Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 79(5): 240, 2022 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-35416530

RESUMEN

Ischemic stroke is a leading cause of death and disability worldwide. Increasing evidence indicates that ischemic stroke is a thromboinflammatory disease in which the contact-kinin pathway has a central role by activating pro-coagulant and pro-inflammatory processes. The blocking of distinct members of the contact-kinin pathway is a promising strategy to control ischemic stroke. Here, a plasma kallikrein and active FXII (FXIIa) inhibitor (sylvestin, contained 43 amino acids, with a molecular weight of 4790.4 Da) was first identified from forest leeches (Haemadipsa sylvestris). Testing revealed that sylvestin prolonged activated partial thromboplastin time without affecting prothrombin time. Thromboelastography and clot retraction assays further showed that it extended clotting time in whole blood and inhibited clot retraction in platelet-rich plasma. In addition, sylvestin prevented thrombosis in vivo in FeCl3-induced arterial and carrageenan-induced tail thrombosis models. The potential role of sylvestin in ischemic stroke was evaluated by transient and permanent middle cerebral artery occlusion models. Sylvestin administration profoundly protected mice from ischemic stroke by counteracting intracerebral thrombosis and inflammation. Importantly, sylvestin showed no signs of bleeding tendency. The present study identifies sylvestin is a promising contact-kinin pathway inhibitor that can proffer profound protection from ischemic stroke without increased risk of bleeding.


Asunto(s)
Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Trombosis , Animales , Inflamación/tratamiento farmacológico , Inflamación/prevención & control , Cininas , Ratones , Accidente Cerebrovascular/tratamiento farmacológico , Tromboinflamación , Trombosis/tratamiento farmacológico
2.
Phytother Res ; 36(12): 4587-4603, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35916377

RESUMEN

Andrographolide(ADE) has been demonstrated to inhibit tumor growth through direct cytotoxicity on tumor cells. However, its potential activity on tumor microenvironment (TME) remains unclear. Tumor-associated macrophages (TAMs), composed mainly of M2 macrophages, are the key cells that create an immunosuppressive TME by secretion of cytokines, thus enhancing tumor progression. Re-polarized subpopulations of macrophages may represent vital new therapeutic alternatives. Our previous studies showed that ADE possessed anti-metastasis and anoikis-sensitization effects. Here, we demonstrated that ADE significantly suppressed M2-like polarization and enhanced M1-like polarization of macrophages. Moreover, ADE inhibited the migration of M2 and tube formation in HUVECs under M2 stimulation. In vivo studies showed that ADE restrained the growth of MDA-MB-231 and HCC1806 human breast tumor xenografts and 4T-1 mammary gland tumors through TAMs. Wnt5a/ß-catenin pathway and MMPs were particularly associated with ADE's regulatory mechanisms to M2 according to RNA-seq and bioinformatics analysis. Moreover, western blot also verified the expressions of these proteins were declined with ADE exposure. Among the cytokines released by M2, PDGF-AA and CCL2 were reduced. Our current findings for the first time elucidated that ADE could modulate macrophage polarization and function through Wnt5a signaling pathway, thereby playing its role in inhibition of triple-negative breast cancer.


Asunto(s)
Neoplasias de la Mama , Diterpenos , Vía de Señalización Wnt , Femenino , Humanos , beta Catenina , Neoplasias de la Mama/tratamiento farmacológico , Microambiente Tumoral , Macrófagos Asociados a Tumores , Diterpenos/farmacología , Células Endoteliales de la Vena Umbilical Humana , Células MDA-MB-231 , Animales
3.
J Cell Mol Med ; 25(2): 686-700, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33225580

RESUMEN

Adoptive immunotherapy is a new potential method of tumour therapy, among which anti-CD19 chimeric antigen receptor T-cell therapy (CAR-T cell), is a typical treatment agent for haematological malignancies. Previous clinical trials showed that the quality and phenotype of CAR-T cells expanded ex vivo would seriously affect the tumour treatment efficacy. Although magnetic beads are currently widely used to expand CAR-T cells, the optimal expansion steps and methods have not been completely established. In this study, the differences between CAR-T cells expanded with anti-CD3/CD28 mAb-coated beads and those expanded with cell-based aAPCs expressing CD19/CD64/CD86/CD137L/mIL-15 counter-receptors were compared. The results showed that the number of CD19-specific CAR-T cells with a 4-1BB and CD28 co-stimulatory domain was much greater with stimulation by aAPCs than that with beads. In addition, the expression of memory marker CD45RO was higher, whereas expression of exhausted molecules was lower in CAR-T cells expanded with aAPCs comparing with the beads. Both CAR-T cells showed significant targeted tumoricidal effects. The CAR-T cells stimulated with aAPCs secreted apoptosis-related cytokines. Moreover, they also possessed marked anti-tumour effect on NAMALWA xenograft mouse model. The present findings provided evidence on the safety and advantage of two expansion methods for CAR-T cells genetically modified by piggyBac transposon system.


Asunto(s)
Antígenos CD19/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Animales , Western Blotting , Antígenos CD8/metabolismo , Línea Celular Tumoral , Electroporación , Citometría de Flujo , Humanos , Inmunoterapia Adoptiva/métodos , Células K562 , Masculino , Ratones , Ratones SCID , Plásmidos/genética , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Biol Chem ; 293(51): 19600-19612, 2018 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-30333226

RESUMEN

Several clinical immunotherapy trials with cytokine-induced killer (CIK) cells have been reported. However, molecular evidence of cell expansion, acquisition of tumor cytotoxicity, and safety of CIK cells is required before putting them to clinical use. Here, we performed dynamic transcriptomic analyses of CIKs generated from primary peripheral blood mononuclear cells exposed to interferon-γ, OKT3, and interleukin-2. CIK mRNAs were extracted and sequenced at days 0, 1, 7, and 14 and subjected to bioinformatics analyses. Using weighted correlation network analysis (WGCNA), we identified two major gene modules that mediate immune cell activation and mitosis. We found that activation and cytotoxicity of CIK cells likely rely on cluster of differentiation 8 (CD8) and its protein partner LCK proto-oncogene, Src family tyrosine kinase (LCK). A time-course series analysis revealed that CIK cells have relatively low immunogenicity because of decreased expression of some self-antigens. Importantly, we identified several crucial activating receptors and auxiliary adhesion receptors expressed on CIK cells that may function as tumor sensors. Interestingly, cytotoxicity-associated genes, including those encoding PRF1, GZMB, FASL, and several cytokines, were up-regulated in mature CIK cells. Most immune-checkpoint molecules and inflammatory tumor-promoting factors were down-regulated in the CIK cells, suggesting efficacy and safety in future clinical trials. Notably, insulin-like growth factor 1 (IGF-1) was highly expressed in CIK cells and may promote cytotoxicity, although it also could facilitate tumorigenesis. The transcriptomic atlas of CIK cells presented here may inform efforts to improve CIK-associated tumor cytotoxicity and safety in clinical trials.


Asunto(s)
Células Asesinas Inducidas por Citocinas/metabolismo , Perfilación de la Expresión Génica , Ciclo Celular/genética , Ciclo Celular/inmunología , Línea Celular , Células Asesinas Inducidas por Citocinas/citología , Células Asesinas Inducidas por Citocinas/inmunología , Humanos , Inmunoterapia/efectos adversos , Familia de Multigenes/genética , Familia de Multigenes/inmunología , Proto-Oncogenes Mas , Seguridad , Análisis de Secuencia de ARN
5.
J Neurooncol ; 145(2): 211-222, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31605296

RESUMEN

PURPOSE: Glioma is one of the lethal cancers which needs effective therapeutic target. TRIM44 has been found playing a carcinogenic role in human tumors such as breast cancer and ovarian cancer. However, the pathophysiological significance of TRIM44 in glioma is still unclear. METHODS: Quantitative-PCR and western blot were used to assess the expression of TRIM44 in glioma cells. For cell proliferation, Brdu incorporation and colony formation assays were performed. By Caspase 3 staining and FACS analysis, we revealed that TRIM44 knockdown induced glioma cell apoptosis. A BALB/c nude mouse xenograft model and following immunohistochemical (IHC) staining enables us to explore the effect of TRIM44 deletion on glioma growth in vivo. Western blot of p21, p27 and AKT indicated the possible role of TRIM44 in regulation AKT pathway in glioma. RESULTS: TRIM44 was significantly elevated in glioma cells, and high expression of TRIM44 is related to poor prognostic of glioma patients. TRIM44 knockdown by shRNAs inhibit glioma cell proliferation, migration, induced cell cycle disruption and further cellular apoptosis in vitro. As well, TRIM44 inactivation obviously inhibit tumor growth in xenograft model. Furthermore, the negative cell cycle regulators p21/p27 are significantly upregulated, while AKT which is known as the main regulator of p21/p27 is inactivated in TRIM44-dificient cells. These results suggested that TRIM44 inactivation disrupted cell cycle progression and inhibit cell proliferation through AKT/p21/p27 pathway in glioma. CONCLUSION: TRIM44 was associated with oncogenic potential of glioma. Targeting TRIM44 might be beneficial for glioma therapy.


Asunto(s)
Glioma/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Transducción de Señal , Proteínas de Motivos Tripartitos/metabolismo , Animales , Neoplasias Encefálicas , Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Células HEK293 , Humanos , Ratones Endogámicos BALB C , Proteínas Proto-Oncogénicas c-akt/metabolismo
6.
Med Sci Monit ; 25: 2756-2763, 2019 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-30982828

RESUMEN

BACKGROUND The NKX2 gene family is made up of core transcription factors that are involved in the morphogenesis of the vertebrate heart. NKx2-5 plays a pivotal role in mouse cardiogenesis, and mutations in NKx2-5 result in an abnormal structure and function of the heart, including atrial septal defect and cardiac electrophysiological abnormalities. MATERIAL AND METHODS To investigate the genetic variation of NKX2-5 in Chinese patients with sporadic atrial septal defect, we sequenced the full length of the NKX2-5 gene in the participants of the study. Four hundred thirty-nine patients and 567 healthy unrelated individuals were recruited. Genomic DNA was extracted from the peripheral blood leukocytes of the participants. DNA samples from the participants were amplified by multiplex PCR and sequenced on an Illumina HiSeq platform. Variations were detected by comparison with a standard reference genome and annotation with a variant effect predictor. RESULTS Thirty variations were detected in Chinese patients with sporadic atrial septal defect, and 6 single nucleotide polymorphisms (SNPs) had a frequency greater than 1%. Among the 30 variations, the SNPs rs2277923 and rs3729753 were extremely prominent, with a high frequency and odds ratio in patients. CONCLUSIONS Single nucleotide variations are the prominent genetic variations of NKX2-5 in Chinese patients with sporadic atrial septal defect. The SNPs rs2277923 and rs3729753 are prominent single nucleotide variations (SNVs) in Chinese patients with sporadic atrial septal defect.


Asunto(s)
Defectos del Tabique Interatrial/genética , Proteína Homeótica Nkx-2.5/genética , Pueblo Asiatico/genética , Secuencia de Bases , China/epidemiología , Análisis Mutacional de ADN , Femenino , Genes Homeobox , Defectos del Tabique Interatrial/sangre , Defectos del Tabique Interatrial/epidemiología , Defectos del Tabique Interatrial/metabolismo , Proteína Homeótica Nkx-2.5/sangre , Proteína Homeótica Nkx-2.5/metabolismo , Humanos , Masculino , Mutación , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN/métodos , Factores de Transcripción/genética
7.
Cell Tissue Bank ; 18(1): 53-59, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28138805

RESUMEN

This study aimed to determine the efficacy and safety of human umbilical cord-derived mesenchymal stem cell (HUC-MSC) transplantation for treating elderly vascular dementia (VaD). Ten VaD patients (average age, 73.88 years old) were treated. HUC-MSCs were isolated, cultured, stem cell-marked, and qualified and administered as a 3-course intravenous infusion to these patients. The Mini-Mental State Exam (MMSE) and the Activities of Daily Living Index (Barthel Index scoring system) were used to assess the cognitive function and daily living activity improvements in these patients before transplantation (T0), 3 months after transplantation (T1), and 6 months after transplantation (T2). The MMSE and Barthel Index scores were 15.80 ± 5.49 and 42.00 ± 9.33 points at T0, respectively, and were significantly different when compared with those at T1 (19.20 ± 6.39 and 49.20 ± 10.86 points, respectively, P < 0.05), whereas there was no difference when compared with those at T2 (14.00 ± 6.55 and 40.70 ± 10.37 points, respectively, P > 0.05). HUC-MSC transplantation was safe and feasible for VaD and improved early cognitive functions and daily living activities in VaD patients to a certain extent, thus improving patients' quality of life.


Asunto(s)
Demencia Vascular/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Cordón Umbilical/citología , Anciano , Separación Celular , Células Cultivadas , Cognición , Demencia Vascular/fisiopatología , Femenino , Humanos , Masculino , Trasplante de Células Madre Mesenquimatosas/métodos , Persona de Mediana Edad , Calidad de Vida
8.
Tumour Biol ; 37(8): 10317-27, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26842926

RESUMEN

Unlike heterogeneous tumor cells, cancer-associated fibroblasts (CAF) are genetically more stable which serve as a reliable target for tumor immunotherapy. Fibroblast activation protein (FAP) which is restrictively expressed in tumor cells and CAF in vivo and plays a prominent role in tumor initiation, progression, and metastasis can function as a tumor rejection antigen. In the current study, we have constructed artificial FAP(+) stromal cells which mimicked the FAP(+) CAF in vivo. We immunized a breast cancer mouse model with FAP(+) stromal cells to perform immunotherapy against FAP(+) cells in the tumor microenvironment. By forced expression of FAP, we have obtained FAP(+) stromal cells whose phenotype was CD11b(+)/CD34(+)/Sca-1(+)/FSP-1(+)/MHC class I(+). Interestingly, proliferation capacity of the fibroblasts was significantly enhanced by FAP. In the breast cancer-bearing mouse model, vaccination with FAP(+) stromal cells has significantly inhibited the growth of allograft tumor and reduced lung metastasis indeed. Depletion of T cell assays has suggested that both CD4(+) and CD8(+) T cells were involved in the tumor cytotoxic immune response. Furthermore, tumor tissue from FAP-immunized mice revealed that targeting FAP(+) CAF has induced apoptosis and decreased collagen type I and CD31 expression in the tumor microenvironment. These results implicated that immunization with FAP(+) stromal cells led to the disruption of the tumor microenvironment. Our study may provide a novel strategy for immunotherapy of a broad range of cancer.


Asunto(s)
Neoplasias de la Mama/patología , Fibroblastos/inmunología , Gelatinasas/inmunología , Inmunoterapia/métodos , Proteínas de la Membrana/inmunología , Serina Endopeptidasas/inmunología , Microambiente Tumoral/inmunología , Animales , Western Blotting , Neoplasias de la Mama/inmunología , Modelos Animales de Enfermedad , Endopeptidasas , Femenino , Técnica del Anticuerpo Fluorescente , Inmunización , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
9.
Cell Tissue Res ; 357(3): 571-82, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24906288

RESUMEN

Amyotrophic lateral sclerosis (ALS) is an adult-onset progressive neurodegenerative disease involving degeneration of motor neurons in the central nervous system. Stem cell treatment is a potential therapy for this fatal disorder. The human amniotic membrane (HAM), an extremely rich and easily accessible tissue, has been proposed as an attractive material in cellular therapy and regenerative medicine because of its advantageous characteristics. In the present study, we evaluate the long-term effects of a cellular treatment by intravenous administration of human amniotic mesenchymal stem cells (hAMSCs) derived from HAM into a hSOD1(G93A) mouse model. The mice received systemic administration of hAMSCs or phosphate-buffered saline (PBS) at the onset, progression and symptomatic stages of the disease. hAMSCs were detected in the spinal cord at the final stage of the disease, in the form of isolates or clusters and were negative for ß-tubulin III and GFAP. Compared with the treatment with PBS, multiple hAMSC transplantations significantly retarded disease progression, extended survival, improved motor function, prevented motor neuron loss and decreased neuroinflammation in mice. These findings demonstrate that hAMSC transplantation is a promising cellular treatment for ALS.


Asunto(s)
Amnios/citología , Esclerosis Amiotrófica Lateral/terapia , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Esclerosis Amiotrófica Lateral/fisiopatología , Animales , Conducta Animal , Movimiento Celular , Supervivencia Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inflamación/patología , Inyecciones Intravenosas , Masculino , Ratones , Actividad Motora , Neuronas Motoras/patología
10.
Tumour Biol ; 35(3): 1997-2007, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24104501

RESUMEN

Generation of cytokine-induced killer (CIK) cells is an emerging approach in adoptive donor lymphocyte infusion for patients with a wide range of tumors. However, our previous in vitro studies have shown that the killing efficacy of CIK cells against lung cancer was lower than other tumor cells, while the underlying mechanisms are not clear. We explored the feasibility to improve CIK cells mediated cytotoxicity against lung cancer. Interleukin (IL)-15 is a pleiotropic cytokine that stimulates cytolytic activity and cytokine secretion of NK cells, which may enhance the cytotoxic activity of CIK cells. In this study, we intended to stimulate the CIK cells by IL-2 in combination with IL-15 in cell expansion to achieve enhanced cytotoxicity against lung cancer cells. The different phenotypes of IL-2 or combination of IL-2 and IL-15 stimulated cytokine-induced killer cells were determined, and the improved cytotoxicity of IL-2 and IL-15 induced CIK cells against lung adenocarcinoma were evaluated both in vitro and in vivo. CIK cells stimulated with both IL-2 and IL-15 has shown greater proliferative potential than CIK cells treated with IL-2 alone. IL-15 induction also has driven the expansion of CD3+CD56+ subset and significantly enhanced cytotoxicity against tumor cells. Further analysis has demonstrated that CIKIL-2&IL-15 injected mice models have shown significant tumor regression and lower expression level of CyclinD1 in tumor tissue. This study has provided preclinical evidences that CIKIL-2&IL-15 with enhanced cytotoxicity may offer alternative treatment option for patients with lung cancer.


Asunto(s)
Adenocarcinoma/inmunología , Células Asesinas Inducidas por Citocinas/inmunología , Inmunoterapia Adoptiva/métodos , Interleucina-15/inmunología , Interleucina-2/inmunología , Neoplasias Pulmonares/inmunología , Adenocarcinoma/terapia , Adenocarcinoma del Pulmón , Animales , Apoptosis/inmunología , Técnicas de Cultivo de Célula/métodos , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Inmunohistoquímica , Neoplasias Pulmonares/terapia , Ratones , Ratones Endogámicos BALB C , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Tumour Biol ; 35(2): 1539-50, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24127039

RESUMEN

Gallbladder cancer (GBC) is one leading cause of cancer-related death worldwide. WW domain-containing oxidoreductase (WWOX) is a tumor suppressor gene which can suppress proliferation of a variety of tumors. However, little was known about the relationships between WWOX and gallbladder cancer. In the current study, we intended to investigate the tumor suppressive role of WWOX in gallbladder malignant cells both in vitro and in vivo, and explore the potential mechanism of tumor toxic function of WWOX. Our results have shown that WWOX triggerred apoptosis in GBC cells and increased the expression of P73 and PUMA in cytoplasm. We also have found that Bax has been upregulated after overexpression of WWOX, whereas, Bcl-2 was downregulated by WWOX. To further validate the results in vivo, we evaluated the tumor suppressive role of WWOX in mouse model of gallbladder cancer. The results have shown that the proliferation of the tumor was inhibited after delivery of WWOX, and the expressions of P73 and PUMA were upregulated in target tissues. The mice models administrated with WWOX have shown better prognosis than mice in negative control groups. The results from our study indicated that WWOX could be used as a therapeutic agent in the gene therapy of gallbladder cancer.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteínas de Unión al ADN/biosíntesis , Neoplasias de la Vesícula Biliar/genética , Proteínas Nucleares/biosíntesis , Oxidorreductasas/genética , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Supresoras de Tumor/genética , Animales , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular , Proteínas de Unión al ADN/genética , Neoplasias de la Vesícula Biliar/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Proteínas Nucleares/genética , Oxidorreductasas/biosíntesis , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteína Tumoral p73 , Oxidorreductasa que Contiene Dominios WW
12.
Nat Commun ; 15(1): 36, 2024 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-38167296

RESUMEN

While canonical Wnt signaling is well recognized for its crucial regulatory functions in cell fate decisions, the role of non-canonical Wnt signaling in adult stem cells remains elusive and contradictory. Here, we identified Mcam, a potential member of the non-canonical Wnt signaling, as an important negative regulator of mammary gland epithelial cells (MECs) by genome-scale CRISPR-Cas9 knockout (GeCKO) library screening. Loss of Mcam increases the clonogenicity and regenerative capacity of MECs, and promotes the proliferation, differentiation, and ductal morphogenesis of mammary epithelial in knockout mice. Mechanically, Mcam knockout recruits and polarizes macrophages through the Il4-Stat6 axis, thereby promoting secretion of the non-canonical Wnt ligand Wnt5a and its binding to the non-canonical Wnt signaling receptor Ryk to induce the above phenotypes. These findings reveal Mcam roles in mammary gland development by orchestrating communications between MECs and macrophages via a Wnt5a/Ryk axis, providing evidences for non-canonical Wnt signaling in mammary development.


Asunto(s)
Proteínas Wnt , Vía de Señalización Wnt , Ratones , Animales , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteína Wnt-5a/genética , Proteína Wnt-5a/metabolismo , Diferenciación Celular , Morfogénesis , Ratones Noqueados , Macrófagos/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo
13.
Tumour Biol ; 34(3): 1667-78, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23430583

RESUMEN

Bladder cancer (BC) is one of the most common human malignancies that account for major death in the world. Apoptin that is derived from chicken anemia virus (CAV) has displayed tumor-specific cytotoxic activity in a variety of human carcinomas. However, the magical function of apoptin in bladder carcinoma cell lines has not been identified yet. In our study, we delivered apoptin into bladder-originating T24, EJ, and HCV29 cell lines by adenovirus system. The selective cytotoxic effect of apoptin was determined by cell viability assay, active caspase-3 measurement, and annexin V/PI double staining. Importantly, we have examined the differential expression patterns of tumor-associated genes including Ki67, C-erbB-2, Rb, and nm23 by flow cytometry and western blot in vitro. In an animal study, apoptin was infused into animal models by AAV system, and immunohistochemistry and quantitative real-time PCR (qRT-PCR) were employed to validate results in vivo. The results indicated that apoptin could selectively induce apoptosis in bladder tumorigenic cells coupled with tumor-specific nucleus accumulation in vitro. Interestingly, apoptin could downregulate expression levels of Ki67 and C-erbB-2 and upregulate the expression of Rb both in vitro and in vivo. Moreover, the animal models treated with AAV-apoptin have shown smaller tumor volumes and displayed better prognosis than controls. In conclusion, apoptin could selectively induce apoptosis in bladder tumor cells through altering expression profiles of tumor-associated genes.


Asunto(s)
Apoptosis , Biomarcadores de Tumor/metabolismo , Proteínas de la Cápside/metabolismo , Proliferación Celular , Transcriptoma , Neoplasias de la Vejiga Urinaria/patología , Animales , Biomarcadores de Tumor/genética , Western Blotting , Proteínas de la Cápside/genética , Virus de la Anemia del Pollo , Dependovirus/genética , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Genes erbB-2/genética , Vectores Genéticos , Humanos , Técnicas para Inmunoenzimas , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nucleósido Difosfato Quinasas NM23/genética , Nucleósido Difosfato Quinasas NM23/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tasa de Supervivencia , Trasplante Homólogo , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/mortalidad , Neoplasias de la Vejiga Urinaria/prevención & control
14.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 44(6): 931-4, 2013 Nov.
Artículo en Zh | MEDLINE | ID: mdl-24490505

RESUMEN

UNLABELLED: OBJECTITVE: To investigate the effect of human umbilici mesenchymal cells (HUMSCs) implantation on the brain derived neurotrophic factor (BDNF) expression in diabetic foot rats. METHODS: SD rats were divided into three groups (n = 12): normal group, diadetic foot model group and HUMSC treatment group. Diabetic foot model in rats was established, then prepared HUMSC were implanted on the diabetic foot ulcers in rats, and control ones were administrated with saline only. The area of ulceration, sensory function, BDNF expression and its localization were determined by using morphology, physiological function measurement, RT-PCR and immunohistochemistry assay. RESULT: Siglificantly decreased area of ulceration in diabetic foot rats of HUMSC implantation group was observed. This was simultaneously companied with the sensory function improvement (P < 0.05). RT-PCR showed that BDNF mRNA expression was significantly up regulated (P < 0.05). BDNF immunstaining was located in epithelia tissue and the protein level of BDNF was markedly increased (P < 0.05). CONCLUSION: HUMSC implantation maybe an effective strategy on the treatment of ulceration in diabetic foot rats, and the possible mechanism may involve in BDNF expression.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Pie Diabético/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Cordón Umbilical/citología , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Diabetes Mellitus Experimental/complicaciones , Pie Diabético/metabolismo , Humanos , Masculino , Ratas , Ratas Sprague-Dawley , Cicatrización de Heridas/fisiología
15.
Front Cell Dev Biol ; 11: 1302578, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37854068

RESUMEN

[This corrects the article DOI: 10.3389/fcell.2023.1293109.].

16.
Cell Regen ; 12(1): 13, 2023 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-37138165

RESUMEN

Adipose tissues are essential for actively regulating systemic energy balance, glucose homeostasis, immune responses, reproduction, and longevity. Adipocytes maintain dynamic metabolic needs and possess heterogeneity in energy storage and supply. Overexpansion of adipose tissue, especially the visceral type, is a high risk for diabetes and other metabolic diseases. Changes in adipocytes, hypertrophy or hyperplasia, contribute to the remodeling of obese adipose tissues, accompanied by abundant immune cell accumulation, decreased angiogenesis, and aberrant extracellular matrix deposition. The process and mechanism of adipogenesis are well known, however, adipose precursors and their fate decision are only being defined with recent information available to decipher how adipose tissues generate, maintain, and remodel. Here, we discuss the key findings that identify adipose precursors phenotypically, with special emphasis on the intrinsic and extrinsic signals in instructing and regulating the fate of adipose precursors under pathophysiological conditions. We hope that the information in this review lead to novel therapeutic strategies to combat obesity and related metabolic diseases.

17.
Front Oncol ; 13: 1122669, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36726382

RESUMEN

Background: Expansion and activation of cytotoxic T lymphocytes (CTLs) in vitro represents a promising immunotherapeutic strategy, and CTLs can be primed by dendritic cells (DCs) loaded with tumor-associated antigens (TAAs) transformed by recombinant adeno-associated virus (rAAV). This study aimed to explore the impact of rAAV-DC-induced CTLs on prognosis of CRC and to explore factors associated with prognosis. Methods: This prospective observational study included patients operated for CRC at Yan'an Hospital Affiliated to Kunming Medical University between 2016 and 2019. The primary outcome was progression-free survival (PFS), secondary outcomes were overall survival (OS) and adverse events. Totally 49 cases were included, with 29 and 20 administered rAAV-DC-induced CTL and chemotherapy, respectively. Results: After 37-69 months of follow-up (median, 54 months), OS (P=0.0596) and PFS (P=0.0788) were comparable between two groups. Mild fever occurred in 2 (6.9%) patients administered CTL infusion. All the chemotherapy group experienced mild-to-moderate adverse effects, including vasculitis (n=20, 100%), vomiting (n=5, 25%), nausea (n=17, 85%) and fatigue (n=17, 85%). Conclusions: Lymphatic metastasis (hazard ratio [HR]=4.498, 95% confidence interval [CI]: 1.290-15.676; P=0.018) and lower HLA-I expression (HR=0.294, 95%CI: 0.089-0.965; P=0.044) were associated with poor OS in the CTL group. CTLs induced by rAAV-DCs might achieve comparable effectiveness in CRC patients compare to chemotherapy, cases with high tumor-associated HLA-I expression and no lymphatic metastasis were more likely to benefit from CTLs.

18.
Int Immunopharmacol ; 124(Pt B): 110974, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37757633

RESUMEN

CAR-T targeting CD19 have achieved significant effects in the treatment of B-line leukemia and lymphoma. However, the treated patients frequently relapsed and could not achieve complete remission. Therefore, improving the proliferation and cytotoxicity of CAR-T cells, reducing exhaustion and enhancing infiltration capacity are still issues to be solved. The IL-7 has been shown to enhance the memory characteristics of CAR-T cells, but the specific mechanism has yet to be elaborated. miRNAs play an important role in T cell activity. However, whether miRNA is involved in the activation of CAR-T cells by IL-7 has not yet been reported. Our previous study had established the 3rd generation CAR-T cells. The present study further found that IL-7 significantly increased the proliferation of anti-CD19 CAR-T cells, the ratio of CD4 + CAR + cells and the S phase of cell cycle. In vivo study NAMALWA xenograft model showed that IL-7-stimulated CAR-T cells possessed stronger tumoricidal efficiency. Further we validated that IL-7 induced CAR-T cells had low expression of CDKN1A and high expression of miRNA-98-5p. Additionally, CDKN1A was associated with miRNA-98-5p. Our results, for the first time, suggested IL-7 could conspicuously enhance the proliferation of CAR-T cells through miRNA-98-5p targeting CDKN1A expression, which should be applied to CAR-T production.


Asunto(s)
MicroARNs , Receptores Quiméricos de Antígenos , Humanos , Receptores Quiméricos de Antígenos/metabolismo , Inmunoterapia Adoptiva/métodos , Interleucina-7/genética , Interleucina-7/metabolismo , MicroARNs/genética , Proliferación Celular , Antígenos CD19/genética , Antígenos CD19/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo
19.
Ann Transplant ; 27: e935488, 2022 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-35347104

RESUMEN

BACKGROUND Cardiac allograft rejection is still a crucial barrier to achieving satisfactory outcomes after surgery. In this study, we propose to find candidate biomarkers from endomyocardial biopsy (EMB) and peripheral blood (PB) samples for efficient diagnosis and treatment of cardiac allograft rejection. MATERIAL AND METHODS Microarray datasets were obtained from the Gene Expression Omnibus (GEO). Differentially expressed genes (DEGs) of cardiac allograft rejection patients and control subjects from EMB and PB samples were screened using the online tool GEO2R. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of all samples' DEGs were performed with the DAVID online tool. Protein-protein interaction (PPI) networks were constructed and visualized using Cytoscape and the top 10 hub genes were selected. Finally, the most highly enriched GO and KEGG pathways of the top 10 hub genes were determined. RESULTS A total of 57 502 genes from EMB samples and 131 624 genes from PB samples were identified. Gene characteristics and enrichment analysis indicated that both EMB and PB samples contained DEGs involved in antigen presentation, immune cells activation, inflammatory process, and cellular injuries. In EMB samples, there were some DEGs related to heart tissue injury and cardiac malfunction. Moreover, DEGs that regulates hypoxia-induced factors and erythrocyte function in response of ischemia and hypoxia stress were present in PB samples but were absent in EMB samples. CONCLUSIONS The screened differentially expressed genes (DEGs) from EMB and PB samples of patients with cardiac graft rejection are potential candidate biomarkers of diagnosis and treatment.


Asunto(s)
Biología Computacional , Trasplante de Corazón , Aloinjertos , Biomarcadores , Biopsia , Biología Computacional/métodos , Perfilación de la Expresión Génica/métodos , Trasplante de Corazón/efectos adversos , Humanos , Donantes de Tejidos
20.
Front Cell Dev Biol ; 10: 876054, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35478960

RESUMEN

The therapeutic effects and mechanism of umbilical cord mesenchymal stem cells (UC-MSC) on kidney injury in MRL/Ipr mice were studied. UC-MSC, methylprednisolone (MP), and their combination were used to treat MRL/Ipr mice. The therapeutic effects were evaluated by renal function assessment, and HE, PAS, and Masson staining were carried out on renal tissues and visualized by electron microscopy. Subsequently, podocyte injury was detected by the presence of podocin in renal tissues by immunofluorescence. To further explore the mechanism, serum TGF-ß1 was measured, and TGF-ß1, p-Smad3, and TRAF6 in the renal tissue were detected by Western blotting. In vitro, TGF-ß1 was used to stimulate podocytes, and the podocyte activity and changes in synaptopodin were observed after UC-MSC treatment. Significant improvements in renal function and pathological injury were observed in the UC-MSC group compared to the lupus nephritis (LN) model group. UC-MSC and MP treatment improved podocyte injury in MRL/Ipr mice. Western blot examination showed a significant increase in TGF-ß1, p-Smad3, and TRAF6 expression in renal tissues of the LN model group, while significant downregulation of those proteins was observed in the UC-MSC group. After TGF-ß1 stimulation in vitro, podocyte activity decreased, and UC-MSC treatment improved podocyte activity and restored synaptopodin expression. UC-MSC therapy could improve the deterioration of renal function and the pathological changes of the renal tissues in MRL/Ipr mice. Our study suggested that UC-MSC may improve kidney injury and podocyte injury in LN mice by inhibiting the TGF-ß1 pathway.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA