Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Hematology ; 29(1): 2293494, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38095304

RESUMO

OBJECTIVES: Acute undifferentiated leukemia (AUL) is a clinical rare leukemia with an overall poor prognosis. Currently, there are no well-established treatment guidelines for AUL, further exploration of optimal treatment options is now required. METHODS: We report an AUL patient who was complicated by a NRAS mutation and del5q was admitted to our hospital and we present the clinical features. In addition, we conducted a literature review. RESULTS: The "VA" scheme combines agents Venetoclax and Azacitidine that have synergistic therapeutic effect with a tolerable safety profile. There is no previous report of the "VA" scheme employed in AUL treatment. An AUL patient who was complicated by a NRAS mutation and del5q was admitted to our hospital. The "VA" scheme was administrated, and complete remission (CR) was achieved at the end of the first cycle. The patient then underwent HLA-identical sibling allogeneic hematopoietic stem cell transplantation. DISCUSSION: The "VA" scheme has been extensively used in AML treatment, but its application in AUL treatment has not yet been reported. This study is the first to report an AUL patient treated with the "VA" scheme and achieved CR. Our result preliminarily suggested the effectiveness and safety of the "VA" scheme in AUL treatment, but validation is required in more clinical samples. The "VA" scheme provides a new treatment option for AUL patients and deserves further clinical promotion.


Assuntos
Leucemia Mieloide Aguda , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Compostos Bicíclicos Heterocíclicos com Pontes/uso terapêutico , Sulfonamidas/uso terapêutico , Azacitidina/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico
2.
Front Biosci (Landmark Ed) ; 28(9): 195, 2023 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-37796705

RESUMO

BACKGROUND: Acute myeloid leukemia (AML) is a recurrence-prone hematologic malignancy. The advent of molecularly targeted therapies provides new opportunities to enhance the effectiveness of AML treatments. Venetoclax, a selective inhibitor of the anti-apoptotic protein Bcl-2, has shown promising results; however, resistance often arises due to elevated expression of the Mcl-1 protein, among other factors. Overcoming this resistance to improve therapeutic outcomes is a pressing issue that requires further investigation. Studies have demonstrated that oridonin, by inhibiting AKT signaling that regulates Mcl-1 expression, can effectively suppress tumor cell growth. This study aims to investigate whether oridonin can synergistically enhance the anti-leukemic effects of venetoclax and explore the underlying mechanisms behind this effect. METHODS: In vitro experiments were performed to evaluate the effects of the combination of oridonin and venetoclax on AML cell proliferation, apoptosis, cell cycle distribution, and mitochondrial membrane potential. Transcriptome sequencing was used to elucidate the molecular mechanisms underlying the synergistic induction of AML cell apoptosis by the combination therapy. Western blotting and reverse transcription quantitative polymerase chain reaction (RT-qPCR) techniques were used to validate the findings. Additionally, an AML mouse model was established to observe the synergistic anti-AML effects of venetoclax combined with oridonin in vivo. RESULTS: Both venetoclax and oridonin individually exhibited inhibitory effects on AML cell proliferation, resulted in cell cycle arrest, and induced cell apoptosis. Moreover, combination of the two drugs resulted in a synergistic effect. We also observed that oridonin inhibited AKT phosphorylation, upregulated the expression of Bim and Bax proteins, facilitated Mcl-1 degradation, and enhanced the apoptotic effects of venetoclax in AML cells. Finally, in vivo experiments demonstrated that the combination of oridonin and venetoclax effectively inhibited the growth of AML xenograft tumors in mice and prolonged the survival time of tumor-bearing mice. CONCLUSIONS: Oridonin and venetoclax synergistically promote AML cell apoptosis by inhibiting AKT signaling.


Assuntos
Leucemia Mieloide Aguda , Proteínas Proto-Oncogênicas c-akt , Animais , Humanos , Camundongos , Apoptose , Linhagem Celular Tumoral , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo
3.
Environ Toxicol ; 38(10): 2487-2498, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37466197

RESUMO

Nanoplastics (NPs) has become a worrying serious environmental problem. However, the toxicological effects and mechanisms of NPs on hematopoiesis are still unknown. To this end, male C57BL/6J mice were directly exposed to the serial concentration gradient of polystyrene NPs (PSNPs, 0, 30, 60, and 120 µg d), respectively, for 42 days by intragastric administration. Results show that PSNPs were clearly visible in bone tissues, meanwhile, induced the count of major blood indicators (WBC, RBC, and LYM) decreased. H&E staining displayed that exposed to PSNPs can cause hematopoietic damage of BM and extramedullary hematopoiesis in spleen. Flow cytometry result show that the proportion of LSK represented a dose-dependent significantly decreased after PSNPs exposure. Further research found that PSNPs can cause the systemic oxidative stress occurs manifested as MDA accumulated. In addition, as the dose of PSNPs increased, the fluorescence intensity of Keap1 and p53 in femur sections gradually increased, meanwhile, the expression of cell oxeiptosis signal pathway Keap1/PGAM5/AIFM1 and the cell senescence signal pathway p53/p21 was all increased, markedly. Overall, our study demonstrated that PSNPs exposure caused oxidative stress, potentially resulting in cell oxeiptosis and senescence to develop haematotoxicity in C57BL/6J mice.


Assuntos
Microplásticos , Poliestirenos , Animais , Camundongos , Masculino , Poliestirenos/toxicidade , Proteína 1 Associada a ECH Semelhante a Kelch , Camundongos Endogâmicos C57BL , Proteína Supressora de Tumor p53 , Fator 2 Relacionado a NF-E2
4.
Food Chem Toxicol ; 177: 113817, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37164248

RESUMO

Nanoplastics is a major environmental concern and may cause potential harm to organisms. Previous studies have found that exposure to nanoplastics inhibited hematopoietic function, however, the effect of polystyrene nanoplastics (PSNPs) on the human CD34+ hematopoietic stem/progenitor cells (HSPCs) and its underlying mechanism remains unknown. In this study, the toxic effects were evaluated and the metabolites changes were systematically analyzed using the metabolomics study in combination with multivariate statistical analysis in HSPCs with PSNPs treatment. The results show that PSNPs could be uptake by cells, significantly decrease cell viability and cause cell membrane damage manifested as increased LDH release in cellular supernatant. Besides, the colony formation assay shows that PSNPs exposure can inhibit the proliferation and differentiation of HSPCs. Meanwhile, we found that PSNPs disturbed the metabolic activity, including amino acids, SCFAs, organic acids, fatty acids and carbohydrates, and mainly affect citrate cycle (TCA cycle) metabolism pathway. Those findings are helpful in evaluating the toxicity mechanisms and providing guidance in the selection of potential metabolism-related biomarkers of hematopoietic damage caused by nanoplastics exposure.


Assuntos
Microplásticos , Poliestirenos , Humanos , Poliestirenos/toxicidade , Poliestirenos/metabolismo , Microplásticos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Sobrevivência Celular , Metabolômica
5.
Ann Transl Med ; 10(22): 1236, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36544631

RESUMO

Background: Currently, there is no satisfactory treatment available for esophageal squamous cell carcinoma (ESCC), and thus, there is a pressing need to develop effective drugs. Chaetoglobosin E, a cytochalasan alkaloid derived from metabolites of Chaetomium madrasense 375, is a chaetoglobosin with intense anti-tumor activity. Therefore, revealing its anti-tumor mechanism for the application of cytochalasans is crucial. Methods: The cytotoxic effect of chaetoglobosin E and cisplatin on esophageal cancer KYSE-30, KYSE-150, and TE-1 cells was detected using cell viability or colony formation assays. The cell cycle, apoptosis, autophagy, invasion, and metastasis were assayed by flow cytometry or western blot. The potential target of chaetoglobosin E was assayed by RNA sequencing (RNA-seq) and large loop prediction software analysis and was assessed by western blot and real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR). The effect of its target on cell pyroptosis was assayed using overexpression and silence experiments. Results: Chaetoglobosin E significantly inhibited the proliferation of KYSE-30, KYSE-150, and TE-1 cells, especially KYSE-30 cells. Our results showed that chaetoglobosin E induced the G2/M phase arrest of KYSE-30 cells, followed by the down-regulation of cyclinB1, CDC2, and p-CDC2, and up-regulation of p21. Moreover, chaetoglobosin E also decreased the anti-apoptotic protein expression of Bcl-2, increased apoptotic expression of Bax, increased autophagy protein expressions of beclin1 and LC3, decreased invasion and metastasis protein expression of E-cadherin, and increased expression of vimentin. The RNA-seq and large loop prediction software analysis results indicated that its potential target might be polo-like kinase 1 (PLK1). Moreover, results also showed that chaetoglobosin E can reverse the PLK1 overexpression plasmid-induced up-regulation of the PLK1 protein. Furthermore, we found that chaetoglobosin E induced pyroptosis via the activation of the gasdermin E (GSDME) protein. Further studies showed that the high expression of PLK1 inactivated the GSDME protein, while the knockdown of PLK1 expression activated the GSDME protein, indicating that chaetoglobosin E induced cell pyroptosis by inhibiting PLK1. Conclusions: This study suggested that chaetoglobosin E may be a novel lead compound to the treatment of ESCC patients by targeting PLK1, and elucidated for the first time that PLK1 was involved in a new pyroptosis mechanism.

6.
Ann Transl Med ; 10(22): 1232, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36544685

RESUMO

Background: Cuproptosis is the recently defined regulatory cell death (RCD) that plays essential roles in tumorigenesis and progression. Long noncoding RNAs (lncRNAs) regulate the gene expression through various means. However, the clinical value of cuproptosis-related lncRNAs in bladder cancer (BLCA) remains poorly described. Methods: We downloaded the transcriptome sequencing data and clinical information from The Cancer Genome Atlas (TCGA) database. Univariate, multivariate, and lasso Cox regression analyses were performed to construct the prognostic risk signature, the predictive accuracy of which was validated in the subsequent independence and stratification analyses. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were used to explore the underlying molecular mechanisms involved in the signature to explore therapeutic vulnerabilities and potential targets in BLCA. Tumor mutational burden (TMB) and tumor immune dysfunction and exclusion (TIDE) were used to estimate the response to immune checkpoint inhibitors (ICIs). We further explored the potential new drug-target candidates based on the half maximal inhibitory concentration for this patient population. Results: Fifteen cuproptosis-related lncRNAs significantly associated with survival were identified to construct the risk signature based on the normalized expression level and regression coefficient of each gene. The patients with BLCA and high-risk scores defined by the signature were associated with worse survival outcomes. The differentially expressed genes (DEGs) between the 2 risk groups had different biological activity. Furthermore, the patients in the low-risk group exhibited a higher TMB index and a lower TIDE score. The sensitivity of multiple antitumor drugs was negatively related to risk score, including AR-42, AS605240, FK866, TAK-715, and tubastatin A, while the sensitivity of some antitumor drugs, such as AMG-706, BX-795, and RO-3306, were positively correlated with risk score. Conclusions: Our study established and verified a novel clinical risk signature with cuproptosis-related lncRNAs that may predict therapy response and prognosis with robust and stable accuracy in patients with BLCA and enhance the personalized management of this patient population.

7.
Transl Androl Urol ; 11(11): 1586-1597, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36507483

RESUMO

Background: The Reporting Items for Practice Guidelines in Healthcare (RIGHT) checklist was developed to improve the reporting quality in clinical practice guidelines (CPGs). CPGs could provide the recommendations for key clinical issues with alternative care options and adherence to them could improve the outcomes. And, high reporting quality CPGs can assist health workers to incorporate the best evidence into the individual practice. There is no evaluation study on the reporting quality of CPGs in bladder cancer (BLCA). This study assessed the reporting quality of CPGs on BLCA and provided new insights for the development of CPGs in this disease. Methods: We conducted a systematic search in multiple literature databases, including PubMed, Wanfang, China National Knowledge Infrastructure (CNKI) and China Biology Medicine (CBM) as well as the medical associations and websites of guideline development organizations. Relevant CPGs published between January 2017 and December 2021 were identified. Four trained investigators independently screened the extracted documents to include all eligible CPGs and evaluated whether the items in the RIGHT checklist were reported in each CPG. Subsequently, the reporting rate of each CPG and item, as well as the mean reporting rate of each domain in the RIGHT checklist was calculated. Results: A total of 23 CPGs related to BLCA were finally included, of which, 22 guidelines were written in English and 1 was published in Chinese. The mean reporting rate of the included CPGs was approximately 65%. The reporting rates of the items in each RIGHT domain were 77% for basic information domain, 75% for recommendations domain, 72% for evidence domain, 69% for background domain, 43% for funding and declaration and management of interest domain, 35% for review and quality assurance domain, and 41% for other information domain. The reporting rate was determined as the mean value in Office Excel 2019. Conclusions: The reporting quality of BLCA CPGs related to the domains of funding and declaration and management of interest domain, review and quality assurance domain, and other information domain is poor and warrants improvement in the future.

8.
Int Immunopharmacol ; 112: 109182, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36058034

RESUMO

Acute myeloid leukemia (AML) is prone to relapse. Targeted therapy with a specific inhibitor of the anti-apoptotic protein Bcl-2 ABT-199 is an effective method for relapsed and refractory patients, but drug resistance is likely, which is primarily related to high Mcl-1 and S100A8 expression. All-trans retinoic acid (ATRA) can inhibit Bcl-2 and Mcl-1 expression. The study purpose was to determine whether ATRA can enhance the antileukemia effect of ABT-199 on AML cells. Our data showed that ATRA combined with ABT-199 exerts a synergistic antileukemic effect by inducing apoptosis and cell cycle arrest in AML. In vivo, combination therapy prolonged the survival of AML xenograft mice. The possible mechanism involves promoting apoptosis through downregulation of S100A8 expression by inhibiting the PI3K/AKT signaling pathway. This study provides a potential treatment strategy and theoretical support for overcoming the clinical ABT-199 resistance problem in AML patients.


Assuntos
Leucemia Mieloide Aguda , Fosfatidilinositol 3-Quinases , Humanos , Camundongos , Animais , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt , Linhagem Celular Tumoral , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/metabolismo , Tretinoína/farmacologia , Tretinoína/uso terapêutico , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo
9.
Ann Transl Med ; 10(8): 490, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35571387

RESUMO

Background: To evaluate whether homoharringtonine (HHT) combined with venetoclax could produce a synergistic anti-acute myeloid leukemia (AML) effect and determine the underlying mechanisms. Methods: The effect of HHT and venetoclax combination on cell viability, apoptosis, and mitochondrial membrane potential was investigated in vitro using AML cell lines and primary cells. High-throughput mRNA sequencing was used to analyze mRNA level changes after the application of HHT and venetoclax on OCI-AML3 cells. Western blotting was used to verify the changes in protein expression within the mitogen-activated protein kinases/extracellular signal-regulated kinase (MAPK/ERK), phosphatidylinositiol 3-kinase (PI3K)/AKT and p53 pathway. The efficacy of HHT and venetoclax in vivo and their effects on survival time were evaluated in a xenograft model established in severe immunodeficiency (NOD/SCID) mice. Results: Venetoclax and HHT synergistically inhibited the proliferation of AML cells, decreased the mitochondrial membrane potential, and promoted AML cell apoptosis in a time- and concentration-dependent manner. Venetoclax combined with HHT increased the expression of the caspase-3, Poly (ADP-ribose) polymerase (PARP), and γH2AX proteins. HHT enhanced the proapoptotic effect of venetoclax by reducing the expression of myeloid cell leukemia sequence 1 (Mcl-1). HHT arrested AML cells in G1 phase of the cell cycle. HHT enhanced the proapoptotic effect of venetoclax by inhibiting the activation of the MAPK/ERK and PI3K/AKT pathways and activating the p53 pathway. In vivo experiments confirmed that the combination of HHT and venetoclax could inhibit the growth of tumors in AML xenotransplanted mice and prolong the survival time of tumor-bearing mice. Conclusions: HHT combined with venetoclax synergistically promoted apoptosis in AML cell lines and primary cells by inhibiting the activation of the MAPK/ERK and PI3K/AKT pathways and activating the p53 pathway.

10.
Ann Transl Med ; 9(18): 1461, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34734013

RESUMO

BACKGROUND: Acute myeloid leukemia (AML) is an aggressive hematologic malignancy. Clinical practice guidelines (CPGs) on the management of AML have great value in clinical practice. However, the reporting quality of CPGs for AML has not yet been evaluated. This is the first study aiming to evaluate the reporting quality of the most recent AML CPGs published worldwide using the Reporting Items for Practice Guidelines in Healthcare (RIGHT) checklist. METHODS: We systematically searched PubMed, Chinese National Knowledge Infrastructure (CNKI), Wanfang, and Chinese Biomedical Literature (CBM) to extract CPGs for AML published between January 2016 and December 2020. Websites for guideline development organizations and medical associations were also searched. Two independent researchers assessed compliance of the guidelines to each of the 35 checklist items and summarized reporting rates for the 7 domains of the RIGHT checklist. RESULTS: We identified 16 guidelines, of which 3 (18.8%) were written in Chinese and 13 (81.3%) were written in English. The average overall reporting rate of the 16 guidelines was 52.9%, and only 7 CPGs (43.8%) had a reporting rate >50%. The average reporting rates of the 7 domains (basic information; background; evidence; recommendations; review and quality assurance; funding, declaration, and management of interests; and other information) were 79.2%, 62.5%, 38.8%, 53.6%, 21.9%, 32.8%, and 43.8%, respectively. For the 35 checklist items, the average reporting rate was 52.9%, and only 16 items had a reporting rate >50%, of which 5 items were reported by all the guidelines. There was 1 item which was not reported by any of the guidelines. CONCLUSIONS: The reporting quality of recently published AML guidelines remains poor. While the recommendations of CPGs have great value in clinical practice, the reporting quality of CPGs for AML still needs to be improved.

11.
Ann Transl Med ; 9(20): 1575, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34790781

RESUMO

BACKGROUND: Acute myeloid leukemia (AML) is a hematological malignancy with a low remission rate and high recurrence rate. Overexpression of the antiapoptotic protein Bcl-2 is associated with a lower overall survival rate in AML patients. Venetoclax (ABT199) is a selective inhibitor of Bcl-2 that has a significant effect in AML, but single-drug resistance often occurs due to the high expression of Mcl-1 protein. Studies have confirmed that chidamide can downregulate the expression levels of Bcl-2 and Mcl-1 and induce apoptosis. METHODS: This study aimed to use AML cell lines and primary cells to study the effects of venetoclax and chidamide combination therapy on AML cell apoptosis, the cell cycle, and changes in related signaling pathways in vitro; establish an AML mouse model to observe the efficacy and survival time of combination therapy in vivo; and analyze the drug effects with multi-omics sequencing technology. The changes in gene and protein expression before and after treatment were examined to clarify the molecular mechanism driving the synergistic effect of the two drugs. RESULTS: (I) Both venetoclax and chidamide promoted apoptosis in AML cell lines and primary cells in a time- and concentration-dependent manner. The effect was further enhanced when the two drugs were combined, and a synergistic effect was observed (combination index <1). (II) At both the mRNA and protein levels, the expression of Mcl-1 was upregulated by venetoclax and downregulated by chidamide, and the expression of Mcl-1 decreased further after combination treatment. (III) Transcriptome sequencing showed that differentially expressed genes in the combination group compared with the venetoclax monotherapy group were mainly enriched in the PI3K-AKT pathway and JAK2/STAT3 pathway. Moreover, qRT-PCR and Western blot confirmed these results. (IV) The combination therapy group exhibited significantly inhibited disease progression and a prolonged survival time among AML mice. CONCLUSIONS: Chidamide combined with venetoclax synergistically promoted apoptosis in AML cell lines and primary cells by inhibiting activation of the PI3K/AKT pathway and JAK2/STAT3 pathway.

12.
Int J Nanomedicine ; 16: 4631-4642, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34262273

RESUMO

PURPOSE: Antibiotic-resistant bacteria are pathogens that have emerged as a serious public health risk. Thus, there is an urgent need to develop a new generation of anti-bacterial materials to kill antibiotic-resistant bacteria. METHODS: Nanosilver-decorated mesoporous organosilica nanoparticles (Ag-MONs) were fabricated for co-delivery of gentamicin (GEN) and nanosilver. After investigating the glutathione (GSH)-responsive matrix degradation and controlled release of both GEN and silver ions, the anti-bacterial activities of Ag-MONs@GEN were systematically determined against several antibiotic-susceptible and antibiotic-resistant bacteria including Escherichia coli, Pseudomonas aeruginosa, Staphylococcus aureus, and Enterococcus faecalis. Furthermore, the cytotoxic profiles of Ag-MONs@GEN were evaluated. RESULTS: The GEN-loaded nanoplatform (Ag-MONs@GEN) showed glutathione-responsive matrix degradation, resulting in the simultaneous controlled release of GEN and silver ions. Ag-MONs@GEN exhibited excellent anti-bacterial activities than Ag-MONs and GEN alone via inducing ROS generation, especially enhancing synergetic effects against four antibiotic-resistant bacteria including Escherichia coli, Pseudomonas aeruginosa, Staphylococcus aureus, and Enterococcus faecalis. Moreover, the IC50 values of Ag-MONs@GEN in L929 and HUVECs cells were 313.6 ± 15.9 and 295.7 ± 12.3 µg/mL, respectively, which were much higher than their corresponding minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) values. CONCLUSION: Our study advanced the development of Ag-MONs@GEN for the synergistic and safe treatment of antibiotic-resistant bacteria.


Assuntos
Resistência Microbiana a Medicamentos/efeitos dos fármacos , Gentamicinas/farmacologia , Glutationa/química , Nanopartículas/química , Dióxido de Silício/química , Prata/química , Animais , Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Liberação Controlada de Fármacos , Sinergismo Farmacológico , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Camundongos , Testes de Sensibilidade Microbiana , Nanopartículas/ultraestrutura
14.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 29(1): 26-31, 2021 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-33554792

RESUMO

OBJECTIVE: To explore the synergistic immunomodulatory mechanism of interferon alpha-1b, interleukin-2 and thalidomide (ITI) regimen on patients with acute myeloid leukemia (AML). METHODS: Sixty eight untreated de novo or relapsed or refractory or maintenance therapy patients with AML admitted in the Affiliated Cancer Hospital of Zhengzhou University and the other 11 medical units from March 2016 to May 2019 were treated with ITI regimen. Peripheral blood specimen per patient was collected into EDTA-K3 anticoagulation vacuum tube before the administration of ITI and 3 months after the treatment; peripheral blood lymphocyte subsets and perforin and Granzyme B expression were analyzed by using flow cytometry; the levels of VEGF, IFN-γ, TNF-α and IL-6 in the plasma were detected by using a cytometric bead array. Thirty-five healthy subjects from the hospital physical examination centre were selected as normal controls. RESULTS: The ratio of CD4+/CD8+ T cells, the percentage of NK cells, the expression of perforin and Granzyme B of NK cells in the peripheral blood of patients with hematological malignancies were lower than those of healthy controls. The level of VEGF, IL-6 and TNF-α in the peripheral plasma were higher than those of the healthy control group, and the difference was statistically significant. The level of IFN-γ was lower, and the difference was not statistically significant. The ratio of CD4+/CD8+ T cells, the percentage of NK cells, the expression of Granzyme B and Perforin of NK cells in peripheral blood were higher after the therapy of thalidomide combined with rhIFNα-1b for 3 months as compared with those before treatment of ITI, the level of the IFN-γ in peripheral plasma was higher while that of VEGF was lower, the difference was statistically significant; after treatment, the ratio of CD3+ CD4+ and CD3+ CD8+ lymphocytes and the level of TNF-α in peripheral blood were higher those that before treatment, IL-6 was lower, while the difference was not statistically significant. CONCLUSION: The ITI regimen can raise the ratio of CD4+/CD8+ T cells and the percentage of natural killer cells, also, can enhance the generation of perforin and granzyme B and the concentration of IFN-γ as well as inhibit the generation of VEGF, suggesting that these activities may enhance the antitumour capacity of patients with AML.


Assuntos
Interleucina-2 , Leucemia Mieloide Aguda , Linfócitos T CD8-Positivos , Humanos , Interferon-alfa , Leucemia Mieloide Aguda/tratamento farmacológico , Perforina , Talidomida
15.
Int J Nanomedicine ; 16: 8323-8334, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34992368

RESUMO

PURPOSE: Photodynamic therapy (PDT) with spatiotemporal controlled and noninvasive advantages has obtained growing attention in cancer treatment. Nevertheless, PDT still suffers from self-aggregation-induced photosensitizer quenching and reactive oxygen species (ROS) scavenging in cancer cells with abundant glutathione (GSH) pools, leading to insufficient performance. METHODS: In this study, we develop a versatile nanocarrier (SSNs) with a disulfide-bond-bridged silica framework for enhanced photo-immunotherapy. Such SSNs spatially confine photosensitizers Ce6 in the matrix to prevent self-aggregation. Under the high GSH level of cancer cells, the disulfide-bond-bridged framework was degradable and triggered the exposure of photosensitizers to oxygen, accelerating the ROS generation during PDT. In addition, GSH depletion via the break of the disulfide-bond increased the ROS level, together resulting in efficient tumor killing outcomes with a considerable immunogenic cell death effect in vitro. Importantly, the SSNs@Ce6 accumulated in the tumor site and exhibited enhanced PDT efficacy with low systemic toxicity in vivo. RESULTS: The GEN-loaded nanoplatform (Ag-MONs@GEN) showed glutathione-responsive matrix degradation, resulting in the simultaneous controlled release of GEN and silver ions. Ag-MONs@GEN exhibited excellent anti-bacterial activities than Ag-MONs and GEN alone, especially enhancing synergetic effects against four antibiotic-resistant bacteria including Escherichia coli, Pseudomonas aeruginosa, Staphylococcus aureus, and Enterococcus faecalis. Moreover, Ag-MONs@GEN showed good biocompatibility on L929 and HUVECS. CONCLUSION: Notably, SSNs@Ce6-mediated PDT completely eradicated 4T1 tumors when combined with the PD-1 checkpoint blockade. Overall, the confinement of photosensitizers in a biodegradable disulfide-bridged-framework provides a promising strategy to unleash the potential of photosensitizers in PDT, especially in combined cancer photo-immunotherapy.


Assuntos
Nanopartículas , Fotoquimioterapia , Linhagem Celular Tumoral , Dissulfetos , Imunoterapia , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico
16.
Autoimmunity ; 51(1): 10-17, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29297233

RESUMO

As a transcription factor, autoimmune regulator (Aire) participates in thymic negative selection and maintains immune tolerance mainly by regulating the ectopic expression of tissue-restricted antigens (TRAs) in medullary thymic epithelial cells (mTECs). Aire is also expressed in dendritic cells (DCs). DCs are professional antigen-presenting cells (APCs) that affect the differentiation of T cells toward distinct subpopulations and participate in the immune response and tolerance, thereby playing an important role in maintaining homeostasis. To determine the role of Aire in maintaining immune tolerance by bone marrow-derived dendritic cells (BMDCs), in the present study we utilized Aire-knockout mice to examine the changes of maturation status and TRAs expression on BMDCs, additionally investigate the differentiation of CD4+ T cells. The results showed that expression of costimulatory molecule and major histocompatibility complex class II (MHC-II) molecule was increased and expression of various TRAs was decreased in BMDCs from Aire-knockout mice. Aire deficiency reduced the differentiation of naïve CD4+ T cells into type 2T helper (Th2) cells and regulatory T cells (Tregs) but enhanced the differentiation of naïve CD4+ T cells into Th1 cells, Th17 cells, and follicular helper T (Tfh) cells. The results demonstrate that Aire expressed by BMDCs plays an important role in the maintenance of homeostasis by regulating TRA expression and the differentiation of T cell subsets.


Assuntos
Células da Medula Óssea , Células Dendríticas , Poliendocrinopatias Autoimunes , Linfócitos T Auxiliares-Indutores , Fatores de Transcrição , Animais , Células da Medula Óssea/imunologia , Células da Medula Óssea/patologia , Células Dendríticas/imunologia , Células Dendríticas/patologia , Camundongos , Camundongos Knockout , Poliendocrinopatias Autoimunes/imunologia , Poliendocrinopatias Autoimunes/patologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/imunologia , Proteína AIRE
17.
Oncol Lett ; 13(5): 3529-3537, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28521455

RESUMO

Toll-like receptor 4 (TLR4) is a pattern recognition receptors, a member of the Toll-like receptor family and it serves a role in innate and acquired immunity. It has previously been reported that TLR4 was overexpressed in a variety of tumor tissues and cells, including colorectal cancer, gastric cancer and ovarian cancer. In the tumor microenvironment, the TLR4 signaling pathway may be activated in order to upregulate forkhead box P3 (Foxp3) expression in regulatory T cells (Tregs), and thus enhance the immunosuppressive function of Tregs. Also, inflammatory cytokine release would be increased, which promotes tumor immune system evasion. Additionally, it has previously been reported that TLR4 activation may induce histone methylation changes at multiple sites. However, the effects of the alterations to histone methylation in the process of TLR4-associated tumor immune system evasion are not currently known. Histone methylation serves a critical role in regulating gene expression. Abnormal histone methylation is closely associated with tumor development and progression. In order to investigate the epigenetic mechanisms underlying Foxp3 regulation by TLR4, the human lung adenocarcinoma cell line A549 was used. In the present study, it was revealed that the expression level of H3K9me1/2 histone lysine demethylase 3A (KDM3A) was significantly increased following TLR 4 activation in the lung adenocarcinoma A549 cell line, whereas silencing of KDM3A expression led to significantly reduced Foxp3 expression under TLR4 regulation. This result suggests that KDM3A participates in TLR4 regulation of Foxp3 transcription. Additional analysis revealed that during nuclear transport of Foxp3, KDM3A may directly bind to the Foxp3 promoter and activate its transcription. This results in increased secretion of Foxp3-downstream inhibitory cytokines, including transforming growth factor-ß1 (TGF-ß1), interleukin 35 (IL-35) and heme oxygenase 1 (HO-1), which have immunosuppressive effects and ultimately facilitate the immune escape of lung cancer cells. From the results, the present study concluded that TLR4 activation promoted the expression of H3K9me1/2 demethylase KDM3A. KDM3A bound directly to the Foxp3 promoter and promoted Foxp3 transcription, thereby inducing the secretion of Foxp3-associated downstream inhibitory cytokines (TGF-ß1, IL-35, and HO-1), ultimately facilitating the immune system evasion of lung adenocarcinoma.

18.
Int Immunopharmacol ; 49: 13-20, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28550730

RESUMO

Autoimmune regulator (Aire) plays an indispensable role in maintaining central immune tolerance by promoting the ectopic expression of tissue-restricted antigens (TRAs) in medullary thymic epithelial cells (mTECs) and dendritic cells (DCs), which lead to the deletion of autoreactive T cells or the induction of Tregs and consequently prevent autoimmune disease development. Curing autoimmune diseases has always been a challenge. DC-based immunotherapy represents a new and effective method to establish tolerance. We attempted to transplant Aire-overexpressing bone marrow-derived DCs (Aire-BMDCs) to treat type 1 diabetes (T1D) and to explore a new strategy for autoimmune disease treatment. We observed that the onset of T1D in recipient mice was delayed; insulin autoantibody (IAA) production was significantly decreased; the structure of islets was protected; and the degree of inflammatory infiltration was lower. Furthermore, we found that Aire-BMDCs can promote apoptosis and induce autoreactive CD4+ T cell clonal anergy, inhibit Th1 and Th17 production, and induce Treg production. These results suggest that transplantation of Aire-BMDCs will be a manipulation and effective method for preventing or treating T1D.


Assuntos
Transplante de Medula Óssea , Células Dendríticas/fisiologia , Diabetes Mellitus Tipo 1/prevenção & controle , Células Epiteliais/imunologia , Imunoterapia/métodos , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Células Th17/imunologia , Timo/imunologia , Fatores de Transcrição/metabolismo , Animais , Células da Medula Óssea/fisiologia , Diferenciação Celular , Células Cultivadas , Anergia Clonal , Células Dendríticas/transplante , Diabetes Mellitus Tipo 1/imunologia , Humanos , Tolerância Imunológica , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição/genética , Proteína AIRE
19.
J Inorg Biochem ; 168: 67-75, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28013066

RESUMO

The antileukemic activity, mechanisms and serum albumin interactions of an arsenomolybdate, K2Na[AsMo6O21(O2CCH2NH3)3]·6H2O (1), was evaluated in the human leukemia HL-60 and U937 cells. The results indicated that 1 could inhibit the proliferation of both leukemia cell lines in a dose-dependent manner with the 50% lethal concentration (IC50) value of 8.61µM for HL-60 and 14.50µM for U937 at 24h, compare to the positive controls, all-trans retinoic acid (ATRA) with IC50 value of 20.76µM and 14.85µM,and As2O3 with IC50 value of 6.40µM and 8.75µM at 24h, respectively (P<0.05). Furthermore, the anti-leukemia activity of compound 1 might be medicated by arresting the leukemic cells in the G1 phase and inducing apoptosis via caspase-3 and bcl-2 regulatory proteins. Spectroscopic techniques results showed that the fluorescence of human serum albumin was quenched by compound 1, and the quenching mechanism was mainly static quenching. Compound 1 might be a potential medicinal candidate against acute promyelocytic leukemia.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Arsenicais/farmacologia , Caspase 3/efeitos dos fármacos , Leucemia Promielocítica Aguda/tratamento farmacológico , Molibdênio/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/química , Arsenicais/síntese química , Arsenicais/química , Western Blotting , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Estabilidade de Medicamentos , Citometria de Fluxo , Fluorescência , Células HL-60 , Humanos , Concentração Inibidora 50 , Molibdênio/química , Células U937
20.
Mol Med Rep ; 13(1): 565-71, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26548750

RESUMO

The autoimmune regulator (Aire) protein is a transcriptional activator that is essential in central immune tolerance, as it regulates the ectopic expression of many tissue­restricted antigens in medullary thymic epithelial cells. Aire expression has also been described in hematopoietic cells, such as monocytes/macrophages and dendritic cells (DCs), in the peripheral immune system. However, the role of Aire expression in peripheral immune system cells, including DCs, remains to be elucidated. In the present study, the effects of secreted cytokines from Aire­overexpressing DCs on cluster of differentiation (CD)4+ T cell subsets were investigated. The dendritic cell line, DC2.4, which overexpresses Aire, was co­cultured with CD4+ T cells from splenocytes using Transwell inserts. The results indicate that Aire­overexpressing cells induce T helper (Th)1 subsets by increasing interleukin (IL)­12 expression, and induce Th17 subsets by upregulating IL­6 and transforming growth factor (TGF)­ß production. In addition, it was observed that increased levels of phosphorylated extracellular signal­regulated kinases and p38 upregulated the expression of cytokines in Aire­overexpressing cells. These data suggest that Aire may have a role in inducing Th1 and Th17 differentiation by upregulating cytokine expression in DCs.


Assuntos
Células Dendríticas/metabolismo , Células Th1/metabolismo , Células Th17/metabolismo , Fatores de Transcrição/metabolismo , Regulação para Cima , Animais , Técnicas de Cocultura , Citocinas , Citometria de Fluxo , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos Endogâmicos C57BL , Fosforilação , Proteína AIRE
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA