Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.386
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Immunol ; 34: 121-49, 2016 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-26735698

RESUMEN

Genomic DNA sequencing technologies have been one of the great advances of the 21st century, having decreased in cost by seven orders of magnitude and opening up new fields of investigation throughout research and clinical medicine. Genomics coupled with biochemical investigation has allowed the molecular definition of a growing number of new genetic diseases that reveal new concepts of immune regulation. Also, defining the genetic pathogenesis of these diseases has led to improved diagnosis, prognosis, genetic counseling, and, most importantly, new therapies. We highlight the investigational journey from patient phenotype to treatment using the newly defined XMEN disease, caused by the genetic loss of the MAGT1 magnesium transporter, as an example. This disease illustrates how genomics yields new fundamental immunoregulatory insights as well as how research genomics is integrated into clinical immunology. At the end, we discuss two other recently described diseases, CHAI/LATAIE (CTLA-4 deficiency) and PASLI (PI3K dysregulation), as additional examples of the journey from unknown immunological diseases to new precision medicine treatments using genomics.


Asunto(s)
Antígeno CTLA-4/genética , Proteínas de Transporte de Catión/genética , Genómica , Enfermedades del Sistema Inmune/genética , Mutación/genética , Fosfatidilinositol 3-Quinasas/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Animales , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Enfermedades del Sistema Inmune/terapia , Masculino , Terapia Molecular Dirigida , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia
2.
Annu Rev Immunol ; 33: 291-353, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25861976

RESUMEN

Ion channels and transporters mediate the transport of charged ions across hydrophobic lipid membranes. In immune cells, divalent cations such as calcium, magnesium, and zinc have important roles as second messengers to regulate intracellular signaling pathways. By contrast, monovalent cations such as sodium and potassium mainly regulate the membrane potential, which indirectly controls the influx of calcium and immune cell signaling. Studies investigating human patients with mutations in ion channels and transporters, analysis of gene-targeted mice, or pharmacological experiments with ion channel inhibitors have revealed important roles of ionic signals in lymphocyte development and in innate and adaptive immune responses. We here review the mechanisms underlying the function of ion channels and transporters in lymphocytes and innate immune cells and discuss their roles in lymphocyte development, adaptive and innate immune responses, and autoimmunity, as well as recent efforts to develop pharmacological inhibitors of ion channels for immunomodulatory therapy.


Asunto(s)
Inmunidad Adaptativa/fisiología , Inmunidad Innata/fisiología , Canales Iónicos/metabolismo , Animales , Canales de Calcio/genética , Canales de Calcio/metabolismo , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Humanos , Hipersensibilidad/genética , Hipersensibilidad/inmunología , Hipersensibilidad/metabolismo , Síndromes de Inmunodeficiencia/tratamiento farmacológico , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Síndromes de Inmunodeficiencia/metabolismo , Inmunoterapia/métodos , Canales Iónicos/genética , Linfocitos/citología , Linfocitos/inmunología , Linfocitos/metabolismo , Mastocitos/inmunología , Mastocitos/metabolismo , Terapia Molecular Dirigida , Mutación , Transducción de Señal
3.
Cell ; 179(4): 829-845.e20, 2019 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-31675496

RESUMEN

The immune microenvironment of hepatocellular carcinoma (HCC) is poorly characterized. Combining two single-cell RNA sequencing technologies, we produced transcriptomes of CD45+ immune cells for HCC patients from five immune-relevant sites: tumor, adjacent liver, hepatic lymph node (LN), blood, and ascites. A cluster of LAMP3+ dendritic cells (DCs) appeared to be the mature form of conventional DCs and possessed the potential to migrate from tumors to LNs. LAMP3+ DCs also expressed diverse immune-relevant ligands and exhibited potential to regulate multiple subtypes of lymphocytes. Of the macrophages in tumors that exhibited distinct transcriptional states, tumor-associated macrophages (TAMs) were associated with poor prognosis, and we established the inflammatory role of SLC40A1 and GPNMB in these cells. Further, myeloid and lymphoid cells in ascites were predominantly linked to tumor and blood origins, respectively. The dynamic properties of diverse CD45+ cell types revealed by this study add new dimensions to the immune landscape of HCC.


Asunto(s)
Carcinoma Hepatocelular/inmunología , Proteínas de Transporte de Catión/genética , Inflamación/inmunología , Neoplasias Hepáticas/inmunología , Glicoproteínas de Membrana/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Comunicación Celular/genética , Comunicación Celular/inmunología , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Células Dendríticas/inmunología , Células Dendríticas/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Inflamación/genética , Inflamación/patología , Antígenos Comunes de Leucocito/inmunología , Hígado/inmunología , Hígado/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Linfocitos/inmunología , Linfocitos/patología , Proteínas de Membrana de los Lisosomas/genética , Macrófagos/inmunología , Macrófagos/patología , Células Mieloides/inmunología , Células Mieloides/patología , Proteínas de Neoplasias/genética , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Transcriptoma/genética , Transcriptoma/inmunología , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
4.
Nat Immunol ; 20(3): 350-361, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30718914

RESUMEN

Despite the known importance of zinc for human immunity, molecular insights into its roles have remained limited. Here we report a novel autosomal recessive disease characterized by absent B cells, agammaglobulinemia and early onset infections in five unrelated families. The immunodeficiency results from hypomorphic mutations of SLC39A7, which encodes the endoplasmic reticulum-to-cytoplasm zinc transporter ZIP7. Using CRISPR-Cas9 mutagenesis we have precisely modeled ZIP7 deficiency in mice. Homozygosity for a null allele caused embryonic death, but hypomorphic alleles reproduced the block in B cell development seen in patients. B cells from mutant mice exhibited a diminished concentration of cytoplasmic free zinc, increased phosphatase activity and decreased phosphorylation of signaling molecules downstream of the pre-B cell and B cell receptors. Our findings highlight a specific role for cytosolic Zn2+ in modulating B cell receptor signal strength and positive selection.


Asunto(s)
Agammaglobulinemia/inmunología , Linfocitos B/inmunología , Proteínas de Transporte de Catión/inmunología , Zinc/inmunología , Agammaglobulinemia/genética , Agammaglobulinemia/metabolismo , Animales , Linfocitos B/metabolismo , Proteínas de Transporte de Catión/deficiencia , Proteínas de Transporte de Catión/genética , Preescolar , Citosol/inmunología , Citosol/metabolismo , Modelos Animales de Enfermedad , Retículo Endoplásmico/inmunología , Retículo Endoplásmico/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Lactante , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Linaje , Zinc/metabolismo
5.
Cell ; 156(4): 730-43, 2014 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-24529376

RESUMEN

Osteoarthritis (OA), primarily characterized by cartilage degeneration, is caused by an imbalance between anabolic and catabolic factors. Here, we investigated the role of zinc (Zn2+) homeostasis, Zn2+ transporters, and Zn(2+)-dependent transcription factors in OA pathogenesis. Among Zn2+ transporters, the Zn2+ importer ZIP8 was specifically upregulated in OA cartilage of humans and mice, resulting in increased levels of intracellular Zn2+ in chondrocytes. ZIP8-mediated Zn2+ influx upregulated the expression of matrix-degrading enzymes (MMP3, MMP9, MMP12, MMP13, and ADAMTS5) in chondrocytes. Ectopic expression of ZIP8 in mouse cartilage tissue caused OA cartilage destruction, whereas Zip8 knockout suppressed surgically induced OA pathogenesis, with concomitant modulation of Zn2+ influx and matrix-degrading enzymes. Furthermore, MTF1 was identified as an essential transcription factor in mediating Zn2+/ZIP8-induced catabolic factor expression, and genetic modulation of Mtf1 in mice altered OA pathogenesis. We propose that the zinc-ZIP8-MTF1 axis is an essential catabolic regulator of OA pathogenesis.


Asunto(s)
Osteoartritis/metabolismo , Osteoartritis/patología , Transducción de Señal , Proteínas ADAM/metabolismo , Anciano , Animales , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Condrocitos/metabolismo , Condrocitos/patología , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Regulación hacia Arriba , Zinc/metabolismo
6.
Mol Cell ; 81(19): 3904-3918.e6, 2021 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-34375581

RESUMEN

Polyamines, small organic polycations, are essential for cell viability, and their physiological levels are homeostatically maintained by post-transcriptional regulation of key biosynthetic enzymes. In addition to de novo synthesis, cells can also take up polyamines; however, identifying cellular polyamine transporters has been challenging. Here we show that the S. cerevisiae HOL1 mRNA is under translational control by polyamines, and we reveal that the encoded membrane transporter Hol1 is a high-affinity polyamine transporter and is required for yeast growth under limiting polyamine conditions. Moreover, we show that polyamine inhibition of the translation factor eIF5A impairs translation termination at a Pro-Ser-stop motif in a conserved upstream open reading frame on the HOL1 mRNA to repress Hol1 synthesis under conditions of elevated polyamines. Our findings reveal that polyamine transport, like polyamine biosynthesis, is under translational autoregulation by polyamines in yeast, highlighting the extensive control cells impose on polyamine levels.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Poliaminas/metabolismo , Biosíntesis de Proteínas , Ribosomas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Transporte Biológico , Proteínas de Transporte de Catión/genética , Regulación Fúngica de la Expresión Génica , Proteínas de Transporte de Membrana/genética , Sistemas de Lectura Abierta , Factores de Iniciación de Péptidos/genética , Factores de Iniciación de Péptidos/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Ribosomas/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crecimiento & desarrollo , Proteínas de Saccharomyces cerevisiae/genética , Factor 5A Eucariótico de Iniciación de Traducción
7.
Cell ; 151(3): 630-44, 2012 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-23101630

RESUMEN

Mitochondrial Ca(2+) (Ca(2+)(m)) uptake is mediated by an inner membrane Ca(2+) channel called the uniporter. Ca(2+) uptake is driven by the considerable voltage present across the inner membrane (ΔΨ(m)) generated by proton pumping by the respiratory chain. Mitochondrial matrix Ca(2+) concentration is maintained five to six orders of magnitude lower than its equilibrium level, but the molecular mechanisms for how this is achieved are not clear. Here, we demonstrate that the mitochondrial protein MICU1 is required to preserve normal [Ca(2+)](m) under basal conditions. In its absence, mitochondria become constitutively loaded with Ca(2+), triggering excessive reactive oxygen species generation and sensitivity to apoptotic stress. MICU1 interacts with the uniporter pore-forming subunit MCU and sets a Ca(2+) threshold for Ca(2+)(m) uptake without affecting the kinetic properties of MCU-mediated Ca(2+) uptake. Thus, MICU1 is a gatekeeper of MCU-mediated Ca(2+)(m) uptake that is essential to prevent [Ca(2+)](m) overload and associated stress.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Calcio/metabolismo , Proteínas de Transporte de Catión/metabolismo , Supervivencia Celular , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Apoptosis , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/genética , Proteínas de Transporte de Catión/química , Proteínas de Transporte de Catión/genética , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Proteínas de Transporte de Membrana Mitocondrial/química , Proteínas de Transporte de Membrana Mitocondrial/genética , Membranas Mitocondriales/metabolismo
8.
Mol Cell ; 75(1): 102-116.e9, 2019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-31128943

RESUMEN

Transcription regulation underlies stem cell function and development. Here, we elucidate an unexpected role of an essential ribogenesis factor, WDR43, as a chromatin-associated RNA-binding protein (RBP) and release factor in modulating the polymerase (Pol) II activity for pluripotency regulation. WDR43 binds prominently to promoter-associated noncoding/nascent RNAs, occupies thousands of gene promoters and enhancers, and interacts with the Pol II machinery in embryonic stem cells (ESCs). Nascent transcripts and transcription recruit WDR43 to active promoters, where WDR43 facilitates releases of the elongation factor P-TEFb and paused Pol II. Knockdown of WDR43 causes genome-wide defects in Pol II release and pluripotency-associated gene expression. Importantly, auxin-mediated rapid degradation of WDR43 drastically reduces Pol II activity, precluding indirect consequences. These results reveal an RNA-mediated recruitment and feedforward regulation on transcription and demonstrate an unforeseen role of an RBP in promoting Pol II elongation and coordinating high-level transcription and translation in ESC pluripotency.


Asunto(s)
Proteínas de Transporte de Catión/genética , Cromatina/química , Regulación del Desarrollo de la Expresión Génica , Células Madre Embrionarias de Ratones/metabolismo , ARN Polimerasa II/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Transcripción Genética , Proteínas de Pez Cebra/genética , Animales , Sitios de Unión , Proteínas de Transporte de Catión/metabolismo , Diferenciación Celular , Línea Celular , Cromatina/metabolismo , Embrión de Mamíferos , Elementos de Facilitación Genéticos , Eliminación de Gen , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Células Madre Embrionarias de Ratones/citología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Factor B de Elongación Transcripcional Positiva/genética , Factor B de Elongación Transcripcional Positiva/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Biosíntesis de Proteínas , Proteolisis , ARN Polimerasa II/metabolismo , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Proteínas de Pez Cebra/metabolismo
9.
Mol Cell ; 73(2): 364-376.e8, 2019 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-30581142

RESUMEN

Mitophagy, a mitochondrial quality control process for eliminating dysfunctional mitochondria, can be induced by a response of dynamin-related protein 1 (Drp1) to a reduction in mitochondrial membrane potential (MMP) and mitochondrial division. However, the coordination between MMP and mitochondrial division for selecting the damaged portion of the mitochondrial network is less understood. Here, we found that MMP is reduced focally at a fission site by the Drp1 recruitment, which is initiated by the interaction of Drp1 with mitochondrial zinc transporter Zip1 and Zn2+ entry through the Zip1-MCU complex. After division, healthy mitochondria restore MMP levels and participate in the fusion-fission cycle again, but mitochondria that fail to restore MMP undergo mitophagy. Thus, interfering with the interaction between Drp1 and Zip1 blocks the reduction of MMP and the subsequent mitophagic selection of damaged mitochondria. These results suggest that Drp1-dependent fission provides selective pressure for eliminating "bad sectors" in the mitochondrial network, serving as a mitochondrial quality surveillance system.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , GTP Fosfohidrolasas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/metabolismo , Dinámicas Mitocondriales , Proteínas Mitocondriales/metabolismo , Mitofagia , Adenosina Trifosfato/metabolismo , Animales , Canales de Calcio/genética , Canales de Calcio/metabolismo , Proteínas de Transporte de Catión/genética , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Dinaminas , Metabolismo Energético , GTP Fosfohidrolasas/genética , Células HEK293 , Células HeLa , Humanos , Potencial de la Membrana Mitocondrial , Proteínas Asociadas a Microtúbulos/genética , Mitocondrias/genética , Mitocondrias/patología , Proteínas Mitocondriales/genética , Mutación , Neuronas/metabolismo , Neuronas/patología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Ratas Sprague-Dawley , Transducción de Señal , Factores de Tiempo , Zinc/metabolismo
10.
Proc Natl Acad Sci U S A ; 121(37): e2407455121, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39240971

RESUMEN

Succinic acid (SA), a dicarboxylic acid of industrial importance, can be efficiently produced by metabolically engineered Mannheimia succiniciproducens. Although the importance of magnesium (Mg2+) ion on SA production has been evident from our previous studies, the role of Mg2+ ion remains largely unexplored. In this study, we investigated the impact of Mg2+ ion on SA production and developed a hyper-SA producing strain of M. succiniciproducens by reconstructing the Mg2+ ion transport system. To achieve this, optimal alkaline neutralizer comprising Mg2+ ion was developed and the physiological effect of Mg2+ ion was analyzed. Subsequently, the Mg2+ ion transport system was reconstructed by introducing an efficient Mg2+ ion transporter from Salmonella enterica. A high-inoculum fed-batch fermentation of the final engineered strain produced 152.23 ± 0.99 g/L of SA, with a maximum productivity of 39.64 ± 0.69 g/L/h. These findings highlight the importance of Mg2+ ions and transportation system optimization in succinic acid production by M. succiniciproducens.


Asunto(s)
Fermentación , Magnesio , Mannheimia , Ácido Succínico , Ácido Succínico/metabolismo , Magnesio/metabolismo , Mannheimia/metabolismo , Mannheimia/genética , Ingeniería Metabólica/métodos , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Proteínas de Transporte de Catión/metabolismo , Proteínas de Transporte de Catión/genética
11.
Proc Natl Acad Sci U S A ; 121(26): e2402538121, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38905240

RESUMEN

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.


Asunto(s)
Homeostasis , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Manganeso , Humanos , Manganeso/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/genética , Proteínas de Transporte de Catión/metabolismo , Proteínas de Transporte de Catión/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Células HEK293 , Hierro/metabolismo
12.
Proc Natl Acad Sci U S A ; 121(40): e2405615121, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39312661

RESUMEN

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.


Asunto(s)
Corteza Auditiva , Neuronas , Sinapsis , Zinc , Animales , Zinc/metabolismo , Corteza Auditiva/metabolismo , Corteza Auditiva/fisiología , Ratones , Sinapsis/metabolismo , Sinapsis/fisiología , Neuronas/metabolismo , Neuronas/fisiología , Proteínas de Transporte de Catión/metabolismo , Proteínas de Transporte de Catión/genética , Estimulación Acústica , Ratones Noqueados , Percepción Auditiva/fisiología , Ratones Endogámicos C57BL , Masculino
13.
Proc Natl Acad Sci U S A ; 121(35): e2402491121, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39163336

RESUMEN

Activating Ca2+-sensitive enzymes of oxidative metabolism while preventing calcium overload that leads to mitochondrial and cellular injury requires dynamic control of mitochondrial Ca2+ uptake. This is ensured by the mitochondrial calcium uptake (MICU)1/2 proteins that gate the pore of the mitochondrial calcium uniporter (mtCU). MICU1 is relatively sparse in the heart, and recent studies claimed the mammalian heart lacks MICU1 gating of mtCU. However, genetic models have not been tested. We find that MICU1 is present in a complex with MCU in nonfailing human hearts. Furthermore, using murine genetic models and pharmacology, we show that MICU1 and MICU2 control cardiac mitochondrial Ca2+ influx, and that MICU1 deletion alters cardiomyocyte mitochondrial calcium signaling and energy metabolism. MICU1 loss causes substantial compensatory changes in the mtCU composition and abundance, increased turnover of essential MCU regulator (EMRE) early on and, later, of MCU, that limit mitochondrial Ca2+ uptake and allow cell survival. Thus, both the primary consequences of MICU1 loss and the ensuing robust compensation highlight MICU1's relevance in the beating heart.


Asunto(s)
Señalización del Calcio , Proteínas de Unión al Calcio , Calcio , Proteínas de Transporte de Catión , Proteínas de Transporte de Membrana Mitocondrial , Miocitos Cardíacos , Animales , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al Calcio/genética , Ratones , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/genética , Humanos , Proteínas de Transporte de Catión/metabolismo , Proteínas de Transporte de Catión/genética , Miocitos Cardíacos/metabolismo , Señalización del Calcio/fisiología , Calcio/metabolismo , Mitocondrias Cardíacas/metabolismo , Canales de Calcio/metabolismo , Canales de Calcio/genética , Ratones Noqueados , Miocardio/metabolismo , Masculino
14.
PLoS Pathog ; 20(8): e1012444, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39173055

RESUMEN

The Ebola virus (EBOV) has emerged as a significant global health concern, notably during the 2013-2016 outbreak in West Africa. Despite the clinical approval of two EBOV antibody drugs, there is an urgent need for more diverse and effective antiviral drugs, along with comprehensive understanding of viral-host interactions. In this study, we harnessed a biologically contained EBOVΔVP30-EGFP cell culture model which could recapitulate the entire viral life cycle, to conduct a genome-wide CRISPR/Cas9 screen. Through this, we identified PIK3C3 (phosphatidylinositide 3-kinase) and SLC39A9 (zinc transporter) as crucial host factors for EBOV infection. Genetic depletion of SLC39A9 and PIK3C3 lead to reduction of EBOV entry, but not impact viral genome replication, suggesting that SLC39A9 and PIK3C3 act as entry factors, facilitating viral entry into host cells. Moreover, PIK3C3 kinase activity is indispensable for the internalization of EBOV virions, presumably through the regulation of endocytic and autophagic membrane traffic, which has been previously recognized as essential for EBOV internalization. Notably, our study demonstrated that PIK3C3 kinase inhibitor could effectively block EBOV infection, underscoring PIK3C3 as a promising drug target. Furthermore, biochemical analysis showed that recombinant SLC39A9 protein could directly bind viral GP protein, which further promotes the interaction of viral GP protein with cellular receptor NPC1. These findings suggests that SLC39A9 plays dual roles in EBOV entry. Initially, it serves as an attachment factor during the early entry phase by engaging with the viral GP protein. Subsequently, SLC39A9 functions an adaptor protein, facilitating the interaction between virions and the NPC1 receptor during the late entry phase, prior to cathepsin cleavage on the viral GP. In summary, this study offers novel insights into virus-host interactions, contributing valuable information for the development of new therapies against EBOV infection.


Asunto(s)
Sistemas CRISPR-Cas , Ebolavirus , Fiebre Hemorrágica Ebola , Internalización del Virus , Animales , Humanos , Proteínas de Transporte de Catión/metabolismo , Proteínas de Transporte de Catión/genética , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Fosfatidilinositol 3-Quinasas Clase III/genética , Ebolavirus/genética , Ebolavirus/fisiología , Ebolavirus/metabolismo , Células HEK293 , Fiebre Hemorrágica Ebola/virología , Fiebre Hemorrágica Ebola/metabolismo , Fiebre Hemorrágica Ebola/genética , Replicación Viral
15.
Blood ; 143(23): 2433-2437, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38518102

RESUMEN

ABSTRACT: Iron-mediated induction of bone morphogenetic protein (BMP)6 expression by liver endothelial cells is essential for iron homeostasis regulation. We used multiple dietary and genetic mouse cohorts to demonstrate a minor functional role for the metal-ion transporter ZIP8 in regulating BMP6 expression under high-iron conditions.


Asunto(s)
Proteína Morfogenética Ósea 6 , Proteínas de Transporte de Catión , Hierro , Animales , Proteína Morfogenética Ósea 6/metabolismo , Proteína Morfogenética Ósea 6/genética , Ratones , Proteínas de Transporte de Catión/metabolismo , Proteínas de Transporte de Catión/genética , Hierro/metabolismo , Células Endoteliales/metabolismo , Ratones Noqueados , Regulación de la Expresión Génica , Hígado/metabolismo , Ratones Endogámicos C57BL , Homeostasis
16.
Blood ; 144(13): 1433-1444, 2024 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-38861671

RESUMEN

ABSTRACT: Ferroportin (Fpn) is the only iron exporter, playing a crucial role in systemic iron homeostasis. Fpn is negatively regulated by its ligand hepcidin, but other potential regulators in physiological and disease conditions remain poorly understood. Diabetes is a metabolic disorder that develops body iron loading with unknown mechanisms. By using diabetic mouse models and human duodenal specimens, we demonstrated that intestinal Fpn expression was increased in diabetes in a hepcidin-independent manner. Protein kinase C (PKC) is hyperactivated in diabetes. We showed that PKCα was required to sustain baseline Fpn expression and diabetes-induced Fpn upregulation in the enterocytes and macrophages. Knockout of PKCα abolished diabetes-associated iron overload. Mechanistically, activation of PKCα increased the exocytotic trafficking of Fpn and decreased the endocytic trafficking of Fpn in the resting state. Hyperactive PKCα also suppressed hepcidin-induced ubiquitination, internalization, and degradation of Fpn. We further observed that iron loading in the enterocytes and macrophages activated PKCα, acting as a novel mechanism to enhance Fpn-dependent iron efflux. Finally, we demonstrated that the loss-of-function of PKCα and pharmacological inhibition of PKC significantly alleviated hereditary hemochromatosis-associated iron overload. Our study has highlighted, to our knowledge, for the first time, that PKCα is an important positive regulator of Fpn and a new target in the control of iron homeostasis.


Asunto(s)
Proteínas de Transporte de Catión , Hemocromatosis , Hepcidinas , Sobrecarga de Hierro , Proteína Quinasa C-alfa , Animales , Sobrecarga de Hierro/metabolismo , Proteína Quinasa C-alfa/metabolismo , Proteína Quinasa C-alfa/genética , Ratones , Humanos , Proteínas de Transporte de Catión/metabolismo , Proteínas de Transporte de Catión/genética , Hemocromatosis/metabolismo , Hemocromatosis/genética , Hemocromatosis/patología , Hepcidinas/metabolismo , Hepcidinas/genética , Ratones Noqueados , Masculino , Hierro/metabolismo , Diabetes Mellitus Experimental/metabolismo , Ratones Endogámicos C57BL , Enterocitos/metabolismo , Enterocitos/patología , Macrófagos/metabolismo
17.
Circ Res ; 135(1): 26-40, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38747181

RESUMEN

BACKGROUND: Calcium (Ca2+) uptake by mitochondria occurs via the mitochondrial Ca2+ uniporter. Mitochondrial Ca2+ uniporter exists as a complex, regulated by 3 MICU (mitochondrial Ca2+ uptake) proteins localized in the intermembrane space: MICU1, MICU2, and MICU3. Although MICU3 is present in the heart, its role is largely unknown. METHODS: We used CRISPR-Cas9 to generate a mouse with global deletion of MICU3 and an adeno-associated virus (AAV9) to overexpress MICU3 in wild-type mice. We examined the role of MICU3 in regulating mitochondrial calcium ([Ca2+]m) in ex vivo hearts using an optical method following adrenergic stimulation in perfused hearts loaded with a Ca2+-sensitive fluorophore. Additionally, we studied how deletion and overexpression of MICU3, respectively, impact cardiac function in vivo by echocardiography and the molecular composition of the mitochondrial Ca2+ uniporter complex via Western blot, immunoprecipitation, and Blue native-PAGE analysis. Finally, we measured MICU3 expression in failing human hearts. RESULTS: MICU3 knock out hearts and cardiomyocytes exhibited a significantly smaller increase in [Ca2+]m than wild-type hearts following acute isoproterenol infusion. In contrast, heart with overexpression of MICU3 exhibited an enhanced increase in [Ca2+]m compared with control hearts. Echocardiography analysis showed no significant difference in cardiac function in knock out MICU3 mice relative to wild-type mice at baseline. However, mice with overexpression of MICU3 exhibited significantly reduced ejection fraction and fractional shortening compared with control mice. We observed a significant increase in the ratio of heart weight to tibia length in hearts with overexpression of MICU3 compared with controls, consistent with hypertrophy. We also found a significant decrease in MICU3 protein and expression in failing human hearts. CONCLUSIONS: Our results indicate that increased and decreased expression of MICU3 enhances and reduces, respectively, the uptake of [Ca2+]m in the heart. We conclude that MICU3 plays an important role in regulating [Ca2+]m physiologically, and overexpression of MICU3 is sufficient to induce cardiac hypertrophy, making MICU3 a possible therapeutic target.


Asunto(s)
Proteínas de Unión al Calcio , Calcio , Ratones Noqueados , Mitocondrias Cardíacas , Proteínas de Transporte de Membrana Mitocondrial , Miocitos Cardíacos , Animales , Femenino , Humanos , Masculino , Ratones , Calcio/metabolismo , Canales de Calcio/metabolismo , Canales de Calcio/genética , Señalización del Calcio , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al Calcio/genética , Cardiomegalia/metabolismo , Cardiomegalia/genética , Proteínas de Transporte de Catión/metabolismo , Proteínas de Transporte de Catión/genética , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/genética , Ratones Endogámicos C57BL , Mitocondrias Cardíacas/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/genética , Miocitos Cardíacos/metabolismo
18.
Mol Cell ; 69(6): 953-964.e5, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29547723

RESUMEN

Plant roots forage the soil for iron, the concentration of which can be dramatically lower than those needed for growth. Soil iron uptake uses the broad metal spectrum IRT1 transporter that also transports zinc, manganese, cobalt, and cadmium. Sophisticated iron-dependent transcriptional regulatory mechanisms allow plants to tightly control the abundance of IRT1, ensuring optimal absorption of iron. Here, we uncover that IRT1 acts as a transporter and receptor (transceptor), directly sensing excess of its non-iron metal substrates in the cytoplasm, to regulate its own degradation. Direct metal binding to a histidine-rich stretch in IRT1 triggers its phosphorylation by the CIPK23 kinase and facilitates the subsequent recruitment of the IDF1 E3 ligase. CIPK23-driven phosphorylation and IDF1-mediated lysine-63 polyubiquitination are jointly required for efficient endosomal sorting and vacuolar degradation of IRT1. Thus, IRT1 directly senses elevated non-iron metal concentrations and integrates multiple substrate-dependent regulations to optimize iron uptake and protect plants from highly reactive metals.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , Proteínas de Transporte de Catión/metabolismo , Metales/metabolismo , Raíces de Plantas/metabolismo , Plantas Modificadas Genéticamente/metabolismo , Arabidopsis/genética , Proteínas de Arabidopsis/genética , Transporte Biológico , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Transporte de Catión/genética , Endocitosis , Regulación de la Expresión Génica de las Plantas , Péptidos y Proteínas de Señalización Intracelular , Metales/toxicidad , Plantas Modificadas Genéticamente/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Estabilidad Proteica , Transporte de Proteínas , Proteolisis , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Transcripción Genética , Ubiquitinación , Vacuolas/metabolismo
19.
Mol Cell ; 72(4): 778-785.e3, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30454562

RESUMEN

Proper control of the mitochondrial Ca2+ uniporter's pore (MCU) is required to allow Ca2+-dependent activation of oxidative metabolism and to avoid mitochondrial Ca2+ overload and cell death. The MCU's gatekeeping and cooperative activation is mediated by the Ca2+-sensing MICU1 protein, which has been proposed to form dimeric complexes anchored to the EMRE scaffold of MCU. We unexpectedly find that MICU1 suppresses inhibition of MCU by ruthenium red/Ru360, which bind to MCU's DIME motif, the selectivity filter. This led us to recognize in MICU1's sequence a putative DIME interacting domain (DID), which is required for both gatekeeping and cooperative activation of MCU and for cell survival. Thus, we propose that MICU1 has to interact with the D-ring formed by the DIME domains in MCU to control the uniporter.


Asunto(s)
Canales de Calcio/metabolismo , Proteínas de Unión al Calcio/metabolismo , Calcio/metabolismo , Proteínas de Transporte de Catión/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Compuestos de Rutenio/farmacología , Animales , Proteínas de Unión al Calcio/genética , Proteínas de Transporte de Catión/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Células HEK293 , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Masculino , Potencial de la Membrana Mitocondrial , Ratones , Ratones Noqueados , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/genética
20.
J Neurosci ; 44(28)2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38830758

RESUMEN

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.


Asunto(s)
Corteza Auditiva , Proteínas de Transporte de Catión , Espinas Dendríticas , Proteínas del Tejido Nervioso , Sinapsis , Animales , Ratones , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Sinapsis/metabolismo , Espinas Dendríticas/metabolismo , Proteínas de Transporte de Catión/metabolismo , Proteínas de Transporte de Catión/genética , Corteza Auditiva/metabolismo , Femenino , Masculino , Ratones Noqueados , Proteínas Portadoras/metabolismo , Proteínas Portadoras/genética , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/metabolismo , Proteínas de Microfilamentos/genética , Potenciales Postsinápticos Excitadores/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA