Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Med Sci Monit ; 28: e937532, 2022 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-36031751

RESUMO

BACKGROUND We sought to further our understanding of the biological characteristics underlying severe COVID-19. MATERIAL AND METHODS RNA sequencing (RNA-Seq) analysis was used to evaluate peripheral blood mononuclear cells from 4 patients with severe COVID-19 and 4 healthy controls. We performed gene expression analyses to detect differentially expressed genes (DEGs). Enrichment analyses were performed to identify their molecular processes and signaling pathways, and the protein-protein interaction network was constructed to extract the core gene cluster. The investigation of protein-chemical interactions and regulatory signatures for specific regulatory checkpoints and powerful chemical agents was then conducted for these essential genes. Finally, we used single-cell RNA-Seq analysis from an online platform to show how these genes were expressed differently, depending on the kind of cell. RESULTS A total of 268 DEGs were found. The biological process of protein ubiquitination was later discovered to be highly influenced by the core gene cluster (ITCH, TRIM21, RNF130, FBXO11, UBE2J1, and ASB16) at the transcriptome level. Six transcription factors, FNIC, FOXA1, YY1, GATA2, MET2A, and FOXC1, as well as miRNAs hsa-miR-1-3p and hsa-miR-27a-3p were identified. We found a potent chemical agent, copper sulfate, may regulate protein ubiquitination genes cooperatively, and the genes regulating protein ubiquitination could be expressed highly on the macrophages. CONCLUSIONS Taken together, we suggest that protein ubiquitination is a crucial functional process in patients with severe COVID-19. This study will give a deeper insight into biological characteristics and progression of COVID-19 and facilitate development of novel therapeutics, leading to significant advancements in the COVID-19 pandemic.


Assuntos
COVID-19 , Proteínas F-Box , MicroRNAs , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Leucócitos Mononucleares , Pandemias , Proteína-Arginina N-Metiltransferases , Transcriptoma , Ubiquitinação
2.
BMC Infect Dis ; 22(1): 651, 2022 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-35902828

RESUMO

BACKGROUND: At present, skeletal tuberculosis (TB) diagnosis is mostly by histopathology, but the positivity rate is low. There is a need to develop new methods for the molecular identification of this disorder. Therefore, we aimed to investigate the clinical utility of quantitative PCR (qPCR)-based diagnosis of skeletal TB from formalin-fixed paraffin-embedded (FFPE) tissues and its comparative evaluation with acid-fast bacillus staining (AFS). METHODS: We detected Mycobacterium tuberculosis (M. tuberculosis/MTB) DNA using qPCR and AFS in FFPE tissue samples from 129 patients suspected of having skeletal TB. The sensitivity, specificity as well as area under the curve (AUC) of qPCR and AFS were calculated. Meanwhile, some factors potentially affecting qPCR and AFS results were investigated. RESULTS: Overall, qPCR outperformed AFS in detecting M. tuberculosis. The AUC of qPCR was higher than that of AFS (0.744 vs.0.561, p < 0.001). Furthermore, decalcification of bone tissues did not affect the sensitivity and specificity of qPCR tests. Whereas it impacted the performance of AFS, decalcification increased AFS's specificity and decreased its sensitivity (p < 0.05). Moreover, qPCR had a significantly larger AUC than AFS in decalcified and non-decalcified groups (0.735/0.756 vs. 0.582/0.534, p < 0.001) respectively. Similarly, the AUC of PCR was more extensive than that of AFS regardless of skeletal TB patients with concomitant pulmonary TB or not (0.929 vs. 0.762; 0.688 vs. 0.524, p < 0.01). CONCLUSIONS: Our data demonstrate that qPCR offers superior accuracy for the detection of mycobacteria in FFPE tissues compared to traditional AFS, indicating its clinical value in osteoarticular TB diagnosis.


Assuntos
Mycobacterium tuberculosis , Tuberculose dos Linfonodos , Formaldeído , Humanos , Mycobacterium tuberculosis/genética , Inclusão em Parafina , Reação em Cadeia da Polimerase em Tempo Real/métodos , Sensibilidade e Especificidade
3.
Biosaf Health ; 4(3): 205-208, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35434596

RESUMO

The immune responses and the function of immune cells among asymptomatic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection cases, especially in immuno-compromised individuals, remain largely unknown. Here we present a case of asymptomatic SARS-CoV-2 infection that lasted for at least 67 days. The patient has administrated Thymalfasin as 1.6 mg per dose every other day from Day 45 to 70, plus 200 mg per dose Arbidol antiviral therapy three doses per day from Day 48 to 57. Throughout the infection, no anti-SARS-CoV-2 specific IgM or IgG antibodies were detected. Instead, the patient showed either a low percentage or an absolute number of non-classical monocytes, dendritic cells (DCs), CD4+ T cells, and regulatory T cells (Tregs), which may account for the clinical feature and absence of antibody response. This case may shed new light on the outbreak management related to control/prevention, treatment, and vaccination of SARS-CoV-2 and other virus infections in immunocompromised individuals.

4.
Mol Ther ; 30(1): 164-174, 2022 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-33992807

RESUMO

Transthyretin (TTR) amyloidosis is a hereditary life-threatening disease characterized by deposition of amyloid fibrils. The main causes of TTR amyloidosis are mutations in the TTR gene that lead to the production of misfolded TTR protein. Reducing the production of toxic protein in the liver is a validated strategy to treat TTR amyloidosis. In this study, we established a humanized mouse model that expresses mutant human TTR (hTTR; V30M) protein in the liver to model TTR amyloidosis. Then, we compared the efficiency of reducing the expression of mutant hTTR by dual adeno-associated virus 8 (AAV8)-mediated split SpCas9 with that by single AAV8-mediated Nme2Cas9 in this model. With two gRNAs targeting different exons, dual AAV-mediated split SpCas9 system achieved efficiencies of 37% and 34% reduction of hTTR mRNA and reporter GFP expression, respectively, in the liver. Surprisingly, single AAV-mediated Nme2Cas9 treatment resulted in 65% and 71% reduction of hTTR mRNA and reporter GFP, respectively. No significant editing was identified in predicted off-target sites in the mouse and human genomes after Nme2Cas9 targeting. Thus, we provide proof of principle for using single AAV-mediated CRISPR-Nme2Cas9 to effectively reduce mutant hTTR expression in vivo, which may translate into gene therapy for TTR amyloidosis.


Assuntos
Neuropatias Amiloides Familiares , Amiloide , Neuropatias Amiloides Familiares/genética , Neuropatias Amiloides Familiares/terapia , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Pré-Albumina/genética
5.
J Dermatol Sci ; 105(1): 18-26, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34930675

RESUMO

BACKGROUND: Dysregulation of miR-486-3p was related to the growth and development of a variety of cancers, but the specific function of miR-486-3p in cutaneous squamous cell carcinoma (cSCC) is not to be confirmed yet. OBJECTIVE: Our present study aimed to validate the potential molecular mechanisms of miR-486-3p in cSCC and the potential of miR-486-3p as a novel target for future treatment. METHODS: Human cSCC samples and normal skin tissues were applied to determine the expression level of miR-486-3p and FLOT2 by fluorescence in situ hybridization (FISH) and quantitative reverse transcription PCR (qRT-PCR), respectively. As well as BALB/C nude mouse tumor model, three cSCC cells lines including HSC-1, HSC-5 and A431 were utilized to demonstrate the potential function of miR-486-3p and FLOT2 in tumorigenesis. RESULTS: Our experimental results showed that miR-486-3p was highly expressed both in tumor samples and cell lines of cSCC. Upregulation of miR-486-3p enhanced the proliferation and migration ability of cSCC cell lines and promoted tumorigenicity in vivo. Furthermore, we confirmed that FLOT2 was a direct targeted gene of miR-486-3p. In contrary to the expression level of miR-486-3p, FLOT2 was low expressed in cSCC patient specimens and cell lines. Knockdown of FLOT2 promoted tumorigenesis of cSCC; whereas FLOT2 reversed the tumor-promoting effect of miR-486-3p. CONCLUSION: Our data exhibited that miR-486-3p exerted its effects on carcinogenesis as an oncogene in cSCC via suppression of FLOT2. This discovery will develop new therapeutic targets of cSCC.


Assuntos
Carcinoma de Células Escamosas/etiologia , Proteínas de Membrana/metabolismo , MicroRNAs/metabolismo , Neoplasias Cutâneas/etiologia , Adulto , Idoso , Animais , Carcinogênese , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/terapia , Linhagem Celular Tumoral , Feminino , Humanos , Masculino , Camundongos Nus , MicroRNAs/antagonistas & inibidores , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Metástase Neoplásica , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/terapia , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Front Immunol ; 12: 745854, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34721415

RESUMO

Alpha 7 nicotinic acetylcholine receptor (α7 nAChR) is critical for the pathogenesis of Escherichia coli (E. coli) K1 meningitis, a severe central nervous system infection of the neonates. However, little is known about how E. coli K1 manipulates α7 nAChR signaling. Here, through employing immortalized cell lines, animal models, and human transcriptional analysis, we showed that E. coli K1 infection triggers releasing of secreted Ly6/Plaur domain containing 1 (SLURP1), an endogenous α7 nAChR ligand. Exogenous supplement of SLURP1, combined with SLURP1 knockdown or overexpression cell lines, showed that SLURP1 is required for E. coli K1 invasion and neutrophils migrating across the blood-brain barrier (BBB). Furthermore, we found that SLURP1 is required for E. coli K1-induced α7 nAChR activation. Finally, the promoting effects of SLURP1 on the pathogenesis of E. coli K1 meningitis was significantly abolished in the α7 nAChR knockout mice. These results reveal that E. coli K1 exploits SLURP1 to activate α7 nAChR and facilitate its pathogenesis, and blocking SLURP1-α7 nAChR interaction might represent a novel therapeutic strategy for E. coli K1 meningitis.


Assuntos
Antígenos Ly/fisiologia , Barreira Hematoencefálica , Infecções por Escherichia coli/microbiologia , Escherichia coli/fisiologia , Meningite devida a Escherichia coli/fisiopatologia , Ativador de Plasminogênio Tipo Uroquinase/fisiologia , Receptor Nicotínico de Acetilcolina alfa7/agonistas , Animais , Antígenos Ly/genética , Linhagem Celular , Líquido Cefalorraquidiano/microbiologia , Células Endoteliais/microbiologia , Escherichia coli/isolamento & purificação , Hipocampo/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Recém-Nascido , Memantina/farmacologia , Meningite devida a Escherichia coli/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/fisiologia , Proteínas Recombinantes/metabolismo , Organismos Livres de Patógenos Específicos , Ativador de Plasminogênio Tipo Uroquinase/genética , Receptor Nicotínico de Acetilcolina alfa7/antagonistas & inibidores , Receptor Nicotínico de Acetilcolina alfa7/deficiência
7.
J Dermatol Sci ; 102(3): 184-192, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34016486

RESUMO

BACKGROUND: Aberrant expression of Armadillo repeat containing 8 (ARMC8) plays crucial roles in tumor growth and metastasis of various cancers. The specific role of ARMC8 in cutaneous squamous cell carcinoma (cSCC) is yet to be elucidated. OBJECTIVE: The present study aimed to investigate the molecular mechanisms of ARMC8 and epithelial-mesenchymal transition (EMT) in cSCC development and provide translational insights for future therapeutics. METHODS: cSCC tumor specimens were used to determine the ARMC8 by immunohistochemistry. Three cSCC cell lines including HSC-1, HSC-5 and A431 as well as BALB/C mouse tumor model was utilized to study the potential mechanisms in tumorigenesis. RESULTS: Our data identified ARMC8 as a direct downstream target of miR-664. We found that ARMC8 was remarkably low expression in cSCC patient specimens and cSCC cell lines. Knockdown of ARMC8 promotes tumorigenic behaviors such as increased cell proliferation, migration and invasion capacities in vitro and enhanced tumorigenicity in xenograft mouse model. Whereas ARMC8 over-expression inhibits tumorigenesis in cSCC. Together, it revealed ARMC8 functions as a tumor suppressor via restraining Wnt/ß-catenin pathway and epithelial-mesenchymal transition in cSCC. CONCLUSION: Our data verifies that aberrant expression of ARMC8 plays a vital role in carcinogenesis of cSCC. And overexpression of ARMC8 will facilitate future development of cSCC therapeutic interventions.


Assuntos
Proteínas do Domínio Armadillo/genética , Carcinogênese/genética , Carcinoma de Células Escamosas/genética , MicroRNAs/metabolismo , Neoplasias Cutâneas/genética , Animais , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Regulação para Baixo , Transição Epitelial-Mesenquimal/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Neoplasias Cutâneas/patologia , Via de Sinalização Wnt/genética , Ensaios Antitumorais Modelo de Xenoenxerto , beta Catenina/metabolismo
8.
J Immunol Res ; 2021: 6657012, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33506057

RESUMO

BACKGROUND: Preoperative chemoradiotherapy (pCRT) is a common and essential therapeutic strategy for patients with locally advanced rectal cancer (LARC), but poor tumor response and therapeutic resistance to chemoradiotherapy have appeared usually among persons and affected those patients' survival prognosis. The resistance to chemoradiotherapy in rectal cancer is difficult to predict. This study was aimed at evaluating the role of V-set and transmembrane domain containing 2 like protein (VSTM2L) in resistance to chemoradiotherapy in rectal cancer. METHODS: Analysis of the GEO profiling datasets of rectal cancer patients receiving pCRT disclosed that VSTM2L as a candidate gene was significantly upregulated in nonresponders of rectal cancer with pCRT. The mRNA and protein expression of VSTM2L was detected by quantitate real-time PCR, western blotting, and immunohistochemistry in six rectal cancer biopsy tissues before pCRT. Furthermore, the rectal cancer patient-derived organoids were cultured to evaluate the association of VSTM2L expression and tumor response to CRT. Overexpression of VSTM2L in cancer cells treated with CRT was analyzed for the function of cell proliferation and viability, clone formation, DNA damage repair, and apoptosis ability. The GSEA and RNA-sequence analysis were used to find the downstream mechanism of VSTM2L overexpression in cells treated with CRT. RESULTS: The mRNA levels of VSTM2L were significantly downregulated in normal rectal tissues compared to tumor tissues and were upregulated in nonresponders of rectal cancer patients receiving pCRT and positively correlated with poor survival prognosis from GEO datasets. High expression of VSTM2L was significantly associated with tumor regression after pCRT (P = 0.030). Moreover, high expression of VSTM2L reduced γ-H2AX expression in rectal cancer patient-derived organoids treated with CRT. The overexpression of VSTM2L in colorectal cancer cells induced resistance to CRT via promoting cell proliferation and inhibiting apoptosis. The molecular mechanism revealed that the overexpression of VSTM2L induced resistance to CRT through downstream IL-4 signaling affecting the progress of cell proliferation and apoptosis. CONCLUSION: The high expression of VSTM2L induced resistance to CRT, and adverse survival outcomes served as a prognostic factor in patients with rectal cancer receiving pCRT, suggesting that VSTM2L high expression may be a potential resistant predictable biomarker for LARC patients receiving pCRT.


Assuntos
Quimiorradioterapia/estatística & dados numéricos , Terapia Neoadjuvante/estatística & dados numéricos , Recidiva Local de Neoplasia/epidemiologia , Proteínas do Tecido Nervoso/metabolismo , Neoplasias Retais/terapia , Adulto , Idoso , Linhagem Celular Tumoral , Conjuntos de Dados como Assunto , Intervalo Livre de Doença , Regulação para Baixo , Resistencia a Medicamentos Antineoplásicos/imunologia , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Interleucina-4/metabolismo , Masculino , Pessoa de Meia-Idade , Terapia Neoadjuvante/métodos , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/imunologia , Recidiva Local de Neoplasia/prevenção & controle , Protectomia , Prognóstico , Tolerância a Radiação/imunologia , Neoplasias Retais/genética , Neoplasias Retais/imunologia , Neoplasias Retais/mortalidade , Transdução de Sinais/imunologia
9.
Transgenic Res ; 30(1): 63-76, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33394315

RESUMO

Taking advantage of their ability to integrate their genomes into the host genome, lentiviruses have been used to rapidly produce transgenic mice in biomedical research. In most cases, transgenes delivered by lentiviral vectors have resisted silencing mediated by epigenetic modifications in mice. However, some studies revealed that methylation caused decreased transgene expression in mice. Therefore, there is conflicting evidence regarding the methylation-induced silencing of transgenes delivered by lentiviral transduction in mice. In this study, we present evidence that the human TTR transgene was silenced by DNA methylation in the liver of a transgenic mouse model generated by lentiviral transduction. The density of methylation on the transgene was increased during reproduction, and the expression of the transgene was completely silenced in mice of the F2 generation. Interestingly, 5-azacytidine (5-AzaC), a methyltransferase inhibitor, potently reactivated the silenced genes in neonatal mice whose hepatocytes were actively proliferating and led to stable transgene expression during development. However, 5-AzaC did not rescue liver transgene expression when administered to adult mice. Moreover, 5-AzaC at the given dose had low developmental toxicity in the newborn mice. In summary, we demonstrate the methylation-induced silencing of an exogenous gene in the liver of a mouse model generated by lentiviral transduction and show that the silenced transgene can be safely and efficiently reactivated by 5-AzaC treatment, providing an alternative way to obtain progeny with stable transgene expression in the case of the methylation of exogenous genes in transgenic mice generated by lentiviral transduction.


Assuntos
Metilação de DNA/genética , Lentivirus/genética , Pré-Albumina/genética , Transgenes/genética , Animais , Animais Recém-Nascidos , Azacitidina/farmacologia , Metilação de DNA/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Vetores Genéticos/efeitos dos fármacos , Humanos , Lentivirus/efeitos dos fármacos , Camundongos , Camundongos Transgênicos/genética
10.
Gut ; 70(10): 1872-1883, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33310751

RESUMO

OBJECTIVE: O-linked N-acetylglucosaminylation (O-GlcNAcylation), controlled by O-GlcNAcase (OGA) and O-GlcNAc transferase (OGT), is an important post-translational modification of eukaryotic proteins and plays an essential role in regulating gut inflammation. Gut microbiota encode various enzymes involved in O-GlcNAcylation. However, the characteristics, abundance and function of these enzymes are unknown. DESIGN: We first investigated the structure and taxonomic distribution of bacterial OGAs and OGTs. Then, we performed metagenomic analysis to explore the OGA genes abundance in health samples and different diseases. Finally, we employed in vitro and in vivo experiments to determine the effects and mechanisms of bacterial OGAs to hydrolyse O-GlcNAcylated proteins in host cells and suppress inflammatory response in the gut. RESULTS: We found OGAs, instead of OGTs, are enriched in Bacteroidetes and Firmicutes, the major bacterial divisions in the human gut. Most bacterial OGAs are secreted enzymes with the same conserved catalytic domain as human OGAs. A pooled analysis on 1999 metagenomic samples encompassed six diseases revealed that bacterial OGA genes were conserved in healthy human gut with high abundance, and reduced exclusively in ulcerative colitis. In vitro studies showed that bacterial OGAs could hydrolyse O-GlcNAcylated proteins in host cells, including O-GlcNAcylated NF-κB-p65 subunit, which is important for activating NF-κB signalling. In vivo studies demonstrated that gut bacteria-derived OGAs could protect mice from chemically induced colonic inflammation through hydrolysing O-GlcNAcylated proteins. CONCLUSION: Our results reveal a previously unrecognised enzymatic activity by which gut microbiota influence intestinal physiology and highlight bacterial OGAs as a promising therapeutic strategy in colonic inflammation.


Assuntos
Colite Ulcerativa/enzimologia , Colite Ulcerativa/genética , N-Acetilglucosaminiltransferases/genética , Animais , Bacteroidetes/enzimologia , Firmicutes/enzimologia , Microbioma Gastrointestinal , Humanos , Metagenômica , Camundongos , N-Acetilglucosaminiltransferases/farmacologia
11.
J Dermatol Sci ; 100(1): 67-74, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32951990

RESUMO

BACKGROUND: Human Apurinic/Apyrimidinic Endonuclease 1 (APE1/REF-1/HAP1) is a multifunction protein involved in the progression of cancer. But the role of APE1 in cutaneous squamous cell carcinoma (cSCC) is unclear. OBJECTIVE: This study is aimed to investigate the basic modulatory mechanism of APE1 in cSCC development and offer a novel potential target for clinical treatment. METHODS: The expression of APE1 in cSCC tissues was detected by western blot and immunohistochemistry (IHC) staining. The function of APE1 and miR-27a in cSCC cells was investigated by cell counting kit-8 (CCK-8) assays, colony formation assays and transwell migration assays. Western blot was used to determine the expression of APE1 in cSCC and epithelial-mesenchymal transition (EMT) markers in HSC-1 and HSC-5 cells with APE1 knockdown or overexpression. Double luciferase reporter assays were performed to confirm the interaction of miR-27a and APE1. RESULTS: We identified that APE1 was significantly upregulated in human cSCC tissues and cSCC cells and its overexpression promoted cell proliferation, migration and the expression of EMT markers in cSCC cells. Mechanistically, miR-27a was predicted and confirmed as the upstream of APE1. Its downregulation also enhanced the proliferation and migration of cSCC cells. Rescue experiments demonstrated that restoration of APE1 expression significantly abolished the inhibition of cell proliferation and migration mediated by miR-27a. CONCLUSION: As a direct gene of miR-27a, APE1 improved cell proliferation and migration to promote the progression of cSCC, which could be considered as a potential therapeutic target for cSCC treatment.


Assuntos
Carcinoma de Células Escamosas/genética , DNA Liase (Sítios Apurínicos ou Apirimidínicos)/genética , Regulação Neoplásica da Expressão Gênica , MicroRNAs/metabolismo , Neoplasias Cutâneas/genética , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Transição Epitelial-Mesenquimal/genética , Feminino , Técnicas de Silenciamento de Genes , Células HaCaT , Humanos , Masculino , MicroRNAs/agonistas , MicroRNAs/antagonistas & inibidores , Pessoa de Meia-Idade , Pele/patologia , Neoplasias Cutâneas/patologia , Regulação para Cima
12.
Dose Response ; 18(2): 1559325820929234, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32547335

RESUMO

BACKGROUND: MiR-664 has been demonstrated to play an important role in dermal diseases. However, the functions of miR-664 in ultraviolet B (UVB) radiation-induced keratinocytes damage remain to be elucidated. OBJECTIVE: The present study aimed to investigate the molecular mechanisms under the UVB-induced keratinocytes damage and provide translational insights for future therapeutics and UVB protection. METHODS: HaCaT cells were transfected with miR-664, either alone or combined with UVB irradiation. Levels of messenger RNA and protein were tested by quantitative real-time polymerase chain reaction and Western blot analyses. Cell proliferation, percentage of apoptotic cells, and expression levels of apoptosis-related factors were measured by Cell Counting Kit-8 assay, flow cytometry assay, and Western blot analysis, respectively. RESULTS: We found that a significant increase in miR-664 was observed in UVB-induced HaCaT cells. Overexpressed miR-664 promoted cell vitalities and suppressed apoptosis of UVB-induced HaCaT cells. Additionally, the loss/gain of armadillo-repeat-containing protein 8 (ARMC8) rescued/blocked the effects of miR-664 on the proliferation of UVB-induced HaCaT cells. CONCLUSIONS: Our data demonstrate that miR-664 functions as a protective regulator in UVB-induced HaCaT cells via regulating ARMC8.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA