Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Clin Exp Immunol ; 215(2): 202-214, 2024 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-37594231

RESUMO

NOD-like receptor family, pyrin domain-containing 3 (NLRP3) is a central protein contributing to human inflammatory disorders, including cryopyrin-associated periodic syndrome and sepsis. However, the molecular mechanisms and functions of NLRP3 activation in various diseases remain unknown. Here, we generated gain-of-function knock-in mice associated with Muckle-Wells syndromes using the Cre-LoxP system allowing for the constitutive T346M mutation of NLRP3 to be globally expressed in all cells under the control of tamoxifen. The mice were treated with tamoxifen for 4 days before determining their genotype by PCR and sequence analysis. In vitro, we found that bone marrow-derived macrophage from homozygous T346M mutation mice displayed a robust ability to produce IL-1ß in response to lipopolysaccharide exposure. Moreover, ASC specks and oligomerization were observed in the homozygous mutant bone marrow-derived macrophages in the presence of lipopolysaccharides alone. Mechanistically, K+ and Ca2+ depletion and mitochondrial depolarization contribute to the hyperactivation of mutant NLRP3. In vivo, homozygous mice carrying the T346M mutation exhibit weight loss and mild inflammation in the resting state. In the lipopolysaccharide-mediated sepsis model, homozygous mutant mice exhibited higher mortality and increased serum circulating cytokine levels, accompanied by serious liver injury. Furthermore, an increase in myeloid cells in the spleen has been suggested to be a risk factor for inducing sepsis sensitivity. Altogether, we describe a cryopyrin-associated syndrome animal model with the T346M mutation of NLRP3 and suggest that the hyperactivated inflammasome aggregated by the mutant NLRP3 lowers the inflammatory response threshold both in vitro and in vivo.


Assuntos
Síndromes Periódicas Associadas à Criopirina , Sepse , Camundongos , Humanos , Animais , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Inflamassomos/metabolismo , Lipopolissacarídeos , Síndromes Periódicas Associadas à Criopirina/genética , Síndromes Periódicas Associadas à Criopirina/metabolismo , Inflamação , Modelos Animais de Doenças , Tamoxifeno , Interleucina-1beta/genética
2.
Heliyon ; 9(9): e20105, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37809969

RESUMO

Carboxyamidotriazole (CAI) was initially considered a non-cytotoxic anticancer agent. However, recently, pronounced anti-inflammatory properties of CAI have been reported. Rheumatoid arthritis (RA) is an autoimmune inflammatory disease characterized by aberrant activation of signaling pathways. Therefore, this study explored the therapeutic effects and potential mechanism of action of CAI on RA in the adjuvant arthritis (AA) model. The results showed that CAI reduced the severity of arthritis in AA rats as demonstrated by inhibited hind paw swelling, reduced body weight, and decreased infiltration of joint pathological inflammatory cells. Importantly, pathological scoring of new blood vessels and immunohistochemical assays revealed that CAI inhibited pannus formation. CAI decreased the expression of pro-angiogenic growth factors, such as vascular epidermal growth factor, basic fibroblast growth factor, and metalloproteinases (MMPs), namely, MMP-1 and MMP-3 in the synovium of AA rats. Furthermore, CAI significantly reduced the increased levels of phosphorylated p38, c-Jun N-terminal kinase (JNK)1/2, and extracellular signal-regulated kinase (ERK)1/2 proteins in AA rats. In addition, the proliferation of fibroblast-like synoviocytes (FLS) was downregulated by CAI both in vivo and in vitro. In conclusion, this investigation illustrates the therapeutic effect of CAI on synovitis and erosion of articular cartilage in RA. Furthermore, the mechanism might involve inhibition of aberrantly activated mitogen-activated protein kinase signaling, as well as a decrease in pro-angiogenic factors, MMP expression, and FLS proliferation.

3.
Clin Rheumatol ; 42(6): 1635-1643, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36418508

RESUMO

OBJECTIVES: This study aimed to explore the changes of four major inflammasomes in adult-onset Still's disease (AOSD) and preliminarily evaluate the therapeutic effect of carboxyamidotriazole (CAI), which has previously been reported to have the significant anti-inflammatory activity. METHOD: The mRNA expressions of proinflammatory cytokines and inflammasome components in peripheral blood mononuclear cells (PBMCs) from AOSD patients and healthy controls (HC) were determined by reverse transcription-quantitative PCR. Poly(dA:dT)-induced AIM2 inflammasome and flagellin-induced NLRC4 inflammasome activation models were established in bone marrow-derived macrophages (BMDMs). The levels of cytokines in serum and culture supernatants were measured by ELISA method. RESULTS: The serum levels of IL-1ß, IL-6, and TNF-α in AOSD patients were significantly higher than those in HC. However, the mRNA expressions of IL-1ß, IL-6, IL-18, and TNF-α in PBMCs did not differ markedly in AOSD patients in comparison with HC. Significantly increased mRNA levels of AIM2, NLRC4, ASC, and caspase-1 were observed in patients with AOSD when compared with HC, while NLRP1 and NLRP3 showed no change in AOSD samples. In addition, CAI treatment could significantly reduce the levels of IL-1ß, IL-6, and TNF-α secreted by AOSD PBMCs and inhibit AIM2 and NLRC4 inflammasomes activation in BMDMs. CONCLUSIONS: Increased levels of proinflammatory cytokines in AOSD might be associated with NLRC4 and AIM2 inflammasomes activation. CAI is likely to have the therapeutic potential for AOSD by inhibiting NLRC4 and AIM2 inflammasomes activation and reducing the proinflammatory cytokines and worthy of further investigation. These results provide new ideas for elucidating the pathogenesis of AOSD and providing specific targeted therapy. Key points • Significantly higher mRNA levels of AIM2 and NLRC4 inflammsome signaling were observed in AOSD patients compared with health controls, indicating that AIM2 and NLRC4 inflammsome activation might be related to the increased proinflammatory cytokines in AOSD. • CAI treatment markedly reduced the secretion levels of cytokines IL-1ß, IL-6, and TNF-α in AOSD PBMCs and inhibited AIM2 and NLRC4 inflammasome activation.


Assuntos
Inflamassomos , Doença de Still de Início Tardio , Adulto , Humanos , Inflamassomos/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Interleucina-6/metabolismo , Leucócitos Mononucleares/metabolismo , Citocinas/metabolismo , Interleucina-1beta/metabolismo , RNA Mensageiro/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteínas de Ligação ao Cálcio , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Proteínas de Ligação a DNA
4.
Clin Cosmet Investig Dermatol ; 15: 1561-1573, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35967917

RESUMO

Background: Silicone implants are currently the most widely used artificial materials in plastic surgery. Capsule formation following implant application is unavoidable. When the capsule is excessively thick and strongly contracted, it can lead to obvious symptoms, clinically known as capsular contracture. Biological factors have always been the focus of research on the capsule formation. As a growth factor, progranulin (PGRN) plays an important regulatory role in wound healing, tissue fibrosis, tumor proliferation and invasion, and inflammation regulation. At present, the research on the capsule mainly involves the regulation of tissue healing and fibrosis under the influence of inflammation. Because PGRN has a regulatory role in these processes, we believe that the study of both can provide a new theoretical basis and intervention sites for monitoring and inhibiting the development of the capsule. Methods: In this experiment, the effects of different surgical operations on the content of PGRN in the surgical site and plasma of rats were detected. Sprague-Dawley (SD) rat dermal fibroblasts were co-cultured by recombinant PGRN. The effects of r-PGRN on fibroblasts were detected by 5-ethynyl-2'-deoxyuridine (EdU) assay, wound healing assay and Western blot assay. Finally, the effect of PGRN on capsule formation and contracture was studied by changing the content of PGRN in the prosthesis in rats after operation. Results: Surgical trauma and silicone implant increased plasma and local PGRN levels in SD rats. PGRN can activate the TGF-ß/SMAD signaling pathway in a dose-dependent manner, thereby promoting fibroblast proliferation, differentiation and migration and inhibiting apoptosis and enhancing cell function, thereby promoting capsule formation and contracture. Conclusion: PGRN promotes the formation and contracture of the silicone implant capsule in SD rats by activating the TGF-ß/SMAD signaling pathway. This discovery may provide new therapeutic targets and detection indicators.

5.
J Plast Reconstr Aesthet Surg ; 75(9): 3595-3602, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35672247

RESUMO

BACKGROUND: Nanofat, as a derivative of adipose tissue, has gradually become a research hotspot in beauty and regenerative medicine. However, the nanofat preparation method has not yet been standardized; it remains unknown whether the aperture of the connector has any influence on the transplantation effect. METHODS: Adipose tissue was mechanically emulsified into nanofat tissue through different connector apertures (1.0, 1.5, and 2.0 mm). Cell survival and apoptosis were measured using the volume of oil droplets, glucose transportation test, flow cytometry, cell counting kit-8 (CCK-8), wound healing assay, transwell migration assay, and fluorescence staining. The expression of adiponectin, GluT4, and PPAR-γ in nanofat-derived stem cells (NFSCs) was detected using quantitative real-time polymerase chain reaction (qRT-PCR). RESULTS: The fineness of nanofat tissue texture decreased with an increase in the aperture connector. The amounts of glucose transferred in the three groups (1, 1.5, and 2 mm) were 4.7 ± 0.894, 6.1 ± 1.026, and 6.9 ± 0.868 mmol/L, respectively. Flow cytometric analysis showed that the proportion of NFSCs in the 2.0 mm group was the highest (91.267±1.210%). Cell proliferation and migration abilities were stronger in the 1.5 and 2.0 mm groups. The numbers of late apoptotic and dead cells in the 2.0 mm group were significantly fewer than those in the two other groups. Expression levels of lipid-related genes were as follows: adiponectin > GluT4 > PPAR-γ in each component. CONCLUSIONS: As nanofat is emulsified, the use of larger aperture connectors (2.0 mm) appeared to decrease the degree of adipocyte lysis and increase the biological activity of adipose tissue.


Assuntos
Adipócitos , Lipídeos , Medicina Regenerativa , Adiponectina , Tecido Adiposo , Emulsões , Glucose , Humanos , Receptores Ativados por Proliferador de Peroxissomo
6.
Transl Oncol ; 15(1): 101253, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34737118

RESUMO

BACKGROUND: Spindle and Kinetochore Associated Complex Subunit 3 (SKA3) is a part of the SKA complex, which plays a key role in cell mitosis. Studies have shown that SKA3 was associated with cancer progression. However, its role in skin cutaneous melanoma (SKCM) remains unclear. Here, we investigated the expression level and prognostic value of SKA3 in SKCM. METHODS: Based on public databases, univariate and multivariate Cox regression analyses were used to investigate the different expression of SKA3 between SKCM and normal tissues. Then, the relationship between SKA3 expression level and prognosis was assessed. PPI network and functional enrichment analysis were performed. ESTIMATE and CIBERSORT were expected to evaluate the SKA3 expression and immune status. CCK8, wound healing, transwell assays and tumor xenograft trial were performed to detect the SKA3 function in cell viability, migration and invasion of the cell lines. RESULTS: The SKA3 was highly expressed in SKCM tissues. SKA3 overexpression was associated with poor survival and immune status. SKA3 knockdown inhibited cell viability, migration and invasion of SKCM cells. CONCLUSION: SKA3 is involved in the progression of SKCM and may serve as a new prognostic biomarker and therapeutic target.

7.
Int J Nanomedicine ; 16: 7943-7957, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34887659

RESUMO

PURPOSE: Inhibition of cellular adaptation to hypoxia can cause persistent inflammation, thereby increasing tissue damage and complicating wound healing in diabetes patients. Regulating cellular adaptation to hypoxic environments can help in effective wound repair. Hypoxia-inducible factor (HIF)-1α is a key regulator of cell hypoxia. Extracellular vesicles (EVs) regulate wound repair. This study investigated the mechanism of HIF-1α overexpression in adipose-derived stem cell extracellular vesicles (ADSCs-hEVs) in the repair of diabetic wounds. MATERIALS AND METHODS: HIF-1α expression in diabetes patients and healthy participants was studied. High-throughput sequencing, GO, and KEGG analysis revealed that ADSCs small extracellular vesicle hypoxia environments may increase HIF-1α expression by affecting cell metabolism, differentiation, and TGF-ß secretion, or by altering the PI3K/AKT pathway. Effect of addition of ADSCs-hEVs on cell proliferation and migration was investigated using Western blotting, EdU assay, transwell assay, and migration. In vivo, after 7, 14, and 21 days, important factors for diabetic wound healing were evaluated by immunohistochemistry, qRT-PCR, Masson staining, and H&E staining. RESULTS: HIF-1α expression decreased in the skin of diabetes patients; interleukin (IL)-6 expression increased, and growth factor-related indexes decreased. ADSCs-hEVs significantly increased the expression and secretion of growth factors, compared with ADSCs-EVs. In vivo, ADSC-hEV treatment accelerated the healing rate and improved the healing quality of diabetic wounds compared with ADSCs-EVs. CONCLUSION: Speed and quality of wound healing increased significantly in the ADSCs-hEVs group, which could inhibit early inflammation while promoting the secretion and expression of growth factors and extracellular matrix-related indexes.


Assuntos
Diabetes Mellitus , Vesículas Extracelulares , Proliferação de Células , Fibroblastos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Fosfatidilinositol 3-Quinases , Células-Tronco , Cicatrização
8.
Drug Des Devel Ther ; 15: 4795-4809, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34876804

RESUMO

OBJECTIVE: Human adipose-derived mesenchymal stem cells (ADSCs) have the potential to be applied to solid organ treatments. However, tissue regeneration is limited by the death of transplanted cells. Ischemia is the main cause of the poor outcome. This study aimed to investigate the effect of berberine (BBR) on ADSCs after fat grafting. METHODS: The antioxidant BBR on apoptosis and autophagy of ADSCs in vitro ischemia model was induced by hypoxia and serum deprivation (HY/SD). The autophagy promoter rapamycin and autophagy inhibitor 3-MA were incubated separately to investigate the crosstalk between autophagy and apoptosis. Pathway inhibitors further verified whether the autophagy and apoptosis were regulated by AMPK/mTor signaling pathway. Fat survival, fibrosis, level of inflammatory cell infiltration, and the effect of angiogenesis after BBR treatment were observed in vivo. RESULTS: BBR could reduce ROS production and reverse the decreasing cell survival rate. HY/SD would induce apoptosis and autophagy in ADSCs, and BBR could alleviate these processes. After interfering with the level of autophagy, we also proved that apoptosis was regulated by autophagy and changed accordingly. The results also indicated that BBR could protect against autophagy and apoptosis of ADSCs through AMPK/mTor pathway. The treated human-derived adipose tissue was transplanted into BALB/c nude mice, and with the intervention of BBR, the fat grafting had a higher survival rate, lower inflammatory cell infiltration and fibrosis level. CONCLUSION: Our present study revealed that BBR was a promising anti-autophagy and apoptosis agent for improving the survival rate of ADSCs during cell transplantation.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Berberina/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Animais , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo
9.
Bioorg Med Chem Lett ; 53: 128415, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34662706

RESUMO

Inflammation and immunity are closely related to the occurrence and development of a variety of immune diseases. Although IL-1ß has been identified as a key cytokine in many immune diseases, safe and specific small molecular IL-1ß releasement inhibitors are still scarce and urgently required in clinic. The investigation prospect of triazoleis limited by its complicated pharmacological effect which exhibited inferior effects on IL-1ß and TNF-α. Herein, 36 novel derivatives were designed and synthesized, and nearly half of the derivatives exhibited much better selectivity on IL-1ß releasement inhibition as well as keep similar inhibitory activities to lead compound. In 20 µM, compound 19 exhibited IL-1ß releasement inhibitory activity (IC50 = 5.489 µM) which closed to the original compound, and 4.5-fold superior selectivity (SI = 4.71) to the lead compound (SI = 0.82). A probable SAR model of triazole derivatives for IL-1ß releasement inhibition and selectivity was also proposed, which might promote the discovery of more effective and specific IL-1ß releasement inhibitors in the future.


Assuntos
Descoberta de Drogas , Interleucina-1beta/antagonistas & inibidores , Triazóis/farmacologia , Relação Dose-Resposta a Droga , Humanos , Interleucina-1beta/imunologia , Estrutura Molecular , Relação Estrutura-Atividade , Triazóis/síntese química , Triazóis/química
10.
J Plast Reconstr Aesthet Surg ; 74(8): 1908-1918, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33358677

RESUMO

BACKGROUND: Human adipose mesenchymal stem cells (hADSCs) show poor survival after transplantation, limiting their clinical application. Tissue regeneration resulting from stem cell treatment may be caused by attenuation of hypoxia-inducible factor-1α (HIF-1α). In this study, we constructed hADSCs stably expressing HIF-1α and investigated the potential effects of HIF-1α expression in the ischemic microenvironment on mitochondrial apoptosis and survival of hADSCs, and studied the mechanisms involved. METHOD: Apoptosis was induced by an ischemic microenvironment in vitro. ADSCs with stable HIF-1α expression were established. Cell survival and apoptosis were observed by CCK-8 assay, western blotting, flow cytometry, and fluorescence staining. ADSCs were subcutaneously transplanted into nude mice in the location where a hypoxia ischemic microenvironment was simulated in vivo. After 1, 3, and 7 d, mitochondrial apoptotic proteins were evaluated by immunohistochemistry and immunofluorescence staining. RESULTS: Exogenous HIF-1α downregulated mitochondrial reactive oxygen species, cytochrome c, caspase-9, and caspase-3, but inhibited mitochondrial membrane potential depolarization and increased the Bcl-2/bax ratio. HIF-1α prevented apoptosis and promoted vascular endothelial growth factor (VEGF) secretion as demonstrated by enzyme-linked immunosorbent assay (ELISA), terminal deoxyribonucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining, and flow cytometry analysis. HIF-1α enhanced the survival of transplanted ADSCs in nude mice. CONCLUSION: We have shown that through inhibition of the mitochondria-mediated apoptotic pathway and promotion of VEGF secretion in hADSCs in an ischemic microenvironment, HIF-1α may potentially be applied in clinical therapy and as an alternative strategy for improving hADSC therapy.


Assuntos
Tecido Adiposo/citologia , Apoptose/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Isquemia/fisiopatologia , Mitocôndrias/metabolismo , Transplante de Células-Tronco , Adulto , Proliferação de Células , Sobrevivência Celular , Feminino , Humanos , Técnicas In Vitro
11.
Exp Ther Med ; 20(2): 1455-1466, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32742379

RESUMO

Carboxyamidotriazole (CAI), originally developed as a non-cytotoxic anti-cancer drug, was shown to have anti-inflammatory activity according to recent studies in a number of animal models of inflammation. However, its mechanism of action has not been characterized. Therefore, the present study was performed to identify the anti-inflammatory action of CAI in lipopolysaccharide (LPS)-induced RAW 264.7 macrophages and to identify the signal transduction pathways involved. The in vitro results revealed that CAI had no direct effect on the activity of cyclooxygenase (COX), suggesting a different anti-inflammatory mechanism compared with that of COX-inhibiting non-steroidal anti-inflammatory drugs. Further investigation in RAW264.7 macrophages revealed that CAI decreased the production of nitric oxide via decreasing the LPS-stimulated expression of inducible nitric oxide synthase, and downregulated both mRNA and protein expression levels of the cytokines tumor necrosis factor-α, interleukin (IL)-1ß, and IL-6. CAI also significantly reduced the increased DNA-binding activity of nuclear factor (NF)-κB induced by LPS stimulation. With respect to the mechanisms involved on NF-κB activity, CAI exhibited suppression of the phosphorylation and degradation of the inhibitor of nuclear factor-κBα (IκB), and decreased the phosphorylation levels of the p65 subunit and its subsequent nuclear translocation. In addition, CAI significantly decreased the phosphorylated forms of p38, JNK and ERK, which were increased following LPS stimulation, while the total expression levels of p38, JNK and ERK remained unaltered. The results in the present study indicate that CAI alleviates the inflammatory responses of RAW 264.7 macrophages in response to LPS stimulation via attenuating the activation of NF-κB and MAPK signaling pathways and decreasing the levels of pro-inflammatory mediators. This offers a novel perspective for understanding the anti-inflammatory mechanism of CAI and suggests its potential use as a therapeutic treatment in inflammatory diseases with excessive macrophage activation.

12.
Med Sci Monit ; 25: 1590-1599, 2019 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-30821276

RESUMO

BACKGROUND Malignant melanoma is recalcitrant to most existing chemotherapies, and aberrant expression of miR-211 plays prominent roles in progression of melanoma. However, the trigger mechanism of aberrant miR-211 expression in melanoma is still elusive. MATERIAL AND METHODS We used qRT-PCR to test miR-211 expression. Cell viability assay and mouse xenograft assay were performed to examine the role of miR-211 on the sensitivity of melanoma cells to cisplatin. The epigenetic modification of miR-211 promoter was assess by DNA methylation analysis and DAC treatment. RESULTS In this study, decreased miR-211 expression was detected. Bisulfite sequencing PCR showed that DNA hypermethylation contributed to the downregulation of miR-211 in melanoma tissues. In melanoma cells, overexpressed 211 could enhance the anticancer effect of cisplatin and restoration of miR-211 rendered susceptibility to cisplatin in cisplatin-resistant cells. And the same result was showed in vivo by mouse xenograft assay. What is more, DAC treatment could increase miR-211 expression and EZH2 expression was increased in cisplatin-resistant cells. MiR-211 could be transcriptionally repressed by EZH2 mediated promoter methylation. CONCLUSIONS Taken together, our findings revealed that epigenetic modification of miR-211 governed melanoma cell chemosensitivity and were involved in the progression of tumorigenesis.


Assuntos
Melanoma/tratamento farmacológico , Melanoma/genética , MicroRNAs/genética , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/genética , Animais , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Sobrevivência Celular/genética , Cisplatino/farmacologia , Metilação de DNA/genética , Resistencia a Medicamentos Antineoplásicos/genética , Epigênese Genética/genética , Regulação Neoplásica da Expressão Gênica/genética , Inativação Gênica , Humanos , Melanoma/metabolismo , Camundongos , Camundongos Nus , MicroRNAs/fisiologia , Neoplasias Cutâneas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Melanoma Maligno Cutâneo
13.
Neuroscience ; 404: 282-296, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30742966

RESUMO

Fragile X mental retardation protein (FMRP), a key determinant of normal brain development and neuronal plasticity, plays critical roles in nucleocytoplasmic shuttling of mRNAs. However, the factors involved in FMRP nuclear localization remain to be determined. Using cross-species sequence comparison, we show that an aspartate in position 132 (D132), located within the conserved nuclear localization signal (NLS) of FMRP, appears in human and other mammals, while glutamate 132 (E132) appears in rodents and birds. Human FMRP-D132E alters the secondary structure of the protein and reduces its nuclear localization, while the reciprocal substitution in mouse FMRP-E132D promotes its nuclear localization. Human FMRP could interact with poly(A)-binding protein 1 (PABP1) which is impeded by the D132E mutation. Reversely, mouse FMRP could not interact with PABP1, but the E132D mutation leads to the FMRP-PABP1 interaction. We further show that overexpression of human FMRP-D132E mutant promotes the formation of cytoplasmic aggregates of PABP1 in human cells, but not of mouse FMRP-E132D in mouse cells. PABP1 knockdown reduces the nuclear localization of human FMRP, but not mouse FMRP. Furthermore, RNase A treatment decreases the PABP1 levels in the anti-V5-immunoprecipitates using the V5-hFMRP-transfected cells, suggesting an interaction between human FMRP and PABP1 in an RNA-dependent fashion. Thus, our data suggest that the FMRP protein with the human-used D132 accommodates a novel protein-RNA-protein interaction which may implicate a connection between FMRP residue transition and neural evolution.


Assuntos
Núcleo Celular/metabolismo , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Proteína I de Ligação a Poli(A)/metabolismo , RNA/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular Tumoral , Proteína do X Frágil da Deficiência Intelectual/química , Proteína do X Frágil da Deficiência Intelectual/genética , Células HEK293 , Humanos , Camundongos , Proteína I de Ligação a Poli(A)/química , Proteína I de Ligação a Poli(A)/genética , Ligação Proteica/fisiologia , Estrutura Secundária de Proteína , RNA/química , RNA/genética , Especificidade da Espécie
14.
Neuroscience ; 349: 64-75, 2017 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-28257890

RESUMO

Fragile X mental retardation protein (FMRP), an important RNA-binding protein responsible for fragile X syndrome, is involved in posttranscriptional control of gene expression that links with brain development and synaptic functions. Here, we reveal a novel role of FMRP in pre-mRNA alternative splicing, a general event of posttranscriptional regulation. Using co-immunoprecipitation and immunofluorescence assays, we identified that FMRP interacts with an alternative-splicing-associated protein RNA-binding protein 14 (RBM14) in a RNA-dependent fashion, and the two proteins partially colocalize in the nuclei of hippocampal neurons. We show that the relative skipping/inclusion ratio of the micro-exon L in the Protrudin gene and exon 10 in the Tau gene decreased in the hippocampus of Fmr1 knockout (KO) mice. Knockdown of either FMRP or RBM14 alters the relative skipping/inclusion ratio of Protrudin and Tau in cultured Neuro-2a cells, similar to that in the Fmr1 KO mice. Furthermore, overexpression of FMRP leads to an opposite pattern of the splicing, which can be offset by RBM14 knockdown. RNA immunoprecipitation assays indicate that FMRP promotes RBM14's binding to the mRNA targets. In addition, overexpression of the long form of Protrudin or the short form of Tau promotes protrusion growth of the retinoic acid-treated, neuronal-differentiated Neuro-2a cells. Together, these data suggest a novel function of FMRP in the regulation of pre-mRNA alternative splicing through RBM14 that may be associated with normal brain function and FMRP-related neurological disorders.


Assuntos
Processamento Alternativo/genética , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Precursores de RNA/genética , Animais , Células Cultivadas , Proteína do X Frágil da Deficiência Intelectual/genética , Hipocampo/metabolismo , Imunoprecipitação/métodos , Camundongos Knockout , Neurônios/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
15.
Mol Neurobiol ; 54(4): 2585-2594, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-26993298

RESUMO

Fragile X mental retardation protein (FMRP), associated with fragile X syndrome, is known as an RNA-binding protein to regulate gene expression at post-transcriptional level in the brain. FMRP is also involved in microRNA (miRNA) biogenesis during the process of precursor miRNA (pre-miRNA) into mature miRNA. However, there is no description of the effect of FMRP on primary miRNA (pri-miRNA) processing. Here, we uncover a novel role of FMRP in pri-miRNA processing via controlling Drosha translation. We show that the expression of DROSHA protein, instead of its messenger RNA (mRNA) transcripts, is downregulated in both the hippocampus of Fmr1-knockout mice and the FMRP-knockdown Neuro-2a cells. Overexpression or knockdown FMRP does not alter Drosha mRNA stability. Immunoprecipitation and polysome analyses demonstrate that FMRP binds to the Drosha mRNA and enhances its translation. Additionally, we show that loss of FMRP in Fmr1-deficient mice results in the accumulation of three in six analyzed pri-miRNAs and the reduction of the corresponding pre-miRNAs and mature miRNAs. Thus, our data suggest that FMRP is involved in pri-miRNA processing via enhancing DROSHA expression that may play an important role in fragile X syndrome.


Assuntos
Proteína do X Frágil da Deficiência Intelectual/metabolismo , MicroRNAs/genética , Biossíntese de Proteínas/genética , Processamento Pós-Transcricional do RNA/genética , Ribonuclease III/genética , Animais , Linhagem Celular Tumoral , Regulação para Baixo/genética , Técnicas de Silenciamento de Genes , Camundongos Knockout , MicroRNAs/metabolismo , Ligação Proteica/genética , Estabilidade de RNA/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ribonuclease III/metabolismo , Regulação para Cima/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA