Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cytotherapy ; 22(4): 214-226, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32305113

RESUMO

Our objective was to summarize the side effect of chimeric antigen receptor (CAR)-T cell therapy in patients with acute lymphocytic leukemia (ALL) and lymphoma. Two independent reviewers extracted relevant data. A total of 35 hematologic malignancy studies with CD19 CAR-T cell were included (1412 participants). Severe cytokine release syndrome (sCRS) proportion was experienced by 18.5% (95% confidence interval [CI], 0.128-0.259; P = 0.000) of 982 patients with the National Cancer Institute/Lee/common terminology criteria for adverse events grading system. The pooled neurotoxicity proportion was 21.7% (95% CI, 0.167-0.287; P = 0.000) of 747 patients with the same grading system. For all of the 25 clinical trials with the same grading system, subgroup analysis was performed. Based on the different disease type, a pooled prevalence of 35.7% was observed with event rate (ER) of 0.358 (95% CI, 0.289-0.434; P = 0.000) for ALL in 12 clinical trials. For lymphoma, a pooled prevalence of 13% was observed with ER of 0.073 (95% CI, 0.028-0.179; P = 0.000) in eight clinical trials. It was demonstrated that the patients who were older than 18 years of age have the lower sCRS incidence of 16.1% (95% CI, 0.110-0.250; P = 0.000) compared with 28.6% of the remaining population who were younger than 18 years of age (95% CI, 0.117-0.462: P = 0.023) in our analysis. Based on the different co-stimulatory domain, the sCRS of 16.5% was observed with ER of 0.175 (95% CI, 0.090-0.312; P = 0.000) for 4-1BB. The sCRS of 22.2% was observed with ER of 0.193 (95% CI, 0.107-0.322; P = 0.000) for CD28. For both the CD28 and 4-1BB, the sCRS of 17.3% was observed with ER of 0.170 (95% CI, 0.067-0.369; P = 0.003). Sub-analysis sCRS of the impact with cell dose and specific disease indication were also demonstrated. Limitations include heterogeneity of study populations, as well as high risk of bias of included studies. These results are helpful for physicians, patients and the other stakeholders to understand the adverse events and to further promote the improvement of CAR-T cell therapy in the future.


Assuntos
Antígenos CD19/imunologia , Terapia Baseada em Transplante de Células e Tecidos/efeitos adversos , Síndrome da Liberação de Citocina/epidemiologia , Neoplasias Hematológicas/terapia , Imunoterapia Adotiva/efeitos adversos , Linfoma/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Receptores de Antígenos Quiméricos/imunologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos CD28/imunologia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Criança , Feminino , Humanos , Imunoterapia Adotiva/métodos , Incidência , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento , Adulto Jovem
2.
Anal Bioanal Chem ; 412(8): 1901-1914, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32030491

RESUMO

Immunoglobin E (IgE)-related allergy constitutes a high proportion in allergic diseases. The production of specific IgE is key to evoking serial cascades and pathological reactions. Thus, targeting IgE is a different therapeutic approach from symptomatic treatments. Monoclonal antibodies (mAbs) against IgE were developed and a humanized antibody, omalizumab, was approved by five countries. It could inhibit the binding of IgE with epsilon receptor I of crystallizable fragment (FcεRI), thus preventing anaphylactic reactions. However, no bioactivity assay, which is the critical quality attribute and should thoroughly reflect the clinical mechanism, has been established to date. In commercial lot release, only the enzyme-linked immunosorbent assay (ELISA) method was applied, which only reflects the binding of omalizumab to IgE but not the subsequent reaction. In scientific research works, human FcεRI-transfected RBL-2H3 cells were used to indicate degranulation based on the detection of ß-hexosaminidase. Nevertheless, this method needs much work to stabilize the response and, hence, is not suitable for routine usage in commercial production and control of antibodies. To evaluate the bioactivity of anti-IgE antibodies including omalizumab using a simple assay that reflects the following mechanism of actions (MOA) after binding, we established an RBL-2H3 cell line transfected with both the α subunit of human FcεRI and nuclear factor-activated T cell (NFAT) response elements, the latter is conjugated with a luciferase gene, which could shed luminescence when substrates exist. The method was proven to possess good specificity, accuracy, linearity, and precision and may be utilized as a supplement to anti-IgE antibody bioactivity assays in terms of development, lot release, stability, and comparability studies. Graphical abstract The mechanism sketch of reporter gene assay for bioactivity determination of anti-IgE antibodies by RBL-2H3/FcεRIα/NFAT-Luc cells (left) and representative curves generated by the reporter gene assay (right).


Assuntos
Anticorpos Anti-Idiotípicos/análise , Bioensaio/métodos , Genes Reporter , Animais , Linhagem Celular Tumoral , Ensaio de Imunoadsorção Enzimática , Humanos
3.
Cytotherapy ; 21(7): 769-781, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31160157

RESUMO

Immunotherapy with chimeric antigen receptor T (CAR-T) cells has proved remarkably effective in recently published clinical trials. In this meta-analysis, we performed a systematic review in terms of the clinical response treated with CAR-T cells in acute lymphoblastic leukemia (ALL), chronic lymphocytic leukemia (CLL) and lymphomas patients. Thirty-eight published clinical studies including 665 patients were eligible for response rate (RR) evaluation. The overall pooled RR of CD19-CAR-T cells was 72% (95% confidence interval: 62-77%). The various clinical parameters were analyzed. RR was 81% in ALL, 68% in lymphoma and 70% in CLL. RR in patients who received interleukin (IL)-2 was 70%, whereas in those who did not receive IL-2, it was 74%. RR was 75% with lymphodepletion and 56% without lymphodepletion. RR with autologous cells was 76% and 57% with allogeneic cells. In conclusion, this meta-analysis showed a high clinical RR of CD19-CAR-T cell-based immunotherapy in patients with refractory B-cell malignancies.


Assuntos
Antígenos CD19/imunologia , Imunoterapia Adotiva/métodos , Leucemia de Células B/imunologia , Leucemia de Células T/imunologia , Receptores de Antígenos Quiméricos/imunologia , Linfócitos T/imunologia , Linfócitos B/imunologia , Linfócitos B/patologia , Ensaios Clínicos como Assunto , Feminino , Humanos , Imunoterapia Adotiva/efeitos adversos , Leucemia de Células B/terapia , Leucemia de Células T/terapia , Linfoma/imunologia , Linfoma/terapia , Linfoma de Células B/imunologia , Linfoma de Células B/terapia , Linfoma de Células T/imunologia , Linfoma de Células T/terapia , Masculino , Receptores de Antígenos Quiméricos/genética , Linfócitos T/metabolismo , Linfócitos T/transplante
4.
Anal Bioanal Chem ; 410(27): 7067-7075, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30178083

RESUMO

IL-6 has an important role in the pathogenesis of autoimmunity and chronic inflammation. Several mAbs that target IL-6 or the IL-6 receptor (IL-6R) have been established and approved for the treatment of various diseases such as multicentric Castleman's disease and rheumatoid arthritis. Quality control of therapeutic antibodies requires accurate determination of bioactivity. However, current cell-based anti-proliferation assays are tedious, time consuming, and result in high variation. We therefore developed a reporter gene assay (RGA) based on an IL-6-dependent DS-1 cell line that stably expressed the reporter luciferase controlled by the serum-induced element (SIE) response element, which was a key element located downstream of the IL-6 signaling pathway. The RGA method demonstrated good performance characteristics after careful optimization, including high specificity, stability, accuracy, precision, and robustness. It also had superior precision and sensitivity. The assay is simple compared with the traditional anti-proliferation assay. This novel RGA based on the IL-6-IL-6R-STAT3 pathway can be useful, in conjunction with the anti-proliferation bioassay, to determine the bioactivity of anti-IL-6/anti-IL-6R therapeutic mAbs. Graphical abstract The mechanism sketch of the reporter gene assay for the bioactivity determination of anti-IL-6/anti-IL-6Rα mAbs.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Avaliação Pré-Clínica de Medicamentos , Interleucina-6/antagonistas & inibidores , Receptores de Interleucina-6/antagonistas & inibidores , Anticorpos Monoclonais Humanizados/imunologia , Engenharia Celular , Linhagem Celular , Proliferação de Células , Avaliação Pré-Clínica de Medicamentos/métodos , Genes Reporter , Humanos , Interleucina-6/imunologia , Luciferases/genética , Luciferases/imunologia , Receptores de Interleucina-6/imunologia , Proteínas Recombinantes/imunologia
5.
J Immunol ; 193(8): 3860-71, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25217164

RESUMO

The germinal center response requires cooperation between Ag-specific T and B lymphocytes, which takes the form of long-lasting cell-cell conjugation in vivo. Signaling lymphocytic activation molecule (SLAM)-associated protein (SAP) is required for stable cognate T-B cell conjugation, whereas SLAM family transmembrane (TM) receptor Ly108 may negatively regulate this process. We show that, other than phosphotyrosine-binding, SAP does not harbor motifs that recruit additional signaling intermediates to stabilize T-B adhesion. Ly108 dampens T cell adhesion to not only Ag-presenting B cells, but also dendritic cells by inhibiting CD3ζ phosphorylation through two levels of regulated Ly108-CD3ζ interactions. Constitutively associated with Src homology 2 domain-containing tyrosine phosphatase-1 even in SAP-competent cells, Ly108 is codistributed with the CD3 complex within a length scale of 100-200 nm on quiescent cells and can reduce CD3ζ phosphorylation in the absence of overt TCR stimulation or Ly108 ligation. When Ly108 is engaged in trans during cell-cell interactions, Ly108-CD3ζ interactions are promoted in a manner that uniquely depends on Ly108 TM domain, leading to more efficient CD3ζ dephosphorylation. Whereas replacement of the Ly108 TM domain still allows the constitutive, colocalization-dependent inhibition of CD3ζ phosphorylation, it abrogates the ligation-dependent Ly108-CD3ζ interactions and CD3ζ dephosphorylation, and it abolishes the suppression on Ag-triggered T-B adhesion. These results offer new insights into how SAP and Ly108 antagonistically modulate the strength of proximal TCR signaling and thereby control cognate T cell-APC interactions.


Assuntos
Antígenos Ly/imunologia , Complexo CD3/imunologia , Adesão Celular/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Animais , Linfócitos B/imunologia , Complexo CD3/metabolismo , Comunicação Celular/imunologia , Células Cultivadas , Células Dendríticas/imunologia , Centro Germinativo/imunologia , Células HEK293 , Humanos , Ativação Linfocitária/imunologia , Camundongos , Fosforilação , Estrutura Terciária de Proteína , Receptores de Antígenos de Linfócitos T/imunologia , Proteínas Adaptadoras da Sinalização Shc , Transdução de Sinais/imunologia , Proteína Associada à Molécula de Sinalização da Ativação Linfocitária , Linfócitos T/imunologia , Domínios de Homologia de src/imunologia
6.
Cytotherapy ; 17(12): 1746-62, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26455275

RESUMO

BACKGROUND AIMS: The aim of this study was to investigate whether active specific immunotherapy (ASI) is able to demonstrate therapeutic efficacy against colorectal cancer. METHODS: We conducted a systematic review of published papers from MEDLINE, the Cochrane Central Register of Controlled Trials, EMBASE, the Wanfang Database, the China Science and Technology Periodical Database and China Journal Net. Published data were extracted independently by two authors who used predefined database templates. The effects of ASI were compared with those of surgery alone, and a pooled analysis was performed with the use of the data from random- or fixed-effect models. RESULTS: Twelve trials matched our inclusion criteria (n = 2993, including 1842 control subjects). The overall analysis showed a significant survival benefit [1-, 2-, 3-, 4-, 5-, 6- and 7-year overall survival (OS), P < 0.05; 10-year OS, P < 0.001] in favor of ASI immunotherapy combined with surgery, but there was not an improvement in the 8- or 9-year OS (P > 0.05). The disease-free survival (DFS) rate was improved after the combination of ASI immunotherapy (2-, 3-, 5- and 10-year DFS, P < 0.05), but no significant improvement was noted for the 1-, 4-, 6-, 7-, 8- or 9-year DFS (P > 0.05). In addition, the disease-specific survival (DSS) was improved at some time points after the combination of ASI immunotherapy and surgery (2-, 3-, 4-, 5- and 6-year DSS, P < 0.05, but not the 1-, 7-, 8- or 9-year DSS, P > 0.05). An improved 2-, 3-, 4-, 5- and 6-year recurrence-free interval (RFI) (P < 0.05) was also observed in patients who received ASI therapy, but this was not observed for the 1-year RFI (P > 0.05). Furthermore, an analysis of the recurrence-free survival (RFS) showed that it was significantly increased in the ASI plus surgery group (1-, 2-, 3-, 4-, 5- and 6-year RFS, P < 0.001). The funnel plots showed that the analyses were relatively reliable and the publication bias was small. CONCLUSIONS: The combination of ASI immunotherapy and surgery was superior in prolonging the overall survival time and enhancing the recurrence-free survival rate compared with surgery alone.


Assuntos
Neoplasias Colorretais/terapia , Imunoterapia/métodos , Neoplasias Colorretais/mortalidade , Intervalo Livre de Doença , Humanos , Recidiva Local de Neoplasia/terapia , Taxa de Sobrevida
7.
Cytotherapy ; 17(7): 956-68, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25824289

RESUMO

BACKGROUND AIMS: In this study, we investigate whether bone marrow mononuclear cells (BM-MNC) or peripheral blood mononuclear cells (PB-MNC) have therapeutic efficacy in type 2 diabetes (T2D). METHODS: Search terms included stem cell, bone marrow cell, peripheral blood cell, umbilical cord blood and T2D in MEDLINE, the Cochrane Controlled Trials Register, EMBASE, the Wanfang Database, the China Science and Technology Periodical Database and China Journal Net. RESULTS: Fifteen trials met our inclusion criteria (n = 497). One group included 266 cases with BM-MNC therapy and the other group contained 231 cases with PB-MNC treatment. Glycosylated hemoglobin was decreased after BM-MNC or PB-MNC therapy compared with that before (12 months: P < 0.001; 6 months: P < 0.001; 3 months: P < 0.05). Fasting plasma glucose was reduced in BM-MNC therapy group compared with control after 12-month follow-up (P < 0.001) and after BM-MNC therapy compared with that before (9 months: P < 0.001) but was not obvious in other stages. Meanwhile, the analysis showed that C-peptide level increased after BM-MNC and PB-MNC therapy compared with the control therapy (12 months: P < 0.001) and with that before therapy (6 months: P < 0.05). Insulin requirement reduction was also observed in patients receiving BM-MNC therapy (3, 6, 9 and 12 months: P < 0.05). CONCLUSIONS: To a certain extent, BM-MNC or PB-MNC therapy for T2D demonstrated superiority of glycemic control, increased insulin biosynthesis and elevated insulin secretion from existing ß-cells and might prevent islet cell loss.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Diabetes Mellitus Tipo 2/terapia , Insulina/biossíntese , Leucócitos Mononucleares/transplante , Transplante de Células-Tronco/métodos , Adulto , Glicemia/análise , Células da Medula Óssea/citologia , Transplante de Medula Óssea , Peptídeo C/sangue , Feminino , Sangue Fetal/citologia , Hemoglobinas Glicadas/análise , Humanos , Insulina/sangue , Células Secretoras de Insulina/citologia , Masculino , Pessoa de Meia-Idade , Células-Tronco/citologia , Transplante Autólogo , Resultado do Tratamento
8.
Cytotherapy ; 16(7): 934-45, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24794183

RESUMO

BACKGROUND: To evaluate the therapeutic efficacy of dendritic cells (DC) alone, cytokine-induced killer (CIK) cells alone and the combination of DC and CIK cells in the treatment of breast cancer, we performed a systemic review of the relevant published clinical studies, collectively referred to as DC-CIK cell therapy. METHODS: Six hundred thirty-three patients with breast cancer were assigned to cohorts, and a meta-analysis was conducted. RESULTS: The treatment of breast cancer with DC-CIK cells was associated with a significantly improved 1-year survival (P = 0.0001). The Karnofsky performance status scale of the patients treated with DC-CIK cells was significantly improved compared with that of the non-DC-CIK group (P < 0.0001). The percentage of T cells (CD3(+), CD4(+) and CD4(+)CD8(+)), CD16(+) monocytes, and CD3(+)CD56(+) natural killer T cells in the peripheral blood of cancer patients was significantly increased (P ≤ 0.05), whereas the percentage of CD4(+)CD25(+) regulatory T cells was not significantly decreased (P = 0.32) in the DC-CIK treatment group compared with the non-DC-CIK group. The levels of interleukin-2, interleukin-12, tumor necrosis factor-α, interferon-γ, and nucleolar organizer region protein in the peripheral blood of cancer patients, which reflect immune function, were significantly increased (P < 0.001) after DC-CIK cell treatment. Furthermore, after DC-CIK treatment, the average levels of the alpha-fetoprotein, cancer antigen embryonic antigen and carbohydrate antigen tumor markers were decreased (P < 0.00001). CONCLUSIONS: DC-CIK cell therapy markedly prolongs survival time, enhances immune function, and improves the efficacy of the treatment of breast cancer patients.


Assuntos
Neoplasias da Mama/terapia , Células Matadoras Induzidas por Citocinas/transplante , Células Dendríticas/transplante , Imunoterapia Adotiva/métodos , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Ensaios Clínicos como Assunto , Feminino , Humanos , Imunoterapia Adotiva/efeitos adversos
9.
Mol Ther ; 21(9): 1668-77, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23877036

RESUMO

Targeting the adaptor protein (transforming growth factor-ß (TGF-ß)-activated protein kinase 1 (TAK1)-binding protein 1) (TAB1)-mediated non-canonical activation of p38α to limit ischemia/reperfusion (I/R) injury after an acute myocardial infarction seems to be attractive since TAB1/p38α interaction occurs specifically in very limited circumstances and possesses unique structural basis. However, so far no TAB1/p38α interaction inhibitor has been reported due to the limited knowledge about the interfaces. In this study, we sought to identify key amino acids essential for the unique mode of interaction with computer-guided molecular simulations and molecular docking. After validation of the predicted three-dimensional (3-D) structure of TAB1/p38α complex, we designed several peptides and evaluated whether they could block TAB1/p38α interaction with selectivity. We found that a cell-permeable peptide worked as a selective TAB1/p38α interaction inhibitor and decreased myocardial I/R injury. To our knowledge, this is the first TAB1/p38α interaction inhibitor.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/química , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Peptídeos/metabolismo , Peptídeos/farmacologia , Sequência de Aminoácidos , Animais , Sítios de Ligação , Modelos Animais de Doenças , Humanos , Modelos Moleculares , Simulação de Acoplamento Molecular , Dados de Sequência Molecular , Traumatismo por Reperfusão Miocárdica/metabolismo , Peptídeos/síntese química , Estrutura Secundária de Proteína , Ratos
10.
Cell Oncol (Dordr) ; 47(1): 141-156, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37639207

RESUMO

OBJECTIVE AND DESIGN: Pancreatic cancer is a highly malignant tumor that is well known for its poor prognosis. Based on glycosylation, we performed integrated quantitative N-glycoproteomics to investigate the synergistic anti-tumor effects of aspirin and gemcitabine on pancreatic cancer cells and explore the potential molecular mechanisms of chemotherapy in pancreatic cancer. METHODS AND RESULTS: Two pancreatic cancer cell lines (PANC-1 and BxPC-3) were treated with gemcitabine, aspirin, and a combination (gemcitabine + aspirin). We found that the addition of aspirin enhanced the inhibitory effect of gemcitabine on the activity of PANC-1 and BxPC-3 cells. Quantitative N-glycoproteome, proteome, phosphorylation, and transcriptome data were obtained from integrated multi-omics analysis to evaluate the anti-tumor effects of aspirin and gemcitabine on pancreatic cancer cells. Mfuzz analysis of intact N-glycopeptide profiles revealed two consistent trends associated with the addition of aspirin, which showed a strong relationship between N-glycosylation and the synergistic effect of aspirin. Further analysis demonstrated that the dynamic regulation of sialylation and high-mannose glycoforms on ECM-related proteins (LAMP1, LAMP2, ITGA3, etc.) was a significant factor for the ability of aspirin to promote the anti-tumor activity of gemcitabine and the drug resistance of pancreatic cancer cells. CONCLUSIONS: In-depth analysis of N-glycosylation-related processes and pathways in pancreatic cancer cells can provide new insight for future studies regarding pancreatic cancer therapeutic targets and drug resistance mechanisms.


Assuntos
Gencitabina , Neoplasias Pancreáticas , Humanos , Desoxicitidina/farmacologia , Desoxicitidina/uso terapêutico , Aspirina/farmacologia , Aspirina/uso terapêutico , Proteômica , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Pancreáticas/patologia , Apoptose
11.
Cell Immunol ; 285(1-2): 42-8, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24055734

RESUMO

The second messenger cAMP is involved in the regulation of many cellular activities partially through modulating the MAPK pathways. The role of cAMP in TGF-ß-mediated adaptive Tregs differentiation remains elusive. In this work, we show that cAMP inhibits antigen-nonspecific proliferation of murine CD4+ T cells without significant promotion of apoptosis. Moreover, cAMP suppresses TGF-ß-induced expression of forkhead transcription factor Foxp3. 6-MB-cAMP, a site-selective activator of PKA, mimics the role of cAMP in TGF-ß-induced Foxp3 expression. Further exploration reveals that TGF-ß activates ERK and JNK, but not p38. cAMP and 6-MB-cAMP block TGF-ß-induced activation of ERK and JNK through transcription-independent manner and transcription-dependent manner, respectively. Since direct inhibition of ERK or JNK activity mimics the effects of cAMP during this process, our work suggests that cAMP suppresses TGF-ß-mediated adaptive Tregs differentiation through, at least partially, inhibiting the activation of ERK and JNK.


Assuntos
AMP Cíclico/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Linfócitos T Reguladores/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Apoptose/imunologia , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Fatores de Transcrição Forkhead/biossíntese , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T Reguladores/imunologia , Transcrição Gênica
12.
Mol Cell Biochem ; 384(1-2): 197-202, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24000012

RESUMO

The receptor for activated C kinase 1 (RACK1), an adaptor protein implicated in the regulation of multiple signaling pathways, has been reported to contribute to the survival of leukemic progenitor cells by enhancing the activity of glycogen synthase kinase 3ß (GSK3ß). However, it remains unknown whether RACK1 also contributes to the oncogenic growth of acute myeloid leukemia (AML) cells. Here, we report that transient or stable silencing of endogenous RACK1 expression by RACK1 short hairpin RNAs (shRNAs) led to impaired proliferation of THP1 AML cells without inducing terminal differentiation. Further exploration revealed that RACK1 loss-of-function resulted in reduced GSK3ß activity. GSK3ß shRNA treatment showed similar effects to RACK1 loss-of-function. Our data collectively suggest that RACK1 contributes to THP1 cell proliferation through, at least partially, enhancing GSK3ß activity. Thus, targeting RACK1 may have some important therapeutic implications in the treatment of AML.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Leucemia Mieloide Aguda/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Superfície Celular/metabolismo , Apoptose/genética , Ciclo Celular/genética , Diferenciação Celular/genética , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Proteínas de Ligação ao GTP/genética , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Humanos , Leucemia Mieloide Aguda/genética , Receptores de Lipopolissacarídeos/biossíntese , Proteínas de Neoplasias/genética , Fagocitose/genética , Interferência de RNA , RNA Interferente Pequeno , Receptores de Quinase C Ativada , Receptores de Superfície Celular/genética , Células U937
13.
Virus Res ; 335: 199201, 2023 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-37595663

RESUMO

Preclinical studies indicate that SARS-CoV-2 nucleocapsid (N)-based vaccines, along with other viral protein(s), confer protection in various animal models against infection by SARS-CoV-2 ancestral virus and variants of concern. However, the optimal vaccination procedure and the role of N-specific host adaptive immune responses remain elusive. Here, we report that intranasal inoculation with replication-deficient human adenovirus type 5 expressing SARS-CoV-2 N protein (Ad5-N) conferred no protection in the lung of female BALB/c mice upon re-encountering the antigen, either by 10-fold Ad5-N re-exposure or sublethal infection of mouse-adapted SARS-CoV-2. By contrast, this procedure led to aggravated lung pathology with more necroptotic CD3+ T cells and Ly6G+ granulocytes, which was associated with the accumulation of IFN-γ-expressing antigen-experienced CD4+ and CD8+ T cells. These findings pre-caution the clinical application of this vaccination procedure. Furthermore, our data suggest that excessive host adaptive immune responses against N protein contributes to COVID-19 pathogenesis.


Assuntos
Adenovírus Humanos , COVID-19 , Humanos , Feminino , Animais , Camundongos , SARS-CoV-2/genética , Linfócitos T CD8-Positivos , Vacinação , Camundongos Endogâmicos BALB C
14.
FEBS Open Bio ; 13(9): 1723-1736, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37400956

RESUMO

Stress-related illnesses are linked to the onset and progression of renal diseases and depressive disorders. To investigate stress-induced changes in the renal transcriptome associated with the development of depressive behaviors, we generated here a chronic social defeat stress (CSDS) model of C57 BL/6 male mice and then performed RNA sequencing of the kidneys to obtain an inflammation-related transcriptome. Administration of the antidepressant drug fluoxetine (10 mg·kg-1 ·day-1 ) during CSDS induction could partially alleviate renal inflammation and reverse CSDS-induced depression-like behaviors. Moreover, fluoxetine also modulated gene expression of stress-related hormone receptors, including prolactin and melanin-concentrating hormone. These results suggest that CSDS can induce gene expression changes associated with inflammation in the kidney of C57 BL/6 male mice, and this inflammation can be treated effectively by fluoxetine.


Assuntos
Antidepressivos , Fluoxetina , Animais , Camundongos , Masculino , Fluoxetina/farmacologia , Antidepressivos/farmacologia , Depressão/tratamento farmacológico , Depressão/metabolismo , Inflamação/tratamento farmacológico , Rim
15.
J Cancer Res Clin Oncol ; 149(19): 17495-17509, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37902853

RESUMO

PURPOSE: Mitogen-activated protein kinases (MAPK), specifically the c-Jun N-terminal kinase (JNK)-MAPK subfamily, play a crucial role in the development of various cancers, including hepatocellular carcinoma (HCC). However, the specific roles of JNK1/2 and their upstream regulators, MKK4/7, in HCC carcinogenesis remain unclear. METHODS: In this study, we performed differential expression analysis of JNK-MAPK components at both the transcriptome and protein levels using TCGA and HPA databases. We utilized Kaplan-Meier survival plots and receiver operating characteristic (ROC) curve analysis to evaluate the prognostic performance of a risk scoring model based on these components in the TCGA-HCC cohort. Additionally, we conducted immunoblotting, apoptosis analysis with FACS and soft agar assays to investigate the response of JNK-MAPK pathway components to various death stimuli (TRAIL, TNF-α, anisomycin, and etoposide) in HCC cell lines. RESULTS: JNK1/2 and MKK7 levels were significantly upregulated in HCC samples compared to paracarcinoma tissues, whereas MKK4 was downregulated. ROC analyses suggested that JNK2 and MKK7 may serve as suitable diagnostic genes for HCC, and high JNK2 expression correlated with significantly poorer overall survival. Knockdown of JNK1 enhanced TRAIL-induced apoptosis in hepatoma cells, while JNK2 knockdown reduced TNF-α/cycloheximide (CHX)-and anisomycin-induced apoptosis. Neither JNK1 nor JNK2 knockdown affected etoposide-induced apoptosis. Furthermore, MKK7 knockdown augmented TNF-α/CHX- and TRAIL-induced apoptosis and inhibited colony formation in hepatoma cells. CONCLUSION: Targeting MKK7, rather than JNK1/2 or MKK4, may be a promising therapeutic strategy to inhibit the JNK-MAPK pathway in HCC therapy.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Carcinoma Hepatocelular/genética , Fator de Necrose Tumoral alfa , Etoposídeo , Anisomicina , MAP Quinase Quinase 7/genética , MAP Quinase Quinase 7/metabolismo , Neoplasias Hepáticas/genética , Apoptose
16.
World J Clin Cases ; 10(27): 9714-9726, 2022 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-36186213

RESUMO

BACKGROUND: Currently, ongoing trials of mesenchymal stem cells (MSC) therapies for coronavirus disease 2019 (COVID-19) have been reported. AIM: In this study, we investigated whether MSCs have therapeutic efficacy in novel COVID-19 patients. METHODS: Search terms included stem cell, MSC, umbilical cord blood, novel coronavirus, severe acute respiratory syndrome coronavirus-2 and COVID-19, applied to PubMed, the Cochrane Controlled Trials Register, EMBASE and Web of Science. RESULTS: A total of 13 eligible clinical trials met our inclusion criteria with a total of 548 patients. The analysis showed no significant decrease in C-reactive protein (CRP) levels after stem cell therapy (P = 0.11). A reduction of D-dimer levels was also not observed in patients after stem cell administration (P = 0.82). Furthermore, interleukin 6 (IL-6) demonstrated no decrease after stem cell therapy (P = 0.45). Finally, we investigated the overall survival (OS) rate after stem cell therapy in COVID-19 patients. There was a significant improvement in OS after stem cell therapy; the OS of enrolled patients who received stem cell therapy was 90.3%, whereas that of the control group was 79.8% (P = 0.02). CONCLUSION: Overall, our analysis suggests that while MSC therapy for COVID-19 patients does not significantly decrease inflammatory markers such as CRP, D-dimer and IL-6, OS is improved.

17.
J Biol Chem ; 285(7): 4931-40, 2010 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-20007324

RESUMO

The existence of cancer stem cells (CSCs) or stem-like cancer cells (SLCCs) is regarded as the cause of tumor formation and recurrence. However, the origin of such cells remains controversial with two competing hypotheses: CSCs are either transformed from tissue adult stem cells or dedifferentiated from transformed progenitor cells. Compelling evidence has determined the chromosomal aneuploidy to be one of the hallmarks of cancer cells, indicating genome instability plays an important role in tumorigenesis, for which CSCs are believed to be the initiator. To gain direct evidence that genomic instability is involved in the induction of SLCCs, we utilized multiple approaches to enhance genomic instability and monitored the percentage of SLCC in cultured cancer cells. Using side population (SP) cells as a marker for SLCC in human nasopharyngeal carcinoma (NPC) and CD133 for human neuroblastoma cells, we found that DNA damage inducers, UV and mitomycin C were capable of increasing SP cells in NPC CNE-2 and neuroblastoma SKN-SH cells. Likewise, either overexpression of a key regulator of cell cycle, Mad2, or knock down of Aurora B, an important kinase in mitosis, or Cdh1, a key E3 ligase in cell cycle, resulted in a significant increase of SP cells in CNE-2. More interestingly, enrichment of SP cells was observed in recurrent tumor tissues as compared with the primary tumor in the same NPC patients. Our study thus suggested that, beside transformation of tissue stem cells leading to CSC generation, genomic instability could be another potential mechanism resulting in SLCC formation, especially at tumor recurrence stage.


Assuntos
Instabilidade Genômica/fisiologia , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/metabolismo , Antígeno AC133 , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Antígenos CD/metabolismo , Aurora Quinase B , Aurora Quinases , Western Blotting , Caderinas/genética , Caderinas/metabolismo , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , Citometria de Fluxo , Instabilidade Genômica/genética , Glicoproteínas/metabolismo , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Cariotipagem , Proteínas Mad2 , Mitomicina/toxicidade , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/metabolismo , Proteínas de Neoplasias/metabolismo , Peptídeos/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Raios Ultravioleta/efeitos adversos
18.
Front Oncol ; 11: 655757, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33987090

RESUMO

Lung cancer is one of the most prevalent cancers in both men and women worldwide. The nucleic acid G4 structures have been implicated in the transcriptional programmes of cancer-related genes in some cancers such as lung cancer. However, the role of the dominant G4 resolvase DHX36 in the progression of lung cancer remains unknown. In this study, by bioinformatic analysis of public datasets (TCGA and GEO), we find DHX36 is an independent prognosis indicator in non-small-cell lung carcinoma (NSCLC) with subtype dependence. The stable lentiviral knockdown of the DHX36 results in accelerated migration and aggregation of the S-phase subpopulation in lung cancer cells. The reduction of DHX36 level de-sensitises the proliferation response of lung cancer cells to chemotherapeutic drugs such as paclitaxel with cell dependence. The knockdown of this helicase leads to promoted tumour growth, demonstrated by a 3D fluorescence spheroid lung cancer model, and the stimulation of cell colony formation as shown by single-cell cultivation. High throughput proteomic array indicates that DHX36 functions in lung cancer cells through regulating multiple signalling pathways including activation of protein activity, protein autophosphorylation, Fc-receptor signalling pathway, response to peptide hormone and stress-activated protein kinase signalling cascade. A causal transcriptomic analysis suggests that DHX36 is significantly associated with mRNA surveillance, RNA degradation, DNA replication and Myc targets. Therefore, we unveil that DHX36 presents clinical significance and plays a role in tumour suppression in lung cancer, and propose a potentially new concept for an anti-cancer therapy based on helicase-specific targeting.

19.
BMC Cancer ; 10: 68, 2010 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-20181293

RESUMO

BACKGROUND: There is increasing evidence that cancers contain their own stem-like cells, and particular attention has been paid to one subset of cancer-stem cells termed side population (SP). Stem cells under normal physical conditions are tightly controlled by their microenvironment, however, the regulatory role of the microenvironment surrounding cancer stem cells is not well characterized yet. In this study we found that the phenotype of SP can be "generated" by macrophage-like cells under conditioned culture. Furthermore the gene regulation pathway involved in cellular reprogramming process was investigated. METHODS: The selection and identification of SP in 50 CNE-2 single cell clones were performed by flow cytometry. The transwell assay and immunofluorescence staining were used to measure migration and cancer stem cell characters of non-SP single clone cells cultured with conditioned medium respectively. The subtraction suppression hybridization (SSH) technique and northern blotting analysis was applied to explore the pluripotency-associated genes under a unique epigenetic sub-microenvironment. RESULTS: Among 50 clones, only one did not possess SP subpopulation while others did. The non-SP cells induced by macrophage-like cells showed more aggressive characters, which increased cell migration compared with the control cells and showed some fraction of SP phenotype. These cells expressed distinguished level of pluripotency-associated genes such as ADP-ribosylation factor-like 6 interacting protein (ARMER), poly (rC) binding protein 1 (PCBP1) and pyruvate dehydrogenase E1-beta subunit (PDHB) when subjected to the environment. CONCLUSION: To our knowledge, this is the first study to demonstrate that non-SP single-clone cells can be induced to generate a SP phenotype when they are cultured with conditioned medium of macrophage-like cells, which is associated with the reactivation of pluripotency-associated genes.


Assuntos
Carcinoma/genética , Carcinoma/patologia , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/patologia , Células-Tronco Neoplásicas/citologia , Técnicas de Cultura de Células/métodos , Linhagem Celular Tumoral , Movimento Celular , Separação Celular , Meios de Cultivo Condicionados/metabolismo , Epigênese Genética , Citometria de Fluxo/métodos , Humanos , Macrófagos/citologia , Microscopia de Fluorescência/métodos , Transplante de Neoplasias , Hibridização de Ácido Nucleico
20.
Rev Neurosci ; 31(4): 427-440, 2020 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-31926107

RESUMO

To evaluate the therapeutic efficacy of passive cellular immunotherapy for glioma, a total of 979 patients were assigned to the meta-analysis. PubMed and the Cochrane Central Register of Controlled Trials were searched initially from February 2018 and updated in April 2019. The overall survival (OS) rates and Karnofsky performance status (KPS) values of patients who underwent passive cellular immunotherapy were compared to those of patients who did not undergo immunotherapy. The proportion of survival rates was also evaluated in one group of clinical trials. Pooled analysis was performed with random- or fixed-effects models. Clinical trials of lymphokine-activated killer cells, cytotoxic T lymphocytes, autologous tumor-specific T lymphocytes, chimeric antigen receptor T cells, cytokine-induced killer cells, cytomegalovirus-specific T cells, and natural killer cell therapies were selected. Results showed that treatment of glioma with passive cellular immunotherapy was associated with a significantly improved 0.5-year OS (p = 0.003) as well as improved 1-, 1.5-, and 3-year OS (p ≤ 0.05). A meta-analysis of 206 patients in one group of clinical trials with 12-month follow-up showed that the overall pooled survival rate was 37.9% (p = 0.003). Analysis of KPS values demonstrated favorable results for the immunotherapy arm (p < 0.001). Thus, the present meta-analysis showed that passive cellular immunotherapy prolongs survival and improves quality of life for glioma patients, suggesting that it has some clinical benefits.


Assuntos
Glioma/terapia , Imunoterapia Adotiva , Imunoterapia , Resultado do Tratamento , Células Matadoras Induzidas por Citocinas/imunologia , Glioma/imunologia , Humanos , Imunoterapia/métodos , Imunoterapia Adotiva/métodos , Qualidade de Vida , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA