Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
J Immunother ; 2024 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-38980111

RESUMO

Hepatocellular carcinoma (HCC) is the most common type of liver cancer and is associated with high morbidity and mortality rates. The aims of this study were to investigate the immune-promoting action of nucleolar and spindle-associated protein 1 (NUSAP1) and identify an immunotherapy target for HCC. The Cancer Genome Atlas (TCGA) was used to analyze interaction molecules and immune correlation. The interaction between NUSAP1 and SHC binding and spindle associated 1 (SHCBP1) was examined. The role of the SHCBP1/Janus kinase 2/signal transducer and activator of transcription 3 (SHCBP1/JAK2/STAT3) pathway in this process was explored. After co-culture with HCC cell lines, the differentiation of peripheral blood mononuclear cells (PBMCs) into dendritic cells (DC) was evaluated by measuring the expression of surface factors CD1a and CD86. Pathological tissues from 50 patients with HCC were collected to validate the results of cell experiments. The expression levels of CD1a and CD86 in tissues were also determined. The results show that NUSAP1 interacted with SHCBP1 and was positively correlated with DC. In HCC cell lines, an interaction was observed between NUSAP1 and SHCBP1. It was verified that NUSAP1 inhibited the JAK2/STAT3 phosphorylation pathway by blocking SHCBP1. After co-culture, the levels of CD1a and CD86 in PBMC were elevated. In the clinical specimens, CD1a and CD86 expression levels were significantly higher in the high-NUSAP1 group versus the low-NUSAP1 group. In Summary, NUSAP1 enhanced immunity by inhibiting the SHCBP1/JAK2/STAT3 phosphorylation pathway and promoted DC generation and HCC apoptosis. NUSAP1 may be a target of immunotherapy for HCC.

2.
J Cell Biochem ; 125(3): e30521, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38226525

RESUMO

Despite surgical treatment combined with multidrug therapy having made some progress, chemotherapy resistance is the main cause of recurrence and death of gastric cancer (GC). Gastric cancer mesenchymal stem cells (GCMSCs) have been reported to be correlated with the limited efficacy of chemotherapy in GC, but the mechanism of GCMSCs regulating GC resistance needs to be further studied. The gene set enrichment analysis (GSEA) was performed to explore the glycolysis-related pathways heterogeneity across different cell subpopulations. Glucose uptake and lactate production assays were used to evaluate the importance of B7H3 expression in GCMSCs-treated GC cells. The therapeutic efficacy of oxaliplatin (OXA) and paclitaxel (PTX) was determined using CCK-8 and colony formation assays. Signaling pathways altered by GCMSCs-CM were revealed by immunoblotting. The expression of TNF-α in GCMSCs and bone marrow mesenchymal stem cells (BMMSCs) was detected by western blot analysis and qPCR. Our results showed that the OXA and PTX resistance of GC cells were significantly enhanced in the GCMSCs-CM treated GC cells. Acquired OXA and PTX resistance was characterized by increased cell viability for OXA and PTX, the formation of cell colonies, and decreased levels of cell apoptosis, which were accompanied by reduced levels of cleaved caspase-3 and Bax expression, and increased levels of Bcl-2, HK2, MDR1, and B7H3 expression. Blocking TNF-α in GCMSCs-CM, B7H3 knockdown or the use of 2-DG, a key enzyme inhibitor of glycolysis in GC cells suppressed the OXA and PTX resistance of GC cells that had been treated with GCMSCs-CM. This study shows that GCMSCs-CM derived TNF-α could upregulate the expression of B7H3 of GC cells to promote tumor chemoresistance. Our results provide a new basis for the treatment of GC.


Assuntos
Células-Tronco Mesenquimais , Neoplasias Gástricas , Humanos , Linhagem Celular Tumoral , Proliferação de Células , Resistencia a Medicamentos Antineoplásicos/genética , Quimioterapia Combinada , Glicólise , Hansenostáticos/farmacologia , Células-Tronco Mesenquimais/metabolismo , Oxaliplatina/farmacologia , Oxaliplatina/uso terapêutico , Paclitaxel/farmacologia , Paclitaxel/uso terapêutico , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Fator de Necrose Tumoral alfa/metabolismo
3.
Anal Chim Acta ; 1282: 341927, 2023 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-37923412

RESUMO

BACKGROUND: Soluble programmed death-ligand 1 (sPD-L1) is critically involved in breast cancer recurrence and metastasis. However, the clinical application of highly sensitive sPD-L1 assays remains a challenge due to its low abundance in peripheral blood. To address this issue, for the first time, an enzyme-catalyzed electrochemical aptasensing platform was devised, incorporating covalent organic frameworks-gold nanoparticles-antibody-horseradish peroxidase (COFs-AuNPs-Ab-HRP) and polyethyleneimine-functionalized multiwalled carbon nanotubes (MWCNTs-PEI-AuNPs) for the highly specific and ultrasensitive detection of sPD-L1. RESULTS: MWCNTs-PEI-AuNPs possessed an extensive specific surface area and exhibited excellent electrical conductivity, facilitating the immobilization of aptamer and amplifying the signal. COFs modified with AuNPs not only amplified the electrical signal but also proffered a loading platform for the Ab and HRP. The favorable biocompatibility of COFs contributed to the preservation of enzyme activity and stability. HRP acted in synergy with hydrogen peroxide (H2O2) to catalyze the oxidation of hydroquinone (HQ) to benzoquinone (BQ). Subsequently, BQ underwent electrochemical reduction to HQ, inducing an enzymatic redox cycle that amplified the electrochemical signal and enhanced the sensitivity and selectivity of the detection method. The developed aptasensor displayed a liner range for sPD-L1 identification from 1 pg mL-1 to 100 ng mL-1 and the detection limit reached 0.143 pg mL-1 (S/N = 3). SIGNIFICANCE: Paving the way for clinical application, this strategy detected differences in sPD-L1 in cell supernatants and peripheral blood of breast cancer patients with higher sensitivity compared to commercial sPD-L1 ELISA kit. This work demonstrates significant potential in offering reference information for early diagnosis and disease surveillance of breast cancer.


Assuntos
Aptâmeros de Nucleotídeos , Técnicas Biossensoriais , Neoplasias da Mama , Nanopartículas Metálicas , Estruturas Metalorgânicas , Nanotubos de Carbono , Humanos , Feminino , Antígeno B7-H1 , Neoplasias da Mama/diagnóstico , Ouro , Limite de Detecção , Peróxido de Hidrogênio , Técnicas Biossensoriais/métodos , Peroxidase do Rábano Silvestre , Catálise , Técnicas Eletroquímicas/métodos
4.
Gut Microbes ; 15(1): 2234653, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37448101

RESUMO

Diarrhea is a leading cause of morbidity and mortality in children worldwide and represents a major dysbiosis event. Rotavirus has been recognized as a global leading pathogen of diarrhea. This study is aimed at investigating differences in the gut virome between diarrheal children and healthy controls. In 2018, 76 diarrheal fecal samples and 27 healthy fecal samples in Shanghai and 40 diarrheal fecal samples and 19 healthy fecal samples in Taizhou were collected to investigate the composition of the gut virome. Viral metagenomic analyses revealed that the alpha diversity of the diarrheal virome was not significantly different from that of the healthy virome, and the beta diversity had a significant difference between diarrheal and healthy children. The diarrheal virome was mainly dominated by the families Adenoviridae, Astroviridae, Caliciviridae, and Picornaviridae. Meanwhile, the healthy virome also contains phages, including Microviridae and Caudovirales. The high prevalence of diverse enteric viruses in all samples and the little abundance of Microviridae and Caudovirales in diarrheal groups were identified. The study introduced a general overview of the gut virome in diarrheal children, revealed the compositional differences in the gut viral community compared to healthy controls, and provided a reference for efficient treatments and prevention of virus-infectious diarrhea in children.


Assuntos
Microbioma Gastrointestinal , Infecções por Rotavirus , Rotavirus , Humanos , Criança , Rotavirus/genética , Metagenômica , China/epidemiologia , Diarreia/epidemiologia , Fezes
5.
Gastric Cancer ; 26(5): 691-707, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37300724

RESUMO

BACKGROUND: Anti-PD-1 immunotherapy has emerged as an important therapeutic modality in advanced gastric cancer (GC). However, drug resistance frequently develops, limiting its effectiveness. METHODS: The role of gastric cancer mesenchymal stem cells (GCMSCs) in anti-PD-1 resistance was evaluated in vivo in NPGCD34+ or NCGPBMC xenograft mouse model. In addition, we investigated CD8+T cell infiltration and effector function by spectral cytometry and IHC. The effects of GCMSCs conditional medium (GCMSC-CM) on GC cell lines were characterized at the level of the proteome, secretome using western blot, and ELISA assays. RESULTS: We reported that GCMSCs mediated tolerance mechanisms contribute to tumor immunotherapy tolerance. GCMSC-CM attenuated the antitumor activity of PD-1 antibody and inhibited immune response in humanized mouse model. In GC cells under serum deprivation and hypoxia, GCMSC-CM promoted GC cells proliferation via upregulating PD-L1 expression. Mechanistically, GCMSC-derived IL-8 and AKT-mediated phosphorylation facilitated HK2 nuclear localization. Phosphorylated-HK2 promoted PD-L1 transcription by binding to HIF-1α. What is more, GCMSC-CM also induced lactate overproduction in GC cells in vitro and xenograft tumors in vivo, leading to impaired function of CD8+ T cells. Furthermore, CXCR1/2 receptor depletion, CXCR2 receptor antagonist AZD5069 and IL-8 neutralizing antibody application also significantly reversed GCMSCs mediated immunosuppression, restoring the antitumor capacity of PD-1 antibody. CONCLUSIONS: Our findings reveal that blocking GCMSCs-derived IL-8/CXCR2 pathway decreasing PD-L1 expression and lactate production, improving antitumor efficacy of anti-PD-1 immunotherapy, may be of value for the treatment of advanced gastric carcinoma.


Assuntos
Células-Tronco Mesenquimais , Neoplasias Gástricas , Humanos , Animais , Camundongos , Neoplasias Gástricas/metabolismo , Linfócitos T CD8-Positivos , Antígeno B7-H1/metabolismo , Interleucina-8/metabolismo , Leucócitos Mononucleares/patologia , Receptores de Interleucina-8B/metabolismo , Terapia de Imunossupressão , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Linhagem Celular Tumoral , Microambiente Tumoral
6.
Oncol Rep ; 50(1)2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37264961

RESUMO

Gastric cancer tissue­derived mesenchymal stem cells (GC­MSCs) play a critical role in facilitating gastric cancer metastasis. Recently, circular RNAs (circRNAs) and metabolic reprogramming have been found to be extensively involved in the malignant progression of tumors, including gastric cancer. However, the biological role and potential mechanisms of GC­MSC­derived circRNAs in metabolic reprogramming remain elusive. Herein, the expression profiles of circRNAs and mRNAs were compared between GC­MSCs and bone marrow­derived MSCs (BM­MSCs) using microarray analysis. circ_0024107 was identified to mediate GC­MSCs to promote gastric cancer lymphatic metastasis by inducing fatty acid oxidation (FAO) metabolic reprogramming. Mechanistically, circ_0024107 served as a sponge of miR­5572 and miR­6855­5p to elicit the FAO metabolic reprograming of GC­MSCs by upregulating carnitine palmitoyltransferase 1A (CPT1A). In addition, GC­MSCs promoted metastasis which was dependent on the induction of FAO in gastric cancer cells mediated by circ_0024107. The circ_0024107/miR­5572/6855­5p/CPT1A axis was deregulated in gastric cancer tissues and GC­MSCs, and was associated with lymph node metastasis and the prognosis of patients with gastric cancer. Taken together, the findings of the present study suggest the crucial role of FAO metabolic reprogramming mediated by GC­MSC­derived circ_0024107 in synergistically promoting gastric cancer lymphatic metastasis via miR­5572/6855­5p­CPT1A signaling; this suggests that circ_0024107 may be an attractive target for gastric cancer intervention.


Assuntos
MicroRNAs , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/patologia , Metástase Linfática/genética , RNA Circular/genética , RNA Circular/metabolismo , Carnitina O-Palmitoiltransferase/genética , Carnitina O-Palmitoiltransferase/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Ácidos Graxos/genética , Regulação Neoplásica da Expressão Gênica , Proliferação de Células/fisiologia , Linhagem Celular Tumoral
7.
Int Immunopharmacol ; 119: 110121, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37044033

RESUMO

Gastric cancer associated mesenchymal stem cells (GC-MSCs) have been demonstrated to promote gastric cancer progression in a paracrine manner. IL-8 is highly secreted by GC-MSCs and is crucial for their oncogenic function. However, the mechanism underlying the modulation of IL-8 secretion by GC-MSCs has not been well elucidated. In this study, Shbio-human ceRNA array was used to identify dysregulated mRNAs and circRNAs between GC-MSCs and bone marrow derived mesenchymal stem cells (BM-MSCs). IL-8 was validated to be a critical paracrine cytokine for GC-MSCs promoting migration and invasion of gastric cancer cells. circ_0073453 was identified as a novel GC-MSC-derived circRNA which acted as a sponge of miR-146a-5p, thus increasing IL-8 expression and secretion to promote gastric cancer cell metastasis. Furthermore, circ_0073453 modulated IL-8 secretion by GC-MSCs to enhance gastric cancer cells PD-L1 expression to resist cytotoxic CD8+ T cell-killing. circ_0073453/miR-146a-5p/IL-8 axis was deregulated in gastric cancer tissues and associated with prognosis depending on MSC abundance in cancer tissues. Taken together, our findings suggest that circ_0073453/miR-146a-5p/IL-8 axis is critical for GC-MSCs promoting gastric cancer progression. Hence, hsa_circ_0073453 may be a potential therapeutic target for gastric cancer.


Assuntos
MicroRNAs , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/patologia , MicroRNAs/genética , MicroRNAs/metabolismo , Interleucina-8/genética , Linhagem Celular Tumoral , RNA Circular/genética , RNA Circular/metabolismo , Proliferação de Células/genética
8.
Cell Signal ; 106: 110639, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36842523

RESUMO

Chemotherapy resistance in advanced gastric cancer (GC) patients has largely limited the effectiveness of therapy, resulting in disease recurrence and poor prognosis. Gastric cancer derived mesenchymal stem cells (GCMSC) are widely believed to promote GC invasion, metastasis and immune escape via up-regulating programmed death ligand 1 (PD-L1). However, the mechanism by which PD-L1 mediated by GCMSC might regulate the chemoresistance is unknown in GC. Herein, higher half maximal inhibitory concentrations (IC50) and less apoptotic rate were observed in GCMSC conditioned medium (GCMSC-CM) treated GC cells exposed to cisplatin (DDP), along with high expression of multi-drug resistance 1 (MDR1) and DNA repair related genes such as Rad51. The knockdown of PD-L1 reversed the increase of Rad51 mediated by GCMSC-CM, resulting in the increased sensitivity of GC cells to DDP. In addition, inhibition of heat shock protein 90 (HSP90) regulated the expression of PD-L1 and Rad51, revealing the important role of HSP90 in GCMSC-CM mediated DDP resistance. Consistent with the observations in vitro, analysis of patient samples and xenograft models further confirmed that reduction of PD-L1 or HSP90 weakened DDP tolerance mediated by GCMSC-CM, along with decrease of Rad51 and MDR1. In conclusion, we demonstrated that GCMSC-CM enhanced DDP resistance in GC cells through regulating PD-L1-Rad51. It is the first to report this particular mechanism of DDP resistance induced by GCMSC in GC, suggesting a potential therapeutic targets for DDP resistant GC cells.


Assuntos
Células-Tronco Mesenquimais , Neoplasias Gástricas , Humanos , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Neoplasias Gástricas/metabolismo , Antígeno B7-H1/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Linhagem Celular Tumoral , Reparo do DNA , Células-Tronco Mesenquimais/metabolismo , Regulação Neoplásica da Expressão Gênica , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo
9.
Exp Cell Res ; 424(1): 113492, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36702194

RESUMO

Lactate extensively involves in gastric cancer (GC) progression, such as suppressing immune cells function and facilitating tumor angiogenesis. However, it remains unclear whether lactate promotes tumor progression by interacting with mesenchymal stem cells (MSCs), one of the major stroma components in GC. Here, we investigated the influence of lactate on the phenotype and function of MSCs. The migration of MSCs and the expression of several CAF markers in MSCs after lactate treatment were detected. We also evaluated the effect of lactate-primed MSCs on GC cells migration, proliferation, and programmed death ligand 1 (PD-L1) expression. It was found that lactate significantly activated MSCs, and increased fibroblast activation protein (FAP) expression via monocarboxylate transporter 1 (MCT1)/transforming growth factor-beta 1 (TGF-ß1) signaling. In addition, lactate-primed MSCs promoted GC cells migration and proliferation via PD-L1. Inhibiting MCT1 by AZD3965 abrogated lactate induced FAP expression and tumor-promoting potential of MSCs. Therefore, targeting MCT1/TGF-ß1/FAP axis in MSCs may serve as a potential strategy to restrain GC development.


Assuntos
Células-Tronco Mesenquimais , Neoplasias Gástricas , Humanos , Fator de Crescimento Transformador beta1/farmacologia , Fator de Crescimento Transformador beta1/metabolismo , Antígeno B7-H1/metabolismo , Neoplasias Gástricas/patologia , Ácido Láctico/farmacologia , Ácido Láctico/metabolismo , Células-Tronco Mesenquimais/metabolismo , Proliferação de Células
10.
Ageing Res Rev ; 85: 101864, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36707035

RESUMO

Osteoarthritis (OA), a common cause of chronic articular cartilage degeneration, is the main cause of disability in older adults and severely affects quality of life. Multiple factors are involved in the pathogenesis of OA, resulting in imbalance in the homeostasis of the joint cavity microenvironment, which exacerbates the disease. Because of the deficiency of blood vessels and nerves in cartilage, existing therapies to promote cartilage healing are relatively ineffective. Mesenchymal stem cell (MSC)-related therapies have achieved positive outcomes for the treatment of OA, and these beneficial effects have been confirmed to be largely mediated by extracellular vesicles (EVs). MSC-derived EVs (MSC-EVs) have been demonstrated to participate in the regulation of chondrocyte function, to have anti-inflammatory and immunomodulatory effects, and to alleviate metabolic disorders of the extracellular matrix, thereby slowing the progression of OA. In addition, engineered MSC-EVs can enrich therapeutic molecules and optimize administration to enhance their therapeutic effects on OA. A thorough understanding of the endogenous properties of EVs and related engineering strategies could help researchers develop more precise control therapy for OA.


Assuntos
Cartilagem Articular , Vesículas Extracelulares , Células-Tronco Mesenquimais , Osteoartrite , Humanos , Idoso , Qualidade de Vida , Osteoartrite/metabolismo , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/patologia , Cartilagem/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Homeostase , Cartilagem Articular/metabolismo
11.
World J Gastroenterol ; 28(25): 2920-2936, 2022 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-35978873

RESUMO

BACKGROUND: Colorectal cancer (CRC) is one of the most common malignancies worldwide. Given its insidious onset, the condition often already progresses to advanced stage when symptoms occur. Thus, early diagnosis is of great significance for timely clinical intervention, efficacy enhancement, and prognostic improvement. Featuring high throughput, fastness, and rich information, next generation sequencing (NGS) can greatly shorten the detection time, which is a widely used detection technique at present. AIM: To screen specific genes or gene combinations in fecal DNA that are suitable for diagnosis and prognostic prediction of CRC, and to establish a technological platform for CRC screening, diagnosis, and efficacy monitoring through fecal DNA detection. METHODS: NGS was used to sequence the stool DNA of patients with CRC, which were then compared with the genetic testing results of the stool samples of normal controls and patients with benign intestinal disease, as well as the tumor tissues of CRC patients. Specific genes or gene combinations in fecal DNA suitable for diagnosis and prognostic prediction of CRC were screened, and their significances in diagnosing CRC and predicting patients' prognosis were comprehensively evaluated. RESULTS: High mutation frequencies of TP53, APC, and KRAS were detected in the stools and tumor tissues of CRC patients prior to surgery. Contrastively, no pathogenic mutations of the above three genes were noted in the postoperative stools, the normal controls, or the benign intestinal disease group. This indicates that tumor-specific DNA was detectable in the preoperative stools of CRC patients. The preoperative fecal expression of tumor-associated genes can reflect the gene mutations in tumor tissues to some extent. Compared to the postoperative stools and the stools in the two control groups, the pathogenic mutation frequencies of TP53 and KRAS were significantly higher for the preoperative stools (χ 2 = 7.328, P < 0.05; χ 2 = 4.219, P < 0.05), suggesting that fecal TP53 and KRAS genes can be used for CRC screening, diagnosis, and prognostic prediction. No significant difference in the pathogenic mutation frequency of the APC gene was found from the postoperative stools or the two control groups (χ 2 = 0.878, P > 0.05), so further analysis with larger sample size is required. Among CRC patients, the pathogenic mutation sites of TP53 occurred in 16 of 27 preoperative stools, with a true positive rate of 59.26%, while the pathogenic mutation sites of KRAS occurred in 10 stools, with a true positive rate of 37.04%. The sensitivity and negative predictive values of the combined genetic testing of TP53 and KRAS were 66.67% (18/27) and 68.97%, respectively, both of which were higher than those of TP53 or KRAS mutation detection alone, suggesting that the combined genetic testing can improve the CRC detection rate. The mutation sites TP53 exon 4 A84G and EGFR exon 20 I821T (mutation start and stop positions were both 7579436 for the former, while 55249164 for the latter) were found in the preoperative stools and tumor tissues. These "undetected" mutation sites may be new types of mutations occurring during the CRC carcinogenesis and progression, which needs to be confirmed through further research. Some mutations of "unknown clinical significance" were found in such genes as TP53, PTEN, KRAS, BRAF, AKT1, and PIK3CA, whose clinical values is worthy of further exploration. CONCLUSION: NGS-based fecal genetic testing can be used as a complementary technique for the CRC diagnosis. Fecal TP53 and KRAS can be used as specific genes for the screening, diagnosis, prognostic prediction, and recurrence monitoring of CRC. Moreover, the combined testing of TP53 and KRAS genes can improve the CRC detection rate.


Assuntos
Neoplasias Colorretais , Sequenciamento de Nucleotídeos em Larga Escala , Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , DNA , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Mutação , Proteínas Proto-Oncogênicas p21(ras)/genética
12.
Medicine (Baltimore) ; 101(25): e29203, 2022 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-35758348

RESUMO

BACKGROUND: Cumulative evidence suggests that A-kinase interacting protein 1 (AKIP1) plays an important role in tumor progression. However, the prognostic value of AKIP1 expression in various cancers remains unclear. Here, we conducted a meta-analysis to evaluate the prognostic value of AKIP1 expression in patients with cancer. METHODS: The PubMed, Web of Science, EMBASE, CNKI, and Wanfang databases were systematically searched to identify studies in which the effect of AKIP1 expression on prognosis (overall survival or disease-free survival) was investigated. Hazard ratios (HRs) with 95% confidence intervals (CIs) were combined to assess the effect of AKIP1 expression on patient survival. Odds ratios (ORs) with 95% CIs were pooled to estimate the association between AKIP1 expression and clinicopathological characteristics of patients with cancer. RESULTS: Nineteen eligible studies, encompassing 3979 patients, were included in the meta-analysis. AKIP1 expression was negatively associated with overall survival (HR = 1.86, 95% CI: 1.58-2.18, P < .001) and disease-free survival (HR = 1.69, 95% CI: 1.53-1.87, P < .001) in patients with cancer. Moreover, AKIP1 overexpression was positively correlated with adverse clinicopathological features, such as tumor size (OR = 2.22, 95% CI: 1.67-2.94, P < .001), clinical stage (OR = 2.05, 95% CI: 1.45-2.90, P < .001), depth of tumor invasion (OR = 2.98, 95% CI: 2.21-4.02, P < .001), and degree of lymph node metastasis (OR = 2.12, 95% CI: 1.75-2.57, P < .001). CONCLUSIONS: High AKIP1 expression is an unfavorable prognostic biomarker and may serve as a potential therapeutic target in patients with cancer.


Assuntos
Biomarcadores Tumorais , Proteínas Nucleares , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Biomarcadores Tumorais/metabolismo , China , Intervalo Livre de Doença , Humanos , Metástase Linfática , Proteínas Nucleares/metabolismo , Prognóstico
13.
Dis Markers ; 2022: 5470166, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35251373

RESUMO

BACKGROUND: Cell-free DNA (cfDNA) has attracted considerable attention in precision medicine. However, few data are available regarding to the prognostic value of cfDNA variables in CA15-3 normal breast cancer (BC) patients. Here, we aimed at investigating the prognostic value of cfDNA variables including gene mutations in CA15-3 normal BC patients. METHODS: A total of 68 BC patients with normal CA15-3 levels were enrolled. cfDNA concentration and integrity were assessed based on qPCR. cfDNA gene mutations were conducted by using next gene sequencing (NGS). The association between cfDNA variables and the prognosis of patients was analyzed. RESULTS: cfDNA concentration was related to tumor stage (P = 0.002), metastases (P = 0.001), and distant metastases (P < 0.001). The elevated copy number variants (CNV) were found in distant metastasis patients compared with patients without distant metastases (P = 0.008). Nineteen mutant genes were validated in enrolled CA15-3 normal BC patients. Thirty-two patients (47.0%) had single nucleotide variants (SNV), and 13 (19.1%) patients had TP53 mutations (TP53 mut). SNV (P = 0.033) was related to tumor stage, and TP53 mut was related to metastases (P = 0.016) and distant metastases (P = 0.006). In multivariate logistic analysis, cfDNA concentration was associated with metastases (OR = 3.404, 95% CI: 1.074-10.788, P = 0.037) and distant metastases (OR = 13.750, 95% CI: 1.473-128.358, P = 0.021). Cases with high cfDNA levels (>15.6 ng/ml), SNV, and TP53 mut showed worse DFS compared with patients with low cfDNA levels (P < 0.001), without SNV (P = 0.002) and with TP53 wildtype (P < 0.001), respectively. In the multivariate Cox proportional hazard model, cfDNA concentration was an independent predictor of poor survival (HR = 5.786, 95% CI: 1.101-30.407, P = 0.038). CONCLUSIONS: Assessment of cfDNA concentration, CNV, SNV, and TP53 mut could be useful in predicting prognosis for CA15-3 normal BC patients. The cfDNA concentration was an independent predictor prognostic factor in CA15-3 normal BC patients.


Assuntos
Neoplasias da Mama , Ácidos Nucleicos Livres/genética , Variações do Número de Cópias de DNA/genética , Mutação/genética , Prognóstico , Biomarcadores Tumorais/genética , Neoplasias da Mama/patologia , Feminino , Humanos , Pessoa de Meia-Idade , Mucina-1
14.
Dis Markers ; 2022: 8443932, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35126792

RESUMO

METHODS: PubMed, Web of Science, Embase, CNKI, and Wanfang databases were thoroughly searched for eligible studies, in which the relationship between SPHK1 expression and cancer prognosis was evaluated. Hazard ratios (HRs) and 95% confidence intervals (CIs) were pooled to estimate the impact of SPHK1 expression on cancer patients' survival. Odds ratios (ORs) and 95% CIs were combined to assess the association between SPHK1 expression and clinicopathological characteristics of cancer patients. The certainty of evidence was evaluated by Grading of Recommendations, Assessment, Development, and Evaluation (GRADE) criteria. RESULTS: Thirty studies comprising 32 cohorts with 5965 patients were included in this meta-analysis. The outcomes indicated that elevated SPHK1 expression was associated with worse overall survival (OS) (HR = 1.71, 95% CI: 1.45-2.01, P < 0.001) and disease-free survival (DFS) (HR = 1.34, 95% CI: 1.13-1.59, P = 0.001). What is more, SPHK1 overexpression was significantly correlated with certain phenotypes of tumor aggressiveness, such as clinical stage (OR = 2.07, 95% CI: 1.39-3.09, P < 0.001), tumor invasion (OR = 2.16, 95% CI: 1.47-3.18, P < 0.001), lymph node metastasis (OR = 2.04, 95% CI: 1.71-2.44, P < 0.001), and distant metastasis (OR = 3.16, 95% CI: 2.44-4.09, P < 0.001). The quality of the evidence for both OS and DFS was low. CONCLUSIONS: Increased SPHK1 expression is related to poor prognosis in human cancers and may serve as a promising prognostic marker and therapeutic target for malignant patients. However, conclusions need to be treated with caution because of lack of high quality of evidence.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Biomarcadores Tumorais/metabolismo , Neoplasias/diagnóstico , Neoplasias/metabolismo , Neoplasias/mortalidade , Humanos , Prognóstico
15.
Medicine (Baltimore) ; 101(2): e28541, 2022 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-35029210

RESUMO

INTRODUCTION: Pantoea dispersa belongs to the genus Pantoea, which is isolated from Enterobacteriaceae. It has been reported to cause some kinds of infections, but there are few detailed studies on it, especially its characteristics and identification methods, which has caused a lot of trouble in clinical work. PATIENT CONCERNS: A 51-year-old Chinese man was admitted to our hospital with a 7-hour history of progressive abdominal pain. He was previously diagnosed with liver cirrhosis secondary to chronic hepatitis B infection and hepatocellular carcinoma. An emergency hepatic artery embolization for hemostasis was performed under local anesthesia. Forty-eight hours later, the patient presented sudden onset of high fever up to 39.0 °C and chill. DIAGNOSIS: Morphological and phenotypic profiles were performed for preliminary identification for P dispersa. The biochemical features were obtained by VITEK 2 Test Kit. Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry analysis and 16S ribosomal RNA sequencing were performed to accurately identify P dispersa. INTERVENTION: Antibiotic therapy of intravenous ceftazidime was started empirically. The antibiotic treatment was switched to intravenous cefepime at the same time because of suspected ceftazidime treatment failure and microbiological sensitivity. OUTCOMES: The patient remained afebrile, and the second blood culture results were negative. Chest X-ray was normal as well. In order to control the progression of the hepatic lesion, transarterial chemoembolization was performed under local anesthesia. After completion of 14 days of antibiotic treatment, the patient was discharged with no signs of recurrence. CONCLUSION: P dispersa, a gram-negative bacterium rod, were facultative anaerobic, which displayed yellow pigmentation, round, raised, smooth on culture plates. Conventional analysis was difficult to complete its identification. With biochemical tests, matrix-assisted laser desorption/ionization time-of-flight mass spectrometry analysis and 16S ribosomal RNA sequencing, P dispersa can be accurately identified. It will help physicians understand the related clinical manifestations and make timely and effective treatment for patients.


Assuntos
Antibacterianos/uso terapêutico , Carcinoma Hepatocelular/tratamento farmacológico , Ceftazidima/uso terapêutico , Neoplasias Hepáticas/tratamento farmacológico , Pantoea/isolamento & purificação , Ruptura Espontânea/tratamento farmacológico , Quimioembolização Terapêutica , Humanos , Masculino , Pessoa de Meia-Idade , Pantoea/genética , RNA Ribossômico 16S/genética , Ruptura Espontânea/microbiologia , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
16.
Biomark Med ; 15(14): 1299-1308, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34544268

RESUMO

There has been a gradual increase in the incidence of colorectal cancer (CRC) in recent years. Most patients lack obvious early symptoms, but are commonly in mid and advanced stages when the symptoms become evident, with rather high mortalities. Early diagnosis, treatment and recurrence monitoring are crucial to improving the recovery rate of CRC. Studies have shown that tumor-related genes can be detected in the feces of CRC patients. Owing to non-invasiveness, convenient sampling and continuous dynamic monitoring, fecal gene detection may be applicable to CRC screening, diagnosis, prognostic assessment and recurrence monitoring. Herein, we review the research advances in fecal gene detection for CRC diagnosis.


Assuntos
Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/genética , Fezes/citologia , Detecção Precoce de Câncer/métodos , Humanos , Taxa de Sobrevida
17.
Int J Immunopathol Pharmacol ; 35: 20587384211026786, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34190613

RESUMO

Macrophage plays a critical part in host defense, tissue repair, and anti-inflammation; Macrophage reprogramming is responsible for disease development or regression. We aimed to clarify the effect of sinomenine-4-hydroxy-palmitate (C16), on macrophage reprogramming and anti-inflammatory in endotoxemia model. According to a structure modification of SIN (Sinomenine), C16 was found. Then, based on the endotoxin model, the mice liver and kidney toxicity was evaluated and serum cytokines level of IL-6 (Interleukin-6), TNF-α (Tumor necrosis factor-α), and IL-1ß (Interleukin-1ß) were measured by ELISA (Enzyme linked immunosorbent assay). Then, we confirmed the effect of C16 on macrophages reprogramming, we used the flow cytometry to test the effect of C16 on macrophages apoptosis in vitro. Then, iNOS (Inducible nitric oxide synthase), M1-type related cytokines, such as IL-1ß, TNF-α, and M2-type related cytokines, such as Arg-1 (Arginase-1), CD206, Fizz1, and Ym1 was detected, which expressed in ANA-1 and primary peritoneal macrophages. To further explore the molecular mechanism of C16 in reprogramming of macrophages from M1 toward M2 phenotype, the expression of STAT1 (signal transducer and activator of Transcription 1), STAT3, ERK1/2 (extracellular signal regulated kinase1/2), AKT, p38, and its corresponding phosphorylation were determined by western blot. Our results demonstrated that C16 improved the survival rate of LPS- (lipopolysaccharide) challenged mice and decreased the inflammatory cytokines expression; After C16 treatment, the expression of M1 phenotype correlation factors decreased significantly, while the expression of M2 phenotype correlation factors increased significantly at different levels compared with normal group. It indicated that C16 reprogram macrophages phenotype from M1 toward M2 following LPS stimulus. Furthermore, the results also showed that C16 showed anti-inflammatory effect by inhibiting LPS-induced p38, AKT and STAT1 phosphorylation and contributing ERK1/2 activation. C16 promoted macrophage reprogramming toward M2-like phenotype via p-p38/p-AKT or STAT1 signals pathway and C16 might be a valid candidate for inflammatory disease.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Endotoxemia/prevenção & controle , Macrófagos/imunologia , Morfinanos/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Citocinas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Nefropatias/induzido quimicamente , Nefropatias/tratamento farmacológico , Lipopolissacarídeos , Ativação de Macrófagos , Macrófagos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Transdução de Sinais/efeitos dos fármacos , Análise de Sobrevida
18.
Mol Biol Rep ; 48(1): 611-621, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33389529

RESUMO

Previous research has demonstrated a correlation between elevated expression of Fos-related antigen 1 (FRA-1) and malignancies. Nevertheless, the role of FRA-1 in Helicobacter pylori infected gastric cancer cells remains vague. Our study aims to investigate whether FRA-1 plays a role in the apoptosis of MGC-803 induced by H. pylori and possible mechanisms. MGC-803 cells were used in vitro to establish a cell model of H. pylori infection. After stimulation with H. pylori, the expression of FRA-1 was increased in MGC-803 cells. H. pylori infection promoted the apoptosis of MGC-803 cells, and led to cell cycle arrest and increased oxidative stress levels. Furthermore, the knockdown of FRA-1 reinforced these changes. H. pylori decreased the expression of Bcl2, Caspase3 and Caspase9, while increased the level of BAX, Cleaved-Caspase3 and Cleaved-Caspase9; in addition, it led to the decrease of major proteins in Ras/Erk and PI3K/AKT signaling pathway. As expected, these changes were augmented by FRA-1 knockdown. Our results demonstrated that high expression of FRA-1 induced by H. pylori suppresses apoptosis in MGC-803 cells which may be regulated by oxidative stress and cycle arrest through caspase family, Ras/Erk and PI3K/AKT signaling pathway.


Assuntos
Proliferação de Células/genética , Infecções por Helicobacter/genética , Proteínas Proto-Oncogênicas c-fos/genética , Neoplasias Gástricas/genética , Apoptose/genética , Caspase 3/genética , Caspase 9/genética , Pontos de Checagem do Ciclo Celular/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/genética , Infecções por Helicobacter/complicações , Infecções por Helicobacter/microbiologia , Infecções por Helicobacter/patologia , Helicobacter pylori/patogenicidade , Humanos , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Transdução de Sinais/genética , Neoplasias Gástricas/complicações , Neoplasias Gástricas/microbiologia , Neoplasias Gástricas/patologia
19.
Biomark Med ; 13(8): 685-695, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31161775

RESUMO

Esophageal carcinoma (EC) is one of the most common human digestive tract tumors, with high morbidity and mortality. It is necessary to elucidate the mechanism of cancer progression and seek early EC diagnostic markers for prompt detection and intervention. Exosomes are membrane nanovesicles secreted from many nucleated cells, 30-100 nm in diameter, containing various proteins, lipids and nucleic acids. They exist in peripheral blood, urine, ascites and other body fluids, widely engaged with intercellular material exchange and signal communication. Exosomes secreted from EC cells or tissues conduct important functions in tumor growth and progression. The detection and analysis of tumor-derived or tumor-associated exosomes has potential for EC early diagnosis and prognosis assessment. In the present paper, the exosomes' biological behaviors, isolation, detection and functions in EC progression - using as potential biomarkers for EC diagnosis or prognosis - are reviewed.


Assuntos
Neoplasias Esofágicas/patologia , Exossomos , Biomarcadores Tumorais/metabolismo , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/metabolismo , Exossomos/metabolismo , Exossomos/patologia , Humanos
20.
Anal Cell Pathol (Amst) ; 2018: 8941908, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29854577

RESUMO

Our previous findings showed a good therapeutic effect of the combination of suicide gene HSV-TK, nuclide 131I, and magnetic fluid hyperthermia (MFH) on hepatoma by using magnetic nanoparticles as linkers, far better than any monotherapy involved, with no adverse effects. This combination therapy might be an eligible strategy to treat hepatic cancer. However, it is not clear how the combination regimen took the therapeutic effects. In the current study, to explore the possible mechanisms of radionuclide-gene therapy combined with MFH to treat hepatoma at tissue, cellular, and molecular levels and to provide theoretical and experimental data for its clinical application, we examined the apoptosis induction of the combination therapy and investigated the expression of the proteins related to apoptosis such as survivin, livin, bcl-2, p53, and nucleus protein Ki67 involved in cell proliferation, detected VEGF, and MVD involved in angiogenesis of tumor tissues and analyzed the pathologic changes after treatment. The results showed that the combination therapy significantly induced the hepatoma cell apoptosis. The expression of survivin, VEGF, bcl-2, p53, livin, Ki67, and VEGF proteins and microvascular density (MVD) were all decreased after treatment. The therapeutic mechanisms may be involved in the downregulation of Ki67 expression leading to tumor cell proliferation repression and inhibition of survivin, bcl-2, p53, and livin protein expression inducing tumor cell apoptosis, negatively regulating VEGF protein expression, and reducing vascular endothelial cells, which results in tumor angiogenesis inhibition and microvascular density decrease and tumor cell necrosis. These findings offer another basic data support and theoretical foundation for the clinical application of the combination therapy.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Carcinoma Hepatocelular/terapia , Ganciclovir/uso terapêutico , Hipertermia Induzida , Radioisótopos do Iodo/química , Neoplasias Hepáticas/terapia , Nanosferas/química , Timidina Quinase/metabolismo , Anticorpos Monoclonais/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/irrigação sanguínea , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/ultraestrutura , Proliferação de Células/efeitos dos fármacos , Ganciclovir/farmacologia , Células Hep G2 , Humanos , Proteínas Inibidoras de Apoptose/metabolismo , Neoplasias Hepáticas/irrigação sanguínea , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/ultraestrutura , Microvasos/efeitos dos fármacos , Microvasos/patologia , Necrose , Proteínas de Neoplasias/metabolismo , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/patologia , Simplexvirus/metabolismo , Survivina , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA