Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 2485, 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38509117

RESUMEN

Proteasome subunit hRpn13 is partially proteolyzed in certain cancer cell types to generate hRpn13Pru by degradation of its UCHL5/Uch37-binding DEUBAD domain and retention of an intact proteasome- and ubiquitin-binding Pru domain. By using structure-guided virtual screening, we identify an hRpn13 binder (XL44) and solve its structure ligated to hRpn13 Pru by integrated X-ray crystallography and NMR to reveal its targeting mechanism. Surprisingly, hRpn13Pru is depleted in myeloma cells following treatment with XL44. TMT-MS experiments reveal a select group of off-targets, including PCNA clamp-associated factor PCLAF and ribonucleoside-diphosphate reductase subunit M2 (RRM2), that are similarly depleted by XL44 treatment. XL44 induces hRpn13-dependent apoptosis and also restricts cell viability by a PCLAF-dependent mechanism. A KEN box, but not ubiquitination, is required for XL44-induced depletion of PCLAF. Here, we show that XL44 induces ubiquitin-dependent loss of hRpn13Pru and ubiquitin-independent loss of select KEN box containing proteins.


Asunto(s)
Glicoproteínas de Membrana , Complejo de la Endopetidasa Proteasomal , Complejo de la Endopetidasa Proteasomal/metabolismo , Glicoproteínas de Membrana/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Ubiquitina/metabolismo , Citoplasma/metabolismo , Factores de Transcripción
2.
Mol Cell ; 84(3): 522-537.e8, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38151017

RESUMEN

The anti-cancer target hRpn13 is a proteasome substrate receptor. However, hRpn13-targeting molecules do not impair its interaction with proteasomes or ubiquitin, suggesting other critical cellular activities. We find that hRpn13 depletion causes correlated proteomic and transcriptomic changes, with pronounced effects in myeloma cells for cytoskeletal and immune response proteins and bone-marrow-specific arginine deiminase PADI4. Moreover, a PROTAC against hRpn13 co-depletes PADI4, histone deacetylase HDAC8, and DNA methyltransferase MGMT. PADI4 binds and citrullinates hRpn13 and proteasomes, and proteasomes from PADI4-inhibited myeloma cells exhibit reduced peptidase activity. When off proteasomes, hRpn13 can bind HDAC8, and this interaction inhibits HDAC8 activity. Further linking hRpn13 to transcription, its loss reduces nuclear factor κB (NF-κB) transcription factor p50, which proteasomes generate by cleaving its precursor protein. NF-κB inhibition depletes hRpn13 interactors PADI4 and HDAC8. Altogether, we find that hRpn13 acts dually in protein degradation and expression and that proteasome constituency and, in turn, regulation varies by cell type.


Asunto(s)
Histona Desacetilasas , Péptidos y Proteínas de Señalización Intracelular , FN-kappa B , Arginina Deiminasa Proteína-Tipo 4 , Factores de Transcripción , Humanos , Epigénesis Genética , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteoma/metabolismo , Proteómica , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Transcriptoma , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Arginina Deiminasa Proteína-Tipo 4/metabolismo , Línea Celular Tumoral
3.
Environ Technol ; : 1-21, 2023 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-37955936

RESUMEN

This study developed a novel constructed wetland (CW) coupled with a magnetic field for treating domestic wastewater, and the magnetic field distribution was solved and optimised by the finite element method. Herein, we investigated the effects of optimising magnetic field optimisation and studied its impact on CW treatment performance and the responses of a microbial community. The optimisation results showed that the average magnetic field strength of the CW unit increases from 3 to 8 mT, and the proportion of areas with magnetic field strength greater than 5 mT also increases from 30% to 74%. The water quality analysis results showed that the removal of chemical oxygen demand (COD) and NH4+-N (p < 0.01) was significantly increased by the magnetic field (average 3 mT), increasing by 12.2% and 8.49%, respectively. Moreover, the removal of COD and NH4+-N (p < 0.01) was more significantly increased by M-VFCW(O) (average 8 mT), increasing by 15.58% and 49.1%, respectively. The magnetic field application shifted significantly the abundance of dominant bacteria in CWs. Relative abundance of dominant bacteria such as Proteobacteria (63.3%), Firmicutes (4.72%) and Actinobacteria (2.11%) that played an important role in organics removal and nitrification and denitrification-related bacteria such as Nitrospirae (1.48%) and Planctomycetes (9.58%) significantly promoted in M-VFCW(O). These results suggest that introducing a magnetic field into CWs may improve organics and nitrogen removal via the biological process, and the optimisation of the magnetic field was significant in enhancing the performance of VFCWs.

4.
BMC Infect Dis ; 22(1): 34, 2022 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-34991517

RESUMEN

BACKGROUND: Sepsis is a leading cause of pediatric morbidity and mortality worldwide. The aim of this study was to explore the association of decreased mitochondrial respiratory chain enzyme activities with the risk for pediatric sepsis, and explore their association with mortality among affected children. METHODS: A total of 50 incident cases with sepsis and 49 healthy controls participated in this study. The level of serum coenzyme Q10 was measured by high-performance liquid chromatography, and selected mitochondrial respiratory chain enzymes in WBC were measured using spectrophotometric. Logistic regression models were used to estimate odds ratio (OR) and 95% confidence interval (CI). RESULTS: The levels of CoQ10, complex II, complex I + III and FoF1-ATPase were significantly higher in healthy controls than in children with sepsis (p < 0.001, = 0.004, < 0.001 and < 0.001, respectively). In children with sepsis, levels of CoQ10 and complex I + III were significantly higher in survived cases than in deceased cases (p < 0.001). Per 0.05 µmol/L, 50 nmol/min.mg and 100 nmol/min.mg increment in CoQ10, complex I + III and FoF1-ATPase were associated with significantly lowered risk of having sepsis, even after adjusting for confounding factors (OR = 0.85, 0.68 and 0.04, p = 0.001, < 0.001 and < 0.001, respectively). Per 0.05 µmol/L and 50 nmol/min.mg increment in CoQ10 and complex I + III was associated with significantly lowered risk of dying from sepsis during hospitalization, and significance retained after adjustment (OR = 0.73 and 0.76, 95% CI: 0.59 to 0.90 and 0.64 to 0.89, p = 0.004 and 0.001, respectively) in children with sepsis. CONCLUSIONS: Our findings indicate the promising predictive contribution of low serum CoQ10 and complex I + III to the risk of pediatric sepsis and its associated mortality during hospitalization among Chinese children. Trial registration The trial was registered with www.chictr.org.cn , number ChiCTR-IOR-15006446 on May 05, 2015. Retrospectively registered.


Asunto(s)
Sepsis , Niño , China/epidemiología , Transporte de Electrón , Humanos , Sepsis/epidemiología
5.
Nat Commun ; 12(1): 7318, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34916494

RESUMEN

Proteasome substrate receptor hRpn13 is a promising anti-cancer target. By integrated in silico and biophysical screening, we identified a chemical scaffold that binds hRpn13 with non-covalent interactions that mimic the proteasome and a weak electrophile for Michael addition. hRpn13 Pru domain binds proteasomes and ubiquitin whereas its DEUBAD domain binds deubiquitinating enzyme UCHL5. NMR revealed lead compound XL5 to interdigitate into a hydrophobic pocket created by lateral movement of a Pru ß-hairpin with an exposed end for Proteolysis Targeting Chimeras (PROTACs). Implementing XL5-PROTACs as chemical probes identified a DEUBAD-lacking hRpn13 species (hRpn13Pru) present naturally with cell type-dependent abundance. XL5-PROTACs preferentially target hRpn13Pru, causing its ubiquitination. Gene-editing and rescue experiments established hRpn13 requirement for XL5-PROTAC-triggered apoptosis. These data establish hRpn13 as an anti-cancer target for multiple myeloma and introduce an hRpn13-targeting scaffold that can be optimized for preclinical trials against hRpn13Pru-producing cancer types.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Mieloma Múltiple/metabolismo , Ubiquitinación , Apoptosis , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Mieloma Múltiple/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional , Ubiquitina/metabolismo
6.
Med Sci Monit ; 27: e932518, 2021 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-34689148

RESUMEN

BACKGROUND We designed an association study among 267 cases of children with sepsis and 283 healthy controls, by genotyping 9 variants in the VDR gene. MATERIAL AND METHODS This was a hospital-based, case-control, genetic association study. In addition to 3 genetic modes of inheritance, haplotype and interaction analyses were employed to examine the prediction of VDR gene for pediatric sepsis. Effect-size estimates are expressed as odds ratio (OR) and 95% confidence interval (CI). RESULTS Two variants in the VDR gene, rs2107301 and rs2189480, were found to play a leading role in susceptibility to sepsis in children. The mutant homozygotes of rs2107301 (CC) and rs2189480 (CC) were associated with a reduced risk of sepsis compared with the corresponding wild homozygotes (OR: 0.44 and 0.43, 95% CI: 0.21-0.92 and 0.23-0.81, p: 0.03 and 0.009, respectively). The mutations of rs2107301-C and rs2189480-C alleles were associated with reduced sepsis risk. Haplotype C-C-C-C-C-T-C-A-G in the VDR gene was significantly associated with a 0.59-fold decreased risk of sepsis (95% CI: 0.12-0.76, p: 0.02). In the haplotype-phenotype analysis, significant association was noted for high-density lipoprotein, even after simulation correction (psim <0.05). CONCLUSIONS Taken together, our findings indicate that the VDR gene may be a sepsis-susceptibility gene in Chinese Han children.


Asunto(s)
Receptores de Calcitriol/genética , Sepsis/genética , Estudios de Casos y Controles , Niño , Preescolar , China , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Lactante , Polimorfismo de Nucleótido Simple
7.
Structure ; 28(5): 495-506.e3, 2020 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-32160516

RESUMEN

Rpn13/Adrm1 is recruited to the proteasome by PSMD1/Rpn2, where it serves as a substrate receptor that binds preferentially to K48-linked ubiquitin chains, an established signal for protein proteolysis. Here, we use NMR to solve the structure of hRpn13 Pru:hRpn2 (940-953):K48-diubiquitin. Surprisingly, hRpn2-bound hRpn13 selects a dynamic, extended conformation of K48-diubiquitin that is unique from previously determined structures. NMR experiments on free K48-diubiquitin demonstrate the presence of the reported "closed" conformation observed by crystallography, but also this more extended state, in which the hRpn13-binding surface is exposed. This extended K48-diubiquitin conformation is defined by interactions between L73 from G76-linked (distal) ubiquitin and a Y59-centered surface of K48-linked (proximal) ubiquitin. Furthermore, hRpn13 exchanges between the two ubiquitins within 100 ms, although prefers the proximal ubiquitin due to interactions with the K48 linker region. Altogether, these data lead to a revised model of how ubiquitinated substrates interact with the proteasome.


Asunto(s)
Hexosiltransferasas/química , Hexosiltransferasas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Complejo de la Endopetidasa Proteasomal/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Hexosiltransferasas/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Lisina/metabolismo , Modelos Moleculares , Complejos Multiproteicos/química , Resonancia Magnética Nuclear Biomolecular , Complejo de la Endopetidasa Proteasomal/genética , Conformación Proteica
8.
ACS Omega ; 3(9): 11917-11929, 2018 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-30288466

RESUMEN

Substitution of the m,p-chloro groups of bis-benzylidinepiperidone RA190 for p-nitro, generating RA183, enhanced covalent drug binding to Cys88 of RPN13. Treatment of cancer cell lines with RA183 inhibited ubiquitin-mediated protein degradation, resulting in rapid accumulation of high-molecular-weight polyubiquitinated proteins, blockade of NFκB signaling, endoplasmic reticulum stress, an unfolded protein response, production of reactive oxygen species, and apoptotic cell death. High-grade ovarian cancer, triple-negative breast cancer, and multiple myeloma cell lines were particularly vulnerable to RA183. RA183 stabilized a tetraubiquitin-linked firefly luciferase reporter protein in cancer cell lines and mice, demonstrating in vitro and in vivo proteasomal inhibition, respectively. However, RA183 was rapidly cleared from plasma, likely reflecting its rapid degradation to the active compound RA9, as seen in human liver microsomes. Intraperitoneal administration of RA183 inhibited proteasome function and orthotopic tumor growth in mice bearing human ovarian cancer model ES2-luc ascites or syngeneic ID8-luc tumor.

9.
Nat Commun ; 8: 15540, 2017 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-28598414

RESUMEN

Proteasome-ubiquitin receptor hRpn13/Adrm1 binds and activates deubiquitinating enzyme Uch37/UCHL5 and is targeted by bis-benzylidine piperidone RA190, which restricts cancer growth in mice xenografts. Here, we solve the structure of hRpn13 with a segment of hRpn2 that serves as its proteasome docking site; a proline-rich C-terminal hRpn2 extension stretches across a narrow canyon of the ubiquitin-binding hRpn13 Pru domain blocking an RA190-binding surface. Biophysical analyses in combination with cell-based assays indicate that hRpn13 binds preferentially to hRpn2 and proteasomes over RA190. hRpn13 also exists outside of proteasomes where it may be RA190 sensitive. RA190 does not affect hRpn13 interaction with Uch37, but rather directly binds and inactivates Uch37. hRpn13 deletion from HCT116 cells abrogates RA190-induced accumulation of substrates at proteasomes. We propose that RA190 targets hRpn13 and Uch37 through parallel mechanisms and at proteasomes, RA190-inactivated Uch37 cannot disassemble hRpn13-bound ubiquitin chains.


Asunto(s)
Antineoplásicos/química , Compuestos de Bencilideno/química , Hexosiltransferasas/metabolismo , Glicoproteínas de Membrana/metabolismo , Neoplasias/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Antineoplásicos/farmacología , Compuestos de Bencilideno/farmacología , Biofisica , Ensayos de Selección de Medicamentos Antitumorales , Regulación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Neoplasias/tratamiento farmacológico , Prolina/química , Unión Proteica , Dominios Proteicos
10.
PLoS One ; 10(10): e0140518, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26466095

RESUMEN

Rpn13 is a proteasome ubiquitin receptor that has emerged as a therapeutic target for human cancers. Its ubiquitin-binding activity is confined to an N-terminal Pru (pleckstrin-like receptor for ubiquitin) domain that also docks it into the proteasome, while its C-terminal DEUBAD (DEUBiquitinase ADaptor) domain recruits deubiquitinating enzyme Uch37 to the proteasome. Bis-benzylidine piperidone derivatives that were found to bind covalently to Rpn13 C88 caused the accumulation of polyubiquitinated proteins as well as ER stress-related apoptosis in various cancer cell lines, including bortezomib-resistant multiple myeloma lines. We find that a 38-amino acid peptide derived from the C-terminus of proteasome PC repeat protein hRpn2/PSMD1 binds to hRpn13 Pru domain with 12 nM affinity. By using NMR, we identify the hRpn13-interacting amino acids in this hRpn2 fragment, some of which are conserved among eukaryotes. Importantly, we find the hRpn2-derived peptide to immunoprecipitate endogenous Rpn13 from 293T cells, and to displace it from the proteasome. These findings indicate that this region of hRpn2 is the primary binding site for hRpn13 in the proteasome. Moreover, the hRpn2-derived peptide was no longer able to interact with endogenous hRpn13 when a strictly conserved phenylalanine (F948 in humans) was replaced with arginine or a stop codon, or when Y950 and I951 were substituted with aspartic acid. Finally, over-expression of the hRpn2-derived peptide leads to an increased presence of ubiquitinated proteins in 293T cells. We propose that this hRpn2-derived peptide could be used to develop peptide-based strategies that specifically target hRpn13 function in the proteasome.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Fragmentos de Péptidos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Secuencia de Aminoácidos , Secuencia Conservada , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular , Glicoproteínas de Membrana/química , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Complejo de la Endopetidasa Proteasomal/química , Complejo de la Endopetidasa Proteasomal/genética , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Alineación de Secuencia
11.
J Biol Chem ; 290(7): 4010-21, 2015 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-25542899

RESUMEN

APOBEC3G (A3G) is a DNA cytidine deaminase (CD) that demonstrates antiviral activity against human immunodeficiency virus 1 (HIV-1) and other pathogenic virus. It has an inactive N-terminal CD1 virus infectivity factor (Vif) protein binding domain (A3G-CD1) and an actively catalytic C-terminal CD2 deamination domain (A3G-CD2). Although many studies on the structure of A3G-CD2 and enzymatic properties of full-length A3G have been reported, the mechanism of how A3G interacts with HIV-1 single-stranded DNA (ssDNA) is still not well characterized. Here, we reported a crystal structure of a novel A3G-CD2 head-to-tail dimer (in which the N terminus of the monomer H (head) interacts with the C terminus of monomer T (tail)), where a continuous DNA binding groove was observed. By constructing the A3G-CD1 structural model, we found that its overall fold was almost identical to that of A3G-CD2. We mutated the residues located in or along the groove in monomer H and the residues in A3G-CD1 that correspond to those seated in or along the groove in monomer T. Then, by performing enzymatic assays, we confirmed the reported key elements and the residues in A3G necessary to the catalytic deamination. Moreover, we identified more than 10 residues in A3G essential to DNA binding and deamination reaction. Therefore, this dimer structure may represent a structural model of full-length A3G, which indicates a possible binding mode of A3G to HIV-1 ssDNA.


Asunto(s)
Citidina Desaminasa/química , Citidina Desaminasa/metabolismo , ADN de Cadena Simple/metabolismo , Pliegue de Proteína , Multimerización de Proteína , Desaminasa APOBEC-3G , Catálisis , Dominio Catalítico , Cristalografía por Rayos X , Desaminación , Humanos , Modelos Moleculares , Unión Proteica , Conformación Proteica
12.
Mol Cell Proteomics ; 11(1): M111.012054, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21969612

RESUMEN

α-Scorpion toxins constitute a family of peptide modulators that induce a prolongation of the action potential of excitable cells by inhibiting voltage-gated sodium channel inactivation. Although they all adopt a conserved structural scaffold, the potency and phylogentic preference of these toxins largely vary, which render them an intriguing model for studying evolutionary diversification among family members. Here, we report molecular characterization of a new multigene family of α-toxins comprising 13 members (named MeuNaTxα-1 to MeuNaTxα-13) from the scorpion Mesobuthus eupeus. Of them, five native toxins (MeuNaTxα-1 to -5) were purified to homogeneity from the venom and the solution structure of MeuNaTxα-5 was solved by nuclear magnetic resonance. A systematic functional evaluation of MeuNaTxα-1, -2, -4, and -5 was conducted by two-electrode voltage-clamp recordings on seven cloned mammalian voltage-gated sodium channels (Na(v)1.2 to Na(v)1.8) and the insect counterpart DmNa(v)1 expressed in Xenopus oocytes. Results show that all these four peptides slow inactivation of DmNa(v)1 and are inactive on Na(v)1.8 at micromolar concentrations. However, they exhibit differential specificity for the other six channel isoforms (Na(v)1.2 to Na(v)1.7), in which MeuNaTxα-4 shows no activity on these isoforms and thus represents the first Mesobuthus-derived insect-selective α-toxin identified so far with a half maximal effective concentration of 130 ± 2 nm on DmNa(v)1 and a half maximal lethal dose of about 200 pmol g(-1) on the insect Musca domestica; MeuNaTxα-2 only affects Na(v)1.4; MeuNaTxα-1 and MeuNaTxα-5 have a wider range of channel spectrum, the former active on Na(v)1.2, Na(v)1.3, Na(v)1.6, and Na(v)1.7, whereas the latter acting on Na(v)1.3-Na(v)1.7. Remarkably, MeuNaTxα-4 and MeuNaTxα-5 are two nearly identical peptides differing by only one point mutation at site 50 (A50V) but exhibit rather different channel subtype selectivity, highlighting a switch role of this site in altering the target specificity. By the maximum likelihood models of codon substitution, we detected nine positively selected sites (PSSs) that could be involved in functional diversification of Mesobuthus α-toxins. The PSSs include site 50 and other seven sites located in functional surfaces of α-toxins. This work represents the first thorough investigation of evolutionary diversification of α-toxins derived from a specific scorpion lineage from the perspectives of sequence, structure, function, and evolution.


Asunto(s)
Venenos de Escorpión/química , Venenos de Escorpión/genética , Escorpiones/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Cromatografía Líquida de Alta Presión , Variación Genética , Moscas Domésticas , Espectroscopía de Resonancia Magnética , Datos de Secuencia Molecular , Filogenia , Venenos de Escorpión/toxicidad , Canales de Sodio/efectos de los fármacos , Canales de Sodio/fisiología , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
13.
PLoS One ; 6(7): e22981, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21829569

RESUMEN

The production of recombinant proteins in a large scale is important for protein functional and structural studies, particularly by using Escherichia coli over-expression systems; however, approximate 70% of recombinant proteins are over-expressed as insoluble inclusion bodies. Here we presented an efficient method for generating soluble proteins from inclusion bodies by using two steps of denaturation and one step of refolding. We first demonstrated the advantages of this method over a conventional procedure with one denaturation step and one refolding step using three proteins with different folding properties. The refolded proteins were found to be active using in vitro tests and a bioassay. We then tested the general applicability of this method by analyzing 88 proteins from human and other organisms, all of which were expressed as inclusion bodies. We found that about 76% of these proteins were refolded with an average of >75% yield of soluble proteins. This "two-step-denaturing and refolding" (2DR) method is simple, highly efficient and generally applicable; it can be utilized to obtain active recombinant proteins for both basic research and industrial purposes.


Asunto(s)
Proteínas Fluorescentes Verdes/metabolismo , Metaloproteinasa 12 de la Matriz/metabolismo , Pliegue de Proteína , Replegamiento Proteico , Proteínas Recombinantes/metabolismo , Proteínas Represoras/metabolismo , Dicroismo Circular , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas Fluorescentes Verdes/aislamiento & purificación , Humanos , Cuerpos de Inclusión , Metaloproteinasa 12 de la Matriz/aislamiento & purificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Represoras/aislamiento & purificación
14.
Acta Pharmacol Sin ; 30(8): 1107-14, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19617896

RESUMEN

AIM: Previous studies have demonstrated that glycine (GLY) markedly reduces lipopolysaccharide (LPS)-induced myocardial injury.However, the mechanism of this effect is still unclear. The present study investigated the effect of GLY on cytosolic calcium concentration([Ca2+]c) and tumor necrosis factor-alpha (TNFalpha) production in cardiomyocytes exposed to LPS, as well as whether the glycine-gated chloride channel is involved in this process. METHODS: Neonatal rat cardiomyocytes were isolated, and the [Ca2+]c and TNFalpha levels were determined by using Fura-2 and a Quantikine enzyme-linked immunosorbent assay, respectively. The distribution of the GLY receptor and GLY-induced currents in cardiomyocytes were also investigated using immunocytochemistry and the whole-cell patch-clamp technique, respectively. RESULTS: LPS at concentrations ranging from 10 ng/mL to 100 microg/mL significantly stimulated TNFalpha production. GLY did not inhibit TNFalpha production induced by LPS at concentrations below 10 ng/mL but did significantly decrease TNFalpha release stimulated by 100 microg/mL LPS and prevented an LPS-induced increase in [Ca2+]c, which was reversed by strychnine, a glycine receptor antagonist. GLY did not block the isoproterenol-induced increase in [Ca2+]c, but did prevent the potassium chloride-induced increase in [Ca2+]c in cardiomyocytes.Strychnine reversed the inhibition of the KCl-stimulated elevation in [Ca2+]c by GLY. In chloride-free buffer, GLY had no effect on the dipotassium hydrogen phosphate-induced increase in [Ca2+]c. Furthermore, GLY receptor alpha1 and beta subunit-immunoreactive spots were observed in cardiomyocytes, and GLY-evoked currents were blocked by strychnine. CONCLUSION: Cardiomyocytes possess the glycine-gated chloride channel, through which GLY prevents the increase in [Ca2+]c and inhibits the TNFalpha production induced by LPS at high doses in neonatal rat cardiomyocytes.


Asunto(s)
Calcio/metabolismo , Glicina/farmacología , Miocitos Cardíacos/efectos de los fármacos , Receptores de Glicina/agonistas , Receptores de Glicina/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Cardiotónicos/farmacología , Células Cultivadas , Glicinérgicos/farmacología , Isoproterenol/farmacología , Lipopolisacáridos/efectos adversos , Miocitos Cardíacos/metabolismo , Fosfatos/metabolismo , Cloruro de Potasio/metabolismo , Compuestos de Potasio/metabolismo , Ratas , Ratas Sprague-Dawley , Estricnina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...