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













Base de datos
Intervalo de año de publicación
1.
Hum Immunol ; 85(3): 110798, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38569354

RESUMEN

BACKGROUND: Antigen-specific T-cell immunity is provided by dendritic cells (DCs), which are specialized antigen-presenting cells. Furthermore, they establish a link between innate and adaptive immune responses. Currently, DC modification is a new approach for the therapy of several disorders. During solid organ transplantation, Everolimus, which is a mammalian target of rapamycin (mTOR) inhibitor, was initially utilized to suppress the immune system's functionality. Due to the intervention of Everolimus in various signaling pathways in cells and its modulatory properties on the immune system, this study aims to investigate the effect of treatment with Everolimus on the maturation and expression of immune checkpoint genes in monocyte-derived DCs. METHODS: To isolate monocytes from PBMCs, the CD14 marker was used via the MACS method. Monocytes were cultured and induced to differentiate into monocyte-derived DCs by utilizing GM-CSF and IL-4 cytokines. On the fifth day, immature DCs were treated with Everolimus and incubated for 24 h. On the sixth day, the flow cytometry technique was used to investigate the effect of Everolimus on the phenotypic characteristics of DCs. In the end, the expression of immune checkpoint genes in both the Everolimus-treated and untreated DCs groups was assessed using the real-time PCR method. RESULTS: The findings of this research demonstrated that the administration of Everolimus to DCs led to a notable rise in human leukocyte antigen (HLA)-DR expression and a decrease in CD11c expression. Furthermore, there was a significant increase in the expression of immune checkpoint molecules, namely CTLA-4, VISTA, PD-L1, and BTLA, in DCs treated with Everolimus. CONCLUSION: The findings of this study show that Everolimus can target DCs and affect their phenotype and function in order to shift them toward a partially tolerogenic state. However, additional research is required to gain a comprehensive understanding of the precise impact of Everolimus on the activation status of DCs.

2.
Cytotherapy ; 26(5): 427-435, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38483358

RESUMEN

BACKGROUND AIMS: Acute myeloid leukemia (AML) is classified as a hematologic malignancy characterized by the proliferation of immature blood cells within the bone marrow (BM), resulting in an aberrant and unregulated cellular growth. The primary therapeutic modalities for AML include chemotherapy and hematopoietic stem cell transplantation. However, it is important to note that these treatments are accompanied by important adverse effects and mortality rates. Therefore, the need for more effective treatment options seems necessary, and dendritic cell (DC) vaccine therapy can be one of these options. In this study, we aim to investigate the effectiveness of DC vaccination therapy for the management of AML. METHODS: PubMed, Scopus, ProQuest, Web of Science, and Google Scholar databases were searched for this systematic review. The articles were evaluated based on the inclusion criteria of this study and initially compared in terms of titles or abstracts. Finally, the articles related to the topic of this review were obtained in full text. The complete remission and partial remission, survival, correlative immune assays, and health-related metrics were used to evaluate this cellular immunotherapy effectiveness. The quality of the studies was assessed independently using the Cochrane risk-of-bias tools. The compiled data were input into a standard Excel spreadsheet. Each domain was evaluated as having either a "low risk," "high risk," or "unclear risk" of bias. RESULTS: Among the 3986 studies that were determined, a total of 11 correlated trials were selected for inclusion in this systematic review. DC vaccine therapy was effective in inducing complete and partial remission, and stabilization of the disease. Additionally, it was discovered that the treatment strengthened the immune system as seen by increased levels of CD4+ and CD8+ T cells, Th1 cytokines, WT1-specific T cells, and activated NK cells. CONCLUSION: We conducted a systematic review that supports the use of DC vaccine therapy as an effective treatment for AML. The therapy demonstrated potentials in achieving remission, enhancing the immune system function, and increasing overall survival. However, more studies are required to improve the methods of preparing and delivering the DC vaccine, and to confirm its long-term safety and effectiveness.


Asunto(s)
Vacunas contra el Cáncer , Células Dendríticas , Leucemia Mieloide Aguda , Humanos , Leucemia Mieloide Aguda/terapia , Leucemia Mieloide Aguda/inmunología , Células Dendríticas/inmunología , Vacunas contra el Cáncer/uso terapéutico , Vacunas contra el Cáncer/inmunología , Vacunación/métodos , Inmunoterapia/métodos
3.
Pathol Res Pract ; 253: 155012, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38071887

RESUMEN

Pancreatic Ductal Adenocarcinoma (PDAC) ranks among the most prevalent gastrointestinal malignancies, with risk factors including smoking, alcohol abuse, diabetes mellitus, obesity, age, family history, and genetic predisposition. Extensive research has focused on unraveling biomarkers and molecular intricacies associated with PDAC. Leveraging data from the Gene Expression Omnibus microarray and single-cell RNA sequencing datasets, our study identified ITGB4 and C19orf33 as potentially differentially expressed genes in PDAC samples when contrasted with non-malignant tissues. Notably, these genes exhibited a strong correlative expression pattern, primarily within ductal cells. Gene Expression Profiling Interactive Analysis corroborated our findings, further confirming the correlation between ITGB4 and C19orf33. Additionally, we conducted experiments involving two pivotal PDAC-related cell lines, MIA PaCa-2 and PANC-1, treated with oxaliplatin and 5-Fluorouracil. We also assessed the expression of these candidate genes in PDAC samples in comparison to adjacent normal tissues. Our findings revealed that C19orf33 is upregulated in PDAC samples, and treatment of PDAC cells with chemotherapeutic agents led to a correlated decrease in the expression of both ITGB4 and C19orf33. These co-expressed and correlated genes are implicated in relevant signaling pathways, suggesting shared biological activities that may contribute to the promotion of metastasis within malignant ductal cells. This study identifies ITGB4 and C19orf33 as key genes potentially shedding light on the molecular mechanisms driving tumorigenesis and metastasis in PDAC. These genes hold promise as potential diagnostic and therapeutic targets, offering valuable insights into the management of this challenging disease.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias Pancreáticas/patología , Carcinoma Ductal Pancreático/patología , Perfilación de la Expresión Génica , Análisis de Secuencia de ARN , Regulación Neoplásica de la Expresión Génica , Integrina beta4/genética , Integrina beta4/metabolismo
4.
Ther Adv Chronic Dis ; 14: 20406223231153572, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37035097

RESUMEN

Auto-immune diseases are a form of chronic disorders in which the immune system destroys the body's cells due to a loss of tolerance to self-antigens. Systemic lupus erythematosus (SLE), identified by the production of autoantibodies in different body parts, is one of the most well-known examples of these diseases. Although the etiology of SLE is unclear, the disease's progression may be affected by genetic and environmental factors. As studies in twins provide adequate evidence for genetic involvement in the SLE, other phenomena such as metallization, histone modifications, and alterations in the expression of noncoding RNAs (ncRNAs) also indicate the involvement of epigenetic factors in this disease. Among all the epigenetic alterations, ncRNAs appear to have the most crucial contribution to the pathogenesis of SLE. The ncRNAs' length and size are divided into three main classes: micro RNAs, long noncoding RNAs (LncRNA), and circular RNAs (circRNAs). Accumulating evidence suggests that dysregulations in these ncRNAs contributed to the pathogenesis of SLE. Hence, clarifying the function of these groups of ncRNAs in the pathophysiology of SLE provides a deeper understanding of the disease. It also opens up new opportunities to develop targeted therapies for this disease.

5.
Biodegradation ; 34(4): 325-340, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36840888

RESUMEN

Tetracycline (TC) is a widely used antibiotic with a complex aromatic chemical structure and is highly resistant to biodegradation. In this study, an SBR equipped with a vertical axially rotating biological bed (SBR-VARB) was used for the biodegradation and mineralization of TC. SBR-VARB showed high efficiency in removing TC (97%), total phenolic compounds (TP) (95%), and COD (85%) under optimal operating conditions (TC = 50 mg/L, HRT = 1.75 d, and OLR = 36 g COD/m3 d). The SBR-VARB was able to treat higher concentrations of TC in shorter HRT than reported in previous studies. The contribution of VARB to improve SBR efficiency in removing TC, TP, and COD was 16, 36, and 48%, respectively. Intermediate compounds formed during the biodegradation of TC were identified using GC-MS under the optimal operating conditions of the bioreactor. These are mainly organic compounds with linear chemical structures. Based on the complete biodegradation of TC under the optimal operating conditions of the bioreactor, 93% and 36% of the chlorine and nitrogen atoms in the chemical structure of TC appeared in the wastewater, respectively. According to the sequence analysis of 16SrDNA, Pseudomonas sp., Kocuria Polaris, and Staphylococcus sp. were identified in the biofilm of VARB and the suspended biomass of the bioreactor. Therefore, SBR-VARB showed high efficiency in the biodegradation and mineralization of TC and can be used as a suitable option for treating wastewater containing antibiotics and other toxic compounds.


Asunto(s)
Compuestos Heterocíclicos , Aguas Residuales , Antibacterianos/metabolismo , Tetraciclina/metabolismo , Compuestos Orgánicos , Reactores Biológicos , Eliminación de Residuos Líquidos , Biodegradación Ambiental
6.
Gene ; 855: 147086, 2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-36535461

RESUMEN

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.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Humanos , Paclitaxel/farmacología , Caspasa 3 , Proteína X Asociada a bcl-2 , Línea Celular Tumoral , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/genética , Osteosarcoma/metabolismo , Apoptosis/genética , Neoplasias Óseas/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Inmunoglobulinas/metabolismo , Inmunoglobulinas/farmacología
7.
Curr Mol Med ; 23(6): 521-526, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35692146

RESUMEN

Breast cancer (BC) is one of the prevalent diseases and causes of death in women, and its incidence rate is increasing in numerous developed and developing countries. The common approach to BC therapy is surgery, followed by radiation therapy or chemotherapy, which doesn't lead to acceptable outcomes in many patients. Therefore, developing innovative strategies for treating BC is essential for the most effective therapy. The immunotherapy of BC is a promising and attractive strategy that can increase the immune system's capacity to recognize and kill the tumor cells, inhibit the recurrence of the tumors, and develop new metastatic sites. The blockade of immune checkpoints is the most attractive and promising strategy for cancer immunotherapy. The cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) is a cellsurface glycoprotein expressed by stimulated T cells and has pivotal roles in cell cycle modulation, cytokine generation, and regulation of T cell proliferation. Currently, anti- CTLA-4 agents such as monoclonal antibodies (Ipilimumab and tremelimumab) are broadly applied as therapeutic agents in clinical studies of different cancers. The anti- CTLA-4 antibodies, alone or combined with other therapeutic agents, remarkably increased the tumor-suppressive effects of the immune system and improved the prognosis of cancer. The immune checkpoint inhibitors may represent promising options for BC treatment as in monotherapy or in combination with other conventional treatments. In this review, we discuss the role of CTLA-4 and its therapeutic potential by inhibitors of immune checkpoints in BC therapeutics.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Antígeno CTLA-4 , Neoplasias de la Mama/tratamiento farmacológico , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Linfocitos T , Citocinas , Inmunoterapia
8.
Adv Med Sci ; 67(2): 353-363, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36116207

RESUMEN

Antigen recognition and presentation are highlighted as the first steps in developing specialized antigen responses. Dendritic cells (DCs) are outstanding professional antigen-presenting cells (APCs) responsible for priming cellular immunity in pathological states, including cancer. However, the diminished or repressed function of DCs is thought to be a substantial mechanism through which tumors escape from the immune system. In this regard, DCs obtained from breast cancer (BC) patients represent a notably weakened potency to encourage specific T-cell responses. Additionally, impaired DC-T-cell cross-talk in BC facilitates the immune evade of cancer cells and is connected with tumor advancement, immune tolerance, and adverse prognosis for patients. In this review we aim to highlight the available knowledge on DC-T-cell interactions in BC aggressiveness and show its therapeutic potential in BC treatment.


Asunto(s)
Neoplasias de la Mama , Linfocitos T , Humanos , Femenino , Células Dendríticas
9.
Molecules ; 27(11)2022 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-35684481

RESUMEN

It is estimated that there were 18.1 million cancer cases worldwide in 2018, with about 9 million deaths. Proper diagnosis of cancer is essential for its effective treatment because each type of cancer requires a specific treatment procedure. Cancer therapy includes one or more approaches such as surgery, radiotherapy, chemotherapy, and immunotherapy. In recent years, immunotherapy has received much attention and immune checkpoint molecules have been used to treat several cancers. These molecules are involved in regulating the activity of T lymphocytes. Accumulated evidence shows that targeting immune checkpoint regulators like PD-1/PD-L1 and CTLA-4 are significantly useful in treating cancers. According to studies, these molecules also have pivotal roles in the chemoresistance of cancer cells. Considering these findings, the combination of immunotherapy and chemotherapy can help to treat cancer with a more efficient approach. Among immune checkpoint molecules, the B7 family checkpoints have been studied in various cancer types such as breast cancer, myeloma, and lymphoma. In these cancers, they cause the cells to become resistant to the chemotherapeutic agents. Discovering the exact signaling pathways and selective targeting of these checkpoint molecules may provide a promising avenue to overcome cancer development and therapy resistance. Highlights: (1) The development of resistance to cancer chemotherapy or immunotherapy is the main obstacle to improving the outcome of these anti-cancer therapies. (2) Recent investigations have described the involvement of immune checkpoint molecules in the development of cancer therapy resistance. (3) In the present study, the molecular participation of the B7 immune checkpoint family in anticancer therapies has been highlighted. (4) Targeting these immune checkpoint molecules may be considered an efficient approach to overcoming this obstacle.


Asunto(s)
Antineoplásicos , Neoplasias , Receptor de Muerte Celular Programada 1 , Antineoplásicos/uso terapéutico , Humanos , Proteínas de Punto de Control Inmunitario , Inmunoterapia/métodos , Neoplasias/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores
10.
Exp Mol Pathol ; 125: 104753, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35235816

RESUMEN

BACKGROUND: Breast cancer (BC) has been known as the most common type of cancer worldwide and resulted in death among women. BC is usually resistant to standard therapies that are significant problems in managing BC patients. miR-200c belongs to the miRNA family, which is considered as a tumor suppressor with reduced expression levels in various kinds of cancer like BC. Increased expression of miR-200c has been reported as a potent inhibitor of drug resistance and tumor advancement. The purpose of this paper is to examine the outcome of miR-200c restoring on enhancing the BC cells' sensitivity to Doxorubicin through downregulating the MDR1 expression. METHODS: Initially, MDA-MB-231 cells were transfected with miR-200c to perform functional analyses. After that, MTT assay was performed to investigate the viability of the cell. Finally, qRT-PCR was used to assess gene expression. RESULTS: According to the results, the miR-200c expression was downregulated in BC cells compared to control. Moreover, the cell viability was reduced in transfected cells via regulation in gene expression associated with apoptosis. Furthermore, miR-200c could increase the BC cells' sensitivity to Doxorubicin by reducing the MDR1 gene expression. CONCLUSION: Hence, this study's findings recommend that miR-200c can consider as a method of therapy for the treatment of BC.


Asunto(s)
Neoplasias de la Mama , Doxorrubicina , MicroARNs , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Línea Celular Tumoral , Doxorrubicina/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/genética
11.
Biomed Pharmacother ; 148: 112735, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35193040

RESUMEN

Autophagy is conserved cellular machinery that degrades un-usable proteins and cellular components and has a crucial role in the pathogenesis and drug resistance of various diseases such as lung cancer (LC). Multiple types of endogenous molecules (i.e. miRNAs) have been found to regulate multiple biological processes, such as autophagy. Dysfunction of these molecules is associated with the onset and progression of a variety of human malignancies. Several studies had shown that some miRNAs could mediate autophagy activity in LC cells, which would affect drug resistance as a major problem in LC therapy. Therefore, identifying the underlying molecular targets of miRNAs and their function in autophagy pathways could develop new treatment interventions for LC patients. In this review, we will summarize the interplay between miRNAs, autophagy, and drug resistance of LC patients, as well as the genes and molecular pathways that are involved.


Asunto(s)
Autofagia/genética , Resistencia a Antineoplásicos/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , MicroARNs/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/metabolismo , MicroARNs/metabolismo , Transducción de Señal/genética
12.
Int J Rheum Dis ; 25(3): 259-271, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34994525

RESUMEN

Autoimmune diseases, especially among young people in the US, are one of the leading causes of morbidity and death. The immune responses are the fundamental pathogenicity of autoimmune disorders. The equilibrium between stimulatory and inhibitory signals is critical for the stimulation, migration, survival, and T cell-related immune responses. The B7 family can substantially regulate T cell-mediated immune responses. Nevertheless, recent breakthroughs in immune checkpoint blockade in cancer immunotherapy have facilitated autoimmune diseases, especially among the prone populations. In the current study, we tried to concisely review the role of the B7 family in regulating immune reactions and the influence of immune checkpoint inhibitors on autoimmunity development.


Asunto(s)
Enfermedades Autoinmunes/terapia , Autoinmunidad , Antígeno B7-H1/farmacología , Inmunoterapia/métodos , Linfocitos T/inmunología , Enfermedades Autoinmunes/inmunología , Humanos
13.
Front Immunol ; 12: 734956, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34603316

RESUMEN

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.


Asunto(s)
Antígeno B7-H1/metabolismo , Neoplasias Encefálicas/inmunología , Linfocitos T CD8-positivos/inmunología , Glioma/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Medicina de Precisión , Escape del Tumor , Microambiente Tumoral/inmunología , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/genética , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Linfocitos T CD8-positivos/metabolismo , Toma de Decisiones Clínicas , Resistencia a Antineoplásicos , Genómica , Glioma/tratamiento farmacológico , Glioma/genética , Glioma/metabolismo , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Linfocitos Infiltrantes de Tumor/metabolismo , Clasificación del Tumor , Transducción de Señal , Análisis de la Célula Individual , Microambiente Tumoral/genética
14.
Int J Mol Sci ; 22(19)2021 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-34638729

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Receptores Inmunológicos/inmunología , Escape del Tumor , Linfocitos T CD8-positivos/patología , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Inmunoterapia , Linfocitos Infiltrantes de Tumor/patología , Neoplasias/patología , Neoplasias/terapia
15.
Molecules ; 26(8)2021 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-33920054

RESUMEN

Over the past decade, there have been remarkable advances in understanding the signaling pathways involved in cancer development. It is well-established that cancer is caused by the dysregulation of cellular pathways involved in proliferation, cell cycle, apoptosis, cell metabolism, migration, cell polarity, and differentiation. Besides, growing evidence indicates that extracellular matrix signaling, cell surface proteoglycans, and angiogenesis can contribute to cancer development. Given the genetic instability and vast intra-tumoral heterogeneity revealed by the single-cell sequencing of tumoral cells, the current approaches cannot eliminate the mutating cancer cells. Besides, the polyclonal expansion of tumor-infiltrated lymphocytes in response to tumoral neoantigens cannot elicit anti-tumoral immune responses due to the immunosuppressive tumor microenvironment. Nevertheless, the data from the single-cell sequencing of immune cells can provide valuable insights regarding the expression of inhibitory immune checkpoints/related signaling factors in immune cells, which can be used to select immune checkpoint inhibitors and adjust their dosage. Indeed, the integration of the data obtained from the single-cell sequencing of immune cells with immune checkpoint inhibitors can increase the response rate of immune checkpoint inhibitors, decrease the immune-related adverse events, and facilitate tumoral cell elimination. This study aims to review key pathways involved in tumor development and shed light on single-cell sequencing. It also intends to address the shortcomings of immune checkpoint inhibitors, i.e., their varied response rates among cancer patients and increased risk of autoimmunity development, via applying the data from the single-cell sequencing of immune cells.


Asunto(s)
Inmunoterapia , Neoplasias/inmunología , Neoplasias/terapia , Oncogenes , Análisis de Secuencia de ADN , Transducción de Señal , Análisis de la Célula Individual , Animales , Humanos , Neoplasias/genética , Neoplasias/patología , Transducción de Señal/genética
16.
Int J Mol Sci ; 22(5)2021 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-33800752

RESUMEN

The members of the B7 family, as immune checkpoint molecules, can substantially regulate immune responses. Since microRNAs (miRs) can regulate gene expression post-transcriptionally, we conducted a scoping review to summarize and discuss the regulatory cross-talk between miRs and new B7 family immune checkpoint molecules, i.e., B7-H3, B7-H4, B7-H5, butyrophilin like 2 (BTNL2), B7-H6, B7-H7, and immunoglobulin like domain containing receptor 2 (ILDR2). The current study was performed using a six-stage methodology structure and Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guideline. PubMed, Embase, Scopus, Cochrane, ProQuest, and Google Scholar were systematically searched to obtain the relevant records to 5 November 2020. Two authors independently reviewed the obtained records and extracted the desired data. After quantitative and qualitative analyses, we used bioinformatics approaches to extend our knowledge about the regulatory cross-talk between miRs and the abovementioned B7 family members. Twenty-seven articles were identified that fulfilled the inclusion criteria. Studies with different designs reported gene-miR regulatory axes in various cancer and non-cancer diseases. The regulatory cross-talk between the aforementioned B7 family molecules and miRs might provide valuable insights into the pathogenesis of various human diseases.


Asunto(s)
Antígenos B7/fisiología , Proteínas de Punto de Control Inmunitario/fisiología , MicroARNs/fisiología , Antígenos B7/genética , Antígenos B7/inmunología , Coagulación Sanguínea , Quimiocinas/fisiología , Activación de Complemento , Biología Computacional/métodos , Enfermedad/genética , Redes Reguladoras de Genes , Humanos , Proteínas de Punto de Control Inmunitario/genética , Proteínas de Punto de Control Inmunitario/inmunología , MicroARNs/genética , Transducción de Señal
17.
Adv Pharm Bull ; 10(2): 264-270, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32373495

RESUMEN

Purpose: Helicobacter pylori is recognized as one of the prevalent causes of human gastricinfection. In the present study, the role of mixed immunization with H. pylori lipopolysaccharide(LPS) and recombinant cytotoxin-associated gene A (rCagA) as a stimulator of host immuneresponses was determined. Methods: BALB/c mice were immunized with different formulations by the systemic administrationat 14-day intervals. The effects of the formulations plus CpG adjuvants were assessed before andpost-immunization in separated studies. Moreover, the expression of Th1/Th2 cytokines wasquantified in sera of immunized mice using reverse transcription polymerase chain reaction (RTPCR)test and the protein levels confirmed with enzyme linked immunosorbent assay (ELISA).Finally, the specific antibody levels in sera were studied by ELISA and the tendency of cellularresponse was examined by IgG1/IgG2a ratio. Results: Data of Western blotting verified the presence of constructed protein. Analysisof lymphocyte proliferation showed that CpG-conjugated rCagA increases lymphocytesproliferation compared to the control group. Also, it was shown that formulations containing LPSand rCagA promote a Th1 response indicated by interferon-gamma expression and induced Th1/Th2 balance. Additionally, the specific IgG1, total IgG and IgG2a levels elevated in response toall treatments. Ultimately, the IgG2a/IgG1 ratio in the mice immunized with rCagA-containingformulations increased. Conclusion: These results indicated that rCagA protein carried with CpG adjuvant not onlymaintained its antigenicity throughout the experiment but also induced robust Th1-biasedimmune responses. Therefore, it holds promise for the production of an efficient vaccine against H. pylori infection.

18.
J Gastrointest Cancer ; 51(1): 267-270, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31073863

RESUMEN

BACKGROUND: Genetic and epigenetic changes have strong role in the development of gastric cancer. The mutation of the MIR129-2 gene is one of the major causes in many cancers, especially gastric cancer. The aim of this study was to investigate the methylation changes of the MIR129-2 gene in tumor and normal tissue of patients with gastric cancer. METHOD: In this study, 50 gastric cancer patients with Iranian Azari ethnic origin without any familial relations were included. Genomic DNAs was extracted from the tumoral and normal tissues. Then the promotor regions of the MIR129-2 gene were analyzed by methylation-specific PCR (MSP) to evaluate the presence or absence of methylated CpG sites. RESULTS: There was a statistically significant difference in methylation level of MIR129-2 gene between tumoral and normal tissues. It was observed that 84 out of 100 CpG cites were methylated in tumoral tissues in compression to 13 out of 100 CpG cites in normal tissues. CONCLUSION: MIR129-2 gene was hypermethylated in tumoral tissues, suggesting that methylation is involved in the development of gastric cancer.


Asunto(s)
Metilación de ADN , MicroARNs/genética , Neoplasias Gástricas/genética , Femenino , Humanos , Masculino , MicroARNs/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología
19.
J Cell Biochem ; 120(6): 8863-8883, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30556298

RESUMEN

For many years, cancer therapy has appeared to be a challenging issue for researchers and physicians. By the introduction of novel methods in immunotherapy, the prospect of cancer therapy even more explained than before. Cytokine-induced killer (CIK) cell-based immunotherapy demonstrated to have potentiality in improving clinical outcomes and relieving major side effects of standard treatment options. In addition, given the distinctive features such as high safety, low toxicity effects on healthy cells, numerous clinical trials conducted on CIK cells. Due to the shortcomings that observed in CIK cell immunotherapy alone, arising a tendency to make modifications (combined modality therapy or combination therapy) including the addition of various types of cytokines, genetic engineering, combination with immune checkpoints, and so on. In this review, we have tried to bring forth the latest immunotherapy methods and their overview. We have discussed the combination therapies with CIK cells and the conducted clinical trials. This helps the future studies to use integrated therapies with CIK cells as a promising treatment of many types of cancers.


Asunto(s)
Células Asesinas Inducidas por Citocinas/trasplante , Neoplasias/terapia , Ensayos Clínicos como Asunto , Terapia Combinada , Células Asesinas Inducidas por Citocinas/inmunología , Ingeniería Genética , Humanos , Inmunoterapia , Neoplasias/inmunología
20.
Int J Reprod Biomed ; 14(7): 459-64, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27525330

RESUMEN

BACKGROUND: Human leukocyte antigen-G (HLA-G) is a non-classical class I molecule highly expressed by extravillous cytotrophoblast cells. Due to a single base pair deletion, its function can be compensated by other isoforms. Investigating the frequency of null allele in Recurrent Miscarriage (RM) subjects could be useful in understanding the relationship between frequency of this allele and RM in a given population. OBJECTIVE: This study aimed to determine the frequency of HLA-G*0105N null allele and its potential association with down-regulation of HLA-G in subjects with RM. MATERIALS AND METHODS: Western blotting was used to assess the level of HLA-G protein expression. For investigating the frequency of HLA-G*0105N null allele in RM subjects, PCR-RFLP method was used. Exon 3 of HLA-G gene was amplified by polymerase chain reaction (PCR). Subsequently, PpuM-1 enzyme was employed to digest the PCR products and fragments were analyzed using gel electrophoresis. RESULTS: Digestion using restriction enzyme showed the presence of heterozygous HLA-G*0105N null allele in 10% of the test population. Western blotting results confirmed the decrease in expression of HLA-G in the placental tissue of subjects with RM compared to subjects who could give normal birth. CONCLUSION: The frequency of heterozygous HLA-G*0105N null allele was high to some extent in subjects with RM. The mutation rate in subjects suggested that there is a significant association between RM and frequency of mutations in this allele.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA