Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(26): e2402538121, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38905240

RESUMO

Intracellular sensors detect changes in levels of essential metals to initiate homeostatic responses. But, a mammalian manganese (Mn) sensor is unknown, representing a major gap in understanding of Mn homeostasis. Using human-relevant models, we recently reported that: 1) the primary homeostatic response to elevated Mn is upregulation of hypoxia-inducible factors (HIFs), which increases expression of the Mn efflux transporter SLC30A10; and 2) elevated Mn blocks the prolyl hydroxylation of HIFs by prolyl hydroxylase domain (PHD) enzymes, which otherwise targets HIFs for degradation. Thus, the mammalian mechanism for sensing elevated Mn likely relates to PHD inhibition. Moreover, 1) Mn substitutes for a catalytic iron (Fe) in PHD structures; and 2) exchangeable cellular levels of Fe and Mn are comparable. Therefore, we hypothesized that elevated Mn directly inhibits PHD by replacing its catalytic Fe. In vitro assays using catalytically active PHD2, the primary PHD isoform, revealed that Mn inhibited, and Fe supplementation rescued, PHD2 activity. However, a mutation in PHD2 (D315E) that selectively reduced Mn binding without substantially impacting Fe binding or enzymatic activity resulted in complete insensitivity of PHD2 to Mn in vitro. Additionally, hepatic cells expressing full-length PHD2D315E were less sensitive to Mn-induced HIF activation and SLC30A10 upregulation than PHD2wild-type. These results: 1) define a fundamental Mn sensing mechanism for controlling Mn homeostasis-elevated Mn inhibits PHD2, which functions as a Mn sensor, by outcompeting its catalytic Fe, and PHD2 inhibition activates HIF signaling to up-regulate SLC30A10; and 2) identify a unique mode of metal sensing that may have wide applicability.


Assuntos
Homeostase , Prolina Dioxigenases do Fator Induzível por Hipóxia , Manganês , Humanos , Manganês/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia/genética , Proteínas de Transporte de Cátions/metabolismo , Proteínas de Transporte de Cátions/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Células HEK293 , Ferro/metabolismo
2.
Proc Natl Acad Sci U S A ; 121(40): e2405615121, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39312661

RESUMO

Stimulus-specific adaptation is a hallmark of sensory processing in which a repeated stimulus results in diminished successive neuronal responses, but a deviant stimulus will still elicit robust responses from the same neurons. Recent work has established that synaptically released zinc is an endogenous mechanism that shapes neuronal responses to sounds in the auditory cortex. Here, to understand the contributions of synaptic zinc to deviance detection of specific neurons, we performed wide-field and 2-photon calcium imaging of multiple classes of cortical neurons. We find that intratelencephalic (IT) neurons in both layers 2/3 and 5 as well as corticocollicular neurons in layer 5 all demonstrate deviance detection; however, we find a specific enhancement of deviance detection in corticocollicular neurons that arises from ZnT3-dependent synaptic zinc in layer 2/3 IT neurons. Genetic deletion of ZnT3 from layer 2/3 IT neurons removes the enhancing effects of synaptic zinc on corticocollicular neuron deviance detection and results in poorer acuity of detecting deviant sounds by behaving mice.


Assuntos
Córtex Auditivo , Neurônios , Sinapses , Zinco , Animais , Zinco/metabolismo , Córtex Auditivo/metabolismo , Córtex Auditivo/fisiologia , Camundongos , Sinapses/metabolismo , Sinapses/fisiologia , Neurônios/metabolismo , Neurônios/fisiologia , Proteínas de Transporte de Cátions/metabolismo , Proteínas de Transporte de Cátions/genética , Estimulação Acústica , Camundongos Knockout , Percepção Auditiva/fisiologia , Camundongos Endogâmicos C57BL , Masculino
3.
J Neurosci ; 44(28)2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38830758

RESUMO

Shank3 is a synaptic scaffolding protein that assists in tethering and organizing structural proteins and glutamatergic receptors in the postsynaptic density of excitatory synapses. The localization of Shank3 at excitatory synapses and the formation of stable Shank3 complexes is regulated by the binding of zinc to the C-terminal sterile-alpha-motif (SAM) domain of Shank3. Mutations in the SAM domain of Shank3 result in altered synaptic function and morphology, and disruption of zinc in synapses that express Shank3 leads to a reduction of postsynaptic proteins important for synaptic structure and function. This suggests that zinc supports the localization of postsynaptic proteins via Shank3. Many regions of the brain are highly enriched with free zinc inside glutamatergic vesicles at presynaptic terminals. At these synapses, zinc transporter 3 (ZnT3) moves zinc into vesicles where it is co-released with glutamate. Alterations in ZnT3 are implicated in multiple neurodevelopmental disorders, and ZnT3 knock-out (KO) mice-which lack synaptic zinc-show behavioral deficits associated with autism spectrum disorder and schizophrenia. Here we show that male and female ZnT3 KO mice have smaller dendritic spines and miniature excitatory postsynaptic current amplitudes than wildtype (WT) mice in the auditory cortex. Additionally, spine size deficits in ZnT3 KO mice are restricted to synapses that express Shank3. In WT mice, synapses that express both Shank3 and ZnT3 have larger spines compared to synapses that express Shank3 but not ZnT3. Together these findings suggest a mechanism whereby presynaptic ZnT3-dependent zinc supports postsynaptic structure and function via Shank3 in a synapse-specific manner.


Assuntos
Córtex Auditivo , Proteínas de Transporte de Cátions , Espinhas Dendríticas , Proteínas do Tecido Nervoso , Sinapses , Animais , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/genética , Sinapses/metabolismo , Espinhas Dendríticas/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Proteínas de Transporte de Cátions/genética , Córtex Auditivo/metabolismo , Feminino , Masculino , Camundongos Knockout , Proteínas de Transporte/metabolismo , Proteínas de Transporte/genética , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/metabolismo , Proteínas dos Microfilamentos/genética , Potenciais Pós-Sinápticos Excitadores/fisiologia
4.
J Biol Chem ; 300(3): 105732, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38336290

RESUMO

The manganese (Mn) export protein SLC30A10 is essential for Mn excretion via the liver and intestines. Patients with SLC30A10 deficiency develop Mn excess, dystonia, liver disease, and polycythemia. Recent genome-wide association studies revealed a link between the SLC30A10 variant T95I and markers of liver disease. The in vivo relevance of this variant has yet to be investigated. Using in vitro and in vivo models, we explore the impact of the T95I variant on SLC30A10 function. While SLC30A10 I95 expressed at lower levels than T95 in transfected cell lines, both T95 and I95 variants protected cells similarly from Mn-induced toxicity. Adeno-associated virus 8-mediated expression of T95 or I95 SLC30A10 using the liver-specific thyroxine binding globulin promoter normalized liver Mn levels in mice with hepatocyte Slc30a10 deficiency. Furthermore, Adeno-associated virus-mediated expression of T95 or I95 SLC30A10 normalized red blood cell parameters and body weights and attenuated Mn levels and differential gene expression in livers and brains of mice with whole body Slc30a10 deficiency. While our in vivo data do not indicate that the T95I variant significantly compromises SLC30A10 function, it does reinforce the notion that the liver is a key site of SLC30A10 function. It also supports the idea that restoration of hepatic SLC30A10 expression is sufficient to attenuate phenotypes in SLC30A10 deficiency.


Assuntos
Substituição de Aminoácidos , Proteínas de Transporte de Cátions , Dependovirus , Fígado , Manganês , Mutação , Animais , Camundongos , Peso Corporal , Encéfalo/metabolismo , Proteínas de Transporte de Cátions/deficiência , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Linhagem Celular , Dependovirus/genética , Eritrócitos , Estudo de Associação Genômica Ampla , Hepatócitos/metabolismo , Fígado/citologia , Fígado/metabolismo , Hepatopatias/genética , Hepatopatias/metabolismo , Manganês/metabolismo , Intoxicação por Manganês/metabolismo , Fenótipo , Regiões Promotoras Genéticas , Globulina de Ligação a Tiroxina/genética
5.
Genomics ; 116(1): 110768, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38128703

RESUMO

The myometrium, composed of the inner circular muscle (CM) and outer longitudinal muscle (LM), is crucial in establishing and maintaining early pregnancy. However, the molecular mechanisms involved are not well understood. In this study, we identified the transcriptomic features of the CM and LM collected from the mesometrial (M) and anti-mesometrial (AM) sides of the pig uterus on day 18 of pregnancy during the placentation initiation phase. Some genes in the cellular zinc ion level regulatory pathways (MT-1A, MT-1D, MT-2B, SLC30A2, and SLC39A2) were spatially and highly enriched in uterine CM at the mesometrial side. In addition, the histone modification profiles of H3K27ac and H3K4me3 in uterine CM and LM collected from the mesometrial side were characterized. Genomic regions associated with the expression of genes regulating the cellular zinc ion level were detected. Moreover, six highly linked variants in the H3K27ac-enriched region of the pig SLC30A2 gene were identified and found to be significantly associated with the total number born at the second parity (P < 0.05). In conclusion, the genes in the pathways of cellular zinc homeostasis and their regulatory elements identified have implications for pig reproduction trait improvement and warrant further investigations.


Assuntos
Epigenômica , Miométrio , Gravidez , Feminino , Suínos , Animais , Miométrio/metabolismo , Útero/metabolismo , Homeostase , Zinco/metabolismo
6.
J Reprod Dev ; 70(5): 338-342, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39048372

RESUMO

Zinc is an essential trace element for various physiological functions, including reproduction. The influx/efflux of zinc ions is regulated by zinc transporters (Zip1-14 and ZnT1-8, 10). However, the precise roles of zinc transporters and zinc dynamics in reproductive functions are unknown. In this study, ZnT3/Slc30a3 gene knockout (KO) mice were used to analyze the role of ZnT3. In ZnT3 KO mice, intracellular zinc ions in oocytes/zygotes were significantly reduced compared to those in controls, and free zinc ions did not accumulate in the oocyte cytoplasm. However, fertilization of these oocytes and the average litter size were comparable to those of control mice. Our results suggest that ZnT3 plays an important role in the accumulation of zinc ions in oocytes but not in the developmental ability of mice. ZnT3 KO mice will be useful for examining zinc dynamics in oocytes and other tissues.


Assuntos
Proteínas de Transporte de Cátions , Camundongos Knockout , Oócitos , Zinco , Animais , Oócitos/metabolismo , Zinco/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Proteínas de Transporte de Cátions/genética , Camundongos , Feminino , Masculino , Tamanho da Ninhada de Vivíparos
7.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34433664

RESUMO

The trace element zinc is essential for many aspects of physiology. The mitochondrion is a major Zn2+ store, and excessive mitochondrial Zn2+ is linked to neurodegeneration. How mitochondria maintain their Zn2+ homeostasis is unknown. Here, we find that the SLC-30A9 transporter localizes on mitochondria and is required for export of Zn2+ from mitochondria in both Caenorhabditis elegans and human cells. Loss of slc-30a9 leads to elevated Zn2+ levels in mitochondria, a severely swollen mitochondrial matrix in many tissues, compromised mitochondrial metabolic function, reductive stress, and induction of the mitochondrial stress response. SLC-30A9 is also essential for organismal fertility and sperm activation in C. elegans, during which Zn2+ exits from mitochondria and acts as an activation signal. In slc-30a9-deficient neurons, misshapen mitochondria show reduced distribution in axons and dendrites, providing a potential mechanism for the Birk-Landau-Perez cerebrorenal syndrome where an SLC30A9 mutation was found.


Assuntos
Proteínas de Transporte de Cátions/farmacologia , Proteínas de Ciclo Celular/farmacologia , Mitocôndrias/metabolismo , Fatores de Transcrição/farmacologia , Zinco/metabolismo , Animais , Axônios/metabolismo , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/fisiologia , Proteínas de Transporte de Cátions/genética , Proteínas de Ciclo Celular/genética , Dendritos/metabolismo , Feminino , Técnicas de Inativação de Genes , Células HeLa , Homeostase , Humanos , Masculino , Potencial da Membrana Mitocondrial , Mutação , Espermatozoides/fisiologia , Fatores de Transcrição/genética
8.
J Struct Biol ; 215(1): 107926, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36464198

RESUMO

Zinc transporter 8 (ZnT8) is mainly expressed in pancreatic islet ß cells and is responsible for H+-coupled uptake (antiport) of Zn2+ into the lumen of insulin secretory granules. Structures of human ZnT8 and its prokaryotic homolog YiiP have provided structural basis for constructing a plausible transport cycle for Zn2+. However, the mechanistic role that protons play in the transport process remains unclear. Here we present a lumen-facing cryo-EM structure of ZnT8 from Xenopus tropicalis (xtZnT8) in the presence of Zn2+ at a luminal pH (5.5). Compared to a Zn2+-bound xtZnT8 structure at a cytosolic pH (7.5), the low-pH structure displays an empty transmembrane Zn2+-binding site with a disrupted coordination geometry. Combined with a Zn2+-binding assay our data suggest that protons may disrupt Zn2+ coordination at the transmembrane Zn2+-binding site in the lumen-facing state, thus facilitating Zn2+ release from ZnT8 into the lumen.


Assuntos
Eucariotos , Prótons , Humanos , Microscopia Crioeletrônica , Concentração de Íons de Hidrogênio , Zinco
9.
Am J Physiol Gastrointest Liver Physiol ; 325(3): G251-G264, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37461848

RESUMO

Manganese (Mn) is essential but neurotoxic at elevated levels. Under physiological conditions, Mn is primarily excreted by the liver, with the intestines playing a secondary role. Recent analyses of tissue-specific Slc30a10 or Slc39a14 knockout mice (SLC30A10 and SLC39A14 are Mn transporters) revealed that, under physiological conditions: 1) excretion of Mn by the liver and intestines is a major pathway that regulates brain Mn; and surprisingly, 2) the intestines compensate for loss of hepatic Mn excretion in controlling brain Mn. The unexpected importance of the intestines in controlling physiological brain Mn led us to determine the role of hepatic and intestinal Mn excretion in regulating brain Mn during elevated Mn exposure. We used liver- or intestine-specific Slc30a10 knockout mice as models to inhibit hepatic or intestinal Mn excretion. Compared with littermates, both knockout strains exhibited similar increases in brain Mn after elevated Mn exposure in early or later life. Thus, unlike physiological conditions, both hepatic and intestinal Mn excretion are required to control brain Mn during elevated Mn exposure. However, brain Mn levels of littermates and both knockout strains exposed to elevated Mn only in early life were normalized in later life. Thus, hepatic and intestinal Mn excretion play compensatory roles in clearing brain Mn accumulated by early life Mn exposure. Finally, neuromotor assays provided evidence consistent with a role for hepatic and intestinal Mn excretion in functionally modulating Mn neurotoxicity during Mn exposure. Put together, these findings substantially enhance understanding of the regulation of brain Mn by excretion.NEW & NOTEWORTHY This article shows that, in contrast with expectations from prior studies and physiological conditions, excretion of manganese by the intestines and liver is equally important in controlling brain manganese during human-relevant manganese exposure. The results provide foundational insights about the interorgan mechanisms that control brain manganese homeostasis at the organism level and have important implications for the development of therapeutics to treat manganese-induced neurological disease.


Assuntos
Proteínas de Transporte de Cátions , Manganês , Camundongos , Animais , Humanos , Manganês/toxicidade , Proteínas de Transporte de Cátions/metabolismo , Fígado/metabolismo , Camundongos Knockout , Encéfalo/metabolismo
10.
Biochem Genet ; 61(6): 2203-2221, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37103601

RESUMO

Genetics plays a role in the development of gestational diabetes mellitus (GDM), which poses serious risks to pregnant women and their children. Several studies have demonstrated a link between GDM susceptibility and rs13266634 C/T polymorphism in SLC30A8 gene and rs1111875 C/T and rs5015480 C/T, which are located near the linkage disequilibrium block containing the IDE, HHEX, and KIF11 genes. However, the results are conflicting. Therefore, we aimed to investigate the association between susceptibility to GDM and HHEX and SLC30A8 gene polymorphisms. PubMed, Web of Science, EBSCO, CNKI, Wanfang Data, VIP, and SCOPUS were used to search for research articles. The quality of the selected literature was evaluated using the Newcastle-Ottawa scale. A meta-analysis was performed using Stata 15.1. Allelic, dominant, recessive, homozygote, and heterozygote models were used for the analysis. Nine articles with 15 studies were included. (1) Four studies about HHEX rs1111875 showed that the C allele was associated with the susceptibility to GDM; (2) three studies on HHEX rs5015480 indicated that the C allele in rs5015480 was significantly associated with GDM; (3) eight studies about SLC30A8 rs13266634 showed that the C allele was significantly associated with the susceptibility to GDM; and (4) a subgroup analysis showed that the rs5015480 polymorphism in HHEX and rs13266634 polymorphism in SLC30A8 gene were associated with GDM susceptibility in Asians. The meta-analysis provided evidence that the C allele in rs1111875 and rs5015480 in HHEX and rs13266634 in SLC30A8 can increase the risk of GDM.PROSPERO registration number CRD42022342280.


Assuntos
Diabetes Mellitus Tipo 2 , Diabetes Gestacional , Criança , Humanos , Feminino , Gravidez , Diabetes Gestacional/genética , Genótipo , Diabetes Mellitus Tipo 2/genética , Polimorfismo de Nucleotídeo Único , Alelos , Predisposição Genética para Doença , Transportador 8 de Zinco/genética , Fatores de Transcrição/genética , Proteínas de Homeodomínio/genética
11.
Am J Physiol Gastrointest Liver Physiol ; 322(1): G79-G92, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34786983

RESUMO

The essential metal manganese (Mn) induces incurable neurotoxicity at elevated levels that manifests as parkinsonism in adults and fine motor and executive function deficits in children. Studies on Mn neurotoxicity have largely focused on the role and mechanisms of disease induced by elevated Mn exposure from occupational or environmental sources. In contrast, the critical role of excretion in regulating Mn homeostasis and neurotoxicity has received less attention although 1) studies on Mn excretion date back to the 1920s; 2) elegant radiotracer Mn excretion assays in the 1940s to 1960s established the routes of Mn excretion; and 3) studies on patients with liver cirrhosis in the 1990s to 2000s identified an association between decreased Mn excretion and the risk of developing Mn-induced parkinsonism in the absence of elevated Mn exposure. Notably, the last few years have seen renewed interest in Mn excretion largely driven by the discovery that hereditary Mn neurotoxicity due to mutations in SLC30A10 or SLC39A14 is caused, at least in part, by deficits in Mn excretion. Quite remarkably, some of the recent results on SLC30A10 and SLC39A14 provide explanations for observations made ∼40-50 years ago. The goal of the current review is to integrate the historic studies on Mn excretion with more contemporary recent work and provide a comprehensive state-of-the-art overview of Mn excretion and its role in regulating Mn homeostasis and neurotoxicity. A related goal is to discuss the significance of some of the foundational studies on Mn excretion so that these highly consequential earlier studies remain influential in the field.


Assuntos
Homeostase/efeitos dos fármacos , Manganês/toxicidade , Metais/metabolismo , Proteínas de Transporte de Cátions/efeitos dos fármacos , Proteínas de Transporte de Cátions/genética , Humanos , Mutação/efeitos dos fármacos , Mutação/genética , Transtornos Parkinsonianos/tratamento farmacológico
12.
Biochem Biophys Res Commun ; 634: 175-181, 2022 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-36244116

RESUMO

Oxygen is essential for aerobic organisms, but generates reactive oxygen species (ROS), which can cause cellular dysfunction by damaging cellular molecules. Many genes are involved in the regulation of ROS; however, much attention has not focused on them. To identify these genes, we screened for mutants with an altered sensitivity to oxidative stress in the nematode Caenorhabditis elegans. We isolated a novel mutant, oxy-7(qa5004) which showed an increased sensitivity to ROS in C. elegans. oxy-7 showed increased production of ROS and decreased longevity due to its increased oxidative stress. Genetic analysis revealed that oxy-7 has a causative mutation in Y71H2AM.9, a homologue of SLC30A9 which encodes a zinc transporter in mitochondria. We further showed that knockdown of human SLC30A9 caused increased ROS production in human cells as well. These results suggested an important role of mitochondrial zinc homeostasis in the regulation of ROS.


Assuntos
Proteínas de Caenorhabditis elegans , Proteínas de Transporte de Cátions , Nematoides , Animais , Humanos , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Estresse Oxidativo/genética , Longevidade/genética , Nematoides/metabolismo , Mutação , Fatores de Transcrição/genética , Proteínas de Ciclo Celular/genética , Proteínas de Transporte de Cátions/genética
13.
Mol Carcinog ; 61(5): 454-471, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35049094

RESUMO

Prostate cancer (PCa) initiation and progression uniquely modify the prostate milieu to aid unrestrained cell proliferation. One salient modification is the loss of the ability of prostate epithelial cells to accumulate high concentrations of zinc; however, molecular alterations associated with loss of zinc accumulating capability in malignant prostate cells remain poorly understood. Herein, we assessed the stage-specific expression of zinc transporters (ZNTs) belonging to the ZNT (SLC30A) and Zrt- and Irt-like protein (ZIP) (SLC39A) solute-carrier family in the prostate tissues of different genetically engineered mouse models (GEMM) of PCa (TMPRSS2-ERG.Ptenflox/flox , Hi-Myc+/- , and transgenic adenocarcinoma of mouse prostate), their age-matched wild-type controls, and 104 prostate core biopsies from human patients with different pathological lesions. Employing immunohistochemistry, differences in the levels of protein expression and spatial distribution of ZNT were evaluated as a function of the tumor stage. Results indicated that the expression of zinc importers (ZIP1, ZIP2, and ZIP3), which function to sequester zinc from circulation and prostatic fluid, was low to negligible in the membranes of the malignant prostate cells in both GEMM and human prostate tissues. Regarding zinc exporters (ZNT1, ZNT2, ZNT9, and ZNT10) that export excess zinc into the extracellular spaces or intracellular organelles, their expression was low in normal prostate glands of mice and humans; however, it was significantly upregulated in prostate adenocarcinoma lesions in GEMM and PCa patients. Together, our findings provide new insights into altered expression of ZNTs during the progression of PCa and indicate that changes in zinc homeostasis could possibly be an early-initiation event during prostate tumorigenesis and a likely prevention/intervention target.


Assuntos
Adenocarcinoma , Proteínas de Transporte de Cátions , Neoplasias da Próstata , Adenocarcinoma/genética , Carcinogênese/genética , Proteínas de Transporte , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Transformação Celular Neoplásica , Humanos , Masculino , Próstata/metabolismo , Neoplasias da Próstata/genética , Zinco/metabolismo
14.
Am J Med Genet A ; 188(8): 2360-2366, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35751429

RESUMO

Joubert syndrome (JS), a well-established ciliopathy, is characterized by the distinctive molar tooth sign on brain MRI, ataxia, and neurodevelopmental features. Other manifestations can include polydactyly, accessory frenula, renal, or liver disease. Here, we report individuals meeting criteria for JS with de novo heterozygous variants in SLC30A7 (Chr1p21.2). The first individual is a female with history of unilateral postaxial polydactyly, classic molar tooth sign on MRI, macrocephaly, ataxia, ocular motor apraxia, neurodevelopmental delay, and precocious puberty. Exome sequencing detected a de novo heterozygous missense variant in SLC30A7: NM_133496.5: c.407 T > C, (p.Val136Ala). The second individual had bilateral postaxial polydactyly, molar tooth sign, macrocephaly, developmental delay, and an extra oral frenulum. A de novo deletion-insertion variant in SLC30A7, c.490_491delinsAG (p.His164Ser) was found. Both de novo variants affect highly conserved residues. Variants were not identified in known Joubert genes for either case. SLC30A7 has not yet been associated with a human phenotype. The SLC30 family of zinc transporters, like SLC30A7, permit cellular efflux of zinc, and although it is expressed in the brain its functions remain unknown. Published data from proteomic studies support SLC30A7 interaction with TCTN3, another protein associated with JS. The potential involvement of such genes in primary cilia suggest a role in Sonic Hedgehog signaling. SLC30A7 is a candidate JS-associated gene. Future work could be directed toward further characterization of SLC30A7 variants and understanding its function.


Assuntos
Anormalidades Múltiplas , Proteínas de Transporte de Cátions/genética , Anormalidades do Olho , Doenças Renais Císticas , Megalencefalia , Polidactilia , Anormalidades Múltiplas/diagnóstico , Anormalidades Múltiplas/genética , Ataxia , Cerebelo/anormalidades , Cerebelo/diagnóstico por imagem , Anormalidades do Olho/diagnóstico , Anormalidades do Olho/genética , Feminino , Proteínas Hedgehog , Humanos , Doenças Renais Císticas/diagnóstico , Doenças Renais Císticas/genética , Proteômica , Retina/anormalidades , Zinco
15.
Biometals ; 35(5): 955-965, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35834148

RESUMO

This study is to examine the effects of single nucleotide polymorphisms (SNPs) of SLC30A and SLC39A on seminal plasma zinc concentration. Blood and seminal plasma samples were collected from outpatients. SNPs of zinc transporters were analyzed by next Generation sequencing technology, and seminal plasma zinc concentration were determined by inductively coupled plasma optical emission spectrometry. Our date showed nine SNPs (SLC30A8 rs2466295, rs2466294, SLC30A10 c.-160 C>G, SLC39A8 rs9331, rs9705, rs151392, rs151393, SLC39A11 rs9912126, SLC39A14 rs1051708) were significantly associated with seminal plasma zinc concentration, and 14 SNPs (SLC30A8 rs2466295, rs2466294, SLC30A10 c.-160 C>G, SLC39A6 rs148550301, SLC39A8 rs9331, rs9705, rs151392, rs151393, SLC39A11 rs9912126, rs61736066, rs36041371 and SLC39A14 rs1051708, rs76963096, rs17060854) were found to be significantly associated with total zinc per ejaculate. The seminal plasma zinc concentrations and total zinc per ejaculate were associated with the number of SNPs, and decreased significantly when five SNPs (SLC39A8 rs9331, rs9705, rs151392, rs151393 and SLC39A14 rs1051708) were a combination of homozygous genotype. Our findings suggest that different zinc transporter SNPs may significantly affect seminal plasma zinc levels.


Assuntos
Polimorfismo de Nucleotídeo Único , Sêmen , Proteínas de Transporte , Humanos , Polimorfismo de Nucleotídeo Único/genética , Zinco , Transportador 8 de Zinco/genética
16.
Neurol Sci ; 43(2): 1441-1445, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34993656

RESUMO

Exogenous manganese (Mn) intoxication leads only to neurotoxicity, whereas inherited hypermanganesemia additionally can cause cirrhosis and polycythemia. We report two affected siblings in a family from South India with severe dysarthria, without dysphagia, generalized dystonia, and characteristic "cock-walk" gait which are clinical clues. Genetic study showed homozygous mutation in the first exon of solute carrier family 30 member 10 (SLC30A10) gene (c.134T>C) confirming the diagnosis of inherited hypermanganesaemia with dystonia 1 (HMNDYT1). Characteristic brain MRI finding is involvement of pontine tegmentum on T1 axial images (due to affliction of central tegmental tract [CTT]) with sparing of ventral pons giving rise to "horseshoe moustache" sign. Symmetric hyperintensities in dentate nucleus, globus pallidus, and putamen while relatively sparing caudate nucleus on T1 without signal intensity abnormalities on T2 images are highly suggestive of hypermanganesaemia. Axial diffusion tensor imaging confirmed the "horseshoe moustache" sign to be constituted by the affected CTT. Hypermanganesaemia-induced CTT involvement in T1 needs to be differentiated from the other more common pediatric causes of CTT affliction which are evident on T2 or diffusion weighted images. Identification is crucial as it is a treatable disorder of metal deposition amenable to chelation.


Assuntos
Proteínas de Transporte de Cátions , Imagem de Tensor de Difusão , Criança , Marcha , Humanos , Imageamento por Ressonância Magnética , Manganês/metabolismo , Manganês/toxicidade
17.
IUBMB Life ; 73(5): 784-799, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33715270

RESUMO

The epigenetic abnormality is believed as a major driver for cancer initiation. Histone modification plays a vital role in tumor formation and progression. Particularly, alteration in histone acetylation has been highly associated with gene expression, cell cycle, as well as carcinogenesis. By analyzing glioblastoma (GBM)-related microarray from the GEO database and conducting chromatin immunoprecipitation-sequencing (ChIP-seq), we discovered that solute carrier family 30 member 3 (SLC30A3), a super enhancer (SE)-regulated factor, was significantly reduced in GBM tissues. Furthermore, histone deacetylase 1 (HDAC1), overexpressed in GBM tissues, could inhibit SLC30A3 expression by promoting histone H3K27ac deacetylation modification of the SE region of SLC30A3. Our functional validation revealed that SLC30A3 can inhibit the growth and metastatic spread of GBM cells in vitro and in vivo, and can activate the MAPK signaling pathway to promote apoptosis of GBM cells. Moreover, overexpression of HDAC1 resulted in a significant increase in DNA replication activity, a significant decline in apoptosis and cell cycle arrest in GBM cells. In a word, these findings indicate that combined epigenetic targeting of SLC30A3 by HDAC1 and SE is potentially therapeutically feasible in GBM.


Assuntos
Neoplasias Encefálicas/patologia , Proteínas de Transporte de Cátions/genética , Glioblastoma/patologia , Código das Histonas , Histona Desacetilase 1/fisiologia , Proteínas de Neoplasias/fisiologia , Acetilação , Adulto , Idoso , Animais , Apoptose , Neoplasias Encefálicas/genética , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , DNA de Neoplasias/genética , Transição Epitelial-Mesenquimal , Feminino , Glioblastoma/genética , Xenoenxertos , Sequenciamento de Nucleotídeos em Larga Escala , Histona Desacetilase 1/genética , Humanos , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Pessoa de Meia-Idade , Metástase Neoplásica , Proteínas de Neoplasias/genética , Fenótipo , Processamento de Proteína Pós-Traducional , Adulto Jovem
18.
Biometals ; 34(3): 573-588, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33713241

RESUMO

Manganese (Mn), an essential metal, can be toxic at elevated levels. In 2012, the first inherited cause of Mn excess was reported in patients with mutations in SLC30A10, a Mn efflux transporter. To explore the function of SLC30A10 in vitro, the current study used CRISPR/Cas9 gene editing to develop a stable SLC30A10 mutant Hep3B hepatoma cell line and collagenase perfusion in live mice to isolate primary hepatocytes deficient in Slc30a10. We also compared phenotypes of primary vs. non-primary cell lines to determine if they both serve as reliable in vitro models for the known physiological roles of SLC30A10. Mutant SLC30A10 Hep3B cells had increased Mn levels and decreased viability when exposed to excess Mn. Transport studies indicated a reduction of 54Mn import and export in mutant cells. While impaired 54Mn export was hypothesized given the essential role for SLC30A10 in cellular Mn export, impaired 54Mn import was unexpected. Whole genome sequencing did not identify any additional mutations in known Mn transporters in the mutant Hep3B mutant cell line. We then evaluated 54Mn transport in primary hepatocytes cultures isolated from genetically altered mice with varying liver Mn levels. Based on results from these experiments, we suggest that the effects of SLC30A10 deficiency on Mn homeostasis can be interrogated in vitro but only in specific types of cell lines.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Modelos Biológicos , Animais , Proteínas de Transporte de Cátions/deficiência , Proteínas de Transporte de Cátions/genética , Linhagem Celular , Hepatócitos/metabolismo , Homeostase , Humanos , Manganês/análise , Manganês/metabolismo , Camundongos , Camundongos Knockout
19.
Proc Natl Acad Sci U S A ; 115(32): E7642-E7649, 2018 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-30038024

RESUMO

SLC30A8 encodes a zinc transporter that is primarily expressed in the pancreatic islets of Langerhans. In ß-cells it transports zinc into insulin-containing secretory granules. Loss-of-function (LOF) mutations in SLC30A8 protect against type 2 diabetes in humans. In this study, we generated a knockin mouse model carrying one of the most common human LOF mutations for SLC30A8, R138X. The R138X mice had normal body weight, glucose tolerance, and pancreatic ß-cell mass. Interestingly, in hyperglycemic conditions induced by the insulin receptor antagonist S961, the R138X mice showed a 50% increase in insulin secretion. This effect was not associated with enhanced ß-cell proliferation or mass. Our data suggest that the SLC30A8 R138X LOF mutation may exert beneficial effects on glucose metabolism by increasing the capacity of ß-cells to secrete insulin under hyperglycemic conditions.


Assuntos
Diabetes Mellitus Tipo 2/genética , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Transportador 8 de Zinco/genética , Alelos , Animais , Glicemia , Diabetes Mellitus Tipo 2/metabolismo , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Humanos , Hiperglicemia/sangue , Hiperglicemia/induzido quimicamente , Hiperglicemia/metabolismo , Secreção de Insulina , Mutação com Perda de Função , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peptídeos/farmacologia , Receptor de Insulina/antagonistas & inibidores , Receptor de Insulina/metabolismo , Transportador 8 de Zinco/metabolismo
20.
Int J Mol Sci ; 22(9)2021 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-33925953

RESUMO

In the last years, research has shown that zinc ions play an essential role in the physiology of brain function. Zinc acts as a potent neuromodulatory agent and signaling ions, regulating healthy brain development and the function of both neurons and glial cells. Therefore, the concentration of zinc within the brain and its cells is tightly controlled. Zinc transporters are key regulators of (extra-) cellular zinc levels, and deregulation of zinc homeostasis and zinc transporters has been associated with neurodegenerative and neuropsychiatric disorders. However, to date, the presence of specific family members and their subcellular localization within brain cells have not been investigated in detail. Here, we analyzed the expression of all zinc transporters (ZnTs) and Irt-like proteins (ZIPs) in the rat brain. We further used primary rat neurons and rat astrocyte cell lines to differentiate between the expression found in neurons or astrocytes or both. We identified ZIP4 expressed in astrocytes but significantly more so in neurons, a finding that has not been reported previously. In neurons, ZIP4 is localized to synapses and found in a complex with major postsynaptic scaffold proteins of excitatory synapses. Synaptic ZIP4 reacts to short-term fluctuations in local zinc levels. We conclude that ZIP4 may have a so-far undescribed functional role at excitatory postsynapses.


Assuntos
Proteínas de Transporte/metabolismo , Neurônios/metabolismo , Zinco/metabolismo , Animais , Astrócitos/metabolismo , Encéfalo/metabolismo , Proteínas de Transporte/fisiologia , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Linhagem Celular , Feminino , Expressão Gênica/genética , Homeostase/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Tecido Nervoso/metabolismo , Gravidez , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley , Transcriptoma/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA