Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 134
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Insect Biochem Mol Biol ; 169: 104125, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38616030

RESUMEN

Voltage-dependent anion channel 2 (VDAC2) is an important channel protein that plays a crucial role in the host response to viral infection. The receptor for activated C kinase 1 (RACK1) is also a key host factor involved in viral replication. Our previous research revealed that Bombyx mori VDAC2 (BmVDAC2) and B. mori RACK1 (BmRACK1) may interact with Bombyx mori nucleopolyhedrovirus (BmNPV), though the specific molecular mechanism remains unclear. In this study, the interaction between BmVDAC2 and BmRACK1 in the mitochondria was determined by various methods. We found that BmNPV p35 interacts directly with BmVDAC2 rather than BmRACK1. BmNPV infection significantly reduced the expression of BmVDAC2, and activated the mitochondrial apoptosis pathway. Overexpression of BmVDAC2 in BmN cells inhibited BmNPV-induced cytochrome c (cyto c) release, decrease in mitochondrial membrane potential as well as apoptosis. Additionally, the inhibition of cyto c release by BmVDAC2 requires the involvement of BmRACK1 and protein kinase C. Interestingly, overexpression of p35 inhibited cyto c release during mitochondrial apoptosis in a RACK1 and VDAC2-dependent manner. Even the mutant p35, which loses Caspase inhibitory activity, could still bind to VDAC2 and inhibit cyto c release. In summary, our results indicated that BmNPV p35 interacts with the VDAC2-RACK1 complex to regulate apoptosis by inhibiting cyto c release. These findings confirm the interaction between BmVDAC2 and BmRACK1, the interaction between p35 and the VDAC2-RACK1 complex, and a novel target that BmNPV p35 regulates apoptosis in Bombyx mori via interaction with the BmVDAC2-BmRACK1 complex. The result provide an initial exploration of the function of this interaction in the BmNPV-induced mitochondrial apoptosis pathway.


Asunto(s)
Apoptosis , Bombyx , Proteínas de Insectos , Nucleopoliedrovirus , Receptores de Cinasa C Activada , Animales , Bombyx/virología , Bombyx/metabolismo , Bombyx/genética , Nucleopoliedrovirus/fisiología , Receptores de Cinasa C Activada/metabolismo , Receptores de Cinasa C Activada/genética , Proteínas de Insectos/metabolismo , Proteínas de Insectos/genética , Canal Aniónico 2 Dependiente del Voltaje/metabolismo , Canal Aniónico 2 Dependiente del Voltaje/genética , Mitocondrias/metabolismo
2.
Cell Commun Signal ; 22(1): 247, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38689280

RESUMEN

BACKGROUND: Renal fibrosis is a prevalent manifestation of chronic kidney disease (CKD), and effective treatments for this disease are currently lacking. Myofibroblasts, which originate from interstitial fibroblasts, aggregate in the renal interstitium, leading to significant accumulation of extracellular matrix and impairment of renal function. The nonreceptor tyrosine kinase c-Abl (encoded by the Abl1 gene) has been implicated in the development of renal fibrosis. However, the precise role of c-Abl in this process and its involvement in fibroblast-myofibroblast transition (FMT) remain poorly understood. METHODS: To investigate the effect of c-Abl in FMT during renal fibrosis, we investigated the expression of c-Abl in fibrotic renal tissues of patients with CKD and mouse models. We studied the phenotypic changes in fibroblast or myofibroblast-specific c-Abl conditional knockout mice. We explored the potential targets of c-Abl in NRK-49F fibroblasts. RESULTS: In this study, fibrotic mouse and cell models demonstrated that c-Abl deficiency in fibroblasts mitigated fibrosis by suppressing fibroblast activation, fibroblast-myofibroblast transition, and extracellular matrix deposition. Mechanistically, c-Abl maintains the stability of the RACK1 protein, which serves as a scaffold for proteins such as c-Abl and focal adhesion kinase at focal adhesions, driving fibroblast activation and differentiation during renal fibrosis. Moreover, specifically targeting c-Abl deletion in renal myofibroblasts could prove beneficial in established kidney fibrosis by reducing RACK1 expression and diminishing the extent of fibrosis. CONCLUSIONS: Our findings suggest that c-Abl plays a pathogenic role in interstitial fibrosis through the regulation of RACK1 protein stabilization and myofibroblast differentiation, suggesting a promising strategy for the treatment of CKD.


Asunto(s)
Fibroblastos , Fibrosis , Miofibroblastos , Proteínas Proto-Oncogénicas c-abl , Receptores de Cinasa C Activada , Transducción de Señal , Animales , Proteínas Proto-Oncogénicas c-abl/metabolismo , Proteínas Proto-Oncogénicas c-abl/genética , Miofibroblastos/metabolismo , Miofibroblastos/patología , Humanos , Ratones , Fibroblastos/metabolismo , Fibroblastos/patología , Receptores de Cinasa C Activada/genética , Receptores de Cinasa C Activada/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Quinasa 1 de Adhesión Focal/genética , Riñón/patología , Riñón/metabolismo , Masculino , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/genética , Ratones Noqueados , Ratones Endogámicos C57BL
3.
J Biol Chem ; 300(3): 105719, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38311171

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by dysregulation of the expression and processing of the amyloid precursor protein (APP). Protein quality control systems are dedicated to remove faulty and deleterious proteins to maintain cellular protein homeostasis (proteostasis). Identidying mechanisms underlying APP protein regulation is crucial for understanding AD pathogenesis. However, the factors and associated molecular mechanisms regulating APP protein quality control remain poorly defined. In this study, we show that mutant APP with its mitochondrial-targeting sequence ablated exhibited predominant endoplasmic reticulum (ER) distribution and led to aberrant ER morphology, deficits in locomotor activity, and shortened lifespan. We searched for regulators that could counteract the toxicity caused by the ectopic expression of this mutant APP. Genetic removal of the ribosome-associated quality control (RQC) factor RACK1 resulted in reduced levels of ectopically expressed mutant APP. By contrast, gain of RACK1 function increased mutant APP level. Additionally, overexpression of the ER stress regulator (IRE1) resulted in reduced levels of ectopically expressed mutant APP. Mechanistically, the RQC related ATPase VCP/p97 and the E3 ubiquitin ligase Hrd1 were required for the reduction of mutant APP level by IRE1. These factors also regulated the expression and toxicity of ectopically expressed wild type APP, supporting their relevance to APP biology. Our results reveal functions of RACK1 and IRE1 in regulating the quality control of APP homeostasis and mitigating its pathogenic effects, with implications for the understanding and treatment of AD.


Asunto(s)
Enfermedad de Alzheimer , Precursor de Proteína beta-Amiloide , Proteínas de Drosophila , Endorribonucleasas , Receptores de Cinasa C Activada , Animales , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas Serina-Treonina Quinasas , Receptores de Cinasa C Activada/genética , Receptores de Cinasa C Activada/metabolismo , Drosophila melanogaster , Modelos Animales de Enfermedad , Endorribonucleasas/genética , Endorribonucleasas/metabolismo
4.
PLoS Negl Trop Dis ; 18(1): e0011872, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38190388

RESUMEN

BACKGROUND: Gut epithelium is the first natural barrier against Trichinella spiralis larval invasion, but the mechanism by which larval penetration of gut epithelium is not completely elucidated. Previous studies showed that proteases secreted by T. spiralis intestinal infective larvae (IIL) degraded tight junctions (TJs) proteins of gut epithelium and mediated larval invasion. A new T. spiralis serine proteinase (TsSPc) was identified in the IIL surface proteins and ES proteins, rTsSPc bound to the intestinal epithelial cell (IECs) and promoted larval invasion of IECs. The aim of this study was to characterize the interacted proteins of TsSPc and IECs, and to investigate the molecular mechanisms of TsSPc mediating larval invasion of gut mucosa. METHODOLOGY/PRINCIPAL FINDING: IIFT results showed natural TsSPc was detected in infected murine intestine at 6, 12 hours post infection (hpi) and 3 dpi. The results of GST pull-down, mass spectrometry (MS) and Co-IP indicated that rTsSPc bound and interacted specifically with receptor for activated protein C kinase 1 (RACK1) in Caco-2 cells. rTsSPc did not directly hydrolyze the TJs proteins. qPCR and Western blot showed that rTsSPc up-regulated RACK1 expression, activated MAPK/ERK1/2 pathway, reduced the expression levels of gut TJs (occludin and claudin-1) and adherent protein E-cad, increased the paracellular permeability and damaged the integrity of intestinal epithelial barrier. Moreover, the RACK1 inhibitor HO and ERK1/2 pathway inhibitor PD98059 abolished the rTsSPc activating ERK1/2 pathway, they also inhibited and abrogated the rTsSPc down-regulating expression of occludin, claudin-1 and E-cad in Caco-2 monolayer and infected murine intestine, impeded larval invasion and improved intestinal epithelial integrity and barrier function, reduced intestinal worm burdens and alleviated intestinal inflammation. CONCLUSIONS: rTsSPc bound to RACK1 receptor in gut epithelium, activated MAPK/ERK1/2 pathway, decreased the expression of gut epithelial TJs proteins and disrupted the epithelial integrity, consequently mediated T. spiralis larval invasion of gut epithelium. The results are valuable to understand T. spiralis invasion mechanism, and TsSPc might be regarded as a vaccine target against T. spiralis invasion and infection.


Asunto(s)
Trichinella spiralis , Triquinelosis , Humanos , Animales , Ratones , Larva/fisiología , Serina Proteasas/genética , Células CACO-2 , Claudina-1/metabolismo , Sistema de Señalización de MAP Quinasas , Ocludina/metabolismo , Proteínas del Helminto/metabolismo , Células Epiteliales/metabolismo , Ratones Endogámicos BALB C , Mucosa Intestinal/metabolismo , Receptores de Cinasa C Activada/metabolismo , Proteínas de Neoplasias/genética
5.
Cell Oncol (Dordr) ; 47(1): 113-128, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37578594

RESUMEN

BACKGROUND: Metabolic reprogramming is closely related to the development of gastric cancer (GC), which remains as the fourth leading cause of cancer-related death worldwide. As a tumor suppressor for GC, whether receptor for activated C-kinase 1 (RACK1) play a modulatory role in metabolic reprogramming remains largely unclear. METHODS: GC cell lines and cell-derived xenograft mouse model were used to identify the biological function of RACK1. Flow cytometry and Seahorse assays were applied to examine cell cycle and oxygen consumption rate (OCR), respectively. Western blot, real-time PCR and autophagy double fluorescent assays were utilized to explore the signaling. Immunohistochemistry was performed to detect the expression of RACK1 and other indicators in tissue sections. RESULTS: Loss of RACK1 facilitated the viability, colony formation, cell cycle progression and OCR of GC cells in a glutamine-dependent manner. Further investigation revealed that RACK1 knockdown inhibited the lysosomal degradation of Alanine-serine-cysteine amino acid transporter 2 (ASCT2). Mechanistically, depletion of RACK1 remarkably decreased PTEN expression through up-regulating miR-146b-5p, leading to the activation of AKT/mTOR signaling pathway which dampened autophagy flux subsequently. Moreover, knockdown of ASCT2 could reverse the promotive effect of RACK1 depletion on GC tumor growth both in vitro and in vivo. Tissue microarray confirmed that RACK1 was negatively correlated with the expression of ASCT2 and p62, as well as the phosphorylation of mTOR. CONCLUSION: Together, our results demonstrate that the suppressive function of RACK1 in GC is associated with ASCT2-mediated glutamine metabolism, and imply that targeting RACK1/ASCT2 axis provides potential strategies for GC treatment.


Asunto(s)
Neoplasias Gástricas , Humanos , Animales , Ratones , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Glutamina/metabolismo , Línea Celular Tumoral , Serina-Treonina Quinasas TOR/metabolismo , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Receptores de Cinasa C Activada/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo
6.
Acta Trop ; 251: 107112, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38157925

RESUMEN

Toxoplasma gondii (T. gondii) surface antigen 1 (SAG1) is crucial for tachyzoite invasion into host cells. However, the role of SAG1 in interaction with host cells remains unknown. The primary objective of this study was to analyze and validate the interaction between SAG1 and host cells. RACK1, an intracellular multifunctional protein, was identified as a SAG1 binding partner in host cells. Furthermore, the expression of RACK1 is manipulated by SAG1, and depletion of RACK1 negatively regulated host cell viability. These results imply that through interaction with RACK1, SAG1 preserves the viability of host cells to satisfy the survival needs of T. gondii. Our findings suggest a novel role for SAG1 in intracellular parasitism.


Asunto(s)
Proteínas Protozoarias , Toxoplasma , Antígenos de Protozoos/genética , Receptores de Cinasa C Activada/genética , Receptores de Cinasa C Activada/metabolismo , Antígenos de Superficie/metabolismo , Anticuerpos Antiprotozoarios
7.
Chem Biol Interact ; 388: 110830, 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38103880

RESUMEN

Microglial polarization modulation has been considered the potential therapeutic strategy for relieving cognitive impairment in sepsis survivors. Rosmarinic acid (RA), a water-soluble polyphenolic natural compound, processes a strong protective effect on various types of neurological disorders including Parkinson's disease, depression, and anxiety. However, its role and potential molecular mechanisms in sepsis-associated cognitive impairment remain unclear. To investigate the preventive and therapeutic effect of RA on sepsis-associated cognitive impairment and elucidate the potential mechanism of RA on regulating microglial polarization, we established a CLP-induced cognitive impairment model in mice and a lipopolysaccharide-induced microglia polarization cell model in BV-2. RACK1 siRNA was designed to identify the potential molecular mechanism of RACK1 on microglial polarization. The preventive and therapeutic effect of RA on cognitive impairment followed by PET-CT and behavioral tests including open-field test and tail suspension test. RACK1/HIF-1α pathway and microglial morphology in the hippocampus or BV-2 cells were measured. The results showed that RA significantly ameliorated the CLP-induced depressive and anxiety-like behaviors and promoted whole-brain glucose uptake in mice. Moreover, RA markedly improved CLP-induced hippocampal neuron loss and microglial activation by inhibiting microglial M1 polarization. Furthermore, experiments showed RACK1 was involved in the regulation of LPS-induced microglial M1 polarization via HIF-1α, and RA suppressed lipopolysaccharide or sepsis-associated microglial M1 polarization via RACK1/HIF-1α pathway (rescued the decrease of RACK1 and increase of HIF-1α). Taken together, RA could be a potential preventive and therapeutic medication in improving cognitive impairment through RACK1/HIF-1α pathway-regulated microglial polarization.


Asunto(s)
Disfunción Cognitiva , Ácido Rosmarínico , Sepsis , Animales , Ratones , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/metabolismo , Lipopolisacáridos/toxicidad , Lipopolisacáridos/metabolismo , Microglía , Tomografía Computarizada por Tomografía de Emisión de Positrones , Receptores de Cinasa C Activada/efectos de los fármacos , Receptores de Cinasa C Activada/metabolismo , Ácido Rosmarínico/farmacología , Ácido Rosmarínico/uso terapéutico , Sepsis/complicaciones , Sepsis/tratamiento farmacológico , Transducción de Señal , Subunidad alfa del Factor 1 Inducible por Hipoxia/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo
8.
Shanghai Kou Qiang Yi Xue ; 32(4): 385-390, 2023 Aug.
Artículo en Chino | MEDLINE | ID: mdl-38044732

RESUMEN

PURPOSE: To investigate the expression of tissue-active protein kinase C receptor 1 (RACK1) and epithelin glycoprotein 40 (EGP40) in oral squamous cell carcinoma (OSCC) and their relationship with clinicopathological features and prognosis. METHODS: A total of 103 patients with OSCC who were admitted to Shangrao People's Hospital from January 2016 to February 2019 were prospectively selected as the research subjects. All patients underwent radical resection of OSCC and were followed up for 3 years. Immunohistochemistry was used to detect the positive expression levels of RACK1 and EGP40 in cancer tissues and adjacent tissues. The positive expression of RACK1 and EGP40 in cancer tissues and adjacent tissues were compared. The relationship between the positive expression level of RACK1 and EGP40 in cancer tissues of OSCC patients and clinicopathological parameters was analyzed. Factors affecting postoperative recurrence and metastasis in OSCC patients were analyzed. The relationship between the expression of RACK1 and EGP40 in cancer tissues and postoperative disease-free survival of OSCC patients was analyzed. SPSS 18.0 software package was used for statistical analysis of the data. RESULTS: The positive expression rate of RACK1 and EGP40 in cancer tissues was significantly higher than those in adjacent tissues (P<0.05). The positive expression rate of RACK1 and EGP40 in cancer tissues of OSCC patients with poorly differentiated, stage III, cervical lymph node metastasis, and infiltrating vessels was significantly higher than that in patients with moderate and high differentiation, stage II, no cervical lymph node metastasis, and no infiltrating vessels(P<0.05). The positive expression rate of RACK1 in cancer tissue of OSCC patients in T3 stage was significantly higher than that in T2 stage(P<0.05). Cox multivariate regression analysis showed pathological grade (RR=6.290, 95%CI: 2.588-15.287), cervical lymph node metastasis(RR=5.995, 95%CI: 2.467-14.571), RACK1 positive rate (RR=4.495, 95%CI: 1.850-10.925) and EGP40 positive rate (RR=4.559, 95%CI: 1.876-11.079) were factors affecting the recurrence and metastasis of OSCC patients after surgery(P<0.05). The disease-free survival curve of patients with negative expression of RACK1 was significantly better than that of patients with positive expression (P<0.05). The disease-free survival curve of patients with negative expression of EGP40 was significantly better than that of patients with positive expression (P<0.05). CONCLUSIONS: The expression of RACK1 and EGP40 in cancer tissues of OSCC patients is related to clinicopathological parameters and prognosis. Patients with positive expression of RACK1 and EGP40 have a high risk of recurrence and metastasis after surgery.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Humanos , Carcinoma de Células Escamosas/cirugía , Carcinoma de Células Escamosas/patología , Granulinas , Metástasis Linfática , Neoplasias de la Boca/cirugía , Neoplasias de la Boca/patología , Proteínas de Neoplasias/metabolismo , Pronóstico , Receptores de Cinasa C Activada/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello
9.
Acta Neuropathol Commun ; 11(1): 200, 2023 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-38111057

RESUMEN

TAR DNA-binding protein 43 (TDP-43) and Fused in Sarcoma/Translocated in Sarcoma (FUS) are ribonucleoproteins associated with pathogenesis of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). Under physiological conditions, TDP-43 and FUS are predominantly localized in the nucleus, where they participate in transcriptional regulation, RNA splicing and metabolism. In disease, however, they are typically mislocalized to the cytoplasm where they form aggregated inclusions. A number of shared cellular pathways have been identified that contribute to TDP-43 and FUS toxicity in neurodegeneration. In the present study, we report a novel pathogenic mechanism shared by these two proteins. We found that pathological FUS co-aggregates with a ribosomal protein, the Receptor for Activated C-Kinase 1 (RACK1), in the cytoplasm of spinal cord motor neurons of ALS, as previously reported for pathological TDP-43. In HEK293T cells transiently transfected with TDP-43 or FUS mutant lacking a functional nuclear localization signal (NLS; TDP-43ΔNLS and FUSΔNLS), cytoplasmic TDP-43 and FUS induced co-aggregation with endogenous RACK1. These co-aggregates sequestered the translational machinery through interaction with the polyribosome, accompanied by a significant reduction of global protein translation. RACK1 knockdown decreased cytoplasmic aggregation of TDP-43ΔNLS or FUSΔNLS and alleviated associated global translational suppression. Surprisingly, RACK1 knockdown also led to partial nuclear localization of TDP-43ΔNLS and FUSΔNLS in some transfected cells, despite the absence of NLS. In vivo, RACK1 knockdown alleviated retinal neuronal degeneration in transgenic Drosophila melanogaster expressing hTDP-43WT or hTDP-43Q331K and improved motor function of hTDP-43WT flies, with no observed adverse effects on neuronal health in control knockdown flies. In conclusion, our results revealed a novel shared mechanism of pathogenesis for misfolded aggregates of TDP-43 and FUS mediated by interference with protein translation in a RACK1-dependent manner. We provide proof-of-concept evidence for targeting RACK1 as a potential therapeutic approach for TDP-43 or FUS proteinopathy associated with ALS and FTLD.


Asunto(s)
Esclerosis Amiotrófica Lateral , Demencia Frontotemporal , Degeneración Lobar Frontotemporal , Sarcoma , Animales , Humanos , Esclerosis Amiotrófica Lateral/patología , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Células HEK293 , Neuronas Motoras/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Demencia Frontotemporal/patología , Degeneración Lobar Frontotemporal/patología , Biosíntesis de Proteínas , Sarcoma/metabolismo , Sarcoma/patología , Proteína FUS de Unión a ARN/genética , Proteína FUS de Unión a ARN/metabolismo , Receptores de Cinasa C Activada/genética , Receptores de Cinasa C Activada/metabolismo , Proteínas de Neoplasias/genética
10.
Cell Death Dis ; 14(10): 685, 2023 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-37848434

RESUMEN

The receptor for activated C kinase 1 (RACK1) is a key scaffolding protein with multifunctional and multifaceted properties. By mediating protein-protein interactions, RACK1 integrates multiple intracellular signals involved in the regulation of various physiological and pathological processes. Dysregulation of RACK1 has been implicated in the initiation and progression of many tumors. However, the exact function of RACK1 in cancer cellular processes, especially in proliferation, remains controversial. Here, we show that RACK1 is required for breast cancer cell proliferation in vitro and tumor growth in vivo. This effect of RACK1 is associated with its ability to enhance ß-catenin stability and activate the canonical WNT signaling pathway in breast cancer cells. We identified PSMD2, a key component of the proteasome, as a novel binding partner for RACK1 and ß-catenin. Interestingly, although there is no interaction between RACK1 and ß-catenin, RACK1 binds PSMD2 competitively with ß-catenin. Moreover, RACK1 prevents ubiquitinated ß-catenin from binding to PSMD2, thereby protecting ß-catenin from proteasomal degradation. Collectively, our findings uncover a novel mechanism by which RACK1 increases ß-catenin stability and promotes breast cancer proliferation.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/patología , beta Catenina/metabolismo , Vía de Señalización Wnt/fisiología , Proliferación Celular , Línea Celular Tumoral , Factor 2 Asociado a Receptor de TNF/metabolismo , Receptores de Cinasa C Activada/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo
11.
Am J Physiol Renal Physiol ; 325(6): F870-F884, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37823194

RESUMEN

Neuropilin 1 (NRP1) is a single-channel transmembrane glycoprotein whose role and mechanism in renal fibrosis remain incompletely elucidated. Therefore, we investigated the effect of NRP1 on renal fibrosis and its potential mechanism. NRP1 expression in the renal sections from patients with chronic kidney disease (CKD) and a unilateral ureteral obstruction (UUO) mouse model was detected. Nrp1 overexpression or knockdown plasmid was transfected into mice, TKPTS mouse kidney proximal tubular epithelial cells (TECs), and rat kidney fibroblasts, after which pathological injury evaluation and fibrosis marker detection were conducted. The direct interaction of the receptor of activated protein C kinase 1 (RACK1) with NRP1 was validated by immunoprecipitation and Western blot analysis. We found that the upregulated renal NRP1 expression in patients with CKD was located in proximal TECs, consistent with the degree of interstitial fibrosis. In the UUO mouse model, NRP1 expression was upregulated in the kidney, and overexpression of Nrp1 increased the mRNA and protein expression of fibronectin (Fn) and α-smooth muscle actin (α-SMA), whereas Nrp1 knockdown significantly reduced Fn and α-SMA expression and downregulated the inflammatory response. NRP1 promoted transforming growth factor ß1 (TGF-ß1)-induced profibrotic responses in the TKPTS cells and fibroblasts, and Nrp1 knockdown partially reversed these responses. Immunoprecipitation combined with liquid chromatography-tandem mass spectrometry verified that NRP1 can directly bind to RACK1, and Rack1 knockdown reversed the NRP1-induced fibrotic response. In summary, NRP1 may enhance the TGF-ß1 pathway by binding to RACK1, thus promoting renal fibrosis.NEW & NOTEWORTHY Although a few studies have confirmed the correlation between neuropilin 1 (NRP1) and renal diseases, the mechanism of NRP1 in renal fibrosis remains unclear. Here, we investigated the effects of NRP1 on renal fibrosis through in vitro and in vivo experiments and explored the possible downstream mechanisms. We found that NRP1 can stimulate the TGF-ß1 signaling pathway, possibly by binding to RACK1, thereby promoting renal fibrosis.


Asunto(s)
Enfermedades Renales , Neuropilina-1 , Receptores de Cinasa C Activada , Insuficiencia Renal Crónica , Obstrucción Ureteral , Animales , Humanos , Ratones , Ratas , Células Epiteliales/metabolismo , Fibrosis , Riñón/metabolismo , Enfermedades Renales/patología , Proteínas de Neoplasias/metabolismo , Neuropilina-1/genética , Neuropilina-1/metabolismo , Receptores de Cinasa C Activada/genética , Receptores de Cinasa C Activada/metabolismo , Insuficiencia Renal Crónica/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Obstrucción Ureteral/patología
12.
Cell Death Differ ; 30(11): 2382-2392, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37828084

RESUMEN

Receptor for activated C kinase 1 (RACK1) has been confirmed to take part in multiple biological events and the mechanism supporting abnormal RACK1 expression in ovarian cancer (OC) remains to be characterized. Here, we identified Smad ubiquitin regulatory factor 2 (SMURF2) as a bona fide E3 ligase of RACK1 in OC. SMURF2 effectively added the K6, K33 and K48 ubiquitin chains to the RACK1, resulting in polyubiquitination and instability of RACK1. PCAF promoted acetylation of RACK1 at K130, leading to SMURF2-mediated RACK1 ubiquitination inhibited and promote OC progression. The expression levels of SMURF2 and RACK1 were negatively correlated. SMURF2 was abnormal low expression in human ovarian cancer, resulting in decreased ubiquitination of RACK1 and increased stability, which promoted OC progression, and strongly associated with poor patients' prognosis. In general, our results demonstrated that SMURF2 plays a pivotal role in stabilizing RACK1, which in turn facilitates tumorigenesis in OC, suggesting that SMURF2-RACK1 axis may prove to be potential targets for the treatment of OC.


Asunto(s)
Neoplasias Ováricas , Ubiquitina , Femenino , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Procesamiento Proteico-Postraduccional , Receptores de Cinasa C Activada/genética , Receptores de Cinasa C Activada/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
13.
Neuron ; 111(24): 3988-4005.e11, 2023 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-37820724

RESUMEN

Fragile X messenger ribonucleoprotein 1 protein (FMRP) deficiency leads to fragile X syndrome (FXS), an autism spectrum disorder. The role of FMRP in prenatal human brain development remains unclear. Here, we show that FMRP is important for human and macaque prenatal brain development. Both FMRP-deficient neurons in human fetal cortical slices and FXS patient stem cell-derived neurons exhibit mitochondrial dysfunctions and hyperexcitability. Using multiomics analyses, we have identified both FMRP-bound mRNAs and FMRP-interacting proteins in human neurons and unveiled a previously unknown role of FMRP in regulating essential genes during human prenatal development. We demonstrate that FMRP interaction with CNOT1 maintains the levels of receptor for activated C kinase 1 (RACK1), a species-specific FMRP target. Genetic reduction of RACK1 leads to both mitochondrial dysfunctions and hyperexcitability, resembling FXS neurons. Finally, enhancing mitochondrial functions rescues deficits of FMRP-deficient cortical neurons during prenatal development, demonstrating targeting mitochondrial dysfunction as a potential treatment.


Asunto(s)
Trastorno del Espectro Autista , Síndrome del Cromosoma X Frágil , Enfermedades Mitocondriales , Humanos , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Trastorno del Espectro Autista/metabolismo , Neuronas/metabolismo , Neurogénesis , Enfermedades Mitocondriales/metabolismo , Receptores de Cinasa C Activada/genética , Receptores de Cinasa C Activada/metabolismo , Proteínas de Neoplasias/metabolismo , Factores de Transcripción/metabolismo
14.
J Virol ; 97(10): e0074723, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37712706

RESUMEN

IMPORTANCE: Respiratory syncytial virus (RSV) matrix (M) protein is indispensable for virion assembly and release. It is localized to the nucleus during early infection to perturb host transcription. However, the function of RSV M protein in other cellular activities remains poorly understood. In this study, several interferon response-associated host factors, including RACK1, were identified by proteomic analysis as RSV M interactors. Knockdown of RACK1 attenuates RSV-restricted IFN signaling leading to enhanced host defense against RSV infection, unraveling a role of M protein in antagonizing IFN response via association with RACK1. Our study uncovers a previously unrecognized mechanism of immune evasion by RSV M protein and identifies RACK1 as a novel host factor recruited by RSV, highlighting RACK1 as a potential new target for RSV therapeutics development.


Asunto(s)
Receptores de Cinasa C Activada , Infecciones por Virus Sincitial Respiratorio , Virus Sincitial Respiratorio Humano , Proteínas de la Matriz Viral , Humanos , Interferones , Proteínas de Neoplasias/genética , Proteínas , Proteómica , Receptores de Cinasa C Activada/metabolismo , Infecciones por Virus Sincitial Respiratorio/metabolismo , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/genética , Proteínas de la Matriz Viral/metabolismo
15.
Cancer Sci ; 114(10): 3900-3913, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37519194

RESUMEN

Colorectal cancer (CRC) metastasis plays a crucial role in disease progression, yet the regulatory mechanisms underlying metastasis remain incompletely understood. Isobutyric acid (IBA), a short-chain fatty acid found at high levels in serum of CRC patients, has been shown to be a critical metabolite influencing CRC proliferation. However, its role in tumor metastasis remains unknown. Here, utilizing liquid chromatography tandem mass spectrometry (LC-MS/MS) analysis, we found that levels of IBA were significantly higher in patients with distant organ metastasis of CRC than in those without. Furthermore, IBA promoted CRC metastasis both in vitro and in vivo. Mass spectrometry, immunofluorescence, and cellular thermal shift assay revealed that IBA interacts with RACK1. Mechanistically, IBA binding to and activating RACK1 promotes regulation of downstream Akt and FAK signaling and CRC metastasis. Collectively, our study highlights the critical interplay between IBA and RACK1 and its impact on tumor metastasis. This study suggests that targeting the IBA-RACK1 signaling axis may be an effective therapeutic strategy for controlling CRC metastasis.


Asunto(s)
Neoplasias Colorrectales , Espectrometría de Masas en Tándem , Humanos , Línea Celular Tumoral , Cromatografía Liquida , Neoplasias Colorrectales/patología , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Metástasis de la Neoplasia , Movimiento Celular , Receptores de Cinasa C Activada/metabolismo , Proteínas de Neoplasias/metabolismo
16.
New Phytol ; 239(5): 1804-1818, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37301989

RESUMEN

Brassinosteroids (BRs) are a group of plant-specific steroid hormones, which induces the rapid nuclear localization of the positive transcriptional factors BRASSINAZOLE RESISTANT1/2 (BZR1/2). However, the mechanisms underlying the regulation of nucleocytoplasmic shuttling of BZR1 remain to be fully elucidated. In this study, we show that the scaffold protein Receptor for Activated C Kinase 1 (RACK1) from Arabidopsis is involved in BR signaling cascades through mediating the nuclear localization of BZR1, which is tightly retained in the cytosol by the conserved scaffold protein 14-3-3s. RACK1 can interact with BZR1 and competitively decrease the 14-3-3 interaction with BZR1 in cytosol, which efficiently enhances the nuclear localization of BZR1. 14-3-3 also retains RACK1 in cytosol through their interaction. Conversely, BR treatment enhances the nuclear localization of BZR1 by disrupting the 14-3-3 interaction with RACK1 and BZR1. Our study uncovers a new mechanism that integrates two kinds of conserved scaffold proteins (RACK1 and 14-3-3) coordinating BR signaling event.


Asunto(s)
Proteínas de Arabidopsis , Arabidopsis , Fitosteroles , Proteínas de Arabidopsis/metabolismo , Proteínas de Unión al ADN/metabolismo , Arabidopsis/metabolismo , Núcleo Celular/metabolismo , Transducción de Señal , Reguladores del Crecimiento de las Plantas/metabolismo , Brasinoesteroides/metabolismo , Fitosteroles/metabolismo , Regulación de la Expresión Génica de las Plantas , Receptores de Cinasa C Activada/metabolismo
17.
Cell Rep ; 42(5): 112456, 2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37126448

RESUMEN

The regulation of translation in astrocytes, the main glial cells in the brain, remains poorly characterized. We developed a high-throughput proteomics screen for polysome-associated proteins in astrocytes and focused on ribosomal protein receptor of activated protein C kinase 1 (RACK1), a critical factor in translational regulation. In astrocyte somata and perisynaptic astrocytic processes (PAPs), RACK1 preferentially binds to a number of mRNAs, including Kcnj10, encoding the inward-rectifying potassium (K+) channel Kir4.1. By developing an astrocyte-specific, conditional RACK1 knockout mouse model, we show that RACK1 represses production of Kir4.1 in hippocampal astrocytes and PAPs. Upregulation of Kir4.1 in the absence of RACK1 increases astrocytic Kir4.1-mediated K+ currents and volume. It also modifies neuronal activity attenuating burst frequency and duration. Reporter-based assays reveal that RACK1 controls Kcnj10 translation through the transcript's 5' untranslated region. Hence, translational regulation by RACK1 in astrocytes represses Kir4.1 expression and influences neuronal activity.


Asunto(s)
Astrocitos , Neuroglía , Animales , Ratones , Astrocitos/metabolismo , Ratones Noqueados , Neuroglía/metabolismo , Neuronas , Receptores de Cinasa C Activada/metabolismo , Ribosomas
18.
Acta Pharmacol Sin ; 44(10): 1948-1961, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37225849

RESUMEN

Parkinson's disease (PD) is a common neurodegenerative motor disorder characterized by a dramatic reduction in pars compacta of substantia nigra dopaminergic neurons and striatal dopamine (DA) levels. Mutations or deletions in the PARK7/DJ-1 gene are associated with an early-onset familial form of PD. DJ-1 protein prevents neurodegeneration via its regulation of oxidative stress and mitochondrial function as well as its roles in transcription and signal transduction. In this study, we investigated how loss of DJ-1 function affected DA degradation, ROS generation and mitochondrial dysfunction in neuronal cells. We showed that loss of DJ-1 significantly increased the expression of monoamine oxidase (MAO)-B but not MAO-A in both neuronal cells and primary astrocytes. In DJ-1-knockout (KO) mice, MAO-B protein levels in the substantia nigra (SN) and striatal regions were significantly increased. We demonstrated that the induction of MAO-B expression by DJ-1 deficiency depended on early growth response 1 (EGR1) in N2a cells. By coimmunoprecipitation omics analysis, we found that DJ-1 interacted with receptor of activated protein C kinase 1 (RACK1), a scaffolding protein, and thus inhibited the activity of the PKC/JNK/AP-1/EGR1 cascade. The PKC inhibitor sotrastaurin or the JNK inhibitor SP600125 completely inhibited DJ-1 deficiency-induced EGR1 and MAO-B expression in N2a cells. Moreover, the MAO-B inhibitor rasagiline inhibited mitochondrial ROS generation and rescued neuronal cell death caused by DJ-1 deficiency, especially in response to MPTP stimulation in vitro and in vivo. These results suggest that DJ-1 exerts neuroprotective effects by inhibiting the expression of MAO-B distributed at the mitochondrial outer membrane, which mediates DA degradation, ROS generation and mitochondrial dysfunction. This study reveals a mechanistic link between DJ-1 and MAO-B expression and contributes to understanding the crosslinks among pathogenic factors, mitochondrial dysfunction and oxidative stress in PD pathogenesis.


Asunto(s)
Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Ratones , Animales , Enfermedad de Parkinson/metabolismo , Monoaminooxidasa/genética , Monoaminooxidasa/metabolismo , Monoaminooxidasa/farmacología , Regulación hacia Arriba , Especies Reactivas de Oxígeno/metabolismo , Neuronas Dopaminérgicas/metabolismo , Transducción de Señal , Enfermedades Neurodegenerativas/metabolismo , Receptores de Cinasa C Activada/genética , Receptores de Cinasa C Activada/metabolismo , Receptores de Cinasa C Activada/farmacología , Proteína Desglicasa DJ-1/genética , Proteína Desglicasa DJ-1/metabolismo
19.
Cell Death Dis ; 14(5): 333, 2023 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-37210387

RESUMEN

Unbalanced protein homeostasis (proteostasis) networks are frequently linked to tumorigenesis, making cancer cells more susceptible to treatments that target proteostasis regulators. Proteasome inhibition is the first licensed proteostasis-targeting therapeutic strategy, and has been proven effective in hematological malignancy patients. However, drug resistance almost inevitably develops, pressing for a better understanding of the mechanisms that preserve proteostasis in tumor cells. Here we report that CD317, a tumor-targeting antigen with a unique topology, was upregulated in hematological malignancies and preserved proteostasis and cell viability in response to proteasome inhibitors (PIs). Knocking down CD317 lowered Ca2+ levels in the endoplasmic reticulum (ER), promoting PIs-induced proteostasis failure and cell death. Mechanistically, CD317 interacted with calnexin (CNX), an ER chaperone protein that limits calcium refilling via the Ca2+ pump SERCA, thereby subjecting CNX to RACK1-mediated autophagic degradation. As a result, CD317 decreased the level of CNX protein, coordinating Ca2+ uptake and thus favoring protein folding and quality control in the ER lumen. Our findings reveal a previously unrecognized role of CD317 in proteostasis control and imply that CD317 could be a promising target for resolving PIs resistance in the clinic.


Asunto(s)
Antígeno 2 del Estroma de la Médula Ósea , Inhibidores de Proteasoma , Proteostasis , Humanos , Calnexina/metabolismo , Supervivencia Celular , Chaperonas Moleculares/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Inhibidores de Proteasoma/farmacología , Receptores de Cinasa C Activada/genética , Receptores de Cinasa C Activada/metabolismo , Antígeno 2 del Estroma de la Médula Ósea/genética , Antígeno 2 del Estroma de la Médula Ósea/metabolismo
20.
J Integr Plant Biol ; 65(7): 1703-1716, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36939002

RESUMEN

Plants have adopted versatile scaffold proteins to facilitate the crosstalk between multiple signaling pathways. Leaf senescence is a well-programmed developmental stage that is coordinated by various external and internal signals. However, the functions of plant scaffold proteins in response to senescence signals are not well understood. Here, we report that the scaffold protein RACK1A (RECEPTOR FOR ACTIVATED C KINASE 1A) participates in leaf senescence mediated by ethylene signaling via the coordination of the EIN3-miR164-ORE1 transcriptional regulatory cascade. RACK1A is a novel positive regulator of ethylene-mediated leaf senescence. The rack1a mutant exhibits delayed leaf senescence, while transgenic lines overexpressing RACK1A display early leaf senescence. Moreover, RACK1A promotes EIN3 (ETHYLENE INSENSITIVE 3) protein accumulation, and directly interacts with EIN3 to enhance its DNA-binding activity. Together, they then associate with the miR164 promoter to inhibit its transcription, leading to the release of the inhibition on downstream ORE1 (ORESARA 1) transcription and the promotion of leaf senescence. This study reveals a mechanistic framework by which RACK1A promotes leaf senescence via the EIN3-miR164-ORE1 transcriptional cascade, and provides a paradigm for how scaffold proteins finely tune phytohormone signaling to control plant development.


Asunto(s)
Proteínas de Arabidopsis , Arabidopsis , Receptores de Cinasa C Activada , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Etilenos/metabolismo , Regulación de la Expresión Génica de las Plantas , Hojas de la Planta/metabolismo , Senescencia de la Planta , Receptores de Cinasa C Activada/genética , Receptores de Cinasa C Activada/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA