Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
BMC Biol ; 22(1): 12, 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38273307

RESUMEN

BACKGROUND: Many viruses enter host cells by hijacking endosomal trafficking. CapZ, a canonical actin capping protein, participates in endosomal trafficking, yet its precise role in endocytosis and virus infection remains elusive. RESULTS: Here, we showed that CapZ was transiently associated with early endosomes (EEs) and was subsequently released from the matured EEs after the fusion of two EEs, which was facilitated by PI(3)P to PI(3,5)P2 conversion. Vacuolin-1 (a triazine compound) stabilized CapZ at EEs and thus blocked the transition of EEs to late endosomes (LEs). Likewise, artificially tethering CapZ to EEs via a rapamycin-induced protein-protein interaction system blocked the early-to-late endosome transition. Remarkably, CapZ knockout or artificially tethering CapZ to EEs via rapamycin significantly inhibited flaviviruses, e.g., Zika virus (ZIKV) and dengue virus (DENV), or beta-coronavirus, e.g., murine hepatitis virus (MHV), infection by preventing the escape of RNA genome from endocytic vesicles. CONCLUSIONS: These results indicate that the temporal association of CapZ with EEs facilitates early-to-late endosome transition (physiologically) and the release of the viral genome from endocytic vesicles (pathologically).


Asunto(s)
Fosfatos de Fosfatidilinositol , Infección por el Virus Zika , Virus Zika , Animales , Humanos , Ratones , Endocitosis/fisiología , Endosomas/metabolismo , Sirolimus/farmacología , Sirolimus/metabolismo , Vesículas Transportadoras , Internalización del Virus , Infección por el Virus Zika/metabolismo
2.
Mar Drugs ; 21(1)2023 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-36662219

RESUMEN

Dysfunctional autophagy is associated with various human diseases, e.g., cancer. The discovery of small molecules modulating autophagy with therapeutic potential could be significant. To this end, we screened the ability of a series of metabolites isolated from marine microorganisms to modulate autophagy. Anhydrodebromoaplysiatoxin (ADAT), a metabolite yielded by the marine red algae Gracilaria coronopifolia, inhibited autophagosome-lysosome fusion in mammalian cells, thereby inducing the accumulation of autophagosomes. Treatment of cells with ADAT alkalinized lysosomal pH. Interestingly, ADAT also activated the mTOR/p70S6K/FoxO3a signaling pathway, likely leading to the inhibition of autophagy induction. ADAT had little effect on apoptosis. Our results suggest that ADAT is a dichotomic autophagy inhibitor that inhibits both late-stage (autophagosome-lysosome fusion) and early-stage (autophagy induction) autophagy.


Asunto(s)
Autofagosomas , Autofagia , Animales , Humanos , Autofagosomas/metabolismo , Lisosomas , Mamíferos , Transducción de Señal
3.
Semin Cell Dev Biol ; 101: 12-19, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31563390

RESUMEN

Autophagy, an evolutional conserved lysosomal degradation process, has been implicated to play an important role in cellular defense against a variety of microbial infection. Interestingly, numerous studies found that some pathogens, especially positive-single-strand RNA viruses, actually hijacked autophagy machinery to promote virus infection within host cells, facilitating different stages of viral life cycle, from replication, assembly to egress. Enterovirus, a genus of positive-strand RNA virus, can cause various human diseases and is one of main public health threat globally, yet no effective clinical intervention is available for enterovirus infection. Here we summarized recent literature on how enteroviruses regulate and utilize autophagy process to facilitate their propagation in the host cells. The studies on the interplay between enterovirus and autophagy not only shed light on the molecular mechanisms underlying how enterovirus hijacks cellular components and pathway for its own benefits, but also provide therapeutic option against enterovirus infection.


Asunto(s)
Autofagia/inmunología , Enterovirus/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos
4.
J Cell Physiol ; 236(10): 6932-6947, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33682133

RESUMEN

Autophagy, an evolutionarily conserved lysosomal degradation pathway, is known to regulate a variety of physiological and pathological processes. At present, the function and the precise mechanism of autophagy regulation in kidney and renal cells remain elusive. Here, we explored the role of ERK1 and ERK2 (referred as ERK1/2 hereafter) in autophagy regulation in renal cells in response to hypoglycemia. Glucose starvation potently and transiently activated ERK1/2 in renal cells, and this was concomitant with an increase in autophagic flux. Perturbing ERK1/2 activation by treatment with inhibitors of RAF or MEK1/2, via the expression of a dominant-negative mutant form of MEK1/2 or RAS, blocked hypoglycemia-mediated ERK1/2 activation and autophagy induction in renal cells. Glucose starvation also induced the accumulation of reactive oxygen species in renal cells, which was involved in the activation of the ERK1/2 cascade and the induction of autophagy in renal cells. Interestingly, ATG13 and FIP200, the members of the ULK1 complex, contain the ERK consensus phosphorylation sites, and glucose starvation induced an association between ATG13 or FIP200 and ERK1/2. Moreover, the expression of the phospho-defective mutants of ATG13 and FIP200 in renal cells blocked glucose starvation-induced autophagy and rendered cells more susceptible to hypoglycemia-induced cell death. However, the expression of the phospho-mimic mutants of ATG13 and FIP200 induced autophagy and protected renal cells from hypoglycemia-induced cell death. Taken together, our results demonstrate that hypoglycemia activates the ERK1/2 signaling to regulate ATG13 and FIP200, thereby stimulating autophagy to protect the renal cells from hypoglycemia-induced cell death.


Asunto(s)
Proteínas Relacionadas con la Autofagia/metabolismo , Autofagia , Glucosa/deficiencia , Hipoglucemia/enzimología , Riñón/enzimología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Relacionadas con la Autofagia/genética , Glucemia/metabolismo , Células HEK293 , Células HeLa , Humanos , Hipoglucemia/sangre , Hipoglucemia/patología , Riñón/patología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
5.
FASEB J ; 34(4): 5193-5207, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32067276

RESUMEN

One of the critical regulatory mechanisms for cell cycle progression is the timely degradation of CDK inhibitors, including p21Cip1 and p27Kip1 . VCP/p97, an AAA-ATPase, is reported to be overexpressed in many types of cancers. Here, we found that treatment of MCF-7 human breast cancer cells with DBeQ, a VCP inhibitor, or VCP knockdown in MCF-7 cells arrested cells at G1 phase, accompanied with the blockage of both p21 and p27 degradation. Whereas, double knockdown of p21 and p27 in MCF-7 cells rendered cells refractory to DBeQ-induced G1 arrest. Moreover, inhibition or knockdown of VCP or UFD1, one of VCP's co-factors, in MCF-7, NIH3T3, or HEK293T cells blocked the nuclear export of p27 during earlier G1 phase after mitogen stimulation. We also identified the nuclear localization sequence (NLS) of VCP, and found that adding back wild-type VCP, not the NLS-deleted VCP mutant, restored the nuclear export and degradation of p27 in VCP knockout MCF-7 cells. Importantly, we found that VCP inhibition sensitized breast cancer cells to the treatment of several anticancer therapeutics both in vitro and in vivo. Taken together, our study not only uncovers the mechanisms underlying VCP-mediated cell proliferation control but also provides potential therapeutic option for cancer treatment.


Asunto(s)
Transporte Activo de Núcleo Celular , Neoplasias de la Mama/patología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Fase G1 , Fase S , Proteína que Contiene Valosina/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteolisis , Células Tumorales Cultivadas , Proteína que Contiene Valosina/genética , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Cell Biol Toxicol ; 37(5): 695-713, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33486680

RESUMEN

Autophagy is a conserved lysosomal degradation process, and abnormal autophagy has been associated with various pathological processes, e.g., neurodegeneration, cancer, and pathogen infection. Small chemical modulators of autophagy show the potential to treat autophagy-associated diseases. Diterpenoids, nature products found in various plants, exhibit a wide range of bioactivity, and we have recently isolated and characterized over 150 diterpenoids from Isodon species distributed in China. Here, we applied a high-content fluorescence imaging-based assay to assess these diterpenoids' ability to affect autophagic flux in HeLa cells. We found that enanderinanin J, an ent-kauranoid dimer, is an autophagy inhibitor, manifested by its ability to increase lysosomal pH and inhibit the fusion between autophagosomes and lysosomes. Autophagy has been shown to be either positively or negatively involved in the life cycle of Zika virus (ZIKV), Japanese encephalitis virus (JEV), Dengue virus (DENV), and enterovirus-A71 (EV-A71). We found that enanderinanin J significantly inhibited the infection of ZIKV, DENV, JEV, or EV-A71. Interestingly, although ATG5 knockdown inhibited ZIKV or JEV infection, enanderinanin J further inhibited the infection of ZIKV or JEV in ATG5-knockdown cells. Taken together, our data indicate that enanderinanin J inhibits autophagosome-lysosome fusion and is a potential antiviral agent.


Asunto(s)
Diterpenos , Isodon , Infección por el Virus Zika , Virus Zika , Antivirales/farmacología , Autofagia , Diterpenos/farmacología , Células HeLa , Humanos
7.
Exp Cell Res ; 394(1): 111989, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32283065

RESUMEN

All-trans retinoic acid (ATRA) is an anti-cancer differentiation therapy agent effective for acute promyelocytic leukemia (APL) but not acute myeloid leukemia (AML) in general. Using the HL-60 human non-APL AML model where ATRA causes nuclear enrichment of c-Raf that drives differentiation and G1/G0 cell cycle arrest, we now observe that c-Raf in the nucleus showed novel interactions with several prominent regulators of the cell cycle and cell differentiation. One is cyclin-dependent kinase 2 (Cdk2). ATRA treatment caused c-Raf to dissociate from Cdk2. This was associated with enhanced binding of Cdk2 with retinoic acid receptor α (RARα). Consistent with this novel Raf/CDK2/RARα axis contributing to differentiation, CD38 expression per cell, which is transcriptionally regulated by a retinoic acid response element (RARE), is enhanced. The RB tumor suppressor, a fundamental regulator of G1 cell cycle progression or arrest, was also targeted by c-Raf in the nucleus. RB and specifically the S608 phosphorylated form (pS608RB) complexed with c-Raf. ATRA treatment induced S608RB-hypophosphorylation associated with G1/G0 cell cycle arrest and release of c-Raf from RB. We also found that nuclear c-Raf interacted with SMARCD1, a pioneering component of the SWI/SNF chromatin remodeling complex. ATRA treatment diminished the amount of this protein bound to c-Raf. The data suggest that ATRA treatment to HL-60 human cells re-directed c-Raf from its historically pro-proliferation functions in the cytoplasm to pro-differentiation functions in the nucleus.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Leucemia Promielocítica Aguda/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-raf/efectos de los fármacos , Tretinoina/farmacología , Antineoplásicos/farmacología , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Células HL-60 , Humanos , Leucemia Promielocítica Aguda/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-raf/metabolismo , Receptor alfa de Ácido Retinoico/efectos de los fármacos , Receptor alfa de Ácido Retinoico/metabolismo
8.
Biochem Biophys Res Commun ; 510(3): 427-434, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30732855

RESUMEN

Autophagy and endocytosis are important pathways regulating macromolecule recycling and regeneration. Small molecule inhibitors are utilized to modulate these pathways and to treat autophagy-related diseases. Vacuolin-1 is a small molecule that can potently and reversibly inhibit autophagy by activating Rab5. In addition, Vacuolin-1 can be applied to inhibit exocytosis in a variety of cell types. Here we report that Vacuolin-1 significantly reduces small interference RNA (siRNA)-mediated gene silencing delivered by liposome transfection reagent or lipid nanoparticles in Hela cells. Vacuolin-1 exhibits the strongest inhibition effect among a few autophagy inhibitors including Chloroquine, Wortmannin, and Bafilomycin A1. We found that siRNAs are over-accumulated intracellularly and colocalized with a late endosome marker Rab7 in Vacuolin-1 treated cells, suggesting Vacuolin-1 inhibits the cytoplasmic release of lipid siRNA complexes from late endosomes. We propose that Vacuolin-1 could potentially be used to control the effects of lipid nanoparticle-based RNAi and gene therapy drugs.


Asunto(s)
Autofagia/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Interferencia de ARN/efectos de los fármacos , Exocitosis , Células HeLa , Humanos , Lípidos , Liposomas , Nanopartículas , ARN Interferente Pequeño/análisis , ARN Interferente Pequeño/metabolismo , Transfección , Proteínas de Unión al GTP rab/análisis , Proteínas de Unión a GTP rab7
9.
Exp Cell Res ; 370(2): 478-489, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-29990474

RESUMEN

Autophagy is an evolutionarily conserved lysosomal degradation process, and is involved in various cellular processes. Here we studied the role of two pore channel 2 (TPC2), a lysosomal non-selective Na+/Ca2+ channel, in autophagy progression. We found that TPC overexpression in 4T1 mouse breast cancer cell line or in HeLa human cervical cancer cell line inhibited the fusion between autophagosome and lysosome, resulting in the accumulation of autophagosomes accompanied with increased lysosomal pH and TFEB nuclear localization. Interestingly, we also found that extracellular vesicle (EV) secretion was markedly decreased in TPC2 overexpressing cells but was induced in TPC2 knockdown cells. In addition, migration of TPC2 knockdown cells, not TPC2 overexpressing cells, was inhibited. Taken together, these results support a role of TPC2 in autophagy progression and EV trafficking in cancer cells.


Asunto(s)
Canales de Calcio/metabolismo , Núcleo Celular/metabolismo , Vesículas Extracelulares/metabolismo , Lisosomas/metabolismo , Autofagosomas/fisiología , Autofagia/fisiología , Células HeLa , Humanos , Transducción de Señal/fisiología
10.
J Asian Nat Prod Res ; 21(10): 977-984, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29873248

RESUMEN

Two new ent-clerodane diterpenoids, named isoscoparins R and S (1 and 2), were isolated from the aerial parts of Isodon scoparius. Their structures were characterized mainly by analyzing the NMR and HRESIMS data, and the relative configuration of compound 1 was determined by single-crystal X-ray diffraction. Compound 2 showed weak activity as an autophagic inhibitor.


Asunto(s)
Diterpenos de Tipo Clerodano/química , Diterpenos de Tipo Clerodano/farmacología , Isodon/química , Antineoplásicos Fitogénicos/química , Autofagia/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Medicamentos Herbarios Chinos/química , Células HEK293 , Células HeLa , Humanos , Inmunosupresores/farmacología , Espectroscopía de Resonancia Magnética , Estructura Molecular , Componentes Aéreos de las Plantas/química , Espectrometría de Masa por Ionización de Electrospray , Difracción de Rayos X
11.
Biochim Biophys Acta Mol Cell Res ; 1864(6): 957-967, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28007458

RESUMEN

High intracellular levels of reactive oxygen species (ROS) cause oxidative stress that results in numerous pathologies, including cell death. Transient potential receptor melastatin-2 (TRPM2), a Ca2+-permeable cation channel, is mainly activated by intracellular adenosine diphosphate ribose (ADPR) in response to oxidative stress. Here we studied the role and mechanisms of TRPM2-mediated Ca2+ influx on oxidative stress-induced cell death in cancer cells. We found that oxidative stress activated the TRPM2-Ca2+-CaMKII cascade to inhibit early autophagy induction, which ultimately led to cell death in TRPM2 expressing cancer cells. On the other hand, TRPM2 knockdown switched cells from cell death to autophagy for survival in response to oxidative stress. Moreover, we found that oxidative stress activated the TRPM2-CaMKII cascade to further induce intracellular ROS production, which led to mitochondria fragmentation and loss of mitochondrial membrane potential. In summary, our data demonstrated that oxidative stress activates the TRPM2-Ca2+-CaMKII-ROS signal loop to inhibit autophagy and induce cell death.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Calcio/metabolismo , Estrés Oxidativo , Transducción de Señal , Canales Catiónicos TRPM/metabolismo , Animales , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Humanos , Canales Catiónicos TRPM/genética
13.
J Am Chem Soc ; 139(1): 156-170, 2017 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-27936653

RESUMEN

Cyclic adenosine diphosphoribose (cADPR), an endogenous nucleotide derived from nicotinamide adenine dinucleotide (NAD+), mobilizes Ca2+ release from endoplasmic reticulum (ER) via ryanodine receptors (RyRs), yet the bridging protein(s) between cADPR and RyRs remain(s) unknown. Here we synthesized a novel photoaffinity labeling (PAL) cADPR agonist, PAL-cIDPRE, and subsequently applied it to purify its binding proteins in human Jurkat T cells. We identified glyceraldehyde 3-phosphate dehydrogenase (GAPDH) as one of the cADPR binding protein(s), characterized the binding affinity between cADPR and GAPDH in vitro by surface plasmon resonance (SPR) assay, and mapped cADPR's binding sites in GAPDH. We further demonstrated that cADPR induces the transient interaction between GAPDH and RyRs in vivo and that GAPDH knockdown abolished cADPR-induced Ca2+ release. However, GAPDH did not catalyze cADPR into any other known or novel compound(s). In summary, our data clearly indicate that GAPDH is the long-sought-after cADPR binding protein and is required for cADPR-mediated Ca2+ mobilization from ER via RyRs.


Asunto(s)
Adenosina Difosfato Ribosa/metabolismo , Proteínas Portadoras/metabolismo , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Etiquetas de Fotoafinidad/metabolismo , Adenosina Difosfato Ribosa/química , Proteínas Portadoras/química , Células Cultivadas , Clonación Molecular , Gliceraldehído-3-Fosfato Deshidrogenasas/química , Gliceraldehído-3-Fosfato Deshidrogenasas/genética , Humanos , Células Jurkat , Ligandos , Modelos Moleculares , Conformación Molecular , Etiquetas de Fotoafinidad/química
14.
Exp Cell Res ; 346(1): 9-16, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27349290

RESUMEN

Nitric oxide (NO) markedly induces cardiomyocyte (CM) differentiation of embryonic stem (ES) cells. Here we examined the role of the Ca(2+) signaling in the NO-induced CM differentiation of mouse ES cells. We found that NO induced intracellular Ca(2+) increases in ES cells in a dose-dependent manner, and application of IP3 pathway antagonists not only significantly inhibited this induced Ca(2+) increase but also abolished NO-induced CM differentiation of ES cells. Subsequently, all 3 types of inositol 1, 4, 5-trisphosphate (IP3) receptors (IP3Rs) in mouse ES cells were individually or triply knocked down. Interestingly, only knockdown of type 3 IP3R (IP3R3) or triple-knockdown of three types of IP3Rs significantly inhibited the NO-induced Ca(2+) increases. Consistently, IP3R3 knockdown blocked the NO-induced CM differentiation of ES cells. CMs derived from IP3R3 knockdown ES cells also showed both structural and functional defects. In summary, our results indicate that the IP3R3-Ca(2+) pathway is required for NO-induced CM differentiation of ES cells.


Asunto(s)
Diferenciación Celular , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Células Madre Embrionarias de Ratones/citología , Miocitos Cardíacos/citología , Óxido Nítrico/farmacología , Animales , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Receptores de Inositol 1,4,5-Trifosfato/genética , Ratones , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células Madre Embrionarias de Ratones/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo
15.
Molecules ; 22(6)2017 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-28555021

RESUMEN

Autophagy is a fundamental cellular degradation process which is essential for cell homeostasis, and dysfunctional autophagy has been associated with a variety of human diseases, such as cancer. Several autophagy chemical modulators have been applied in a number of preclinical or clinical trials against these autophagy related diseases, especially cancer. Small molecule vacuolin-1 potently and reversibly inhibits both endosomal-lysosomal trafficking and autophagosome-lysosome fusion, yet the molecular mechanisms underlying vacuolin-1 mediated autophagy inhibition remain unknown. Here, we first performed the virtual drug screening and identified 14 vacuolin-1 analogues as autophagy inhibitors. Based on these virtual screening results, we further designed and synthesized 17 vacuolin-1 analogues, and found that 13 of them are autophagy inhibitors and a couple of them are as potent as vacuolin-1. In summary, these studies expanded the pool of useful autophagy inhibitors and reveal the structural-activity relationship of vacuolin-1 analogues, which is useful for future development of vacuolin-1 analogues with high potency and for identification of the molecular targets of vacuolin-1.


Asunto(s)
Autofagia/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Compuestos Heterocíclicos de 4 o más Anillos/química , Endosomas/química , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Lisosomas/química
16.
Stem Cells ; 33(9): 2664-73, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26012865

RESUMEN

CD38 is a multifunctional membrane enzyme and the main mammalian ADP-ribosyl cyclase, which catalyzes the synthesis and hydrolysis of cADPR, a potent endogenous Ca(2+) mobilizing messenger. Here, we explored the role of CD38 in the neural differentiation of mouse embryonic stem cells (ESCs). We found that the expression of CD38 was decreased during the differentiation of mouse ESCs initiated by adherent monoculture. Perturbing the CD38/cADPR signaling by either CD38 knockdown or treatment of cADPR antagonists inhibited the neural commitment of mouse ESCs, whereas overexpression of CD38 promoted it. Moreover, CD38 knockdown dampened reactive oxygen species (ROS) production during neural differentiation of ESCs by inhibiting NADPH oxidase activity, while CD38 overexpression enhanced it. Similarly, application of hydrogen peroxide mitigated the inhibitory effects of CD38 knockdown on neural differentiation of ESCs. Taken together, our data indicate that the CD38 signaling pathway is required for neural differentiation of mouse ESCs by modulating ROS production.


Asunto(s)
ADP-Ribosil Ciclasa 1/biosíntesis , Diferenciación Celular/fisiología , Glicoproteínas de Membrana/biosíntesis , Células Madre Embrionarias de Ratones/metabolismo , Neuronas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , ADP-Ribosil Ciclasa 1/genética , Animales , Células Cultivadas , Técnicas de Silenciamiento del Gen , Glicoproteínas de Membrana/genética , Ratones
17.
Chem Rec ; 15(2): 511-23, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25707449

RESUMEN

Cyclic adenosine 5'-diphosphate ribose (cADPR) is a second messenger in the Ca(2+) signaling pathway. To elucidate its molecular mechanism in calcium release, a series of cADPR analogues with modification on ribose, nucleobase, and pyrophosphate have been investigated. Among them, the analogue with the modification of the northern ribose by ether linkage substitution (cIDPRE) exhibits membrane-permeate Ca(2+) agonistic activity in intact HeLa cells, human T cells, mouse cardiac myocytes and neurosecretory PC12 cell lines; thus, cIDPRE and coumarin-caged cIDPRE are valuable probes to investigate the cADPR-mediated Ca(2+) signal pathway.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , ADP-Ribosa Cíclica/química , Sondas Moleculares/química , Miocitos Cardíacos/metabolismo , Animales , Cumarinas/química , ADP-Ribosa Cíclica/síntesis química , Células HeLa , Humanos , Inosina Monofosfato/análogos & derivados , Inosina Monofosfato/química , Células Jurkat , Ratones , Imitación Molecular , Sondas Moleculares/síntesis química , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Células PC12 , Ratas , Relación Estructura-Actividad
18.
J Biol Chem ; 288(33): 24247-63, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23836916

RESUMEN

Autophagy is an evolutionarily conserved lysosomal degradation pathway, yet the underlying mechanisms remain poorly understood. Nicotinic acid adenine dinucleotide phosphate (NAADP), one of the most potent Ca(2+) mobilizing messengers, elicits Ca(2+) release from lysosomes via the two pore channel 2 (TPC2) in many cell types. Here we found that overexpression of TPC2 in HeLa or mouse embryonic stem cells inhibited autophagosomal-lysosomal fusion, thereby resulting in the accumulation of autophagosomes. Treatment of TPC2 expressing cells with a cell permeant-NAADP agonist, NAADP-AM, further induced autophagosome accumulation. On the other hand, TPC2 knockdown or treatment of cells with Ned-19, a NAADP antagonist, markedly decreased the accumulation of autophagosomes. TPC2-induced accumulation of autophagosomes was also markedly blocked by ATG5 knockdown. Interestingly, inhibiting mTOR activity failed to increase TPC2-induced autophagosome accumulation. Instead, we found that overexpression of TPC2 alkalinized lysosomal pH, and lysosomal re-acidification abolished TPC2-induced autophagosome accumulation. In addition, TPC2 overexpression had no effect on general endosomal-lysosomal degradation but prevented the recruitment of Rab-7 to autophagosomes. Taken together, our data demonstrate that TPC2/NAADP/Ca(2+) signaling alkalinizes lysosomal pH to specifically inhibit the later stage of basal autophagy progression.


Asunto(s)
Álcalis/metabolismo , Autofagia , Canales de Calcio/metabolismo , Lisosomas/metabolismo , Fusión de Membrana , Fagosomas/metabolismo , Animales , Autofagia/efectos de los fármacos , Calcio/farmacología , Diferenciación Celular/efectos de los fármacos , Células Madre Embrionarias/citología , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Endosomas/ultraestructura , Células HeLa , Humanos , Concentración de Iones de Hidrógeno/efectos de los fármacos , Lisosomas/efectos de los fármacos , Lisosomas/ultraestructura , Fusión de Membrana/efectos de los fármacos , Ratones , NADP/análogos & derivados , NADP/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fagosomas/efectos de los fármacos , Fagosomas/ultraestructura , Unión Proteica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Proteínas de Unión al GTP rab/efectos de los fármacos , Proteínas de Unión a GTP rab7
19.
J Cell Physiol ; 229(2): 202-12, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23881642

RESUMEN

Human bone marrow-derived mesenchymal stem cells (MSCs) serve as a reservoir for the continuous renewal of various mesenchymal tissues; however, cellular physiology of ion channels is not fully understood. The present study investigated potential roles of large-conductance Ca(2+) -activated potassium (BKCa ) channels and ether-à-go-go potassium (hEag1 or Kv10.1) channels in regulating cell proliferation and differentiation in human MSCs. We found that inhibition of BKCa with paxilline or hEag1 with astemizole, or knockdown of BKCa with shRNAs targeting KCa1.1 or hEag1 channels with shRNAs targeting KCNH1 arrested the cells at G0/G1 phase. In addition, silencing BKCa or hEag1 channels significantly reduced adipogenic differentiation with decrease of lipid accumulation and expression of the adipocyte marker PPARγ, and decreased osteogenic differentiation with reduction of mineral precipitation and osteocalcin. These effects were accompanied with a reduced cyclin D1, cyclin E, p-ERK1/2, and p-Akt. Our results demonstrate that BKCa and hEag1 channels not only regulate cell proliferation, but also participate in the adipogenic and osteogenic differentiations in human MSCs, which indicates that BKCa and hEag1 channels may be essential in maintaining bone marrow physiological function and bone regeneration.


Asunto(s)
Células de la Médula Ósea/citología , Diferenciación Celular , Proliferación Celular , Canales de Potasio Éter-A-Go-Go/metabolismo , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Células Madre Mesenquimatosas/citología , Células de la Médula Ósea/metabolismo , Células Cultivadas , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/genética , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Humanos , Indoles/farmacología , Activación del Canal Iónico , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/antagonistas & inhibidores , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Células Madre Mesenquimatosas/metabolismo , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño
20.
Pflugers Arch ; 466(5): 947-59, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24057349

RESUMEN

Preadipocytes are widely used as an in vitro model to investigate proliferation, adipogenic differentiation, and lipodystrophy; however, cellular physiology and biology are not fully understood in human preadipocytes. The present study was to investigate the expression of transient receptor potential (TRP) channels in human preadipocytes and their potential roles in regulating proliferation and adipogenic differentiation using approaches of confocal microscopy, whole-cell patch voltage-clamp, reverse transcription polymerase chain reaction, Western blot, etc. We found that TRPV2, TRPV4, and TRPM7 channels were abundantly expressed in human preadipocytes. The intracellular Ca(2+) transient activated by the TRPV2 activator probenecid was reversed or prevented by ruthenium red, a TRPV2 blocker. The TRPV4 channel activator, 4α-phorbol 12-13-dicaprinate, enhanced intracellular Ca(2+) oscillations, and the effect was inhibited by the TRPV4 blocker RN-1734. TRPM7 current was recorded with dialysis of Mg(2+)-free pipette solution, which was inhibited by the TRP channel blocker 2-aminoethoxydiphenyl borate and enhanced by acidic extracellular pH. Silencing TRPV2 or TRPM7, but not TRPV4, significantly reduced cell proliferation via inhibiting cyclin D1, cyclin E, and p-ERK1/2. Interestingly, individually silencing these three channels decreased adipogenic differentiation of human preadipocytes by reducing p-Akt kinase. Our results demonstrate for the first time that functional TRPV2, TRPV4, and TRPM7 channels are abundantly expressed in human preadipocytes. TRPV2 and TRPM7, but not TRPV4, regulate cell proliferation via activating cyclin D1, cyclin E, and p-ERK1/2, while they are all involved in adipogenesis in human preadipocytes via phosphorylating Akt kinase.


Asunto(s)
Potenciales de Acción , Adipocitos/metabolismo , Células Madre Adultas/metabolismo , Canales Catiónicos TRPM/metabolismo , Canales Catiónicos TRPV/metabolismo , Adipocitos/citología , Adipocitos/fisiología , Adipogénesis , Células Madre Adultas/citología , Células Madre Adultas/fisiología , Señalización del Calcio , Proliferación Celular , Ciclina D1/metabolismo , Ciclina E/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas , Ésteres del Forbol/farmacología , Probenecid/farmacología , Proteínas Serina-Treonina Quinasas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Rojo de Rutenio/farmacología , Sulfonamidas/farmacología , Canales Catiónicos TRPM/genética , Canales Catiónicos TRPV/agonistas , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA