Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Med Res Rev ; 43(5): 1346-1373, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-36924449

RESUMEN

The P2X7 receptor is an exceptional member of the P2X purinergic receptor family, with its activation requiring high concentrations of extracellular adenosine 5'-triphosphate (ATP) that are often associated with tissue damage and inflammation. In the central nervous system (CNS), it is highly expressed in glial cells, particularly in microglia. In this review, we discuss the role and mechanisms of the P2X7 receptor in mediating neuroinflammation and other pathogenic events in a variety of traumatic CNS damage conditions, which lead to loss of neurological and cognitive functions. We raise the perspective on the steady progress in developing CNS-penetrant P2X7 receptor-specific antagonists that leverage the ATP-P2X7 receptor signaling axis as a potential therapeutic strategy to alleviate traumatic CNS damage and related complications.


Asunto(s)
Sistema Nervioso Central , Receptores Purinérgicos P2X7 , Humanos , Microglía , Antagonistas del Receptor Purinérgico P2X/farmacología , Antagonistas del Receptor Purinérgico P2X/uso terapéutico , Adenosina Trifosfato
2.
Glia ; 71(4): 848-865, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36447422

RESUMEN

Microglial cells are crucial in maintaining central nervous system (CNS) homeostasis and mediating CNS disease pathogenesis. Increasing evidence supports that alterations in the mechanical properties of CNS microenvironments influence glial cell phenotypes, but the mechanisms regulating microglial cell function remain elusive. Here, we examined the mechanosensitive Piezo1 channel in microglial cells, particularly, how Piezo1 channel activation regulates pro-inflammatory activation and production of pro-inflammatory cytokines, using BV2 and primary microglial cells. Piezo1 expression in microglial cells was detected both at mRNA and protein levels. Application of Piezo1 channel activator Yoda1 induced Ca2+ flux to increase intracellular Ca2+ concentration that was reduced by treatment with ruthenium red, a Piezo1 inhibitor, or Piezo1-specific siRNA, supporting that Piezo1 functions as a cell surface Ca2+ -permeable channel. Priming with lipopolysaccharide (LPS) induced microglial cell activation and production of TNF-α and IL-6, which were inhibited by treatment with Yoda1. Furthermore, LPS priming induced the activation of ERK, p38 MAPKs, and NF-κB. LPS-induced activation of NF-κB, but not ERK and p38, was inhibited by treatment with Yoda1. Yoda1-induced inhibition was blunted by siRNA-mediated depletion of Piezo1 expression and, furthermore, treatment with BAPTA-AM to prevent intracellular Ca2+ increase. Collectively, our results support that Piezo1 channel activation downregulates the pro-inflammatory function of microglial cells, especially production of TNF-α and IL-6, by initiating intracellular Ca2+ signaling to inhibit the NF-κB inflammatory signaling pathway. These findings reveal Piezo1 channel activation as a previously unrecognized mechanism regulating microglial cell function, raising an interesting perspective on targeting this molecular mechanism to alleviate neuroinflammation and associated CNS pathologies.


Asunto(s)
Lipopolisacáridos , FN-kappa B , FN-kappa B/metabolismo , Lipopolisacáridos/toxicidad , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-6/metabolismo , Microglía/metabolismo , Transducción de Señal , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
3.
Proteins ; 90(3): 619-624, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34622987

RESUMEN

The P2X7 receptor (P2X7R) is a calcium-permeable cation channel activated by high concentrations of extracellular ATP. It plays a role in vital physiological processes, particularly in innate immunity, and is dysregulated in pathological conditions such as inflammatory diseases, neurodegenerative diseases, mood disorders, and cancers. Structural modeling of the human P2X7R (hP2X7R) based on the recently available structures of the rat P2X7 receptor (rP2XR) in conjunction with molecular docking predicts the orientation of tyrosine at position 288 (Y288) in the extracellular domain to face ATP. In this short communication, we combined site-directed mutagenesis and whole-cell patch-clamp recording to investigate the role of this residue in the hP2X7R function. Mutation of this extracellular residue to amino acids with different properties massively impaired current responses to both ATP and BzATP, suggesting that Y288 is important for normal receptor function. Such a finding facilitates development of an in-depth understanding of the molecular basis of hP2X7R structure-function relationships.


Asunto(s)
Mutagénesis Sitio-Dirigida/métodos , Receptores Purinérgicos P2X7/química , Tirosina/química , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/química , Secuencia de Aminoácidos , Animales , Humanos , Simulación del Acoplamiento Molecular , Mutación , Técnicas de Placa-Clamp , Unión Proteica , Ratas
4.
J Membr Biol ; 255(2-3): 357-361, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35322298

RESUMEN

Large-conductance Ca2+-activated K+ (BKCa) channel and L-type voltage-dependent Ca2+ channel (L-VDCC) play important roles in regulating uterine contractility. The uterus stretch, occurring during pregnancy, is a critical factor to trigger uterine contraction. However, how mechanical stimuli impact the two channels remains unknown. Here we investigated the effects of exposure to mechanical stretches with varying magnitudes and durations on expressions of the two channels in rat uterine smooth muscle cells. Our results show that stretch down-regulates the BKCa channel expression but upregulates the L-VDCC expression. These findings are helpful to better understand the roles of L-VDCC and BKCa channel in stretch-triggered uterine contraction.


Asunto(s)
Canales de Calcio Tipo L , Canales de Potasio de Gran Conductancia Activados por el Calcio , Miocitos del Músculo Liso , Contracción Uterina , Útero , Animales , Canales de Calcio Tipo L/genética , Canales de Calcio Tipo L/metabolismo , Femenino , Canales de Potasio de Gran Conductancia Activados por el Calcio/genética , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Miocitos del Músculo Liso/fisiología , Embarazo , Ratas , Útero/fisiología
5.
Cell Tissue Res ; 388(3): 565-581, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35362831

RESUMEN

Epigenetics plays a critical role in regulating mesenchymal stem cells' (MSCs) fate for tissue repair and regeneration. There is increasing evidence that the inhibition of histone deacetylase (HDAC) isoform 3 can enhance MSC osteogenesis. This study investigated the potential of using a selective HDAC2 and 3 inhibitor, MI192, to promote human dental pulp stromal cells (hDPSCs) bone-like tissue formation in vitro and in vivo within porous Bombyx Mori silk scaffolds. Both 2 and 5 wt% silk scaffolds were fabricated and characterised. The 5 wt% scaffolds possess thicker internal lamellae, reduced scaffold swelling and degradation rates, whilst increased compressive modulus in comparison to the 2 wt% silk scaffold. MI192 pre-treatment of hDPSCs on 5 wt% silk scaffold significantly enhanced hDPSCs alkaline phosphatase activity (ALP). The expression of osteoblast-related genes (RUNX2, ALP, Col1a, OCN) was significantly upregulated in the MI192 pre-treated cells. Histological analysis confirmed that the MI192 pre-treated hDPSCs-silk scaffold constructs promoted bone extracellular matrix (ALP, Col1a, OCN) deposition and mineralisation compared to the untreated group. Following 6 weeks of subcutaneous implantation in nude mice, the MI192 pre-treated hDPSCs-silk scaffold constructs enhanced the vascularisation and extracellular matrix mineralisation compared to untreated control. In conclusion, these findings demonstrate the potential of using epigenetic reprogramming and silk scaffolds to promote hDPSCs bone formation efficacy, which provides evidence for clinical translation of this technology for bone augmentation.


Asunto(s)
Inhibidores de Histona Desacetilasas , Ingeniería de Tejidos , Animales , Benzamidas , Células Cultivadas , Pulpa Dental/metabolismo , Epigénesis Genética , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Isoquinolinas , Ratones , Ratones Desnudos , Osteogénesis/genética , Seda/farmacología , Células del Estroma/metabolismo , Andamios del Tejido
6.
J Cell Physiol ; 236(10): 6897-6906, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33650160

RESUMEN

Vascular stiffening, an early and common characteristic of cardiovascular diseases (CVDs), stimulates vascular smooth muscle cell (VSMC) proliferation which reciprocally accelerates the progression of CVDs. However, the mechanisms by which extracellular matrix stiffness accompanying vascular stiffening regulates VSMC proliferation remain largely unknown. In the present study, we examined the role of the intermediate-conductance Ca2+ -activated K+  (IKCa ) channel in the matrix stiffness regulation of VSMC proliferation by growing A7r5 cells on soft and stiff polydimethylsiloxane substrates with stiffness close to these of arteries under physiological and pathological conditions, respectively. Stiff substrates stimulated cell proliferation and upregulated the expression of the IKCa channel. Stiff substrate-induced cell proliferation was suppressed by pharmacological inhibition using TRAM34, an IKCa channel blocker, or genetic depletion of the IKCa channel. In addition, stiff substrate-induced cell proliferation was also suppressed by reducing extracellular Ca2+ concentration using EGTA or intracellular Ca2+ concentration using BAPTA-AM. Moreover, stiff substrate induced activation of extracellular signal-regulated kinases (ERKs), which was inhibited by treatment with TRAM34 or BAPTA-AM. Stiff substrate-induced cell proliferation was suppressed by treatment with PD98059, an ERK inhibitor. Taken together, these results show that substrates with pathologically relevant stiffness upregulate the IKCa channel expression to enhance intracellular Ca2+ signaling and subsequent activation of the ERK signal pathway to drive cell proliferation. These findings provide a novel mechanism by which vascular stiffening regulates VSMC function.


Asunto(s)
Señalización del Calcio , Proliferación Celular , Dimetilpolisiloxanos/química , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Mecanotransducción Celular , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Animales , Técnicas de Cultivo de Célula , Línea Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Ratas
7.
Stem Cells ; 38(3): 410-421, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31746084

RESUMEN

In this study, we examined the Ca2+ -permeable Piezo1 channel, a newly identified mechanosensing ion channel, in human dental pulp-derived mesenchymal stem cells (MSCs) and hypothesized that activation of the Piezo1 channel regulates MSC migration via inducing ATP release and activation of the P2 receptor purinergic signaling. The Piezo1 mRNA and protein were readily detected in hDP-MSCs from multiple donors and, consistently, brief exposure to Yoda1, the Piezo1 channel-specific activator, elevated intracellular Ca2+ concentration. Yoda1-induced Ca2+ response was inhibited by ruthenium red or GsMTx4, two Piezo1 channel inhibitors, and also by Piezo1-specific siRNA. Brief exposure to Yoda1 also induced ATP release. Persistent exposure to Yoda1 stimulated MSC migration, which was suppressed by Piezo1-specific siRNA, and also prevented by apyrase, an ATP scavenger, or PPADS, a P2 generic antagonist. Furthermore, stimulation of MSC migration induced by Yoda1 as well as ATP was suppressed by PF431396, a PYK2 kinase inhibitor, or U0126, an inhibitor of the mitogen-activated protein kinase MEK/ERK signaling pathway. Collectively, these results suggest that activation of the Piezo1 channel stimulates MSC migration via inducing ATP release and subsequent activation of the P2 receptor purinergic signaling and downstream PYK2 and MEK/ERK signaling pathways, thus revealing novel insights into the molecular and signaling mechanisms regulating MSC migration. Such findings provide useful information for evolving a full understanding of MSC migration and homing and developing strategies to improve MSC-based translational applications.


Asunto(s)
Adenosina Trifosfato/metabolismo , Canales Iónicos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Receptores Purinérgicos P2/metabolismo , Adulto , Movimiento Celular , Niño , Femenino , Humanos , Masculino , Transducción de Señal , Adulto Joven
8.
Purinergic Signal ; 17(3): 331-344, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33987781

RESUMEN

The P2X7 receptor, originally known as the P2Z receptor due to its distinctive functional properties, has a structure characteristic of the ATP-gated ion channel P2X receptor family. The P2X7 receptor is an important mediator of ATP-induced purinergic signalling and is involved the pathogenesis of numerous conditions as well as in the regulation of diverse physiological functions. Functional characterisations, in conjunction with site-directed mutagenesis, molecular modelling, and, recently, structural determination, have provided significant insights into the structure-function relationships of the P2X7 receptor. This review discusses the current understanding of the structural basis for the functional properties of the P2X7 receptor.


Asunto(s)
Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Líquido Extracelular/metabolismo , Receptores Purinérgicos P2X7/química , Receptores Purinérgicos P2X7/metabolismo , Adenosina Trifosfato/genética , Secuencia de Aminoácidos , Animales , Sitios de Unión/fisiología , Humanos , Estructura Secundaria de Proteína , Receptores Purinérgicos P2X7/genética
9.
Int J Mol Sci ; 22(10)2021 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-34069280

RESUMEN

The use of human dental pulp stromal cells (hDPSCs) has gained increasing attention as an alternative stem cell source for bone tissue engineering. The modification of the cells' epigenetics has been found to play an important role in regulating differentiation, with the inhibition of histone deacetylases 3 (HDAC3) being linked to increased osteogenic differentiation. This study aimed to induce epigenetic reprogramming using the HDAC2 and 3 selective inhibitor, MI192 to promote hDPSCs osteogenic capacity for bone regeneration. MI192 treatment caused a time-dose-dependent change in hDPSC morphology and reduction in viability. Additionally, MI192 successfully augmented hDPSC epigenetic functionality, which resulted in increased histone acetylation and cell cycle arrest at the G2/M phase. MI192 pre-treatment exhibited a dose-dependent effect on hDPSCs alkaline phosphatase activity. Quantitative PCR and In-Cell Western further demonstrated that MI192 pre-treatment significantly upregulated hDPSCs osteoblast-related gene and protein expression (alkaline phosphatase, bone morphogenic protein 2, type I collagen and osteocalcin) during osteogenic differentiation. Importantly, MI192 pre-treatment significantly increased hDPSCs extracellular matrix collagen production and mineralisation. As such, for the first time, our findings show that epigenetic reprogramming with the HDAC2 and 3 selective inhibitor MI192 accelerates the osteogenic differentiation of hDPSCs, demonstrating the considerable utility of this MSCs engineering approach for bone augmentation strategies.


Asunto(s)
Benzamidas/farmacología , Pulpa Dental/citología , Inhibidores de Histona Desacetilasas/farmacología , Isoquinolinas/farmacología , Osteogénesis/efectos de los fármacos , Acetilación/efectos de los fármacos , Fosfatasa Alcalina/metabolismo , Benzamidas/administración & dosificación , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/administración & dosificación , Histonas/metabolismo , Humanos , Isoquinolinas/administración & dosificación , Tercer Molar/citología , Osteogénesis/fisiología , Células del Estroma/metabolismo
10.
J Cell Mol Med ; 24(1): 4-12, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31568632

RESUMEN

The transient receptor potential melastatin-related 2 (TRPM2) channel, a reactive oxygen species (ROS)-sensitive cation channel, has been well recognized for being an important and common mechanism that confers the susceptibility to ROS-induced cell death. An elevated level of ROS is a salient feature of ischaemia-reperfusion, chronic cerebral hypo-perfusion and neonatal hypoxia-ischaemia. The TRPM2 channel is expressed in hippocampus, cortex and striatum, the brain regions that are critical for cognitive functions. In this review, we examine the recent studies that combine pharmacological and/or genetic interventions with using in vitro and in vivo models to demonstrate a crucial role of the TRPM2 channel in brain damage by ischaemia-reperfusion, chronic cerebral hypo-perfusion and neonatal hypoxic-ischaemia. We also discuss the current understanding of the underlying TRPM2-dependent cellular and molecular mechanisms. These new findings lead to the hypothesis of targeting the TRPM2 channel as a potential novel therapeutic strategy to alleviate brain damage and cognitive dysfunction caused by these conditions.


Asunto(s)
Hipoxia-Isquemia Encefálica/patología , Hipoxia-Isquemia Encefálica/terapia , Terapia Molecular Dirigida , Daño por Reperfusión/patología , Daño por Reperfusión/terapia , Canales Catiónicos TRPM/metabolismo , Humanos , Hipoxia-Isquemia Encefálica/metabolismo , Recién Nacido , Daño por Reperfusión/metabolismo
11.
J Cell Mol Med ; 24(6): 3739-3744, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32065503

RESUMEN

Mechanical stimulation is an important factor regulating mesenchymal stem cell (MSC) functions such as proliferation. The Ca2+ -activated K+ channel, KCa 3.1, is critically engaged in MSC proliferation but its role in mechanical regulation of MSC proliferation remains unknown. Here, we examined the KCa 3.1 channel expression and its role in rat bone marrow-derived MSC (BMSC) proliferation in response to mechanical stretch. Application of mechanical stretch stimulated BMSC proliferation via promoting cell cycle progression. Such mechanical stimulation up-regulated the KCa 3.1 channel expression and pharmacological or genetic inhibition of the KCa 3.1 channel strongly suppressed stretch-induced increase in cell proliferation and cell cycle progression. These results support that the KCa 3.1 channel plays an important role in transducing mechanical forces to MSC proliferation. Our finding provides new mechanistic insights into how mechanical stimuli regulate MSC proliferation and also a viable bioengineering approach to improve MSC proliferation.


Asunto(s)
Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Estrés Mecánico , Animales , Proliferación Celular , Masculino , Ratas Sprague-Dawley
12.
Purinergic Signal ; 16(4): 485-490, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33029714

RESUMEN

The P2X7 receptor (P2X7R) is an ATP-gated cation channel with a critical role in many physiological and pathological processes, and shows prominent functional differences across mammalian species, exemplified by larger current responses of the rat (r) P2X7R to ATP and its analogue BzATP and a greater sensitivity to agonists compared with the human (h) P2X7R. Here, we showed that substitution of Val87 residue in the extracellular domain of the hP2X7R with isoleucine in the rP2X7R increased the current responses of the hP2X7R to both ATP and BzATP. Conversely, introduction of reciprocal I87V mutation in the rP2X7R led to a noticeable but statistically insignificant reduction in the current responses of the rP2X7R to ATP and BzATP. The mutations did not affect the sensitivity of the human and rat P2X7Rs to ATP and BzATP. These results suggest a contribution of Val/Ile87 in agonist-induced current responses of human and rat P2X7Rs, which helps to better understand the molecular determinants for species-dependent function of the mammalian P2X7Rs.


Asunto(s)
Receptores Purinérgicos P2X7/genética , Receptores Purinérgicos P2X7/metabolismo , Animales , Células HEK293 , Humanos , Mutación , Ratas
13.
Part Fibre Toxicol ; 17(1): 23, 2020 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-32513195

RESUMEN

BACKGROUND: Wide applications of nanoparticles (NPs) have raised increasing concerns about safety to humans. Oxidative stress and inflammation are extensively investigated as mechanisms for NPs-induced toxicity. Autophagy and lysosomal dysfunction are emerging molecular mechanisms. Inhalation is one of the main pathways of exposing humans to NPs, which has been reported to induce severe pulmonary inflammation. However, the underlying mechanisms and, more specifically, the interplays of above-mentioned mechanisms in NPs-induced pulmonary inflammation are still largely obscure. Considered that NPs exposure in modern society is often unavoidable, it is highly desirable to develop effective strategies that could help to prevent nanomaterials-induced pulmonary inflammation. RESULTS: Pulmonary inflammation induced by intratracheal instillation of silica nanoparticles (SiNPs) in C57BL/6 mice was prevented by PJ34, a poly (ADP-ribose) polymerase (PARP) inhibitor. In human lung bronchial epithelial (BEAS-2B) cells, exposure to SiNPs reduced cell viability, and induced ROS generation, impairment in lysosome function and autophagic flux. Inhibition of ROS generation, PARP and TRPM2 channel suppressed SiNPs-induced lysosome impairment and autophagy dysfunction and consequent inflammatory responses. Consistently, SiNPs-induced pulmonary inflammation was prevented in TRPM2 deficient mice. CONCLUSION: The ROS/PARP/TRPM2 signaling is critical in SiNPs-induced pulmonary inflammation, providing novel mechanistic insights into NPs-induced lung injury. Our study identifies TRPM2 channel as a new target for the development of preventive and therapeutic strategies to mitigate nanomaterials-induced lung inflammation.


Asunto(s)
Autofagia/efectos de los fármacos , Lisosomas/efectos de los fármacos , Nanopartículas/toxicidad , Neumonía/inducido químicamente , Poli(ADP-Ribosa) Polimerasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Dióxido de Silicio/toxicidad , Canales Catiónicos TRPM/metabolismo , Animales , Exposición por Inhalación , Lisosomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Tamaño de la Partícula , Neumonía/metabolismo , Neumonía/patología , Transducción de Señal , Propiedades de Superficie
14.
J Cell Physiol ; 234(4): 3647-3660, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30229906

RESUMEN

Transient receptor potential melastatin 2 (TRPM2) channel activation by reactive oxygen species (ROS) plays a critical role in delayed neuronal cell death, responsible for postischemia brain damage via altering intracellular Zn2+ homeostasis, but a mechanistic understanding is still lacking. Here, we showed that H2 O2 induced neuroblastoma SH-SY5Y cell death with a significant delay, dependently of the TRPM2 channel and increased [Zn2+ ]i , and therefore used this cell model to investigate the mechanisms underlying ROS-induced TRPM2-mediated delayed cell death. H2 O2 increased concentration-dependently the [Zn2+ ]i and caused lysosomal dysfunction and Zn2+ loss and, furthermore, mitochondrial Zn2+ accumulation, fragmentation, and ROS generation. Such effects were suppressed by preventing poly(adenosine diphosphate ribose, ADPR) polymerase-1-dependent TRPM2 channel activation with PJ34 and 3,3',5,5'-tetra-tert-butyldiphenoquinone, inhibiting the TRPM2 channel with 2-aminoethoxydiphenyl borate (2-APB) and N-(p-amylcinnamoyl)anthranilic acid, or chelating Zn2+ with N,N,N,N-tetrakis(2-pyridylmethyl)-ethylenediamine (TPEN). Bafilomycin-induced lysosomal dysfunction also resulted in mitochondrial Zn2+ accumulation, fragmentation, and ROS generation that were inhibited by PJ34 or 2-APB, suggesting that these mitochondrial events are TRPM2 dependent and sequela of lysosomal dysfunction. Mitochondrial TRPM2 expression was detected and exposure to ADPR-induced Zn2+ uptake in isolated mitochondria, which was prevented by TPEN. H2 O2 -induced delayed cell death was inhibited by apocynin and diphenyleneiodonium, nicotinamide adenine dinucleotide phosphate hydrogen (NADPH) oxidase (NOX) inhibitors, GKT137831, an NOX1/4-specific inhibitor, or Gö6983, a protein kinase C (PKC) inhibitor. Moreover, inhibition of PKC/NOX prevented H2 O2 -induced ROS generation, lysosomal dysfunction and Zn2+ release, and mitochondrial Zn2+ accumulation, fragmentation and ROS generation. Collectively, these results support a critical role for the TRPM2 channel in coupling PKC/NOX-mediated ROS generation, lysosomal Zn2+ release, and mitochondrial Zn2+ accumulation, and ROS generation to form a vicious positive feedback signaling mechanism for ROS-induced delayed cell death.


Asunto(s)
Peróxido de Hidrógeno/toxicidad , Neuronas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Canales Catiónicos TRPM/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Retroalimentación Fisiológica , Células HEK293 , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Lisosomas/patología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , NADPH Oxidasas/metabolismo , Neuronas/metabolismo , Neuronas/patología , Proteína Quinasa C/metabolismo , Transducción de Señal , Canales Catiónicos TRPM/genética , Factores de Tiempo , Zinc/metabolismo
15.
Glia ; 66(3): 562-575, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29143372

RESUMEN

Amyloid ß (Aß)-induced neuroinflammation plays an important part in Alzheimer's disease (AD). Emerging evidence supports a role for the transient receptor potential melastatin-related 2 (TRPM2) channel in Aß-induced neuroinflammation, but how Aß induces TRPM2 channel activation and this relates to neuroinflammation remained poorly understood. We investigated the mechanisms by which Aß42 activates the TRPM2 channel in microglial cells and the relationships to microglial activation and generation of tumor necrosis factor-α (TNF-α), a key cytokine implicated in AD. Exposure to 10-300 nM Aß42 induced concentration-dependent microglial activation and generation of TNF-α that were ablated by genetically deleting (TRPM2 knockout ;TRPM2-KO) or pharmacologically inhibiting the TRPM2 channel, revealing a critical role of this channel in Aß42 -induced microglial activation and generation of TNF-α. Mechanistically, Aß42 activated the TRPM2 channel via stimulating generation of reactive oxygen species (ROS) and activation of poly(ADPR) polymerase-1 (PARP-1). Aß42 -induced generation of ROS and activation of PARP-1 and TRPM2 channel were suppressed by inhibiting protein kinase C (PKC) and NADPH oxidases (NOX). Aß42 -induced activation of PARP-1 and TRPM2 channel was also reduced by inhibiting PYK2 and MEK/ERK. Aß42 -induced activation of PARP-1 was attenuated by TRPM2-KO and moreover, the remaining PARP-1 activity was eliminated by inhibiting PKC and NOX, but not PYK2 and MEK/ERK. Collectively, our results suggest that PKC/NOX-mediated generation of ROS and subsequent activation of PARP-1 play a role in Aß42 -induced TRPM2 channel activation and TRPM2-dependent activation of the PYK2/MEK/ERK signalling pathway acts as a positive feedback to further facilitate activation of PARP-1 and TRPM2 channel. These findings provide novel insights into the mechanisms underlying Aß-induced AD-related neuroinflammation.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Microglía/metabolismo , Fragmentos de Péptidos/metabolismo , Canales Catiónicos TRPM/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Péptidos beta-Amiloides/administración & dosificación , Animales , Calcio/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Sistema de Señalización de MAP Quinasas/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasas/metabolismo , Necrosis/metabolismo , Fragmentos de Péptidos/administración & dosificación , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Proteína Quinasa C/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Canales Catiónicos TRPM/genética
16.
J Cell Sci ; 129(15): 3008-14, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27311484

RESUMEN

Fresh water protozoa and algae face hypotonic challenges in their living environment. Many of them employ a contractile vacuole system to uptake excessive water from the cytoplasm and expel it to the environment to achieve cellular homeostasis. K(+), a major osmolyte in contractile vacuole, is predicted to create higher osmolarity for water influx. Molecular mechanisms for K(+) permeation through the plasma membrane have been well studied. However, how K(+) permeates organelles such as the contractile vacuole is not clear. Here, we show that the six-transmembrane K(+) channel KCN11 in Chlamydomonas is exclusively localized to contractile vacuole. Ectopic expression of KCN11 in HEK293T cells results in voltage-gated K(+) channel activity. Disruption of the gene or mutation of key residues for K(+) permeability of the channel leads to dysfunction of cell osmoregulation in very hypotonic conditions. The contractile cycle is inhibited in the mutant cells with a slower rate of contractile vacuole swelling, leading to cell death. These data demonstrate a new role for six-transmembrane K(+) channels in contractile vacuole functioning and provide further insights into osmoregulation mediated by the contractile vacuole.


Asunto(s)
Chlamydomonas reinhardtii/metabolismo , Orgánulos/metabolismo , Osmorregulación , Canales de Potasio/metabolismo , Proteínas Algáceas/química , Proteínas Algáceas/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Membrana Celular/metabolismo , Chlamydomonas reinhardtii/genética , Proteínas Mutantes/metabolismo , Mutación/genética , Canales de Potasio/química , Transporte de Proteínas , Vacuolas/metabolismo
17.
Cell Mol Life Sci ; 74(20): 3697-3710, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28534085

RESUMEN

The ability of cells to migrate to the destined tissues or lesions is crucial for physiological processes from tissue morphogenesis, homeostasis and immune responses, and also for stem cell-based regenerative medicines. Cytosolic Ca2+ is a primary second messenger in the control and regulation of a wide range of cell functions including cell migration. Extracellular ATP, together with the cognate receptors on the cell surface, ligand-gated ion channel P2X receptors and a subset of G-protein-coupled P2Y receptors, represents common autocrine and/or paracrine Ca2+ signalling mechanisms. The P2X receptor ion channels mediate extracellular Ca2+ influx, whereas stimulation of the P2Y receptors triggers intracellular Ca2+ release from the endoplasmic reticulum (ER), and activation of both type of receptors thus can elevate the cytosolic Ca2+ concentration ([Ca2+]c), albeit with different kinetics and capacity. Reduction in the ER Ca2+ level following the P2Y receptor activation can further induce store-operated Ca2+ entry as a distinct Ca2+ influx pathway that contributes in ATP-induced increase in the [Ca2+]c. Mesenchymal stem cells (MSC) are a group of multipotent stem cells that grow from adult tissues and hold promising applications in tissue engineering and cell-based therapies treating a great and diverse number of diseases. There is increasing evidence to show constitutive or evoked ATP release from stem cells themselves or mature cells in the close vicinity. In this review, we discuss the mechanisms for ATP release and clearance, the receptors and ion channels participating in ATP-induced Ca2+ signalling and the roles of such signalling mechanisms in mediating ATP-induced regulation of MSC migration.


Asunto(s)
Adenosina Trifosfato/metabolismo , Señalización del Calcio , Movimiento Celular , Células Madre Mesenquimatosas/citología , Animales , Calcio/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Receptores Purinérgicos P2/metabolismo
18.
J Cell Physiol ; 232(2): 287-297, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27403750

RESUMEN

Extracellular ATP and other nucleotides induce autocrine and/or paracrine purinergic signalling via activation of the P2 receptors on the cell surface, which represents one of the most common signalling mechanisms. Mesenchymal stem cells (MSC) are a type of multipotent adult stem cells that have many promising applications in regenerative medicine. There is increasing evidence to show that extracellular nucleotides regulate MSC functions and P2 receptor-mediated purinergic signalling plays an important role in such functional regulation. P2 receptors comprise ligand-gated ion channel P2X receptors and G-protein-coupled P2Y receptors. In this review, we provide an overview of the current understanding with respect to expression of the P2X and P2Y receptors in MSC and their roles in mediating extracellular nucleotide regulation of MSC proliferation, migration and differentiation. J. Cell. Physiol. 232: 287-297, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Espacio Extracelular/metabolismo , Células Madre Mesenquimatosas/metabolismo , Nucleótidos/metabolismo , Receptores Purinérgicos P2/metabolismo , Animales , Diferenciación Celular , Humanos , Células Madre Mesenquimatosas/citología
19.
Stem Cells ; 34(8): 2102-14, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27038239

RESUMEN

ATP is an extrinsic signal that can induce an increase in the cytosolic Ca(2+) level ([Ca(2+) ]c ) in mesenchymal stem cells (MSCs). However, the cognate intrinsic mechanisms underlying ATP-induced Ca(2+) signaling in MSCs is still contentious, and their importance in MSC migration remains unknown. In this study, we investigated the molecular mechanisms underlying ATP-induced Ca(2+) signaling and their roles in the regulation of cell migration in human dental pulp MSCs (hDP-MSCs). RT-PCR analysis of mRNA transcripts and interrogation of agonist-induced increases in the [Ca(2+) ]c support that P2X7, P2Y1 , and P2Y11 receptors participate in ATP-induced Ca(2+) signaling. In addition, following P2Y receptor activation, Ca(2+) release-activated Ca(2+) Orai1/Stim1 channel as a downstream mechanism also plays a significant role in ATP-induced Ca(2+) signaling. ATP concentration-dependently stimulates hDP-MSC migration. Pharmacological and genetic interventions of the expression or function of the P2X7, P2Y1 and P2Y11 receptors, and Orai1/Stim1 channel support critical involvement of these Ca(2+) signaling mechanisms in ATP-induced stimulation of hDP-MSC migration. Taken together, this study provide evidence to show that purinergic P2X7, P2Y1 , and P2Y11 receptors and store-operated Orai1/Stim1 channel represent important molecular mechanisms responsible for ATP-induced Ca(2+) signaling in hDP-MSCs and activation of these mechanisms stimulates hDP-MSC migration. Such information is useful in building a mechanistic understanding of MSC homing in tissue homeostasis and developing more efficient MSC-based therapeutic applications. Stem Cells 2016;34:2102-2114.


Asunto(s)
Adenosina Trifosfato/farmacología , Señalización del Calcio/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Receptores Purinérgicos/metabolismo , Adulto , Niño , Pulpa Dental/citología , Espacio Extracelular/metabolismo , Femenino , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/metabolismo , Molécula de Interacción Estromal 1/metabolismo , Adulto Joven
20.
Purinergic Signal ; 13(1): 135-141, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28025718

RESUMEN

The P2X7 receptor (P2X7R) is important in mediating a range of physiological functions and pathologies associated with tissue damage and inflammation and represents an attractive therapeutic target. However, in terms of their structure-function relationships, the mammalian P2X7Rs remain poorly characterised compared to some of their other P2XR counterparts. In this study, combining cysteine-based cross-linking and whole-cell patch-clamp recording, we examined six pairs of residues (A44/I331, D48/I331, I58/F311, S60/L320, I75/P177 and K81/V304) located in different parts of the extracellular and transmembrane domains of the human P2X7R. These residues are predicted to undergo substantial movement during the transition of the receptor ion channel from the closed to the open state, predictions which are made based on structural homology models generated from the crystal structures of the zebrafish P2X4R. Our results provide evidence that among the six pairs of cysteine mutants, D48C/I133C and K81C/V304C formed disulphide bonds that impaired the channel gating to support the notion that such conformational changes, particularly those in the outer ends of the transmembrane domains, are critical for human P2X7R activation.


Asunto(s)
Activación del Canal Iónico/fisiología , Receptores Purinérgicos P2X7/metabolismo , Cisteína , Humanos , Modelos Moleculares , Conformación Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA