Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cancer Immunol Immunother ; 72(7): 2151-2168, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36828964

RESUMEN

The metabolic stress present in the tumor microenvironment of many cancers can attenuate T cell antitumor activity, which is intrinsically controlled by the mitochondrial plasticity, dynamics, metabolism, and biogenesis within these T cells. Previous studies have reported that the complement C1q binding protein (C1QBP), a mitochondrial protein, is responsible for maintenance of mitochondrial fitness in tumor cells; however, its role in T cell mitochondrial function, particularly in the context of an antitumor response, remains unclear. Here, we show that C1QBP is indispensable for T cell antitumor immunity by maintaining mitochondrial integrity and homeostasis. This effect holds even when only one allele of C1qbp is functional. Further analysis of C1QBP in the context of chimeric antigen receptor (CAR) T cell therapy against the murine B16 melanoma model confirmed the cell-intrinsic role of C1QBP in regulating the antitumor functions of CAR T cells. Mechanistically, we found that C1qbp knocking down impacted mitochondrial biogenesis via the AMP-activated protein kinase (AMPK)/peroxisome proliferator-activated receptor gamma coactivator 1-alpha signaling pathway, as well as mitochondrial morphology via the phosphorylation of mitochondrial dynamics protein dynamin-related protein 1. In summary, our study provides a novel mitochondrial target to potentiate the plasticity and metabolic fitness of mitochondria within T cells, thus improving the immunotherapeutic potential of these T cells against tumors.


Asunto(s)
Mitocondrias , Proteínas Mitocondriales , Linfocitos T , Microambiente Tumoral , Animales , Ratones , Humanos , Xenoinjertos , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos T/metabolismo , Técnicas de Silenciamiento del Gen , Mitocondrias/metabolismo , Transducción de Señal , Inmunoterapia Adoptiva
2.
Pharmacol Res ; 197: 106942, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37775021

RESUMEN

The design of chimeric antigen receptors (CAR) significantly enhances the antitumor efficacy of T cells. Although some CAR-T products have been approved by FDA in treating hematological tumors, adoptive immune therapy still faces many difficulties and challenges in the treatment of solid tumors. In this study, we reported a new strategy to treat solid tumors using a natural killer-like T (NKT) cell line which showed strong cytotoxicity to lyse 15 cancer cell lines, safe to normal cells and had low or no Graft-versus-host activity. We thus named it as universal NKT (UNKT). In both direct and indirect 3D tumor-like organ model, UNKT showed efficient tumor-killing properties, indicating that it could penetrate the microenvironment of solid tumors. In mesothelin (MSLN)-positive tumor cells (SKOV-3 and MCF-7), MSLN targeting CAR modified-UNKT cells had enhanced killing potential against MSLN positive ovarian cancer compared with the wild type UNKT, as well as MSLN-CAR-T cells. Compared with CAR-T, Single-cell microarray 32-plex proteomics revealed CAR-UNKT cells express more effector cytokines, such as perforin and granzyme B, and less interleukin-6 after activation. Moreover, our CAR-UNKT cells featured in more multifunctionality than CAR-T cells. CAR-UNKT cells also demonstrated strong antitumor activity in mouse models of ovarian cancer, with the ability to migrate and infiltrate the tumor without inducing immune memory. The fast-in and -out, enhanced and prolonged tumor killing properties of CAR-UNKT suggested a novel cure option of cellular immunotherapy in the treatment of MSLN-positive solid tumors.


Asunto(s)
Neoplasias Hematológicas , Neoplasias Ováricas , Receptores Quiméricos de Antígenos , Animales , Femenino , Humanos , Ratones , Línea Celular , Mesotelina , Neoplasias Ováricas/terapia , Microambiente Tumoral
3.
Cancer Sci ; 113(3): 875-890, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34978120

RESUMEN

T cells survival, proliferation, and anti-tumor response are closely linked to their mitochondrial health. Complement C1q binding protein (C1QBP) promotes mitochondrial fitness through regulation of mitochondrial metabolism and morphology. However, whether C1QBP regulates T cell survival, proliferation, and anti-tumor immune function remains unclear. Our data demonstrated that C1QBP knockdown induced the accumulation of reactive oxygen species (ROS) and the loss of mitochondrial membrane potential to impair T cell mitochondrial fitness. At the same time, C1QBP insufficiency reduced the recruitment of the anti-apoptotic proteins, including Bcl-2 and Bcl-XL, and repressed caspase-3 activation and poly (ADP-ribose) polymerase cleavage, which consequently accelerated the T cell apoptotic process. In contrast, C1QBP knockdown rendered T cells with relatively weaker proliferation due to the inhibition of AKT/mTOR signaling pathway. To investigate the exact role of C1QBP in anti-tumor response, C1QBP+/- and C1QBP+/+ mice were given a subcutaneous injection of murine MC38 cells. We found that C1QBP deficiency attenuated T cell tumor infiltration and aggravated tumor-infiltrating T lymphocytes (TIL) exhaustion. Moreover, we further clarified the potential function of C1QBP in chimeric antigen receptor (CAR) T cell immunotherapy. Our data showed that C1QBP+/- CAR T cells exhibited relatively weaker anti-tumor response than the corresponding C1QBP+/+ CAR T cells. Given that C1QBP knockdown impairs T cells' anti-apoptotic capacity, proliferation as well as anti-tumor immune function, development of the strategy for potentiation of T cells' mitochondrial fitness through C1QBP could potentially optimize the efficacy of the related immunotherapy.


Asunto(s)
Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Linfocitos T/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Humanos , Inmunoterapia Adoptiva , Linfocitos Infiltrantes de Tumor/citología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones , Proteínas Mitocondriales/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/terapia , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptores Quiméricos de Antígenos , Transducción de Señal , Linfocitos T/citología , Linfocitos T/inmunología
4.
Pharmacol Res ; 175: 106034, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34915126

RESUMEN

Renal carcinoma progresses aggressively in patients with metastatic disease while curative strategies are limited. Here, we constructed a recombinant non-replicating adenovirus (Ad) vaccine encoding an immune activator, CD137L, and a tumor antigen, CAIX, for treating renal carcinoma. In a subcutaneous tumor model, tumor growth was significantly suppressed in the Ad-CD137L/CAIX vaccine group compared with the single vaccine group. The induction and maturity of CD11C+ and CD8+CD11C+ dendritic cell (DC) subsets were promoted in Ad-CD137L/CAIX co-immunized mice. Furthermore, the Ad-CD137L/CAIX vaccine elicited stronger tumor-specific multifunctional CD8+ T cell immune responses as demonstrated by increased proliferation and cytolytic function of CD8+ T cells. Notably, depletion of CD8+ T cells greatly compromised the effective protection provided by Ad-CD137L/CAIX vaccine, suggesting an irreplaceable role of CD8+ T cells for the immunopotency of the vaccine. In both lung metastatic and orthotopic models, Ad-CD137L/CAIX vaccine treatment significantly decreased tumor metastasis and progression and increased the induction of tumor-specific multifunctional CD8+ T cells, in contrast to treatment with the Ad-CAIX vaccine alone. The Ad-CD137L/CAIX vaccine also augmented the tumor-specific multifunctional CD8+ T cell immune response in both orthotopic and metastatic models. These results indicated that Ad-CD137L/CAIX vaccine elicited a potent anti-tumor activity by inducing CD8+DC-mediated multifunctional CD8+ T cell immune responses. The potential strategy of CD137L-based vaccine might be served as a novel treatment for renal carcinoma or other malignant tumors.


Asunto(s)
Ligando 4-1BB/genética , Vacunas contra el Adenovirus/uso terapéutico , Anhidrasa Carbónica IX/genética , Carcinoma de Células Renales/terapia , Neoplasias Renales/terapia , Neoplasias Pulmonares/terapia , Ligando 4-1BB/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Anhidrasa Carbónica IX/inmunología , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/patología , Células Dendríticas/inmunología , Femenino , Células HEK293 , Humanos , Interleucina-6/sangre , Neoplasias Renales/inmunología , Neoplasias Renales/patología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Ratones Endogámicos BALB C
5.
Am J Hematol ; 97(6): 711-718, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35179242

RESUMEN

CD19-targeted chimeric antigen receptor T (CAR-T) cells using murine single-chain variable fragment (scFv) has shown substantial clinical efficacy in treating relapsed/refractory acute lymphoblastic leukemia (R/R ALL). However, potential immunogenicity of the murine scFv domain may limit the persistence of CAR-T cells. In this study, we treated 52 consecutive subjects with R/R ALL with humanized CD19-specific CAR-T cells (hCART19s). Forty-six subjects achieved complete remission (CR) (N = 43) or CR with incomplete count recovery (CRi) (N = 3) within 1 month post infusion. During the follow-up with a median time of 20 months, the 1-year cumulative incidence of relapse was 25% (95% confidence interval [CI] 13-46), and 1-year event-free survival was 45% (95% CI 29-60). To the cutoff date, 20 patients presented CD19+ relapse and 2 had CD19- relapse. Among the 22 relapsed patients, 14 had treatment-mediated and treatment-boosted antidrug antibodies (ADA) as detected in a sensitive and specific cell-based assay. ADA positivity was correlated with the disease relapse risk. ADA-positive patients had a significantly lower CAR copy number than ADA-negative patients at the time of recurrence (p < .001). In conclusion, hCART19s therapy is safe and highly active in R/R ALL patients, and the hCART19s treatment could induce the emergence of ADA, which is related to the recurrence of the primary disease.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras , Receptores Quiméricos de Antígenos , Anticuerpos de Cadena Única , Proteínas Adaptadoras Transductoras de Señales , Animales , Antígenos CD19 , Recuento de Células , Humanos , Ratones , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/uso terapéutico
6.
Dermatol Ther ; 35(8): e15594, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35617452

RESUMEN

Disseminated facial verruca plana is a chronic disorder that causes significant psychological distress. However, safe and effective treatment is lacking. This study aimed to explore the efficacy and safety of 35% glycolic acid (GA) for the treatment of disseminated facial verruca plana. A split-face clinical trial was conducted to explore the efficacy and safety of using chemical peeling with 35% GA for the treatment of disseminated facial verruca plana. One side of the face was applied with 35% GA once every fortnight for a total of three times. Adapalene gel was applied every night to the other side of the face as the control. The clearance rate of lesions was evaluated at different time points. Between June 2020 and December 2020, 30 patients with disseminated verruca plana who visited the Dermatology Hospital of Southern Medical University were enrolled. After three chemical peelings with 35% GA that was applied at 2-week intervals, 15 (50%) patients achieved >70% lesion reduction. The same effective rate in the adapalene gel-treated side of the face was documented in eight patients. Subgroup analysis showed a higher clearance rate in patients with a shorter disease duration. Moreover, concurrent improvements in facial roughness were observed in the 35% GA-treated group. Adverse effects including mild erythema and desquamation were observed during chemical peeling with 35% GA. In conclusion, chemical peeling with 35% GA could be a safe and effective option for treating disseminated facial verruca plana, especially for those who desire skin improvement.


Asunto(s)
Quimioexfoliación , Verrugas , Adapaleno , Quimioexfoliación/efectos adversos , Glicolatos/efectos adversos , Humanos , Resultado del Tratamiento , Verrugas/tratamiento farmacológico
7.
Cancer Sci ; 112(9): 3469-3483, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34157192

RESUMEN

Renal carcinoma shows a high risk of invasion and metastasis without effective treatment. Herein, we developed a chitosan (CS) nanoparticle-mediated DNA vaccine containing an activated factor L-Myc and a tumor-specific antigen CAIX for renal carcinoma treatment. The subcutaneous tumor models were intramuscularly immunized with CS-pL-Myc/pCAIX or control vaccine, respectively. Compared with single immunization group, the tumor growth was significantly suppressed in CS-pL-Myc/pCAIX co-immunization group. The increased proportion and mature of CD11c+ DCs, CD8+ CD11c+ DCs and CD103+ CD11c+ DCs were observed in the splenocytes from CS-pL-Myc/pCAIX co-immunized mice. Furthermore, the enhanced antigen-specific CD8+ T lymphocyte proliferation, cytotoxic T lymphocyte (CTL) responses, and multi-functional CD8+ T cell induction were detected in CS-pL-Myc/pCAIX co-immunization group compared with CS-pCAIX immunization group. Of note, the depletion of CD8 T cells resulted in the reduction of CD8+ T cells or CD8+ CD11c+ DCs and the loss of anti-tumor efficacy induced by CS-pL-Myc/pCAIX vaccine, suggesting the therapeutic efficacy of the vaccine was required for CD8+ DCs and CD103+ DCs mediated CD8+ T cells responses. Likewise, CS-pL-Myc/pCAIX co-immunization also significantly inhibited the lung metastasis of renal carcinoma models accompanied with the increased induction of multi-functional CD8+ T cell responses. Therefore, these results indicated that CS-pL-Myc/pCAIX vaccine could effectively induce CD8+ DCs and CD103+ DCs mediated tumor-specific multi-functional CD8+ T cell responses and exert the anti-tumor efficacy. This vaccine strategy offers a potential and promising approach for solid or metastatic tumor treatment.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Neoplasias/administración & dosificación , Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/inmunología , Anhidrasa Carbónica IX/administración & dosificación , Carcinoma de Células Renales/terapia , Quitosano/química , Células Dendríticas/inmunología , Sistemas de Liberación de Medicamentos/métodos , Inmunidad , Inmunización/métodos , Cadenas alfa de Integrinas/metabolismo , Neoplasias Renales/terapia , Nanopartículas/química , Proteínas Proto-Oncogénicas c-myc/administración & dosificación , Vacunas de ADN/administración & dosificación , Animales , Antígenos de Neoplasias/genética , Anhidrasa Carbónica IX/genética , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/patología , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Humanos , Neoplasias Renales/inmunología , Neoplasias Renales/patología , Ratones , Ratones Endogámicos BALB C , Proteínas Proto-Oncogénicas c-myc/genética , Resultado del Tratamiento , Vacunas de ADN/inmunología
8.
J Cell Mol Med ; 24(18): 10744-10755, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32725966

RESUMEN

Conditionally replicative adenoviruses (CRAds) were promising approach for solid tumour treatment, but its oncolytic efficiency and toxicity are still not satisfactory for further clinical application. Here, we developed the CAIX promotor (CAIXpromotor )-controlled CRAd armed with a tumour suppressor absent in melanoma 2 (AIM2) to enhance its oncolytic potency. The CAIXpromotor -AIM2 adenoviruses (Ad-CAIXpromotor -AIM2) could efficiently express E1A and AIM2 in renal cancer cells. Compared with Ad-CAIXpromotor , Ad-CAIXpromotor -AIM2 significantly inhibited cell proliferation and enhanced cell apoptosis and cell killing, thus resulting in the oncolytic efficiency in 786-O cells or OSRC-2 cells. To explore the therapeutic effect, various Ads were intratumourally injected into OSRC-2-xenograft mice. The tumour growth was remarkably inhibited in Ad-CAIXpromotor -AIM2-treated group as demonstrated by reduced tumour volume and weight with a low toxicity. The inflammasome inhibitor YVAD-CMK resulted in the reduction of anti-tumour activity by Ad-CAIXpromotor -AIM2 in vitro or in vivo, suggesting that inflammasome activation response was required for the enhanced therapeutic efficiency. Furthermore, lung metastasis of renal cancer mice was also suppressed by Ad-CAIXpromotor -AIM2 treatment accompanied by the decreased tumour fossil in lung tissues. These results indicated that the tumour-specific Ad-CAIXpromotor -AIM2 could be applied for human renal cancer therapy. The therapeutic strategy of AIM2-based CRAds could be a potential and promising approach for the therapy of primary solid or metastasis tumours.


Asunto(s)
Adenovirus Humanos/fisiología , Anhidrasa Carbónica IX/genética , Carcinoma de Células Renales/terapia , Proteínas de Unión al ADN/fisiología , Neoplasias Renales/terapia , Viroterapia Oncolítica , Regiones Promotoras Genéticas/genética , Proteínas E1A de Adenovirus/biosíntesis , Proteínas E1A de Adenovirus/genética , Adenovirus Humanos/genética , Clorometilcetonas de Aminoácidos/farmacología , Animales , Carcinoma de Células Renales/secundario , Caspasa 1/análisis , Línea Celular , Línea Celular Tumoral , Citocinas/análisis , Proteínas de Unión al ADN/genética , Genes Sintéticos , Humanos , Inflamasomas/efectos de los fármacos , Neoplasias Renales/patología , Túbulos Renales/citología , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Ensayo de Tumor de Célula Madre , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Exp Cell Res ; 370(2): 561-570, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-30031129

RESUMEN

Absent in melanoma 2 (AIM2) as a tumor suppressor is recently recognized to play an important role in many human solid tumors. However, the biological significance and function of AIM2 in renal cell carcinoma (RCC) remain unknown. In this study, we proposed to investigate the clinicopathological and prognostic significance of AIM2 in RCC patient specimens, to analyze the correlation between AIM2 expression and disease progression, and to further investigate the role and mechanism of AIM2 in malignant behaviors of renal carcinoma by cellular and animal models. The correlation analysis showed that low AIM2 expression was significantly correlated with lymph node metastasis, poor 5-year overall (P < 0.05) and disease-specific survival (P < 0.05) in RCC patients. Over-expression of AIM2 in 786-O or OSRC-2 cells suppressed the cell proliferation, migration and invasion as compared with control. The increased AIM2 expression enhanced the expression of autophagy-related genes (Bcl-2, Beclin-1, LC3-Ⅱ, and ATG-5) and increased the ratio of LC3-Ⅱ/LC3-Ⅰin renal carcinoma cells. Of note, the blockade of autophagy by 3-Methyladenine (3-MA) abrogated the inhibition of cell migration and invasion by over-expressing AIM2, indicated that AIM2 suppressed the malignant behaviors of renal carcinoma by enhancing the induction of autophagy. Furthermore, 3-MA abolished the prevention of tumor growth on nude mouse established by AIM2-OSRC-2 cells. Our results suggested that AIM2 might serve as a novel prognostic indicator as well as a potential therapeutic target for renal carcinoma treatment.


Asunto(s)
Autofagia/fisiología , Carcinoma de Células Renales/metabolismo , Proteínas de Unión al ADN/metabolismo , Metástasis Linfática/patología , Adulto , Anciano , Animales , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Masculino , Melanoma/genética , Ratones Endogámicos BALB C , Persona de Mediana Edad
10.
J Cell Mol Med ; 22(11): 5670-5681, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30160343

RESUMEN

Renal cell carcinoma (RCC) is a high metastasis tumour with less effective treatment available currently. Absent in melanoma 2 (AIM2) as a tumour suppressor might be used as a potential therapeutic target for RCC treatment. Here, we found that AIM2 expression was significantly decreased in RCC patient specimens and renal carcinoma cell lines (786-O and OSRC-2). To establish a safe and effective AIM2 gene delivery system, we formed the nanoparticles consisting of a folate grafted PEI600-CyD (H1) nanoparticle-mediated AIM2 gene (H1/pAIM2) as an effective delivery agent. Delivery of H1/pAIM2 in renal carcinoma cells could remarkably increase the expression of AIM2, and subsequently decrease cell proliferation, migration, and invasion as well as enhance cell apoptosis. In order to evaluate the therapeutic efficacy of AIM2 in vivo, H1/pAIM2 nanoparticles were injected intratumorally into 786-O-xenograft mice. Administration of H1/pAIM2 nanoparticles could inhibit the tumour growth as evidenced by reduced tumour volume and weight. Furthermore, Blockade of inflammasome activation triggered by H1/pAIM2 nanoparticles using inflammasome inhibitor YVAD-CMK abrogated the anti-tumoral activities of H1/AIM2. These results indicated the therapeutic effect of H1/pAIM2 nanoparticles was mainly attributable to its capability to enhance the inflammasome activation. H1/AIM2 nanoparticles might act as an efficient therapeutic approach for RCC treatment.


Asunto(s)
Carcinoma de Células Renales/terapia , Proteínas de Unión al ADN/genética , Técnicas de Transferencia de Gen , Nanopartículas/administración & dosificación , Animales , Apoptosis/efectos de los fármacos , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteínas de Unión al ADN/administración & dosificación , Proteínas de Unión al ADN/química , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Xenoinjertos , Humanos , Inflamasomas/genética , Ratones , Nanopartículas/química , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología
11.
Am J Hematol ; 93(7): 851-858, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29633386

RESUMEN

Chimeric antigen receptor T (CAR-T) cell therapy has shown promising results for relapsed/refractory (R/R) acute lymphoblastic leukemia (ALL). The immune response induced by murine single-chain variable fragment (scFv) of the CAR may limit CAR-T cell persistence and thus increases the risk of leukemia relapse. In this study, we developed a novel humanized scFv from the murine FMC63 antibody. A total of 18 R/R ALL patients with or without prior murine CD19 CAR-T therapy were treated with humanized CD19-targeted CAR-T cells (hCART19s). After lymphodepletion chemotherapy with cyclophosphamide and fludarabine, the patients received a single dose (1 × 106 /kg) of autologous hCART19s infusion. Among the 14 patients without previous CAR-T therapy, 13 (92.9%) achieved complete remission (CR) or CR with incomplete count recovery (CRi) on day 30, whereas 1 of the 3 patients who failed a second murine CAR-T infusion achieved CR after hCART19s infusion. At day 180, the overall and leukemia-free survival rates were 65.8% and 71.4%, respectively. The cumulative incidence of relapse was 22.6%, and the nonrelapse mortality rate was 7.1%. During treatment, 13 patients developed grade 1-2 cytokine release syndrome (CRS), 4 patients developed grade 3-5 CRS, and 1 patient experienced reversible neurotoxicity. These results indicated that hCART19s could induce remission in patients with R/R B-ALL, especially in patients who received a reinfusion of murine CAR-T.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Receptores Quiméricos de Antígenos/uso terapéutico , Terapia Recuperativa/métodos , Adulto , Animales , Antígenos CD19/uso terapéutico , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Recurrencia , Inducción de Remisión , Análisis de Supervivencia , Resultado del Tratamiento
12.
J Biol Chem ; 290(39): 23850-62, 2015 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-26260793

RESUMEN

Neddylation is a posttranslational modification that controls diverse biological processes by covalently conjugating the ubiquitin-like protein NEDD8 to specific targets. Neddylation is commonly mediated by NEDD8-specific enzymes (typical neddylation) and, sometimes, by ubiquitin enzymes (atypical neddylation). Although typical neddylation is known to regulate protein function in many ways, the regulatory mechanisms and biological consequence of atypical neddylation remain largely unexplored. Here we report that NEDD8 conjugates were accumulated in the diseased hearts from mouse models and human patients. Proteotoxic stresses induced typical and atypical neddylation in cardiomyocytes. Loss of NUB1L exaggerated atypical neddylation, whereas NUB1L overexpression repressed atypical neddylation through promoting the degradation of NEDD8. Activation of atypical neddylation accumulated a surrogate misfolded protein, GFPu. In contrast, suppression of atypical neddylation by NUB1L overexpression enhanced GFPu degradation. Moreover, NUB1L depletion accumulated a cardiomyopathy-linked misfolded protein, CryAB(R120G), whereas NUB1L overexpression promoted its degradation through suppressing neddylation of ubiquitinated proteins in cardiomyocytes. Consequently, NUB1L protected cells from proteotoxic stress-induced cell injury. In summary, these data indicate that NUB1L suppresses atypical neddylation and promotes the degradation of misfolded proteins by the proteasome. Our findings also suggest that induction of NUB1L could potentially become a novel therapeutic strategy for diseases with increased proteotoxic stress.


Asunto(s)
Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Factores de Transcripción/metabolismo , Ubiquitinas/metabolismo , Cadena B de alfa-Cristalina/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Sustitución de Aminoácidos , Animales , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Mutación Missense , Proteína NEDD8 , Complejo de la Endopetidasa Proteasomal/genética , Factores de Transcripción/genética , Ubiquitinas/genética , Cadena B de alfa-Cristalina/genética
13.
Cancer Immunol Immunother ; 65(3): 305-14, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26842126

RESUMEN

T cell Ig and ITIM domain (TIGIT) is a newly identified inhibitory receptor expressed on T and natural killer (NK) cells. Cytokine-induced killer (CIK) cells express CD3 and CD56 molecules, and share functional properties with both NK and T cells. However, it remains unknown whether TIGIT is expressed in CIK cells. Here, we show that TIGIT is expressed by CIK cells and interacts with CD155. By blocking TIGIT using an anti-TIGIT functional antibody, we demonstrate that CIK cells display increased proliferation; higher cytotoxic targeting of tumor cells expressing CD155; and higher expression of interferon-γ (IFN-γ), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α). Furthermore, increases in IFN-γ and cytotoxicity by blockade of TIGIT were reduced by blocking DNAX accessory molecule-1 (DNAM-1) signaling, implying that TIGIT exerts immunosuppressive effects by competing with DNAM-1 for the same ligand, CD155. Our results provide evidence that blockade of TIGIT may be a novel strategy to improve the cytotoxic activity of CIK cells.


Asunto(s)
Células Asesinas Inducidas por Citocinas/inmunología , Citotoxicidad Inmunológica , Receptores Inmunológicos/fisiología , Receptores Virales/fisiología , Antígenos de Diferenciación de Linfocitos T/fisiología , Línea Celular Tumoral , Humanos , Transducción de Señal/fisiología
14.
Tumour Biol ; 37(1): 141-50, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26566628

RESUMEN

Estrogen-mediated high reactive oxygen species (ROS) tolerance plays an important role in driving carcinogenesis. ROS overproduction acts as the significant effector to increase genomic instability and transduce redox-related signal pathway. Especially, estrogen-mediated mitochondrial ROS promote the mutations in mitochondrial DNA (mtDNA) and the damage to mitochondrial proteins. Moreover, estrogen-mediated ROS contribute to the alteration of energy metabolism and modulate several redox-sensitive proteins responsible for cell proliferation and anti-apoptosis. On the other hand, estrogen simultaneously performs the antioxidative beneficial functions, which protects cancer cells from the potential cytotoxic effects of estrogen-mediated ROS through activation of nuclear factor-erythroid-2-related factor 2 (Nrf2)/Kelch-like ECH-associated protein 1 (Keap1) antioxidant response. Consequently, estrogen potentiates the high ROS tolerance through increase of ROS production as well as acceleration of ROS elimination, which ultimately results in estrogen-mediated carcinogenesis and malignant transformation. However, this overdependence on antioxidant response system to resist ROS-mediated cytotoxicity also represents the "Achilles' Heel" of estrogen-mediated cancer cells. In other words, the destruction of the high ROS tolerance using antioxidant inhibitors may provide a novel and efficacious measure to selectively eliminate these cancer cells without harming normal cells. Of course, it will be necessary to define the exact situation of ROS homeostasis in the different cellular microenvironment and further decipher the mechanisms of redox regulation, which is consequently used as a new avenue to optimize the clinical therapy for estrogen-mediated cancer.


Asunto(s)
Carcinogénesis/metabolismo , Transformación Celular Neoplásica , Estrógenos/fisiología , Neoplasias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Antineoplásicos/química , Antioxidantes/química , Apoptosis , Supervivencia Celular , ADN Mitocondrial/metabolismo , Homeostasis , Humanos , Ratones , Oxidación-Reducción
15.
Biomed Microdevices ; 18(4): 62, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27393216

RESUMEN

Polymerase Chain Reaction (PCR) is used to amplify a specific segment of DNA through a thermal cycling protocol. The PCR industry is shifting its focus away from macro-scale systems and towards micro-scale devices because: micro-scale sample sizes require less blood from patients, total reaction times are on the order of minutes opposed to hours, and there are cost advantages as many microfluidic devices are manufactured from inexpensive polymers. Some of the fastest PCR devices use continuous flow, but they have all been built of silicon or glass to allow sufficient heat transfer. This article presents a disposable polycarbonate (PC) device that is capable of achieving real-time, continuous flow PCR in a completely disposable polymer device in less than 13 minutes by thermally cycling the sample through an established temperature gradient in a serpentine channel. The desired temperature gradient was determined through simulations and validated by experiments which showed that PCR was achieved. Practical demonstration included amplification of foot-and-mouth disease virus (FMDV) derived cDNA.


Asunto(s)
Diseño de Equipo , Reacción en Cadena de la Polimerasa/instrumentación , Simulación por Computador , ADN , Vidrio/química , Calor , Técnicas Analíticas Microfluídicas , Cemento de Policarboxilato/química , Polímeros/química , Silicio/química
16.
BMC Cancer ; 15: 346, 2015 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-25934296

RESUMEN

BACKGROUND: Previous study showed that mitochondrial ND6 (mitND6) gene missense mutation resulted in NADH dehydrogenase deficiency and was associated with tumor metastasis in several mouse tumor cell lines. In the present study, we investigated the possible role of mitND6 gene nonsense and missense mutations in the metastasis of human lung adenocarcinoma. METHODS: The presence of mitND6 gene mutations was screened by DNA sequencing of tumor tissues from 87 primary lung adenocarcinoma patients and the correlation of the mutations with the clinical features was analyzed. In addition, we constructed cytoplasmic hybrid cells with denucleared primary lung adenocarcinoma cell as the mitochondria donor and mitochondria depleted lung adenocarcinoma A549 cell as the nuclear donor. Using these cells, we studied the effects of mitND6 gene nonsense and missense mutations on cell migration and invasion through wounding healing and matrigel-coated transwell assay. The effects of mitND6 gene mutations on NADH dehydrogenase activity and ROS production were analyzed by spectrophotometry and flow cytometry. RESULTS: mitND6 gene nonsense and missense mutations were detected in 11 of 87 lung adenocarcinoma specimens and was correlated with the clinical features including age, pathological grade, tumor stage, lymph node metastasis and survival rate. Moreover, A549 cell containing mitND6 gene nonsense and missense mutation exhibited significantly lower activity of NADH dehydrogenase, higher level of ROS, higher capacity of cell migration and invasion, and higher pAKT and pERK1/ERK2 expression level than cells with the wild type mitND6 gene. In addition, NADH dehydrogenase inhibitor rotenone was found to significantly promote the migration and invasion of A549 cells. CONCLUSIONS: Our data suggest that mitND6 gene nonsense and missense mutation might promote cell migration and invasion in lung adenocarcinoma, probably by NADH dehydrogenase deficiency induced over-production of ROS.


Asunto(s)
Adenocarcinoma/genética , Movimiento Celular , Neoplasias Pulmonares/genética , NADH Deshidrogenasa/genética , Adenocarcinoma/secundario , Línea Celular Tumoral , Codón sin Sentido , Femenino , Humanos , Neoplasias Pulmonares/patología , Metástasis Linfática , Masculino , Persona de Mediana Edad , Mutación Missense , NADH Deshidrogenasa/metabolismo , Invasividad Neoplásica , Especies Reactivas de Oxígeno/metabolismo
18.
Eur Neurol ; 74(1-2): 28-35, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26139100

RESUMEN

Our knowledge about pathophysiology of intracerebral hemorrhage (ICH) mainly originates from preclinical models of ICH. In this study, cerebral ultrastructure surrounding hematoma and its correlation with clinical severity were investigated in ICH patients. Thirty patients with basal ganglia hemorrhage and 6 control subjects were enrolled. Surgical evacuation was performed for patients with a blood loss >30 ml. Stroke severity was assessed using the Glasgow Coma Scale (GCS) and the National Institute of Health Stroke Scale (NIHSS). Transmission electron microscopy (TEM) was used to evaluate the ultrastructural characteristics of tissue specimens. Neural cells surrounding the hematomas showed evidence of cell swelling and necrosis. Decreased numbers of organelles and mitochondrial cristae were accompanied by cytoplasmic vacuolization, nuclear membrane invagination and breakdown, and intranuclear chromatic agglutination. These changes resulted in disintegration together with malacia, disappearance of the nucleus and nucleolus, and karyopyknosis. More serious ultrastructural damage was seen in patients with greater NIHSS scores, lower GCS scores, and greater bleeding volumes (p < 0.001). These findings suggest that neural cells undergo unfavorable ultrastructural changes that are responsible for dysfunction after ICH.


Asunto(s)
Hemorragia de los Ganglios Basales/patología , Encéfalo/ultraestructura , Adulto , Anciano , Femenino , Escala de Coma de Glasgow , Hematoma/patología , Humanos , Masculino , Microscopía Electrónica de Transmisión , Persona de Mediana Edad , Accidente Cerebrovascular/patología
19.
Biomed Microdevices ; 16(1): 173-80, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24081458

RESUMEN

In this work, an innovative vascular coupling device (VCD) is created to realize quick and reliable vessel anastomosis. Vessel anastomosis is common and often necessary during trauma, replantation and free tissue transfer surgeries. The current method of vessel anastomosis is traditional hand suturing. This technique is time consuming (20-30 min), difficult, and requires complex instruments. Additionally, it requires very skilled surgeons to efficiently perform the operation. To improve the reliability and reduce the amount of time required to connect two vessels, while providing an intima-to-intima anastomosis with no foreign material in contact with the blood flow, as occurs with sutures, a series of VCDs ranging from 1.5 mm to 7 mm inner diameter are designed and fabricated from polytetrafluoroethylene (PTFE) using laser cutting. A set of installation tools are also designed and fabricated to facilitate the VCD application. A series of experiments to test the VCD functionality are performed using both latex tubes and arteries. The results showed that the anastomosis process using VCDs and the installation tools can be completed in 5 min. The coupled VCDs can withstand 20 N tensile force, which is much higher than normal physiology conditions. There is no leakage or significant effects of the VCDs on the flow. A Micro-CT scan and histology images of cadaver arteries coupled with VCDs demonstrated that the VCD keeps the vessels open and does not constrict blood flow, suggesting that the VCD could be used in more advanced testing.


Asunto(s)
Arterias/cirugía , Microcirugia/instrumentación , Anastomosis Quirúrgica , Endotelio Vascular/cirugía , Diseño de Equipo , Humanos , Politetrafluoroetileno/química , Reproducibilidad de los Resultados
20.
Mol Genet Metab Rep ; 39: 101088, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38736698

RESUMEN

Isovaleric acidemia (IVA) is a rare autosomal recessive disorder that manifests as a deficiency of isovaleryl-CoA dehydrogenase (IVD), a key enzyme in leucine metabolism. The clinical presentations associated with IVD deficiency are variable and include feeding intolerance, vomiting, metabolic acidosis, ketonemia, "sweaty feet" odor, lethargy, coma and even death. Tandem mass spectrometry (MS/MS) and gas chromatography-mass spectrometry (GC/MS) methods were used to perform organic acid analysis of blood and urine samples from IVA patients, and the genetic analysis included next generation sequencing (NGS) and Sanger sequencing of the IVD gene. Here, we report the case of an almost seven-year-old male patient from a Chinese family who was asymptomatic during the newborn period, including the clinical manifestations and examination results. Genetic analysis revealed a previously unreported compound heterozygous variant in the IVD gene: c.593G > C (p.W198S) and c.859C > T (p.R287W).

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA