Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cancer Lett ; 562: 216168, 2023 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-37031915

RESUMO

Dendritic cells (DCs) release nanometer-sized membrane vesicles known as dexosomes, containing different molecules, particularly proteins, for presenting antigens, i.e., major histocompatibility complex (MHC)-I/II and CD86. Dexosomes can, directly and indirectly, stimulate antigen-reactive CD8+ and CD4+ T cell responses. Antigen-loaded dexosomes can lead to the development of potent anti-tumoral immune responses. Notably, developing dexosome-based cell-free vaccines could serve as a new vaccination platform in the era of immunotherapy for various cancers. Furthermore, combining dexosomes vaccination strategies with other treatment approaches can considerably increase tumor-specific T cell responses. Herein, we aimed to review how dexosomes interact with immune cells, e.g., CD4+ and CD8+ T cells and natural killer (NK) cells. Besides, we discussed the limitations of this approach and suggested potential strategies to improve its effectiveness for affected patients.


Assuntos
Vacinas Anticâncer , Exossomos , Neoplasias , Humanos , Linfócitos T CD8-Positivos , Células Dendríticas , Neoplasias/terapia , Neoplasias/metabolismo , Imunoterapia , Vacinas Anticâncer/uso terapêutico
2.
Gene ; 855: 147086, 2023 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36535461

RESUMO

BACKGROUND: Osteosarcoma is common type of bone cancer; however, the prognosis of patients with metastatic osteosarcoma is poor. As a new inhibitory immune checkpoint molecule, HHLA2 is upregulated in osteosarcoma. Herein, we studied the significance of tumor-intrinsic HHLA2 in MG-63 growth. Also, we examined the influence of combined therapy of HHLA2 knockdown with paclitaxel on the apoptosis, cell cycle, migration, and stemness of MG-63 cells. METHODS: The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay was performed to study the half-maximal inhibitory concentration (IC50) of paclitaxel and the cytotoxicity of HHLA2-small interfering RNA (siRNA) on MG-63 cells. The apoptosis and cell cycle were analyzed using flow cytometry. The wound-healing and colony formation assays were conducted to investigate the effect of paclitaxel and HHLA2 knockdown on the migration and stemness of MG-63 cells, respectively. QRT-PCR was used to determine the Bax, caspase-3, and Bcl-2 mRNA expression levels. RESULTS: HHLA2 silencing has enhanced the chemosensitivity of MG-63 cells to paclitaxel. Besides, HHLA2 knockdown has increased the paclitaxel-induced cytotoxic effect on MG-63 cells. In terms of stimulating apoptosis, decreasing clonogenicity, halting the cell cycle at the sub G1 phase, and inhibiting migration, tumor-intrinsic HHLA2 silencing has increased these anti-tumor effects of paclitaxel on MG-63 cells. Besides, HHLA2 knockdown has potentiated paclitaxel-mediated Bcl-2 downregulation and paclitaxel-mediated caspase-3 and Bax upregulation in MG-63 cells. CONCLUSION: Tumor-intrinsic HHLA2 knockdown increases the anti-tumoral effect of paclitaxel on MG-63 cells and enhances the chemosensitivity of MG-63 cells to paclitaxel.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Humanos , Paclitaxel/farmacologia , Caspase 3 , Proteína X Associada a bcl-2 , Linhagem Celular Tumoral , Osteossarcoma/tratamento farmacológico , Osteossarcoma/genética , Osteossarcoma/metabolismo , Apoptose/genética , Neoplasias Ósseas/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Imunoglobulinas/metabolismo , Imunoglobulinas/farmacologia
3.
Photodiagnosis Photodyn Ther ; 41: 103212, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36436735

RESUMO

Photodynamic therapy (PDT) is a light-based anti-neoplastic therapeutic approach. Growing evidence indicates that combining conventional anti-cancer therapies with PDT can be a promising approach to treat malignancies. Herein, we aimed to investigate anti-cancer effects of the combination treatment of zinc phthalocyanine (ZnPc)-PDT with tamoxifen (TA) on MDA-MB-231 cells (as a triple-negative breast cancer (TNBC) cell line). For this purpose, we investigated the cytotoxicity of TA and ZnPc-PDT on MDA-MB-231 cells performing the MTT assay. The effect of TA and ZnPc-PDT on the apoptosis of MDA-MB-231 cells was studied using Annexin V/PI and DAPI staining. The wound-healing assay, and colony formation assay were performed to study the effect of TA and ZnPc-PDT on the migration, and clonogenicity of MDA-MB-231 cells, respectively. The qRT-PCR was done to study the gene expression of caspase-8, caspase-9, caspase-3, ZEB1, ROCK1, SNAIL1, CD133, CD44, SOX2, and ABCG2 (ATP-binding cassette sub-family G member 2). Based on our results, monotherapies with TA and ZnPc-PDT can remarkably increase cell cytotoxicity effects, stimulate apoptosis via downregulating Bcl-2 and upregulating caspase-3 and caspase-9, inhibit migration via downregulating SNAIL1 and ZEB1, and suppress clonogenicity via downregulating SOX2 and CD44 in MDA-MB-231 cells. Besides, these monotherapies can downregulate the expression of ABCG2 in MDA-MB-231 cells. Nevertheless, the combination treatment can potentiate the above-mentioned anti-cancer effects compared to monotherapy with TA. Of interest, the combined treatment of TA with ZnPc-PDT can synergically increase cell cytotoxicity effects on MDA-MB-231 cells. In fact, synergistic effects were estimated by calculation of Combination Index (CI); that synergistic outcomes were observed in all groups. Also, this combination treatment can significantly upregulate the caspase-8 gene expression and downregulate ROCK1 and CD133 gene expression in MDA-MB-231 cells. Overall, our results show that ZnPc-PDT can more sensitize the MDA-MB-231 cells to TA treatment. Based on our knowledge and experiment, the synergistic effects of ZnPc-PDT and TA deserve further evaluation in cancer research.


Assuntos
Fotoquimioterapia , Neoplasias de Mama Triplo Negativas , Humanos , Fármacos Fotossensibilizantes/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Caspase 3 , Caspase 9/farmacologia , Caspase 8/farmacologia , Caspase 8/uso terapêutico , Fotoquimioterapia/métodos , Tamoxifeno/farmacologia , Tamoxifeno/uso terapêutico , Linhagem Celular Tumoral , Indóis , Apoptose , Quinases Associadas a rho/farmacologia , Quinases Associadas a rho/uso terapêutico
4.
Front Med (Lausanne) ; 9: 1027758, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36388933

RESUMO

Background: The programmed death-ligand 1 (PD-L1)/PD-1 axis is one of the well-established inhibitory axes in regulating immune responses. Besides the significance of tumor-intrinsic PD-L1 expression in immune evasion, its oncogenic role has been implicated in various malignancies, like non-small cell lung cancer (NSCLC). As small non-coding RNAs, microRNAs (miRs) have pivotal roles in cancer biology. The current study aimed to systematically review the current knowledge about the significance of PD-L1-inhibiting miRs in NSCLC inhibition and their underlying mechanisms. Materials and methods: We conducted the current scoping review based on the PRISMA-ScR statement. We systematically searched Embase, Scopus, Web of Science, PubMed, Ovid, EBSCO, ProQuest, Cochrane Library, African Index Medicus, and Pascal-Francis up to 4 April 2021. We also performed in silico tumor bulk RNA sequencing and single-cell RNA sequencing to further the current knowledge of the non-coding RNA-mediated tumor-intrinsic PD-L1 regulation and the PD-L1/PD-1 axis in NSCLC. Results: The ectopic expression of hsa-miR-194-5p, hsa-miR-326, hsa-miR-526b-3p, hsa-miR-34a-5p, hsa-miR-34c-5p, hsa-miR-138-5p, hsa-miR-377-3p, hsa-let-7c-5p, hsa-miR-200a-3p, hsa-miR-200b-3p, hsa-miR-200c-3p, and hsa-miR-197-3p, as PD-L1-inhibiting miR, inhibits NSCLC development. These PD-L1-inhibiting miRs can substantially regulate the cell cycle, migration, clonogenicity, invasion, apoptosis, tumor chemosensitivity, and host anti-tumoral immune responses. Based on single-cell RNA sequencing results, PD-L1 inhibition might liberate the tumor-infiltrated CD8+ T-cells and dendritic cells (DCs)-mediated anti-tumoral immune responses via disrupting the PD-L1/PD-1 axis. Conclusion: Given the promising preclinical results of these PD-L1-inhibiting miRs in inhibiting NSCLC development, their ectopic expression might improve NSCLC patients' prognosis; however, further studies are needed to translate this approach into clinical practice.

5.
Pathol Res Pract ; 236: 154004, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35834883

RESUMO

Dysregulated cell cycle progression has been implicated in cancer development. Cytarabine can interfere with the S phase of the cell cycle; however, tumoral cells can develop chemoresistance. Specific tumor-suppressive microRNAs (miRs) replacement can arrest the cell cycle and enhance chemosensitivity. Herein, we investigated the effect of hsa-miR-34a-5p replacement and cytarabine on the cell cycle, chemosensitivity, and migration of MDA-MB-231 cells. Our in-silico results have shown that hsa-miR-34a-5p has considerable interactions with ß-catenin, CDK4, CDK6, and cyclin-D1; therefore, hsa-miR-34a-5p replacement could arrest cell cycle at the sub-G1 phase. Our in vitro results have indicated that monotherapies with hsa-miR-34a-5p replacement and cytarabine can substantially arrest the cell cycle at the sub-G1 phase; however, the maximal cell cycle arrest has been observed with the combined therapy. Ectopic overexpression of hsa-miR-34a-5p has remarkably enhanced the chemosensitivity of MDA-MB-231 cells. Also, the combined therapy has considerably suppressed the migration of MDA-MB-231 cells compared to the monotherapies. Although the combination therapy has not remarkably decreased the expression of CDK4, CDK6, and cyclin-D1 compared to monotherapy with cytarabine, the combination therapy has substantially downregulated ß-catenin expression compared to monotherapy with cytarabine. Overall, this combination therapy is a promising approach to arresting the cell cycle and migration of MDA-MB-231 cells.


Assuntos
MicroRNAs , beta Catenina , Ciclo Celular , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Citarabina/farmacologia , Regulação Neoplásica da Expressão Gênica , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , beta Catenina/metabolismo
6.
Mol Biol Rep ; 49(9): 8325-8335, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35666424

RESUMO

BACKGROUND: The combined restoration of tumor-suppressive microRNAs (miRs) has been identified as a promising approach for inhibiting breast cancer development. This study investigated the effect of the combined restoration of miR-424-5p and miR-142-3p on MCF-7 cells and compared the efficacy of the combined therapy with the monotherapies with miR-424-5p and miR-142-3p. METHODS: After transfection of miR-424-5p and miR-142-3p mimics into MCF-7 cells in the combined and separated manner, the proliferation of tumoral cells was assessed by the MTT assay. Also, the apoptosis, autophagy, and cell cycle of the cells were analyzed by flow cytometry. Western blot and qRT-PCR were used to study the expression levels of c-Myc, Bcl-2, Bax, STAT-3, Oct-3, and Beclin-1. RESULTS: Our results have demonstrated that the combined restoration of miR-424-5p and miR-142-3p is more effective in inhibiting tumor proliferation via upregulating Bax and Beclin-1 and downregulating Bcl-2 and c-Myc. Besides, the combined therapy has arrested the cell cycle in the sub-G1 and G2 phases and has suppressed the clonogenicity via downregulating STAT-3 and Oct-3, respectively. CONCLUSION: The combined restoration of miR-424-5p and miR-142-3p is more effective in inhibiting MCF-7 breast cancer development than monotherapies with miR-424-5p and miR-142-3p.


Assuntos
Apoptose , Autofagia , Neoplasias da Mama , Ciclo Celular , MicroRNAs , Apoptose/genética , Autofagia/genética , Proteína Beclina-1/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Células MCF-7 , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
7.
Life Sci ; 297: 120466, 2022 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-35271882

RESUMO

Dendritic cells (DCs) can present tumoral antigens to T-cells and stimulate T-cell-mediated anti-tumoral immune responses. In addition to uptaking, processing, and presenting tumoral antigens to T-cells, co-stimulatory signals have to be established between DCs with T-cells to develop anti-tumoral immune responses. However, most of the tumor-infiltrated immune cells are immunosuppressive in the tumor microenvironment (TME), paving the way for immune evasion of tumor cells. This immunosuppressive TME has also been implicated in suppressing the DC-mediated anti-tumoral immune responses, as well. Various factors, i.e., immunoregulatory cells, metabolic factors, tumor-derived immunosuppressive factors, and inhibitory immune checkpoint molecules, have been implicated in developing the immunosuppressive TME. Herein, we aimed to review the biology of DCs in developing T-cell-mediated anti-tumoral immune responses, the significance of immunoregulatory cells in the TME, metabolic barriers contributing to DCs dysfunction in the TME, tumor-derived immunosuppressive factors, and inhibitory immune checkpoint molecules in DC-based cell therapy outcomes. With reviewing the ongoing clinical trials, we also proposed a novel therapeutic strategy to increase the efficacy of DC-based cell therapy. Indeed, the combination of DC-based cell therapy with monoclonal antibodies against novel immune checkpoint molecules can be a promising strategy to increase the response rate of patients with cancers.


Assuntos
Inibidores de Checkpoint Imunológico , Neoplasias , Células Dendríticas , Humanos , Inibidores de Checkpoint Imunológico/uso terapêutico , Imunoterapia , Neoplasias/metabolismo , Microambiente Tumoral
8.
Immunotherapy ; 14(6): 475-488, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35232264

RESUMO

Gastric cancer (GC) is one of the most frequently diagnosed malignancies. Recent studies have highlighted cellular immunotherapy (CI) as a promising approach for treating this disease. Among the CI-based approaches, adoptive cell therapy and dendritic cell-based vaccination are commonly studied in preclinical and clinical trials. Here we review the current evidence on the potentiality of CI in treating GC, the targets for adoptive cell therapy, ongoing clinical trials, constraints and the future outlook. The results suggest that there is a need to identify novel biomarkers that predict which GC patients will most likely respond to these approaches. Also, CI plus chemotherapy or immune checkpoint inhibitors can improve the survival of patients with late-stage GC. Therefore, this approach can be promising for treating these patients.


Assuntos
Neoplasias Gástricas , Terapia Baseada em Transplante de Células e Tecidos , Células Dendríticas , Humanos , Imunoterapia/métodos , Imunoterapia Adotiva/métodos , Neoplasias Gástricas/terapia , Vacinação
9.
Biomed Pharmacother ; 146: 112588, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35062062

RESUMO

Tumor-associated macrophages (TAMs) are among the abundant cell populations of the tumor microenvironment (TME), which have pivotal roles in tumor development, chemoresistance, immune evasion, and metastasis. Growing evidence indicates that TAMs and the cross-talk between TAMs and tumoral endothelial cells can substantially contribute to tumor angiogenesis, which is considered a vital process for cancer development. Besides, tumoral endothelial cells can regulate the leukocyte infiltration to the TME in solid cancers and contribute to immune evasion. Therefore, targeting the immunosuppressive TAMs and the cross-talk between them can be a promising strategy for improving anti-tumoral immune responses. This review aims to summarize the biology of TAMs, their recently identified roles in tumor development/angiogenesis, and recent advances in macrophage-based cancer immunotherapy approaches for treating cancers.


Assuntos
Imunoterapia/métodos , Neoplasias/imunologia , Microambiente Tumoral , Macrófagos Associados a Tumor/metabolismo , Antineoplásicos Imunológicos/uso terapêutico , Células Endoteliais , Humanos , Neoplasias/tratamento farmacológico , Receptor Cross-Talk , Macrófagos Associados a Tumor/patologia
10.
Eur J Clin Nutr ; 76(5): 647-658, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34584225

RESUMO

Taurine (Tau) has modulatory effects on inflammatory and oxidative stress biomarkers; however, the results of clinical studies are not comprehensive enough to determine the effect of different durations and doses of Tau supplementation on inflammatory and oxidative stress biomarkers. The current study was conducted based on the preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines. For this purpose, PubMed/Medline, Scopus, and Embase databases were systematically searched to obtain the relevant studies published before 30th March 2021. Meta-analysis was performed on controlled clinical trials by using the random-effects method. Non-linear relationship between variables and effect size was performed using dose-response and time-response analyses. The Cochrane Collaboration's tool was used to evaluate the quality of included studies. Tau supplementation can reduce the levels of malondialdehyde (MDA) (SMD = -1.17 µmol/l; 95% CI: -2.08, - 0.26; P = 0.012) and C-reactive protein (CRP) (SMD = -1.95 mg/l; 95% CI: -3.20, - 0.71; P = 0.002). There have been no significant effects of Tau supplementation on the levels of tumor necrosis factors-alpha (TNF-α) (SMD = -0.18 pg/ml; 95% CI: -0.56, 0.21; P = 0.368), and interleukin-6 (IL-6) (SMD = -0.49 pg/ml; 95% CI: -1.13, 0.16; P = 0.141). Besides, Tau has more alleviating effect on oxidative stress and inflammation on 56 days after supplementation (P < 0.05). Tau can decrease the levels of CRP and MDA. Based on the currently available evidence, Tau has no significant effect on the level of TNF-α and IL-6. Eight-week of Tau supplementation has more beneficial effects on inflammatory and oxidative stress biomarkers.


Assuntos
Interleucina-6 , Taurina , Biomarcadores/metabolismo , Proteína C-Reativa/metabolismo , Suplementos Nutricionais , Humanos , Inflamação/tratamento farmacológico , Estresse Oxidativo , Taurina/farmacologia , Taurina/uso terapêutico , Fator de Necrose Tumoral alfa
11.
Cell Mol Neurobiol ; 42(8): 2849-2861, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34661780

RESUMO

Alzheimer's disease (AD), the most frequently diagnosed dementia, is a senile neurodegenerative disorder characterized by amnesia and cognitive dysfunction. Unfortunately, there are still no successful strategies to prevent AD progression. Thus, the vast majority of research focuses on recognizing risk factors for developing and progressing this disease. Human spirochetes, fungi, Borrelia burgdorferi, Chlamydophila pneumoniae, Helicobacter pylori, and human herpes simplex virus type 1 (HSV-1) have all been implicated in the development and progression of AD. Identifying microRNAs (miRs) encoded by DNA viruses has indicated that viruses can be evolved to exploit RNA silencing to regulate host and viral genes. Similar to host miR, v-miR can interact with the 3' untranslated region (UTR) of the target mRNA to regulate gene expression. Although HSV-1 can also encode various miRs, their significance in the development and progression of AD is still unclear. In the present study, utilizing the bioinformatics approach (R software and related packages), we analyzed the differentially expressed genes (DEGs) in AD samples (grey matter) of GSE37263 dataset obtained from the NCBI Gene Expression Omnibus (GEO). Then, the sequences of HSV-1-encoded-miRs were retrieved from miRbase, and their targets were predicted by miRDB. Afterward, the common genes between downregulated DEGs in AD and targets of HSV-1-encoded miRs were identified to shed new light on the relationship between HSV-1 infection and AD development. Our results have indicated that HSV-1-encoded-miRs can target the downregulated DEGs in AD, and these aberrant interactions can offer valuable diagnostic/prognostic biomarkers for affected patients.


Assuntos
Doença de Alzheimer , Herpes Simples , Herpesvirus Humano 1 , MicroRNAs , Regiões 3' não Traduzidas , Doença de Alzheimer/genética , Biomarcadores/metabolismo , Herpes Simples/genética , Herpesvirus Humano 1/genética , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Mensageiro/metabolismo
12.
Curr Med Chem ; 29(19): 3471-3482, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34781858

RESUMO

As a novel group of non-coding RNAs, long non-coding RNA (lncRNAs) can substantially regulate various biological processes. Downregulated tumor-suppressive lncRNAs and upregulated oncogenic lncRNAs (onco-lncRNAs) have been implicated in gastric cancer (GC) development. These dysregulations have been associated with decreased chemosensitivity, inhibited apoptosis, and increased tumor migration in GC. Besides, growing evidence indicates that lncRNAs can be a valuable diagnostic and prognostic biomarker, and their expression levels are substantially associated with the clinicopathological features of affected patients. The current study aims to review the recent findings of the tumor-suppressive lncRNAs and onco-lncRNAs in GC development and highlight their therapeutic, diagnostic, and prognostic values in treating GC cells. Besides, it intends to highlight the future direction of lncRNAs in treating GC.


Assuntos
RNA Longo não Codificante , Neoplasias Gástricas , Biomarcadores Tumorais/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Prognóstico , RNA Longo não Codificante/genética , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/genética
13.
Biomed Pharmacother ; 146: 112537, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34922114

RESUMO

BACKGROUND: Ovarian cancer (OC) is one of the worrisome gynecological cancers worldwide. Given its considerable mortality rate, it is necessary to investigate its oncogenesis. METHODS: In this study, we used systems biology approaches to describe the key gene modules, hub genes, and regulatory drugs associated with serous OC as the novel biomarkers using weighted gene co-expression network analysis (WGCNA). FINDINGS: Our findings have demonstrated that the blue module genes (r = 0.8, p-value = 1e-16) are involved in OC progression. Based on gene enrichment analysis, the genes in this module are frequently involved in biological processes such as the Cyclic adenosine monophosphate (cAMP) signaling pathway and the cellular response to transforming growth factor-beta stimulation. The co-expression network has been built using the correlated module's top hub genes, which are ADORA1, ANO9, CD24P4, CLDN3, CLDN7, ELF3, KLHL14, PRSS8, RASAL1, RIPK4, SERINC2, and WNT7A. Finally, a drug-target network has been built to show the interaction of the FDA-approved drugs with hub genes. CONCLUSIONS: Our results have discovered that ADORA1, ANO9, SERINC2, and KLHL14 are hub genes associated with serous OC. These genes can be considered as novel candidate target genes for treating OC.


Assuntos
Redes Reguladoras de Genes , Neoplasias Ovarianas , Claudinas , Bases de Dados Genéticas , Perfilação da Expressão Gênica/métodos , Humanos , Proteínas de Membrana/genética , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Biologia de Sistemas
14.
Biomed Pharmacother ; 145: 112370, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34862113

RESUMO

AIMS: Besides suppressing anti-tumoral immune responses, tumor-intrinsic inhibitory immune checkpoints have been implicated in tumor development. Herein, we aimed to investigate the significance of tumor-intrinsic CD73, as an inhibitory immune checkpoint, in non-small cell lung cancer (NSCLC) development and propose a novel therapeutic approach. MAIN METHODS: We investigated the cell viability, chemosensitivity, apoptosis, migration, and the cell cycle of A-549 and NCI-H1299 following treatment with cisplatin and CD73-small interfering RNA (siRNA) transfection. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay was used to study the viability of studied groups and chemosensitivity of tumoral cells. Flow cytometry and 4',6-diamidino-2-phenylindole (DAPI) staining were used to investigate the apoptosis of NSCLC cells. Flow cytometry and the wound-healing assay were used to investigate the cell cycle and migration of NSCLC cells, respectively. The mRNA expression levels of c-Myc, caspase 3, ROCK, and MMP-9 were investigated to study the underlying molecular mechanism. KEY FINDINGS: CD73-siRNA transfection has significantly decreased the cell viability and enhanced the chemosensitivity of A-549 and NCI-H1299 cells to cisplatin. CD73-siRNA has considerably stimulated apoptosis, arrested the cell cycle, inhibited tumor migration, downregulated the mRNA expression of c-Myc, MMP-9, and ROCK, and upregulated caspase 3 expression in NSCLC cells. Besides, combined cisplatin therapy with CD73-siRNA transfection has potentiated the aforementioned anti-tumoral effects of cisplatin on NSCLC cells. SIGNIFICANCE: Besides suppressing anti-tumoral immune responses, tumor-intrinsic CD73 can facilitate NSCLC development, and the combined cisplatin therapy with CD73-siRNA transfection can substantially enhance the chemosensitivity of NSCLC to cisplatin and potentiates cisplatin-induced anti-tumoral effects on NSCLC.


Assuntos
5'-Nucleotidase/genética , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Cisplatino/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Células A549 , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Proteínas Ligadas por GPI/genética , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Humanos , Neoplasias Pulmonares/genética , RNA Interferente Pequeno/genética , Transfecção
15.
Life (Basel) ; 11(12)2021 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-34947886

RESUMO

Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-related death worldwide. HCC patients may benefit from liver transplantation, hepatic resection, radiofrequency ablation, transcatheter arterial chemoembolization, and targeted therapies. The increased infiltration of immunosuppressive immune cells and the elevated expression of immunosuppressive factors in the HCC microenvironment are the main culprits of the immunosuppressive nature of the HCC milieu. The immunosuppressive tumor microenvironment can substantially attenuate antitumoral immune responses and facilitate the immune evasion of tumoral cells. Immunotherapy is an innovative treatment method that has been promising in treating HCC. Immune checkpoint inhibitors (ICIs), adoptive cell transfer (ACT), and cell-based (primarily dendritic cells) and non-cell-based vaccines are the most common immunotherapeutic approaches for HCC treatment. However, these therapeutic approaches have not generally induced robust antitumoral responses in clinical settings. To answer to this, growing evidence has characterized immune cell populations and delineated intercellular cross-talk using single-cell RNA sequencing (scRNA-seq) technologies. This review aims to discuss the various types of tumor-infiltrating immune cells and highlight their roles in HCC development. Besides, we discuss the recent advances in immunotherapeutic approaches for treating HCC, e.g., ICIs, dendritic cell (DC)-based vaccines, non-cell-based vaccines, oncolytic viruses (OVs), and ACT. Finally, we discuss the potentiality of scRNA-seq to improve the response rate of HCC patients to immunotherapeutic approaches.

16.
Med Oncol ; 39(1): 9, 2021 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-34761351

RESUMO

The deregulation of microRNAs (miRs) has been identified in tumor development. Indeed, the restoration of tumor-suppressive miRs has been associated with inhibited tumor development in various cancers. Herein, we aimed to evaluate the impact of combined miR-383-5p restoration, as a tumor-suppressive miR, with taxol therapy in suppressing MDA-MB-231 breast cancer development. MDA-MB-231 cell line was restored with miR-383-5p and treated with paclitaxel both in combined and separate manners. The MTT experiment was carried out to measure the cytotoxicity of the therapeutic approaches on the tumoral cells. Besides, flow cytometry was conducted to assess apoptosis and cell cycle status following the treatments. Furthermore, the expression levels of critical factors contributed to tumor proliferation, migration, apoptosis were investigated via the qRT-PCR and western blotting techniques. The outcomes pointed out that the miR-383-5p might substantially enhance the chemosensitivity of MDA-MB-231 to taxol. Besides, miR-383-5p restoration and the combined therapy of miR-383-5p restoration with paclitaxel could remarkably increase apoptosis, decrease cell viability, arrest the cell cycle, inhibit clonogenicity, suppress tumor migration, suppress the PI3K/Akt signaling pathway, and down-regulate PD-L1 expression of BC cells. The restoration of miR-383-5p can enhance the chemosensitivity of MDA-MB-231 cells to taxol. Despite the anti-tumoral effects of miR-383-5p restoration on MDA-MB-231 breast cancer development, the combined therapy of miR-383-5p restoration with paclitaxel can be more effective in repressing MDA-MB-231 breast cancer development.


Assuntos
Neoplasias da Mama , MicroRNAs , Paclitaxel/farmacologia , Apoptose/efeitos dos fármacos , Antígeno B7-H1/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Feminino , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , MicroRNAs/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
17.
Molecules ; 26(22)2021 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-34833970

RESUMO

Photodynamic therapy (PDT) is a light-based cancer therapy approach that has shown promising results in treating various malignancies. Growing evidence indicates that cancer stem cells (CSCs) are implicated in tumor recurrence, metastasis, and cancer therapy resistance in colorectal cancer (CRC); thus, targeting these cells can ameliorate the prognosis of affected patients. Based on our bioinformatics results, SOX2 overexpression is significantly associated with inferior disease-specific survival and worsened the progression-free interval of CRC patients. Our results demonstrate that zinc phthalocyanine (ZnPc)-PDT with 12 J/cm2 or 24 J/cm2 irradiation can substantially decrease tumor migration via downregulating MMP9 and ROCK1 and inhibit the clonogenicity of SW480 cells via downregulating CD44 and SOX2. Despite inhibiting clonogenicity, ZnPc-PDT with 12 J/cm2 irradiation fails to downregulate CD44 expression in SW480 cells. Our results indicate that ZnPc-PDT with 12 J/cm2 or 24 J/cm2 irradiation can substantially reduce the cell viability of SW480 cells and stimulate autophagy in the tumoral cells. Moreover, our results show that ZnPc-PDT with 12 J/cm2 or 24 J/cm2 irradiation can substantially arrest the cell cycle at the sub-G1 level, stimulate the intrinsic apoptosis pathway via upregulating caspase-3 and caspase-9 and downregulating Bcl-2. Indeed, our bioinformatics results show considerable interactions between the studied CSC-related genes with the studied migration- and apoptosis-related genes. Collectively, the current study highlights the potential role of ZnPc-PDT in inhibiting stemness and CRC development, which can ameliorate the prognosis of CRC patients.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Isoindóis/farmacologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Compostos Organometálicos/farmacologia , Fármacos Fotossensibilizantes/farmacologia , Compostos de Zinco/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Neoplasias Colorretais/patologia , Humanos , Células-Tronco Neoplásicas/patologia , Fotoquimioterapia
18.
Int J Mol Sci ; 22(19)2021 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-34638729

RESUMO

Preclinical studies have indicated that T-cell immunoglobulin and ITIM domain (TIGIT) can substantially attenuate anti-tumoral immune responses. Although multiple clinical studies have evaluated the significance of TIGIT in patients with solid cancers, their results remain inconclusive. Thus, we conducted the current systematic review and meta-analysis based on the preferred reporting items for systematic reviews and meta-analyses (PRISMA) to determine its significance in patients with solid cancers. We systematically searched the Web of Science, Embase, PubMed, and Scopus databases to obtain peer-reviewed studies published before September 20, 2020. Our results have shown that increased TIGIT expression has been significantly associated with inferior overall survival (OS) (HR = 1.42, 95% CI: 1.11-1.82, and p-value = 0.01). Besides, the level of tumor-infiltrating TIGIT+CD8+ T-cells have been remarkably associated inferior OS and relapse-free survival (RFS) of affected patients (HR = 2.17, 95% CI: 1.43-3.29, and p-value < 0.001, and HR = 1.89, 95% CI: 1.36-2.63, and p-value < 0.001, respectively). Also, there is a strong positive association between TIGIT expression with programmed cell death-1 (PD-1) expression in these patients (OR = 1.71, 95% CI: 1.10-2.68, and p-value = 0.02). In summary, increased TIGIT expression and increased infiltration of TIGIT+CD8+ T-cells can substantially worsen the prognosis of patients with solid cancers. Besides, concerning the observed strong association between TIGIT and PD-1, ongoing clinical trials, and promising preclinical results, PD-1/TIGIT dual blockade can potentially help overcome the immune-resistance state seen following monotherapy with a single immune checkpoint inhibitor in patients with solid cancers.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos do Interstício Tumoral/imunologia , Proteínas de Neoplasias/imunologia , Neoplasias/imunologia , Receptor de Morte Celular Programada 1/imunologia , Receptores Imunológicos/imunologia , Evasão Tumoral , Linfócitos T CD8-Positivos/patologia , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Imunoterapia , Linfócitos do Interstício Tumoral/patologia , Neoplasias/patologia , Neoplasias/terapia
19.
Front Immunol ; 12: 734956, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34603316

RESUMO

Based on preclinical findings, programmed death-ligand 1 (PD-L1) can substantially attenuate CD8+ T-cell-mediated anti-tumoral immune responses. However, clinical studies have reported controversial results regarding the significance of the tumor-infiltrating CD8+ T-cells/PD-L1 axis on the clinical picture and the response rate of patients with high-grade glial tumors to anti-cancer therapies. Herein, we conducted a systematic review according to the preferred reporting items for systematic reviews and meta-analyses (PRISMA) statements to clarify the clinical significance of the tumor-infiltrating CD8+ T-cells/PD-L1 axis and elucidate the impact of this axis on the response rate of affected patients to anti-cancer therapies. Indeed, a better understanding of the impact of this axis on the response rate of affected patients to anti-cancer therapies can provide valuable insights to address the futile response rate of immune checkpoint inhibitors in patients with high-grade glial tumors. For this purpose, we systematically searched Scopus, Web of Science, Embase, and PubMed to obtain peer-reviewed studies published before 1 January 2021. We have observed that PD-L1 overexpression can be associated with the inferior prognosis of glioblastoma patients who have not been exposed to chemo-radiotherapy. Besides, exposure to anti-cancer therapies, e.g., chemo-radiotherapy, can up-regulate inhibitory immune checkpoint molecules in tumor-infiltrating CD8+ T-cells. Therefore, unlike unexposed patients, increased tumor-infiltrating CD8+ T-cells in anti-cancer therapy-exposed tumoral tissues can be associated with the inferior prognosis of affected patients. Because various inhibitory immune checkpoints can regulate anti-tumoral immune responses, the single-cell sequencing of the cells residing in the tumor microenvironment can provide valuable insights into the expression patterns of inhibitory immune checkpoints in the tumor micromovement. Thus, administrating immune checkpoint inhibitors based on the data from the single-cell sequencing of these cells can increase patients' response rates, decrease the risk of immune-related adverse events development, prevent immune-resistance development, and reduce the risk of tumor recurrence.


Assuntos
Antígeno B7-H1/metabolismo , Neoplasias Encefálicas/imunologia , Linfócitos T CD8-Positivos/imunologia , Glioma/imunologia , Linfócitos do Interstício Tumoral/imunologia , Medicina de Precisão , Evasão Tumoral , Microambiente Tumoral/imunologia , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/genética , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Tomada de Decisão Clínica , Resistencia a Medicamentos Antineoplásicos , Genômica , Glioma/tratamento farmacológico , Glioma/genética , Glioma/metabolismo , Humanos , Inibidores de Checkpoint Imunológico/uso terapêutico , Linfócitos do Interstício Tumoral/metabolismo , Gradação de Tumores , Transdução de Sinais , Análise de Célula Única , Microambiente Tumoral/genética
20.
Biomed Pharmacother ; 143: 112213, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34560556

RESUMO

Tumoral programmed cell death ligand 1 (PD-L1) has been implicated in the immune evasion and development of colorectal cancer. Although monoclonal immune checkpoint inhibitors can exclusively improve the prognosis of patients with microsatellite instability-high (MSI-H) and tumor mutational burden-high (TMB-H) colorectal cancer, specific tumor-suppressive microRNAs (miRs) can regulate multiple oncogenic pathways and inhibit the de novo expression of oncoproteins, like PD-L1, both in microsatellite stable (MSS) and MSI-H colorectal cancer cells. This scoping review aimed to discuss the currently available evidence regarding the therapeutic potentiality of PD-L1-inhibiting miRs for colorectal cancer. For this purpose, the Web of Science, Scopus, and PubMed databases were systematically searched to obtain peer-reviewed studies published before 17 March 2021. We have found that miR-191-5p, miR-382-3p, miR-148a-3p, miR-93-5p, miR-200a-3p, miR-200c-3p, miR-138-5p, miR-140-3p, and miR-15b-5p can inhibit tumoral PD-L1 in colorectal cancer cells. Besides inhibiting PD-L1, miR-140-3p, miR-382-3p, miR-148a-3p, miR-93-5p, miR-200a-3p, miR-200c-3p, miR-138-5p, and miR-15b-5p can substantially reduce tumor migration, inhibit tumor development, stimulate anti-tumoral immune responses, decrease tumor viability, and enhance the chemosensitivity of colorectal cancer cells regardless of the microsatellite state. Concerning the specific, effective, and safe delivery of these miRs, the single-cell sequencing-guided biocompatible-based delivery of these miRs can increase the specificity of miR delivery, decrease the toxicity of traditional nanoparticles, transform the immunosuppressive tumor microenvironment into the proinflammatory one, suppress tumor development, decrease tumor migration, and enhance the chemosensitivity of tumoral cells regardless of the microsatellite state.


Assuntos
Antígeno B7-H1/antagonistas & inibidores , Materiais Biocompatíveis/administração & dosagem , Neoplasias Colorretais/terapia , Técnicas de Transferência de Genes , Terapia Genética , Inibidores de Checkpoint Imunológico/uso terapêutico , MicroRNAs/administração & dosagem , Animais , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Resistencia a Medicamentos Antineoplásicos , Regulação Neoplásica da Expressão Gênica , Humanos , Inibidores de Checkpoint Imunológico/efeitos adversos , MicroRNAs/genética , MicroRNAs/metabolismo , Nanomedicina , Nanopartículas , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA