Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 764
Filtrar
1.
CNS Neurosci Ther ; 30(3): e14691, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38532543

RESUMO

AIMS: Neuronal cell death is a primary factor that determines the outcome after traumatic brain injury (TBI). We previously revealed the importance of receptor for activated C kinase (RACK1), a multifunctional scaffold protein, in maintaining neuronal survival after TBI, but the specific mechanism remains unclear. The aim of this study was to explore the mechanism underlying RACK1-mediated neuroprotection in TBI. METHODS: TBI model was established using controlled cortical impact injury in Sprague-Dawley rats. Genetic intervention and pharmacological inhibition of RACK1 and PERK-autophagy signaling were administrated by intracerebroventricular injection. Western blotting, coimmunoprecipitation, transmission electron microscopy, real-time PCR, immunofluorescence, TUNEL staining, Nissl staining, neurobehavioral tests, and contusion volume assessment were performed. RESULTS: Endogenous RACK1 was upregulated and correlated with autophagy induction after TBI. RACK1 knockdown markedly inhibited TBI-induced autophagy, whereas RACK1 overexpression exerted the opposite effects. Moreover, RACK1 overexpression ameliorated neuronal apoptosis, neurological deficits, and cortical tissue loss after TBI, and these effects were abrogated by the autophagy inhibitor 3-methyladenine or siRNAs targeting Beclin1 and Atg5. Mechanistically, RACK1 interacted with PERK and activated PERK signaling. Pharmacological and genetic inhibition of the PERK pathway abolished RACK1-induced autophagy after TBI. CONCLUSION: Our findings indicate that RACK1 protected against TBI-induced neuronal damage partly through autophagy induction by regulating the PERK signaling pathway.


Assuntos
Lesões Encefálicas Traumáticas , Transdução de Sinais , Ratos , Animais , Ratos Sprague-Dawley , Lesões Encefálicas Traumáticas/metabolismo , Neuroproteção , Apoptose , Autofagia , Receptores de Quinase C Ativada
2.
J Biol Chem ; 300(3): 105719, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38311171

RESUMO

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.


Assuntos
Doença de Alzheimer , Precursor de Proteína beta-Amiloide , Proteínas de Drosophila , Endorribonucleases , Receptores de Quinase C Ativada , Animais , Doença de Alzheimer/metabolismo , Peptídeos 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 Quinases , Receptores de Quinase C Ativada/genética , Receptores de Quinase C Ativada/metabolismo , Drosophila melanogaster , Modelos Animais de Doenças , Endorribonucleases/genética , Endorribonucleases/metabolismo
3.
J Clin Lab Anal ; 38(4): e25012, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38305509

RESUMO

BACKGROUND: RACK1 has been identified as a multifunctional cytosolic protein, and plays a pivotal role in multiple biological responses involved in several kinds of tumors, while its effect in cervical cancer has not been well elucidated yet. The study aimed to investigate the role of RACK1 in cervical cancer occurrence and progression. METHODS: The expression of RACK1 in cervical specimens was measured by immunohistochemical staining and Western blot assay. Transgenic mice were used to detect the role of RACK1 in modulating tumorigenesis in vivo. Cervical carcinoma cell lines were used to explore the underlying mechanisms of RACK1 on the behaviors of tumor cells in vitro. RESULTS: We found that RACK1 expression was upregulated in cancer tissues compared with adjacent tissues, and its expression was gradually increased from cervictis, and cervical intraepithelial neoplasis (CIN) to carcinoma. Genetic overexpression of RACK1 facilitated tumor formation and growth in nude mice. Mechanism studies disclosed that RACK1 over-expression prolonged the G0 /G1 phase by up-regulating the expression of cyclinD1, down-regulating p21 and p27 probably by modulating the phosphorylation of AKT. CONCLUSIONS: Taken together, we concluded that RACK1 stimulates tumorigenesis and progression of cervical cancer via modulating the proliferation of tumor cells, implying that targeting RACK1 may serve as a promising method for cervical cancer therapy.


Assuntos
Neoplasias do Colo do Útero , Humanos , Camundongos , Feminino , Animais , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/patologia , Camundongos Nus , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Carcinogênese , Linhagem Celular Tumoral , Proliferação de Células/genética , Receptores de Quinase C Ativada/genética , Receptores de Quinase C Ativada/farmacologia
4.
PLoS Negl Trop Dis ; 18(1): e0011872, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38190388

RESUMO

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.


Assuntos
Trichinella spiralis , Triquinelose , Humanos , Animais , Camundongos , Larva/fisiologia , Serina Proteases/genética , Células CACO-2 , Claudina-1/metabolismo , Sistema de Sinalização das MAP Quinases , Ocludina/metabolismo , Proteínas de Helminto/metabolismo , Células Epiteliais/metabolismo , Camundongos Endogâmicos BALB C , Mucosa Intestinal/metabolismo , Receptores de Quinase C Ativada/metabolismo , Proteínas de Neoplasias/genética
5.
Cell Oncol (Dordr) ; 47(1): 113-128, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37578594

RESUMO

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.


Assuntos
Neoplasias Gástricas , Humanos , Animais , Camundongos , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Glutamina/metabolismo , Linhagem Celular Tumoral , Serina-Treonina Quinases TOR/metabolismo , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Receptores de Quinase C Ativada/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo
6.
Chem Biol Interact ; 388: 110830, 2024 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-38103880

RESUMO

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.


Assuntos
Disfunção Cognitiva , 60556 , Sepse , Animais , Camundongos , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/metabolismo , Lipopolissacarídeos/toxicidade , Lipopolissacarídeos/metabolismo , Microglia , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Receptores de Quinase C Ativada/efeitos dos fármacos , Receptores de Quinase C Ativada/metabolismo , 60556/farmacologia , 60556/uso terapêutico , Sepse/complicações , Sepse/tratamento farmacológico , Transdução de Sinais , Subunidade alfa do Fator 1 Induzível por Hipóxia/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo
7.
Acta Trop ; 251: 107112, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38157925

RESUMO

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.


Assuntos
Proteínas de Protozoários , Toxoplasma , Antígenos de Protozoários/genética , Receptores de Quinase C Ativada/genética , Receptores de Quinase C Ativada/metabolismo , Antígenos de Superfície/metabolismo , Anticorpos Antiprotozoários
8.
Shanghai Kou Qiang Yi Xue ; 32(4): 385-390, 2023 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-38044732

RESUMO

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.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Neoplasias Bucais , Humanos , Carcinoma de Células Escamosas/cirurgia , Carcinoma de Células Escamosas/patologia , Granulinas , Metástase Linfática , Neoplasias Bucais/cirurgia , Neoplasias Bucais/patologia , Proteínas de Neoplasias/metabolismo , Prognóstico , Receptores de Quinase C Ativada/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço
9.
Acta Neuropathol Commun ; 11(1): 200, 2023 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-38111057

RESUMO

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.


Assuntos
Esclerose Amiotrófica Lateral , Demência Frontotemporal , Degeneração Lobar Frontotemporal , Sarcoma , Animais , Humanos , Esclerose Amiotrófica Lateral/patologia , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Células HEK293 , Neurônios Motores/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Demência Frontotemporal/patologia , Degeneração Lobar Frontotemporal/patologia , Biossíntese de Proteínas , Sarcoma/metabolismo , Sarcoma/patologia , Proteína FUS de Ligação a RNA/genética , Proteína FUS de Ligação a RNA/metabolismo , Receptores de Quinase C Ativada/genética , Receptores de Quinase C Ativada/metabolismo , Proteínas de Neoplasias/genética
10.
Biomolecules ; 13(12)2023 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-38136569

RESUMO

The Ras GTPase-activating protein SH3 domain-binding protein (G3BP) belongs to the highly conserved family of RNA-binding proteins, which has been well-investigated in humans and animals. However, limited study of plant G3BP has been reported, and the precise biological function of the G3BP family has not been elucidated yet. In this study, the Arabidopsis G3BP family, comprising seven members, was comparatively analyzed. Transcriptome analysis showed that most G3BP genes are ubiquitously expressed in various tissues/organs. Transient expression analysis revealed that all G3BPs were presented in the cytoplasm, among which G3BP6 was additionally found in the nucleus. Further study revealed a conserved NLS motif required for the nuclear localization of G3BP6. Additionally, phenotypic analysis revealed that loss-of-function g3bp6 presented late-flowering phenotypes. RNA-sequencing analysis and qRT-PCR assays demonstrated that the expressions of abundant floral genes were significantly altered in g3bp6 plants. We also discovered that overexpression of G3BP6 in the nucleus, rather than in the cytoplasm, propelled bolting. Furthermore, we revealed that the scaffold protein Receptor for Activated C Kinase 1 (RACK1) interacted with and modulated the nuclear localization of G3BP6. Altogether, this study sheds new light on G3BP6 and its specific role in regulating the flowering transition in Arabidopsis.


Assuntos
Proteínas de Arabidopsis , Arabidopsis , Humanos , Animais , Arabidopsis/genética , Arabidopsis/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Reação em Cadeia da Polimerase , Regulação da Expressão Gênica de Plantas , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Receptores de Quinase C Ativada/genética
11.
Neuron ; 111(24): 3988-4005.e11, 2023 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-37820724

RESUMO

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.


Assuntos
Transtorno do Espectro Autista , Síndrome do Cromossomo X Frágil , Doenças Mitocondriais , Humanos , Proteína do X Frágil de Retardo Mental/genética , Transtorno do Espectro Autista/metabolismo , Neurônios/metabolismo , Neurogênese , Doenças Mitocondriais/metabolismo , Receptores de Quinase C Ativada/genética , Receptores de Quinase C Ativada/metabolismo , Proteínas de Neoplasias/metabolismo , Fatores de Transcrição/metabolismo
12.
Cell Death Differ ; 30(11): 2382-2392, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37828084

RESUMO

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.


Assuntos
Neoplasias Ovarianas , Ubiquitina , Feminino , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Processamento de Proteína Pós-Traducional , Receptores de Quinase C Ativada/genética , Receptores de Quinase C Ativada/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
13.
Commun Biol ; 6(1): 1056, 2023 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-37853189

RESUMO

Neuroligins are transmembrane cell adhesion proteins well-known for their genetic links to autism spectrum disorders. Neuroligins can function by regulating the actin cytoskeleton, however the factors and mechanisms involved are still largely unknown. Here, using the Drosophila neuromuscular junction as a model, we reveal that F-Actin assembly at the Drosophila NMJ is controlled through Cofilin signaling mediated by an interaction between DNlg2 and RACK1, factors not previously known to work together. The deletion of DNlg2 displays disrupted RACK1-Cofilin signaling pathway with diminished actin cytoskeleton proteo-stasis at the terminal of the NMJ, aberrant NMJ structure, reduced synaptic transmission, and abnormal locomotion at the third-instar larval stage. Overexpression of wildtype and activated Cofilin in muscles are sufficient to rescue the morphological and physiological defects in dnlg2 mutants, while inactivated Cofilin is not. Since the DNlg2 paralog DNlg1 is known to regulate F-actin assembly mainly via a specific interaction with WAVE complex, our present work suggests that the orchestration of F-actin by Neuroligins is a diverse and complex process critical for neural connectivity.


Assuntos
Proteínas de Drosophila , Drosophila , Animais , Drosophila/genética , Drosophila/metabolismo , Fatores de Despolimerização de Actina/genética , Fatores de Despolimerização de Actina/metabolismo , Actinas/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Transdução de Sinais/fisiologia , Receptores de Quinase C Ativada/genética
14.
Cell Death Dis ; 14(10): 685, 2023 10 17.
Artigo em Inglês | MEDLINE | ID: mdl-37848434

RESUMO

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.


Assuntos
Neoplasias da Mama , Humanos , Feminino , Neoplasias da Mama/patologia , beta Catenina/metabolismo , Via de Sinalização Wnt/fisiologia , Proliferação de Células , Linhagem Celular Tumoral , Fator 2 Associado a Receptor de TNF/metabolismo , Receptores de Quinase C Ativada/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo
15.
Am J Physiol Renal Physiol ; 325(6): F870-F884, 2023 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-37823194

RESUMO

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.


Assuntos
Nefropatias , Neuropilina-1 , Receptores de Quinase C Ativada , Insuficiência Renal Crônica , Obstrução Ureteral , Animais , Humanos , Camundongos , Ratos , Células Epiteliais/metabolismo , Fibrose , Rim/metabolismo , Nefropatias/patologia , Proteínas de Neoplasias/metabolismo , Neuropilina-1/genética , Neuropilina-1/metabolismo , Receptores de Quinase C Ativada/genética , Receptores de Quinase C Ativada/metabolismo , Insuficiência Renal Crônica/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Obstrução Ureteral/patologia
16.
Int Immunopharmacol ; 124(Pt B): 111034, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37820423

RESUMO

Osteoarthritis (OA) is a degenerative joint disease characterized by cartilage degradation and inflammation. The molecular mechanisms underlying OA progression remain incompletely understood. In this study, we investigated the role of STEAP3 (Six Transmembrane Epithelial Antigen of the Prostate 3) in the development of OA. Our results demonstrated that STEAP3 was upregulated in OA cartilage tissues and contributes to the progression of the disease. To elucidate the mechanism, we employed transcriptomic and interaction proteomics analysis, and identified dysregulated genes and pathways associated with STEAP3 overexpression. Specifically, we found that STEAP3 interacted with Rab7A, a protein involved in intracellular trafficking and autophagy, and suppressed its activity. In addition, STEAP3 interacted with activated C kinase 1 (RACK1) and enhanced its activity. Furthermore, our data indicated that the suppression of Rab7A activity by STEAP3 promoted the activation of receptor tyrosine kinases (RTKs) and the promoting effects of RACK1 by STEAP3, both of which in turn activated the MAPK and JAK/STAT signaling pathways. In conclusion, our findings highlighted the role of STEAP3 in promoting OA progression. By inhibiting Rab7A activity and promoting RACK1 activity, STEAP3 enhanced inflammation through the activation of RTKs and subsequent activation of the MAPK and JAK/STAT signaling pathways. Targeting STEAP3 may provide a potential therapeutic strategy for the treatment of OA by modulating these interconnected pathways.


Assuntos
Osteoartrite , Transdução de Sinais , Animais , Masculino , Ratos , Cartilagem/metabolismo , Inflamação , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Osteoartrite/genética , Receptores de Quinase C Ativada/genética
17.
Eur J Immunol ; 53(12): e2350520, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37683186

RESUMO

Inhibition of the co-stimulatory ligand CD40L has shown beneficial effects in many experimental models of autoimmune disease and inflammation. Here, we show that CD40L deficiency in T cells in mice causes a reduction of CD4+ T-cell activation and specifically a strong reduction in IFN-γ-producing Th1 cells. In vitro, we could not reproduce this antigen presenting cell-dependent effects, but found that T-cell CD40L affects cell death and proliferation. We identified receptor of activated C kinase, the canonical PKC binding partner and known to drive proliferation and apoptosis, as a mediator of CD40L reverse signaling. Furthermore, we found that CD40L clustering stabilizes IFN-γ mediated Th1 polarization through STAT1, a known binding partner of receptor of activated C kinase. Together this highlights the importance of both CD40L forward and reverse signaling.


Assuntos
Ligante de CD40 , Ativação Linfocitária , Camundongos , Animais , Receptores de Quinase C Ativada , Células Th1 , Células Apresentadoras de Antígenos , Antígenos CD40 , Linfócitos T CD4-Positivos
18.
Vet Res ; 54(1): 73, 2023 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-37684678

RESUMO

Pasteurella multocida is a gram-negative bacterium that causes serious diseases in a wide range of animal species. Inflammasomes are intracellular multimolecular protein complexes that play a critical role in host defence against microbial infection. Our previous study showed that bovine P. multocida type A (PmCQ2) infection induces NLRP3 inflammasome activation. However, the exact mechanism underlying PmCQ2-induced NLRP3 inflammasome activation is not clear. Here, we show that NLRP3 inflammasome activation is positively regulated by a scaffold protein called receptor for activated C kinase 1 (RACK1). This study shows that RACK1 expression was downregulated by PmCQ2 infection in primary mouse peritoneal macrophages and mouse tissues, and overexpression of RACK1 prevented PmCQ2-induced cell death and reduced the numbers of adherent and invasive PmCQ2, indicating a modulatory role of RACK1 in the cell death that is induced by P. multocida infection. Next, RACK1 knockdown by siRNA significantly attenuated PmCQ2-induced NLRP3 inflammasome activation, which was accompanied by a reduction in the protein expression of interleukin (IL)-1ß, pro-IL-1ß, caspase-1 and NLRP3 as well as the formation of ASC specks, while RACK1 overexpression by pcDNA3.1-RACK1 plasmid transfection significantly promoted PmCQ2-induced NLRP3 inflammasome activation; these results showed that RACK1 is essential for NLRP3 inflammasome activation. Furthermore, RACK1 knockdown decreased PmCQ2-induced NF-κB activation, but RACK1 overexpression had the opposite effect. In addition, the immunofluorescence staining and immunoprecipitation results showed that RACK1 colocalized with NLRP3 and that NEK7 and interacted with these proteins. However, inhibition of potassium efflux significantly attenuated the RACK1-NLRP3-NEK7 interaction. Our study demonstrated that RACK1 plays an important role in promoting NLRP3 inflammasome activation by regulating NF-κB and promoting NLRP3 inflammasome assembly.


Assuntos
Doenças dos Bovinos , Infecções por Pasteurella , Pasteurella multocida , Animais , Bovinos , Camundongos , Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , NF-kappa B , Infecções por Pasteurella/veterinária , Receptores de Quinase C Ativada
19.
J Virol ; 97(10): e0074723, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37712706

RESUMO

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.


Assuntos
Receptores de Quinase C Ativada , Infecções por Vírus Respiratório Sincicial , Vírus Sincicial Respiratório Humano , Proteínas da Matriz Viral , Humanos , Interferons , Proteínas de Neoplasias/genética , Proteínas , Proteômica , Receptores de Quinase C Ativada/metabolismo , Infecções por Vírus Respiratório Sincicial/metabolismo , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sincicial Respiratório Humano/genética , Proteínas da Matriz Viral/metabolismo
20.
PeerJ ; 11: e15873, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37601269

RESUMO

Background: The receptor for activated C kinase 1 (RACK1) expression is associated with clinicopathological characteristics and the prognosis of various cancers; however, the conclusions are controversial. As a result, this study aimed to explore the clinicopathological and prognostic values of RACK1 expression in patients with cancer. Methodology: PubMed, Embase, Web of Science, Cochrane Library, and Scopus were comprehensively explored from their inception to April 20, 2023, for selecting studies on the clinicopathological and prognostic role of RACK1 in patients with cancer that met the criteria for inclusion in this review. Pooled hazard ratios (HRs) and 95% confidence intervals (CIs) were used to assess the prognosis-predictive value of RACK1 expression, while pooled odds ratios (ORs) and 95% CIs were used to evaluate the correlation between RACK1 expression and the clinicopathological characteristics of patients with cancer. The quality of the included studies was evaluated using the Newcastle-Ottawa Scale. Results: Twenty-two studies (13 on prognosis and 20 on clinicopathological characteristics) were included in this systematic review and meta-analysis. The findings indicated that high RACK1 expression was significantly associated with poor overall survival (HR = 1.62; 95% CI, 1.13-2.33; P = 0.009; I2 = 89%) and reversely correlated with disease-free survival/recurrence-free survival (HR = 1.87; 95% CI, 1.22-2.88; P = 0.004; I2 = 0%). Furthermore, increased RACK1 expression was significantly associated with lymphatic invasion/N+ stage (OR = 1.74; 95% CI, 1.04-2.90; P = 0.04; I2 = 79%) of tumors. Conclusions: RACK1 may be a global predictive marker of poor prognosis in patients with cancer and unfavorable clinicopathological characteristics. However, further clinical studies are required to validate these findings.


Assuntos
Proteínas de Neoplasias , Neoplasias , Receptores de Quinase C Ativada , Humanos , Intervalo Livre de Doença , Proteínas de Neoplasias/genética , Neoplasias/diagnóstico , Prognóstico , Receptores de Quinase C Ativada/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...