Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Surg Res ; 263: 44-52, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33631377

RESUMEN

BACKGROUND: The generation of long-term durable tumor immunity and prolonged disease-free survival depends on the ability to generate and support CD8+ central memory T-cells. Microsatellite-stable colon cancer is resistant to currently available immunotherapies; thus, development of novel mechanisms to increase both lymphocyte infiltration and central memory formation are needed to improve outcomes in these patients. We have previously demonstrated that both interleukin-2 (IL-2) and LIGHT (TNFSF14) independently enhance antitumor immune responses and hypothesize that combination immunotherapy may increase the CD8+ central memory T-cell response. METHODS: Murine colorectal cancer tumors were established in syngeneic mice. Tumors were treated with control, soluble, or liposomal IL-2 at established intervals. A subset of animal tumors overexpressed tumor necrosis superfamily factor LIGHT (TNFSF14). Peripheral blood, splenic, and tumor-infiltrating lymphocytes were isolated for phenotypic studies and flow cytometry. RESULTS: Tumors exposed to a combination of LIGHT and IL-2 experienced a decrease in tumor size compared with IL-2 alone that was not demonstrated in wild-type tumors or between other treatment groups. Combination exposure also increased splenic central memory CD8+ cells compared with IL-2 administration alone, while not increasing tumor-infiltrating lymphocytes. In the periphery, the combination enhanced levels of circulating CD8 T-cells and central memory T-cells, while also increasing circulating T-regulatory cells. CONCLUSIONS: Combination of IL-2, whether soluble or liposomal, with exposure to LIGHT results in increased CD8+ central memory cells in the spleen and periphery. New combination immunotherapy strategies that support both effector and memory T-cell functions are critical to enhancing durable antitumor responses and warrant further investigation.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Neoplasias del Colon/terapia , Inmunoterapia/métodos , Interleucina-2/administración & dosificación , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Línea Celular Tumoral/trasplante , Neoplasias del Colon/genética , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Memoria Inmunológica/efectos de los fármacos , Inyecciones Intralesiones , Liposomas , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Proteínas Recombinantes/administración & dosificación , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
2.
J Surg Res ; 233: 57-64, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30502288

RESUMEN

BACKGROUND: Colorectal cancer remains a leading cause of cancer-related mortality worldwide. Metastases to the liver are often present at initial presentation and will form in most patients during their course of disease. We have previously demonstrated that enhanced trafficking and activation of tumor-infiltrating lymphocytes in colorectal liver metastases (CRLM) may improve antitumor immune responses. Thus, development of novel mechanisms to increase lymphocyte infiltration and activation are needed to improve patient outcomes. METHODS: CT26 murine colorectal cancer cells were treated with physiologic levels of the potent inducer of immunogenic cell death mitoxantrone (MTX). An in situ vaccine was created with treated cells in an established model of CRLM. Cells were evaluated by flow cytometry for cell cycle evaluation and calreticulin expression. Splenic and tumor-infiltrating lymphocytes were isolated for phenotypic studies. RESULTS: MTX-treatment of colon cancer cells resulted in a sub-G1 peak, inhibition of G1 cell cycle progression, and increased G2/M cell fractions while simultaneously increasing dynamic exposure of calreticulin on the cell surface (P < 0.05). Vaccination with MTX-treated cells resulted in significant decreases in CRLM formation associated with increased tumor-infiltrating leukocytes that displayed increased expression of the T cell surface activation marker CD69. CONCLUSIONS: Vaccination with MTX-treated primary colon cancer cells enhances tumor-infiltrating lymphocytes and clinical responses in CRLM.


Asunto(s)
Antineoplásicos/administración & dosificación , Vacunas contra el Cáncer/administración & dosificación , Neoplasias Colorrectales/patología , Neoplasias Hepáticas/terapia , Mitoxantrona/administración & dosificación , Animales , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/inmunología , Línea Celular Tumoral/trasplante , Modelos Animales de Enfermedad , Femenino , Humanos , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/secundario , Activación de Linfocitos/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Resultado del Tratamiento
3.
J Immunol ; 194(4): 1639-45, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25560411

RESUMEN

CD28 costimulation is essential for the development of thymic-derived CD4(+)CD25(+)Foxp3(+) regulatory T cells ("tTregs"). E3 ubiquitin ligase Cbl-b has been shown to regulate CD28 dependence of T cell activation. In this paper, we report that the loss of Cbl-b partially but significantly rescues the defective development of tTregs in Cd28(-/-) mice. This partial rescue is independent of IL-2. Mechanistically, Cbl-b binds to Foxp3 upon TCR stimulation and, together with Stub1, targets Foxp3 for ubiquitination and subsequently degradation in the proteasome. As Cbl-b self-ubiquitination and proteasomal degradation is impaired in Cd28(-/-) T cells, the defective development of tTregs in Cd28(-/-) mice may in part be due to increased Foxp3 ubiquitination and degradation targeted by Stub1 and Cbl-b. Treating Cd28(-/-) mice with a proteasome inhibitor completely rescues defective tTreg development in these mice. Therefore, Cbl-b, together with Stub1, ubiquitinate Foxp3, and regulate tTreg development.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Factores de Transcripción Forkhead/inmunología , Activación de Linfocitos/inmunología , Proteínas Proto-Oncogénicas c-cbl/inmunología , Linfocitos T Reguladores/inmunología , Animales , Factores de Transcripción Forkhead/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Timo/citología , Timo/inmunología , Transfección , Ubiquitina-Proteína Ligasas/inmunología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
4.
J Immunol ; 195(9): 4218-27, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26416283

RESUMEN

Previously, we demonstrated that CD28 and CTLA-4 signaling control Casitas-B-lineage lymphoma (Cbl)-b protein expression, which is critical for T cell activation and tolerance induction. However, the molecular mechanism(s) of this regulation remains to be elucidated. In this study, we found that Cbl-b fails to undergo tyrosine phosphorylation upon CD3 stimulation because SHP-1 is recruited to and dephosphorylates Cbl-b, whereas CD28 costimulation abrogates this interaction. In support of this finding, T cells lacking SHP-1 display heightened tyrosine phosphorylation and ubiquitination of Cbl-b upon TCR stimulation, which correlates with decreased levels of Cbl-b protein. The aberrant Th2 phenotype observed in T cell-specific Shp1(-/-) mice is reminiscent of heightened Th2 response in Cblb(-/-) mice. Indeed, overexpressing Cbl-b in T cell-specific Shp1(-/-) T cells not only inhibits heightened Th2 differentiation in vitro, but also Th2 responses and allergic airway inflammation in vivo. Therefore, SHP-1 regulates Cbl-b-mediated T cell responses by controlling its tyrosine phosphorylation and ubiquitination.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 6/fisiología , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Linfocitos T/inmunología , Animales , Antígenos CD28/fisiología , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/fisiología , Ratones , Ratones Endogámicos C57BL , Fosforilación , Proteína Tirosina Fosfatasa no Receptora Tipo 6/química , Células Th2/inmunología , Ubiquitinación
5.
Clin Immunol ; 157(2): 166-74, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25638413

RESUMEN

Lupus nephritis (LN) is a challenging problem that affects 50% of patients with systemic lupus erythematosus (SLE) without effective therapy. Here, we report that A77 1726, the active metabolite of leflunomide, effectively inhibits development of LN and attenuates the generalized autoimmune features. A77 1726 suppresses the expansion of double negative (DN) T cells, and inhibits T and B cell activation. Intriguingly, A77 1726 treatment significantly increases CD4(+)Foxp3(+) regulatory T cells but suppresses potential "pathogenic" IL-17-producing DN T cells in lymph nodes. In vitro experiment shows that A77 1726 potentiates the conversion of naive CD4(+)CD25(-) T cells into CD4(+)CD25(+)Foxp3(+) inducible regulatory T cells (iTregs) by inhibiting Akt. Taken together, our data indicate that the therapeutic effects of A77 1726 in murine LN are mediated, at least in part, by augmenting iTregs which suppress pathogenic IL-17-producing DN T cells through an Akt-dependent mechanism.


Asunto(s)
Compuestos de Anilina/farmacología , Hidroxibutiratos/farmacología , Inmunosupresores/farmacología , Interleucina-17/metabolismo , Riñón/efectos de los fármacos , Nefritis Lúpica/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Animales , Anticuerpos Antinucleares/inmunología , Crotonatos , Modelos Animales de Enfermedad , Interleucina-17/inmunología , Riñón/inmunología , Riñón/patología , Nefritis Lúpica/patología , Ratones , Nitrilos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/inmunología , Toluidinas
6.
J Immunol ; 191(2): 632-9, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23749633

RESUMEN

E3 ubiquitin ligase Casitas-B-lineage lymphoma protein-b (Cbl-b) is critical for establishing the threshold for T cell activation and is essential for induction of T cell anergy. Recent studies suggest that Cbl-b is involved in the development of CD4(+)CD25(+) inducible regulatory T cells (iTregs). In this study, we report that the optimal induction of Foxp3 by naive CD4(+)CD25(-) T cells requires suboptimal TCR triggering. In the absence of Cbl-b, the TCR strength for optimal Foxp3 induction is downregulated in vitro. Using TCR-transgenic Rag(-/-) mice in combination with Cbl-b deficiency, we show that in vivo iTreg development is also controlled by Cbl-b via tuning the TCR strength. Furthermore, we show that Akt-2 but not Akt-1 regulates Foxp3 expression downstream of Cbl-b. Therefore, we demonstrate that Cbl-b regulates the fate of iTregs via controlling the threshold for T cell activation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Activación de Linfocitos , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Linfocitos T Reguladores/inmunología , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Antígenos CD4/metabolismo , Diferenciación Celular/inmunología , Células Cultivadas , Regulación hacia Abajo , Factores de Transcripción Forkhead/metabolismo , Proteínas de Homeodominio/genética , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
7.
BMC Vet Res ; 11: 33, 2015 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-25889187

RESUMEN

BACKGROUND: Haemophilus parasuis (H. parasuis) causes Glässer's disease and multisystem infectious disease. It is one of the major causes of nursery mortality in swine herds. Cefquinome (CEQ) is proposed for the treatment of pigs against respiratory tract infection. However, few studies have investigated the PK/PD characteristics and PK/PD cutoff of this drug against H. parasuis. RESULTS: A total of 213 H. parasuis strains were isolated from diseased pigs in China. The minimal inhibitory concentrations (MICs) of CEQ against these isolates were determined. The MIC(50) and MIC(90) values were 0.125 and 8 mg/L, respectively. An in vitro dynamic PK/PD infection model was used to investigate the antimicrobial effect of CEQ against H. parasuis strain of serotype 5. The target values of CEQ for 3-log(10)-unit and 4-log10-unit decreases effects were the percent time that CEQ concentrations were above the minimum inhibitory concentration (T% > MIC) of 61 and 71 respectively. According to Monte Carlo simulation, the PK/PD cutoff for CEQ against H. parasuis was 0.06 mg/L. The suggested dose regimen was 4 mg/kg/12 h BW. CONCLUSIONS: The value of PK/PD surrogate marker T% > MIC is of great utility in CEQ clinical usage. The very first CEQ PK/PD cutoff provide fundamental data for CEQ breakpoint determination. A more desirable dose regimen against H. parasuis was provided for CEQ using in China district.


Asunto(s)
Antibacterianos/farmacología , Cefalosporinas/farmacocinética , Haemophilus parasuis/efectos de los fármacos , Antibacterianos/administración & dosificación , Cefalosporinas/administración & dosificación , Técnicas In Vitro , Pruebas de Sensibilidad Microbiana/veterinaria , Método de Montecarlo
8.
Biochem Biophys Res Commun ; 450(1): 526-31, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24924627

RESUMEN

It has been documented that caspase-8, a central player in apoptosis, is also crucial for TCR-mediated NF-κB activation. However, whether other caspases are also involved this process is unknown. In this report, we showed that in addition to caspase-8, caspase-9 is required for TCR-mediated NF-κB activation. Caspase-9 induces activation of PKC-θ, phosphorylation of Bcl10 and NF-κB activation in a caspase-3-dependent manner, but it appears that Bcl10 phosphorylation is uncoupled from NF-κB activation. Furthermore, caspase-8 lies upstream of caspase-9 during T cell activation. Therefore, TCR ligation elicits a caspase cascade involving caspase-8, caspase-9 and caspase-3 which initiates PKC-θ-dependent pathway leading to NF-κB activation and PKC-θ-independent Bcl10 phosphorylation which limits NF-kB activity.


Asunto(s)
Caspasas/metabolismo , Isoenzimas/metabolismo , Activación de Linfocitos/fisiología , FN-kappa B/metabolismo , Proteína Quinasa C/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Animales , Células Cultivadas , Humanos , Células Jurkat , Ratones , Ratones Endogámicos C57BL , Complejos Multienzimáticos/metabolismo , Proteína Quinasa C-theta , Linfocitos T
9.
Cancers (Basel) ; 15(11)2023 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-37296966

RESUMEN

We have shown that activin A (activin), a TGF-ß superfamily member, has pro-metastatic effects in colorectal cancer (CRC). In lung cancer, activin activates pro-metastatic pathways to enhance tumor cell survival and migration while augmenting CD4+ to CD8+ communications to promote cytotoxicity. Here, we hypothesized that activin exerts cell-specific effects in the tumor microenvironment (TME) of CRC to promote anti-tumoral activity of immune cells and the pro-metastatic behavior of tumor cells in a cell-specific and context-dependent manner. We generated an Smad4 epithelial cell specific knockout (Smad4-/-) which was crossed with TS4-Cre mice to identify SMAD-specific changes in CRC. We also performed IHC and digital spatial profiling (DSP) of tissue microarrays (TMAs) obtained from 1055 stage II and III CRC patients in the QUASAR 2 clinical trial. We transfected the CRC cells to reduce their activin production and injected them into mice with intermittent tumor measurements to determine how cancer-derived activin alters tumor growth in vivo. In vivo, Smad4-/- mice displayed elevated colonic activin and pAKT expression and increased mortality. IHC analysis of the TMA samples revealed increased activin was required for TGF-ß-associated improved outcomes in CRC. DSP analysis identified that activin co-localization in the stroma was coupled with increases in T-cell exhaustion markers, activation markers of antigen presenting cells (APCs), and effectors of the PI3K/AKT pathway. Activin-stimulated PI3K-dependent CRC transwell migration, and the in vivo loss of activin lead to smaller CRC tumors. Taken together, activin is a targetable, highly context-dependent molecule with effects on CRC growth, migration, and TME immune plasticity.

10.
Clin Immunol ; 143(2): 128-33, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22459706

RESUMEN

Program death-1 (PD-1) has been documented to negatively regulate immune responses. However, the cellular and molecular mechanisms for PD-1-mediated immune suppression have not been fully elucidated. In this study, we show that loss of PD-1 does not lead to defective induction of CD4(+) T cell anergy in vitro and in vivo. Rather, the absence of PD-1 inhibits the development of inducible CD4(+)Foxp3(+) regulatory T cells (iTregs) induced by TGF-ß in vitro. In support of this finding, PD-1 deficiency impairs the generation of iTregs in vivo and leads to development of severe T cell-transfer-induced colitis. Mechanistically, defective iTreg generation in the absence of PD-1 was attributed to the heightened phosphorylation of Akt. Therefore, we first demonstrate that PD-1 controls peripheral T cell tolerance via an anergy-independent but iTreg-dependent mechanism.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Traslado Adoptivo , Animales , Colitis/etiología , Colitis/inmunología , Proteínas de Homeodominio/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Receptor de Muerte Celular Programada 1/deficiencia , Receptor de Muerte Celular Programada 1/genética
11.
J Immunol ; 185(3): 1375-8, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20601598

RESUMEN

Th cells that produce IL-17 (Th17 cells) are a distinct subset of Th cells implicated in several autoimmune diseases. Although CD28-B7 interaction has been shown to be involved in Th17 differentiation in vitro, the role of CTLA-4 in controlling Th17 development is completely unknown. We report in this paper that blocking the CTLA-4-B7 interaction potentiates Th17 cell differentiation in vitro and in vivo. Furthermore, blocking CTLA-4-B7 interaction in vivo confers the susceptibility of experimental autoimmune myocarditis to CD28(-/-) mice or increases the severity of experimental autoimmune myocarditis in wild-type mice. The enhanced disease susceptibility is mediated by heightened Th17 responses. With these results, we are the first to demonstrate that CTLA-4-B7 interaction inhibits Th17 differentiation in vitro and in vivo and suppresses Th17-mediated autoimmunity.


Asunto(s)
Antígenos CD/fisiología , Antígeno B7-1/fisiología , Diferenciación Celular/inmunología , Regulación hacia Abajo/inmunología , Inhibidores de Crecimiento/fisiología , Interleucina-17/fisiología , Linfocitos T Colaboradores-Inductores/inmunología , Secuencia de Aminoácidos , Animales , Antígenos CD/metabolismo , Enfermedades Autoinmunes/etiología , Enfermedades Autoinmunes/patología , Enfermedades Autoinmunes/prevención & control , Antígeno B7-1/genética , Antígeno B7-1/metabolismo , Antígenos CD28/genética , Antígeno CTLA-4 , Células Cultivadas , Regulación hacia Abajo/genética , Predisposición Genética a la Enfermedad , Inhibidores de Crecimiento/antagonistas & inhibidores , Inhibidores de Crecimiento/metabolismo , Interleucina-17/antagonistas & inhibidores , Interleucina-17/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Miocarditis/etiología , Miocarditis/inmunología , Miocarditis/patología , Linfocitos T Colaboradores-Inductores/metabolismo , Linfocitos T Colaboradores-Inductores/patología
12.
Oncogene ; 41(14): 2054-2068, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35177811

RESUMEN

Increased tumor infiltrating lymphocytes (TIL) are associated with improved patient responses to immunotherapy. As a result, there is interest in enhancing lymphocyte trafficking particularly to colon cancers since the majority are checkpoint blockade-resistant and microsatellite stable. Here, we demonstrate that activated T-cells (ATC) armed with anti-CD3 x anti-EGFR bispecific antibody increases TIL and mediate anti-tumor cytotoxicity while decreasing tumor cell viability. Furthermore, treatment induces endogenous anti-tumor immunity that resisted tumor rechallenge and increased memory T-cell subsets in the tumor. When combined with targeted tumor expression of the tumor necrosis factor superfamily member LIGHT, activated T-cell proliferation and infiltration were further enhanced, and human colorectal tumor regressions were observed. Our data indicate that tumor-targeted armed bispecific antibody increases TIL trafficking and is a potentially potent strategy that can be paired with combination immunotherapy to battle microsatellite stable colon cancer. SIGNIFICANCE: Enhancing trafficking of tumor infiltrating lymphocytes (TILs) to solid tumors has been shown to improve outcomes. Unfortunately, few strategies have been successful in the clinical setting for solid tumors, particularly for "cold" microsatellite stable colon cancers. In order to address this gap in knowledge, this study combined TNFSF14/LIGHT immunomodulation with a bispecific antibody armed with activated T-cells targeted to the tumor. This unique T-cell trafficking strategy successfully generated anti-tumor immunity in a microsatellite stable colon cancer model, stimulated T-cell infiltration, and holds promise as a combination immunotherapy for treating advanced and metastatic colorectal cancer.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias del Colon , Anticuerpos Biespecíficos/farmacología , Anticuerpos Biespecíficos/uso terapéutico , Complejo CD3 , Neoplasias del Colon/tratamiento farmacológico , Humanos , Inmunoterapia , Linfocitos T
13.
Biochem Biophys Res Commun ; 400(1): 16-20, 2010 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-20691662

RESUMEN

Akt signaling plays a central role in T cell functions, such as proliferation, apoptosis, and regulatory T cell development. Phosphorylation at Ser(473) in the hydrophobic motif, along with Thr(308) in its activation loop, is considered necessary for Akt function. It is widely accepted that phosphoinositide-dependent kinase 1 (PDK-1) phosphorylates Akt at Thr(308), but the kinase(s) responsible for phosphorylating Akt at Ser(473) (PDK-2) remains elusive. The existence of PDK-2 is considered to be specific to cell type and stimulus. PDK-2 in T cells in response to TCR stimulation has not been clearly defined. In this study, we found that conventional PKC positively regulated TCR-induced Akt Ser(473) phosphorylation. PKC-alpha purified from T cells can phosphorylate Akt at Ser(473) in vitro upon TCR stimulation. Knockdown of PKC-alpha in T-cell-line Jurkat cells reduced TCR-induced phosphorylation of Akt as well as its downstream targets. Thus our results suggest that PKC-alpha is a candidate for PDK-2 in T cells upon TCR stimulation.


Asunto(s)
Proteína Quinasa C-alfa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/enzimología , Animales , Activación Enzimática , Técnicas de Silenciamiento del Gen , Humanos , Células Jurkat , Ratones , Ratones Endogámicos C57BL , Fosforilación , Proteína Quinasa C-alfa/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Receptores de Antígenos de Linfocitos T/agonistas , Serina/genética , Serina/metabolismo
14.
Endocr Relat Cancer ; 26(6): 551-563, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-30999276

RESUMEN

ATC is an aggressive disease with limited therapeutic options due to drug resistance. TRAIL is an attractive anti-cancer therapy that can trigger apoptosis in a cancer cell-selective manner. However, TRAIL resistance is a major clinical obstacle for its use as a therapeutic drug. Previously, we demonstrated that MADD is a cancer cell pro-survival factor that can modulate TRAIL resistance. However, its role, if any, in overcoming TRAIL resistance in ATC is unknown. First, we characterized ATC cell lines as either TRAIL resistant, TRAIL sensitive or moderately TRAIL sensitive and evaluated MADD expression/cellular localization. We determined the effect of MADD siRNA on cellular growth and investigated its effect on TRAIL treatment. We assessed the effect of combination treatment (MADD siRNA and TRAIL) on mitochondrial membrane potential (MMP) and reactive oxygen species (ROS) levels. The effect of combination treatment on tumor growth was assessed in vivo. We found increased levels of MADD in ATC cells relative to Nthy-ori 3-1. MADD protein localizes in the cytosol (endoplasmic reticulum and Golgi body) and membrane. MADD knockdown resulted in spontaneous cell death that was synergistically enhanced when combined with TRAIL treatment in otherwise resistant ATC cells. Combination treatment resulted in a significant reduction in MMP and enhanced generation of ROS indicating the putative mechanism of action. In an orthotopic mouse model of TRAIL-resistant ATC, treatment with MADD siRNA alone reduced tumor growth that, when combined with TRAIL, resulted in significant tumor regressions. We demonstrated the potential clinical utility of MADD knockdown in sensitizing cells to TRAIL-induced apoptosis in ATC.


Asunto(s)
Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/genética , Factores de Intercambio de Guanina Nucleótido/genética , ARN Interferente Pequeño/administración & dosificación , Ligando Inductor de Apoptosis Relacionado con TNF/uso terapéutico , Carcinoma Anaplásico de Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/metabolismo , Silenciador del Gen , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones Desnudos , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Carcinoma Anaplásico de Tiroides/genética , Carcinoma Anaplásico de Tiroides/metabolismo , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/metabolismo
15.
Kidney Int ; 74(12): 1577-81, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19034301

RESUMEN

Previously, we showed that vitamin D receptor gene knockout leads to hyperreninemia independent of calcium metabolism; however, the contribution of parathyroid hormone to renin upregulation remained unclear. Here we separated the role of vitamin D and parathyroid hormone in the regulation of renin expression in vivo by generating transgenic mice that overexpressed the human vitamin D receptor in renin-producing cells using the 4.1 kb Ren-1c gene promoter. Targeting of human vitamin D receptor to the juxtaglomerular cells of the mice was confirmed by immunohistochemistry. Renal renin mRNA levels and plasma renin activity were decreased in these transgenic mice by about 50% and 30%, respectively, with no significant change in blood pressure, calcium, or parathyroid hormone levels. Moreover using vitamin D receptor knockout mice, we found that expression of the human receptor in their juxtaglomerular cells reduced renin expression in these mice without affecting calcium or parathyroid hormone status. Our study shows that suppression of renin expression by 1,25-dihydroxyvitamin D in vivo is independent of parathyroid hormone and calcium.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Aparato Yuxtaglomerular/citología , Receptores de Calcitriol/genética , Renina/genética , Animales , Calcitriol/farmacología , Calcio/farmacología , Humanos , Aparato Yuxtaglomerular/metabolismo , Ratones , Ratones Transgénicos , Hormona Paratiroidea/fisiología , ARN Mensajero/sangre
16.
Cell Death Dis ; 8(2): e2584, 2017 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-28151483

RESUMEN

Immunotherapeutic approaches to manage patients with advanced gastrointestinal malignancies are desired; however, mechanisms to incite tumor-specific immune responses remain to be elucidated. Rose bengal (RB) is toxic at low concentrations to malignant cells and may induce damage-associated molecular patterns; therefore, we investigated its potential as an immunomodulator in colon cancer. Murine and human colon cancer lines were treated with RB (10% in saline/PV-10) for cell cycle, cell death, and apoptosis assays. Damage-associated molecular patterns were assessed with western blot, ELISA, and flow cytometry. In an immunocompetent murine model of colon cancer, we demonstrate that tumors regress upon RB treatment, and that RB induces cell death in colon cancer cells through G2/M growth arrest and predominantly necrosis. RB-treated colon cancer cells expressed distinct hallmarks of immunogenic cell death (ICD), including enhanced expression of calreticulin and heat-shock protein 90 on the cell surface, a decrease in intracellular ATP, and the release of HMGB1. To confirm the ICD phenotype, we vaccinated immunocompetent animals with syngeneic colon cancer cells treated with RB. RB-treated tumors served as a vaccine against subsequent challenge with the same CT26 colon cancer tumor cells, and vaccination with in vitro RB-treated cells resulted in slower tumor growth following inoculation with colon cancer cells, but not with syngeneic non-CT26 cancer cells, suggesting a specific antitumor immune response. In conclusion, RB serves as an inducer of ICD that contributes to enhanced specific antitumor immunity in colorectal cancer.


Asunto(s)
Antineoplásicos/inmunología , Antineoplásicos/farmacología , Muerte Celular/efectos de los fármacos , Muerte Celular/inmunología , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/inmunología , Rosa Bengala/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Calreticulina/metabolismo , Línea Celular Tumoral , Neoplasias del Colon/metabolismo , Células HCT116 , Proteína HMGB1/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Células HT29 , Humanos , Ratones , Ratones Endogámicos BALB C , Necrosis/tratamiento farmacológico , Necrosis/inmunología , Necrosis/metabolismo
17.
Cancer Res ; 77(8): 1880-1891, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28249900

RESUMEN

The majority of patients with colon cancer will develop advanced disease, with the liver being the most common site of metastatic disease. Patients with increased numbers of tumor-infiltrating lymphocytes in primary colon tumors and liver metastases have improved outcomes. However, the molecular factors that could empower antitumor immune responses in this setting remain to be elucidated. We reported that the immunostimulatory cytokine LIGHT (TNFSF14) in the microenvironment of colon cancer metastases associates with improved patient survival, and here we demonstrate in an immunocompetent murine model that colon tumors expressing LIGHT stimulate lymphocyte proliferation and tumor cell-specific antitumor immune responses. In this model, increasing LIGHT expression in the microenvironment of either primary tumors or liver metastases triggered regression of established tumors and slowed the growth of liver metastases, driven by cytotoxic T-lymphocyte-mediated antitumor immunity. These responses corresponded with significant increases in tumor-infiltrating lymphocytes and increased expression of lymphocyte-homing signals in the metastatic tumors. Furthermore, we demonstrated evidence of durable tumor-specific antitumor immunity. In conclusion, increasing LIGHT expression increased T-cell proliferation, activation, and infiltration, resulting in enhanced tumor-specific immune-mediated tumor regressions in primary tumors and colorectal liver metastases. Mechanisms to increase LIGHT in the colon cancer microenvironment warrant further investigation and hold promise as an immunotherapeutic strategy. Cancer Res; 77(8); 1880-91. ©2017 AACR.


Asunto(s)
Neoplasias del Colon/inmunología , Neoplasias del Colon/metabolismo , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/biosíntesis , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología , Animales , Línea Celular Tumoral , Neoplasias del Colon/patología , Femenino , Células HEK293 , Humanos , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/secundario , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Ratones Endogámicos BALB C , Linfocitos T/inmunología
18.
ALTEX ; 34(2): 301-310, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27846345

RESUMEN

Translating in vitro biological data into actionable information related to human health holds the potential to improve disease treatment and risk assessment of chemical exposures. While genomics has identified regulatory pathways at the cellular level, translation to the organism level requires a multiscale approach accounting for intra-cellular regulation, inter-cellular interaction, and tissue/organ-level effects. Tissue-level effects can now be probed in vitro thanks to recently developed systems of three-dimensional (3D), multicellular, "organotypic" cell cultures, which mimic functional responses of living tissue. However, there remains a knowledge gap regarding interactions across different biological scales, complicating accurate prediction of health outcomes from molecular/genomic data and tissue responses. Systems biology aims at mathematical modeling of complex, non-linear biological systems. We propose to apply a systems biology approach to achieve a computational representation of tissue-level physiological responses by integrating empirical data derived from organotypic culture systems with computational models of intracellular pathways to better predict human responses. Successful implementation of this integrated approach will provide a powerful tool for faster, more accurate and cost-effective screening of potential toxicants and therapeutics. On September 11, 2015, an interdisciplinary group of scientists, engineers, and clinicians gathered for a workshop in Research Triangle Park, North Carolina, to discuss this ambitious goal. Participants represented laboratory-based and computational modeling approaches to pharmacology and toxicology, as well as the pharmaceutical industry, government, non-profits, and academia. Discussions focused on identifying critical system perturbations to model, the computational tools required, and the experimental approaches best suited to generating key data.


Asunto(s)
Técnicas de Cultivo de Célula , Simulación por Computador , Biología de Sistemas , Alternativas a las Pruebas en Animales , Animales , Técnicas de Cultivo de Célula/métodos , Sustancias Peligrosas/toxicidad , Humanos , Dispositivos Laboratorio en un Chip , Medición de Riesgo
19.
Cell Death Discov ; 2: 16067, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27648301

RESUMEN

Colon cancer is a leading cause of cancer-related mortality for which targeted therapy is needed; however, trials using apoptosis-inducing ligand monotherapy to overcome resistance to apoptosis have not shown clinical responses. Since colon cancer cells selectively uptake and rapidly metabolize glucose, a property utilized for clinical staging, we investigated mechanisms to alter glucose metabolism in order to selectively target the cancer cells and to overcome evasion of apoptosis. We demonstrate TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) resistance in the majority of human colon cancers tested and utilize the glucose analog 2-deoxy-d-glucose to sensitize TRAIL-resistant gastrointestinal adenocarcinoma cells, and not normal gastrointestinal epithelial cells, to TRAIL-induced apoptosis through enhanced death receptor 5 expression, downstream modulation of MAPK signaling and subsequent miRNA expression modulation by increasing the expression of miR-494 via MEK activation. Further, established human colon cancer xenografts treated with this strategy experience anti-tumor responses. These findings in colon adenocarcinoma support further investigation of manipulation of cellular energetics to selectively overcome resistance to apoptosis and to impart tumor regressions in established colon cancer tumors.

20.
Arch Immunol Ther Exp (Warsz) ; 64(5): 417-23, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27197661

RESUMEN

Program death-1 (PD-1) is a co-inhibitory receptor inducibly expressed on activated T cells. PD-1 has been reported to be associated with the development of several autoimmune diseases including rheumatoid arthritis, but the precise cellular and molecular mechanisms have not been fully elucidated. To study the role of PD-1 in the pathogenesis of rheumatoid arthritis and the possible underlying mechanisms, we performed collagen-induced arthritis (CIA) in C57BL/6 mice. Here, we show that PD-1 deficiency leads to the development of severe CIA in mice. When analyzing T cells from CIA mice ex vivo, we noticed aberrant antigen-specific Th17 responses in mice lacking PD-1. This is possibly due to deregulated activation of PKC-θ and Akt. In support of this notion, treating Pdcd1 (-/-) mice with an inhibitor of PI3-kinase that is upstream of PKC-θ and Akt significantly suppressed the disease severity. Therefore, our data indicate that PD-1 dampens antigen-specific Th17 response, thus inhibiting the disease.


Asunto(s)
Artritis Reumatoide/inmunología , Enfermedades Autoinmunes/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Células Th17/citología , Animales , Artritis/inmunología , Artritis/metabolismo , Artritis Experimental , Artritis Reumatoide/metabolismo , Enfermedades Autoinmunes/metabolismo , Diferenciación Celular , Colágeno/química , Modelos Animales de Enfermedad , Citometría de Flujo , Humanos , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA