells expressing WT TRPM7, cells in which TRPM7's kinase activity was inactivated had faster degradation, elevated ubiquitination, and increased intracellular retention of the channel. Mutational analysis of TRPM7 autophosphorylation sites further revealed a role for Ser-1360 of TRPM7 as a key residue mediating both TRPM7 stability and intracellular trafficking. Additional trafficking roles were uncovered for Ser-1403 and Ser-1567, whose phosphorylation by TRPM7's kinase activity mediated the interaction of the channel with the signaling protein 14-3-3θ. In summary, our results point to a critical role for TRPM7's kinase activity in regulating proteasome-mediated turnover of the TRPM7 channel and controlling its cellular localization in polarized epithelial cells. Overall, these findings improve our understanding of the significance of TRPM7's kinase activity for functional regulation of its channel activity.


Assuntos
Células Epiteliais/metabolismo , Canais de Cátion TRPM/metabolismo , Proteínas 14-3-3/metabolismo , Animais , Polaridade Celular , Células Epiteliais/citologia , Células HEK293 , Humanos , Camundongos , Fosforilação , Ligação Proteica , Proteínas Quinases/análise , Proteínas Quinases/metabolismo , Estabilidade Proteica , Transporte Proteico , Canais de Cátion TRPM/análise
8.
Sci Rep ; 7(1): 15623, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-29142255

RESUMO

In humans, germline mutations in Trpm6 cause autosomal dominant hypomagnesemia with secondary hypocalcemia disorder. Loss of Trpm6 in mice also perturbs cellular magnesium homeostasis but additionally results in early embryonic lethality and neural tube closure defects. To define the mechanisms by which TRPM6 influences neural tube closure, we functionally characterized the role of TRPM6 during early embryogenesis in Xenopus laevis. The expression of Xenopus TRPM6 (XTRPM6) is elevated at the onset of gastrulation and is concentrated in the lateral mesoderm and ectoderm at the neurula stage. Loss of XTRPM6 produced gastrulation and neural tube closure defects. Unlike XTRPM6's close homologue XTRPM7, whose loss interferes with mediolateral intercalation, depletion of XTRPM6 but not XTRPM7 disrupted radial intercalation cell movements. A zinc-influx assay demonstrated that TRPM6 has the potential to constitute functional channels in the absence of TRPM7. The results of our study indicate that XTRPM6 regulates radial intercalation with little or no contribution from XTRPM7 in the region lateral to the neural plate, whereas XTRPM7 is mainly involved in regulating mediolateral intercalation in the medial region of the neural plate. We conclude that both TRPM6 and TRPM7 channels function cooperatively but have distinct and essential roles during neural tube closure.


Assuntos
Desenvolvimento Embrionário/genética , Placa Neural/crescimento & desenvolvimento , Tubo Neural/crescimento & desenvolvimento , Canais de Cátion TRPM/genética , Proteínas de Xenopus/genética , Animais , Cálcio/metabolismo , Movimento Celular/genética , Regulação da Expressão Gênica no Desenvolvimento , Mutação em Linhagem Germinativa/genética , Humanos , Hipercalciúria/genética , Hipercalciúria/metabolismo , Hipercalciúria/patologia , Hipocalcemia/genética , Hipocalcemia/metabolismo , Hipocalcemia/patologia , Magnésio/metabolismo , Nefrocalcinose/genética , Nefrocalcinose/metabolismo , Nefrocalcinose/patologia , Placa Neural/metabolismo , Erros Inatos do Transporte Tubular Renal/genética , Erros Inatos do Transporte Tubular Renal/metabolismo , Erros Inatos do Transporte Tubular Renal/patologia , Xenopus laevis
9.
Sci Rep ; 7: 42739, 2017 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-28220887

RESUMO

TRPM7 and TRPM6 were the first identified bifunctional channels to contain their own kinase domains, but how these channel-kinases are regulated is poorly understood. Previous studies identified numerous phosphorylation sites on TRPM7, but very little is known about TRPM6 phosphorylation or sites on TRPM7 transphosphorylated by TRPM6. Our mass spectrometric analysis of homomeric and heteromeric TRPM7 and TRPM6 channels identified phosphorylation sites on both proteins, as well as several prominent sites on TRPM7 that are commonly modified through autophosphorylation and transphosphorylation by TRPM6. We conducted a series of amino acid substitution analyses and identified S1777, in TRPM7's catalytic domain, and S1565, in TRPM7's exchange domain that mediates kinase dimerization, as potential regulatory sites. The phosphomimetic S1777D substitution disrupted catalytic activity, most likely by causing an electrostatic perturbation at the active site. The S1565D phosphomimetic substitution also inactivated the kinase but did so without interfering with kinase dimerization. Molecular modeling indicates that phosphorylation of S1565 is predicted to structurally affect TRPM7's functionally conserved N/D loop, which is thought to influence the access of substrate to the active site pocket. We propose that phosphorylation of S1565 within the exchange domain functions as a regulatory switch to control TRPM7 catalytic activity.


Assuntos
Processamento de Proteína Pós-Traducional , Proteínas Serina-Treonina Quinases/metabolismo , Canais de Cátion TRPM/metabolismo , Substituição de Aminoácidos , Domínio Catalítico , Células HEK293 , Humanos , Fosforilação , Multimerização Proteica , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Canais de Cátion TRPM/química , Canais de Cátion TRPM/genética
10.
Int J Dev Biol ; 59(7-9): 281-8, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26679946

RESUMO

Magnesium (Mg(2+)) is the second most abundant cellular cation and is essential for all stages of life, from the early embryo to adult. Mg(2+) deficiency causes or contributes to many human diseases, including migraine headaches, Parkinson's disease, Alzheimer's disease, hypotension, type 2 diabetes mellitus and cardiac arrhythmias. Although the concentration of Mg(2+) in the extracellular environment can vary significantly, the total intracellular Mg(2+) concentration is actively maintained within a relatively narrow range (14 - 20 mM) via tight, yet poorly understood, regulation of intracellular Mg(2+)by Mg(2+) transporters and Mg(2+)-permeant ion channels. Recent studies have continued to add to the growing number of Mg(2+) transporters and ion channels involved in Mg(2+) homeostasis, including TRPM6 and TRPM7, members of the transient receptor potential (TRP) ion channel family. Mutations in TRPM6, including amino acid substitutions that prevent its heterooligomerization with TRPM7, occur in the rare autosomal-recessive disease hypomagnesemia with secondary hypocalcemia (HSH). Genetic ablation of either gene in mice results in early embryonic lethality, raising the question of whether these channels' capacity to mediate Mg(2+) influx plays an important role in embryonic development. Here we review what is known of the function of Mg(2+) in early development and summarize recent findings regarding the function of the TRPM6 and TRPM7 ion channels during embryogenesis.


Assuntos
Desenvolvimento Embrionário/fisiologia , Magnésio/metabolismo , Canais de Cátion TRPM/metabolismo , Animais , Camundongos , Camundongos Knockout
11.
Sci Rep ; 5: 18395, 2015 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-26671672

RESUMO

Mutations in protein kinase C substrate 80K-H (PRKCSH), which encodes for an 80 KDa protein named hepatocystin (80K-H, PRKCSH), gives rise to polycystic liver disease (PCLD). Hepatocystin functions as the noncatalytic beta subunit of Glucosidase II, an endoplasmic reticulum (ER)-resident enzyme involved in processing and quality control of newly synthesized glycoproteins. Patients harboring heterozygous germline mutations in PRKCSH are thought to develop renal cysts as a result of somatic loss of the second allele, which subsequently interferes with expression of the TRP channel polycystin-2 (PKD2). Deletion of both alleles of PRKCSH in mice results in embryonic lethality before embryonic day E11.5. Here, we investigated the function of hepatocystin during Xenopus laevis embryogenesis and identified hepatocystin as a binding partner of the TRPM7 ion channel, whose function is required for vertebrate gastrulation. We find that TRPM7 functions synergistically with hepatocystin. Although other N-glycosylated proteins are critical to early development, overexpression of TRPM7 in Xenopus laevis embryos was sufficient to fully rescue the gastrulation defect caused by loss of hepatocystin. We observed that depletion of hepatocystin in Xenopus laevis embryos decreased TRPM7 expression, indicating that the early embryonic lethality caused by loss of hepatocystin is mainly due to impairment of TRPM7 protein expression.


Assuntos
Embrião não Mamífero/embriologia , Desenvolvimento Embrionário/fisiologia , Gástrula/embriologia , Glucosidases/metabolismo , Canais de Cátion TRPM/metabolismo , Proteínas de Xenopus/metabolismo , Animais , Linhagem Celular , Glucosidases/genética , Humanos , Camundongos , Canais de Cátion TRPM/genética , Proteínas de Xenopus/genética , Xenopus laevis
12.
Magnes Res ; 27(1): 1-8, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24721994

RESUMO

Important for energy metabolism, neurotransmission, bone stability, and other cellular functions, Mg(2+) has well-established and undisputedly critical roles in adult tissues. Its contributions to early embryonic development are less clearly understood. For decades it has been known that gestational Mg(2+) deficiency in rodents produces teratogenic effects. More recent studies have linked deficiency in this vital cation to birth defects in humans, including spina bifida, a neural fold closure defect in humans that occurs at an average rate of 1 per 1000 pregnancies. The first suggestion that Mg(2+) may be playing a more specific role in early development arose from studies of the TRPM7 and TRPM6 ion channels. TRPM7 and TRPM6 are divalent-selective ion channels in possession of their own kinase domains that have been implicated in the control of Mg(2+) homeostasis in vertebrates. Disruption of the functions of these ion channels in mice as well as in frogs interferes with gastrulation, a pivotal process during early embryonic development that executes the emergence of the body plan and closure of the neural tube. Surprisingly, gastrulation defects produced by depletion of TRPM7 can be prevented by Mg(2+) supplementation, indicating an essential role for Mg(2+) in gastrulation and neural fold closure. The aim of this review is to summarize the data emerging from molecular genetic, biochemical and electrophysiological studies of TRPM6 and TRPM7 and provide a model of how Mg(2+), through these unique channel-kinases, may be impacting early embryonic development.


Assuntos
Magnésio/metabolismo , Canais de Cátion TRPM/metabolismo , Xenopus laevis/embriologia , Xenopus laevis/metabolismo , Animais , Humanos
13.
Dev Biol ; 386(2): 428-39, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24291744

RESUMO

Transient receptor potential, melastatin-like 7 (Trpm7) is a combined ion channel and kinase implicated in the differentiation or function of many cell types. Early lethality in mice and frogs depleted of the corresponding gene impedes investigation of the functions of this protein particularly during later stages of development. By contrast, zebrafish trpm7 mutant larvae undergo early morphogenesis normally and thus do not have this limitation. The mutant larvae are characterized by multiple defects including melanocyte cell death, transient paralysis, and an ion imbalance that leads to the development of kidney stones. Here we report a requirement for Trpm7 in differentiation or function of dopaminergic neurons in vivo. First, trpm7 mutant larvae are hypomotile and fail to make a dopamine-dependent developmental transition in swim-bout length. Both of these deficits are partially rescued by the application of levodopa or dopamine. Second, histological analysis reveals that in trpm7 mutants a significant fraction of dopaminergic neurons lack expression of tyrosine hydroxylase, the rate-limiting enzyme in dopamine synthesis. Third, trpm7 mutants are unusually sensitive to the neurotoxin 1-methyl-4-phenylpyridinium, an oxidative stressor, and their motility is partially rescued by application of the iron chelator deferoxamine, an anti-oxidant. Finally, in SH-SY5Y cells, which model aspects of human dopaminergic neurons, forced expression of a channel-dead variant of TRPM7 causes cell death. In summary, a forward genetic screen in zebrafish has revealed that both melanocytes and dopaminergic neurons depend on the ion channel Trpm7. The mechanistic underpinning of this dependence requires further investigation.


Assuntos
Diferenciação Celular/fisiologia , Neurônios Dopaminérgicos/citologia , Atividade Motora/genética , Proteínas Serina-Treonina Quinases/genética , Canais de Cátion TRPM/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento , 1-Metil-4-fenilpiridínio/toxicidade , Análise de Variância , Animais , Linhagem Celular , Primers do DNA/genética , Desferroxamina/farmacologia , Eletrorretinografia , Larva/crescimento & desenvolvimento , Melanócitos/metabolismo , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Mutação/genética , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tirosina 3-Mono-Oxigenase/metabolismo , Peixe-Zebra/genética
14.
Biochem J ; 445(3): 441-8, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22587440

RESUMO

The TRPM7 (transient receptor potential melastatin 7) channel has been shown to play a pivotal role in cell survival during brain ischaemia as well as in the survival of other cell types challenged with apoptotic stimuli. Ca(2+) is thought to be central to the channel's ability to regulate ROS (reactive oxygen species) production. However, channel-mediated entry of Mg(2+) and Zn(2+) have also been implicated in cell death. In the present study, we show that depletion of TRPM7 by RNA interference in fibroblasts increases cell resistance to apoptotic stimuli by decreasing ROS levels in an Mg(2+)-dependent manner. Depletion of TRPM7 lowered cellular Mg(2+), decreased the concentration of ROS and lessened p38 MAPK (mitogen-activated protein kinase) and JNK (c-Jun N-terminal kinase) activation as well as decreased caspase 3 activation and PARP [poly(ADP-ribose) polymerase] cleavage in response to apoptotic stimuli. Re-expression of TRPM7 or of a kinase-inactive mutant of TRPM7 in TRPM7-knockdown cells increased cellular Mg(2+) and ROS levels, as did expression of the Mg(2+) transporter SLC41A2 (solute carrier family 41 member 2). In addition, expression of SLC41A2 increased the sensitivity of TRPM7-knockdown cells to apoptotic stimuli and boosted ROS generation in response to cell stress. Taken together, these data uncover an essential role for Mg(2+) in TRPM7's control of cell survival and in the regulation of cellular ROS levels.


Assuntos
Magnésio/metabolismo , Canais de Cátion TRPM/metabolismo , Animais , Apoptose , Proteínas de Transporte de Cátions/metabolismo , Sobrevivência Celular , Técnicas de Silenciamento de Genes , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Estresse Oxidativo , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células Swiss 3T3 , Canais de Cátion TRPM/antagonistas & inibidores , Canais de Cátion TRPM/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
Sci Rep ; 1: 146, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22180838

RESUMO

TRPM6 is crucial for human Mg2+ homeostasis as patients carrying TRPM6 mutations develop hypomagnesemia and secondary hypocalcemia (HSH). However, the activation mechanism of TRPM6 has remained unknown. Here we demonstrate that phosphatidylinositol-4,5-bisphophate (PIP2) controls TRPM6 activation and Mg2+ influx. Stimulation of PLC-coupled M1-receptors to deplete PIP2 potently inactivates TRPM6. Translocation of over-expressed 5-phosphatase to cell membrane to specifically hydrolyze PIP2 also completely inhibits TRPM6. Moreover, depolarization-induced-activation of the voltage-sensitive-phosphatase (Ci-VSP) simultaneously depletes PIP2 and inhibits TRPM6. PLC-activation induced PIP2-depletion not only inhibits TRPM6, but also abolishes TRPM6-mediated Mg2+ influx.Furthermore, neutralization of basic residues in the TRP domain leads to nonfunctional or dysfunctional mutants with reduced activity by PIP2, suggesting that they are likely to participate in interactions with PIP2.Our data indicate that PIP2 is required for TRPM6 channel function; hydrolysis of PIP2 by PLC-coupled hormones/agonists may constitute an important pathway for TRPM6 gating, and perhaps Mg2+ homeostasis.


Assuntos
Magnésio/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Canais de Cátion TRPM/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Linhagem Celular , Células HEK293 , Homeostase , Humanos , Ativação do Canal Iônico , Cinética , Dados de Sequência Molecular , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Serina-Treonina Quinases , Estrutura Terciária de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Canais de Cátion TRPM/antagonistas & inibidores , Canais de Cátion TRPM/genética , Fosfolipases Tipo C/metabolismo
16.
PLoS One ; 6(8): e24291, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21897875

RESUMO

Increasing evidence has pointed to activated type I interferon signaling in tumors. However, the molecular basis for such activation and its role in tumorigenesis remain unclear. In the current studies, we report that activation of type I interferon (IFN) signaling in tumor cells is primarily due to elevated secretion of the type I interferon, IFN-ß. Studies in oncogene-transformed cells suggest that oncogenes such as Ras and Src can activate IFN-ß signaling. Significantly, elevated IFN-ß signaling in Ras-transformed mammary epithelial MCF-10A cells was shown to contribute to Ras transformation as evidenced by morphological changes, anchorage-independent growth, and migratory properties. Our results demonstrate for the first time that the type I IFN, IFN-ß, contributes to Ras transformation and support the notion that oncogene-induced cytokines play important roles in oncogene transformation.


Assuntos
Transformação Celular Neoplásica , Interferon beta/metabolismo , Proteína Oncogênica p21(ras)/metabolismo , Transdução de Sinais , Animais , Linhagem Celular Tumoral , Movimento Celular , Citocinas/metabolismo , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Genes src/genética , Humanos , Camundongos , Células NIH 3T3 , Ratos , Ubiquitinas/metabolismo
17.
Development ; 138(10): 2035-47, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21471152

RESUMO

Neural tube closure is a critical morphogenetic event that is regulated by dynamic changes in cell shape and behavior. Although previous studies have uncovered a central role for the non-canonical Wnt signaling pathway in neural tube closure, the underlying mechanism remains poorly resolved. Here, we show that the missing in metastasis (MIM; Mtss1) protein, previously identified as a Hedgehog response gene and actin and membrane remodeling protein, specifically binds to Daam1 and couples non-canonical Wnt signaling to neural tube closure. MIM binds to a conserved domain within Daam1, and this interaction is positively regulated by Wnt stimulation. Spatial expression of MIM is enriched in the anterior neural plate and neural folds, and depletion of MIM specifically inhibits anterior neural fold closure without affecting convergent extension movements or mesoderm cell fate specification. Particularly, we find that MIM is required for neural fold elevation and apical constriction along with cell polarization and elongation in both the superficial and deep layers of the anterior neural plate. The function of MIM during neural tube closure requires both its membrane-remodeling domain and its actin-binding domain. Finally, we show that the effect of MIM on neural tube closure is not due to modulation of Hedgehog signaling in the Xenopus embryo. Together, our studies define a morphogenetic pathway involving Daam1 and MIM that transduces non-canonical Wnt signaling for the cytoskeletal changes and membrane dynamics required for vertebrate neural tube closure.


Assuntos
Tubo Neural/embriologia , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriologia , Xenopus laevis/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Sequência Conservada , Citoesqueleto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Dados de Sequência Molecular , Tubo Neural/metabolismo , Ligação Proteica , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteínas de Xenopus/genética , Xenopus laevis/genética
18.
Biochem J ; 434(3): 513-21, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-21208190

RESUMO

TRPM7 (transient receptor potential melastatin 7) is a Ca²+- and Mg²+-permeant ion channel in possession of its own kinase domain. As a kinase, the protein has been linked to the control of actomyosin contractility, whereas the channel has been found to regulate cell adhesion as well as cellular Mg²+ homoeostasis. In the present study we show that depletion of TRPM7 by RNA interference in fibroblasts alters cell morphology, the cytoskeleton, and the ability of cells to form lamellipodia and to execute polarized cell movements. A pulldown-purification assay revealed that knockdown of TRPM7 prevents cells from activating Rac and Cdc42 (cell division cycle 42) when stimulated to migrate into a cellular wound. Re-expression of TRPM7 reverses these phenotypic changes, as does, unexpectedly, expression of a kinase-inactive mutant of TRPM7. Surprisingly, expression of the Mg²+ transporter SLC41A2 (solute carrier family 41 member 2) is also effective in restoring the change in cell morphology, disruption of the cytoskeleton and directional cell motility caused by depletion of the channel-kinase. The results of the present study uncover an essential role for Mg²+ in the control of TRPM7 over the cytoskeleton and its ability to regulate polarized cell movements.


Assuntos
Movimento Celular , Polaridade Celular , Fibroblastos/fisiologia , Canais de Cátion TRPM/fisiologia , Células 3T3 , Actomiosina/fisiologia , Adenoviridae/genética , Animais , Proteínas de Transporte de Cátions/biossíntese , Cátions Bivalentes , Adesão Celular , Citoesqueleto/metabolismo , Adesões Focais/metabolismo , Técnicas de Silenciamento de Genes , Vetores Genéticos , Magnésio/fisiologia , Camundongos , Interferência de RNA , Canais de Cátion TRPM/biossíntese , Canais de Cátion TRPM/genética
19.
Dev Biol ; 350(2): 348-57, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21145885

RESUMO

During gastrulation, cells in the dorsal marginal zone polarize, elongate, align and intercalate to establish the physical body axis of the developing embryo. Here we demonstrate that the bifunctional channel-kinase TRPM7 is specifically required for vertebrate gastrulation. TRPM7 is temporally expressed maternally and throughout development, and is spatially enriched in tissues undergoing convergent extension during gastrulation. Functional studies reveal that TRPM7's ion channel, but not its kinase domain, specifically affects cell polarity and convergent extension movements during gastrulation, independent of mesodermal specification. During gastrulation, the non-canonical Wnt pathway via Dishevelled (Dvl) orchestrates the activities of the GTPases Rho and Rac to control convergent extension movements. We find that TRPM7 functions synergistically with non-canonical Wnt signaling to regulate Rac activity. The phenotype caused by depletion of the Ca(2+)- and Mg(2+)-permeant TRPM7 is suppressed by expression of a dominant negative form of Rac, as well as by Mg(2+) supplementation or by expression of the Mg(2+) transporter SLC41A2. Together, these studies demonstrate an essential role for the ion channel TRPM7 and Mg(2+) in Rac-dependent polarized cell movements during vertebrate gastrulation.


Assuntos
Desenvolvimento Embrionário , Gastrulação , Canais de Cátion TRPM/fisiologia , Proteínas de Xenopus/fisiologia , Xenopus laevis/embriologia , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Movimento Celular , Proteínas Desgrenhadas , Magnésio/farmacologia , Mesoderma/fisiologia , Morfogênese , Fosfoproteínas/fisiologia , Canais de Cátion TRPM/análise , Proteínas de Xenopus/análise , Proteínas rac de Ligação ao GTP/fisiologia
20.
Curr Pharm Biotechnol ; 12(1): 42-53, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20932259

RESUMO

Unique among ion channels, TRPM6 and TRPM7 garnered much interest upon their discovery as the first ion channels to possess their own kinase domain. Soon after their identification, the two proteins were quickly linked to the regulation of magnesium homeostasis. However, study of their physiological functions in mouse and zebrafish have revealed expanding roles for these channel-kinases that include skeletogenesis and melanopore formation, thymopoiesis, cell adhesion, and neural fold closure during early development. In addition, mutations in the TRPM6 gene constitute the underlying genetic defect in hypomagnesemia with secondary hypocalcemia, a rare autosomal-recessive disease characterized by low serum magnesium accompanied by hypocalcemia. Depletion of TRPM7 expression in brain, on the other hand, proved successful in mitigating much of the cellular devastation that accompanies oxygen-glucose deprivation during ischemia. The aim of this review is to summarize the data emerging from molecular genetic, biochemical, electrophysiological, and pharmacological studies of these unique channel-kinases.


Assuntos
Proteínas Nucleares/metabolismo , Proteínas Quinases/metabolismo , Canais de Cátion TRPM/metabolismo , Animais , Cátions/metabolismo , Processos de Crescimento Celular , Modelos Animais de Doenças , Células HEK293 , Homeostase , Humanos , Canais Iônicos/metabolismo , Magnésio/sangue , Magnésio/metabolismo , Camundongos , Proteínas Nucleares/agonistas , Proteínas Nucleares/antagonistas & inibidores , Proteínas Serina-Treonina Quinases , Canais de Cátion TRPM/agonistas , Canais de Cátion TRPM/antagonistas & inibidores , Canais de Cátion TRPM/genética , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...