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













Base de datos
Intervalo de año de publicación
1.
Gut ; 73(3): 509-520, 2024 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-37770128

RESUMEN

OBJECTIVE: Liver metastases are often resistant to immune checkpoint inhibitor therapy (ICI) and portend a worse prognosis compared with metastases to other locations. Regulatory T cells (Tregs) are one of several immunosuppressive cells implicated in ICI resistance of liver tumours, but the role played by Tregs residing within the liver surrounding a tumour is unknown. DESIGN: Flow cytometry and single-cell RNA sequencing were used to characterise hepatic Tregs before and after ICI therapy. RESULTS: We found that the murine liver houses a Treg population that, unlike those found in other organs, is both highly proliferative and apoptotic at baseline. On administration of αPD-1, αPD-L1 or αCTLA4, the liver Treg population doubled regardless of the presence of an intrahepatic tumour. Remarkably, this change was not due to the preferential expansion of the subpopulation of Tregs that express PD-1. Instead, a subpopulation of CD29+ (Itgb1, integrin ß1) Tregs, that were highly proliferative at baseline, doubled its size in response to αPD-1. Partial and full depletion of Tregs identified CD29+ Tregs as the prominent niche-filling subpopulation in the liver, and CD29+ Tregs demonstrated enhanced suppression in vitro when derived from the liver but not the spleen. We identified IL2 as a critical modulator of both CD29+ and CD29- hepatic Tregs, but expansion of the liver Treg population with αPD-1 driven by CD29+ Tregs was in part IL2-independent. CONCLUSION: We propose that CD29+ Tregs constitute a unique subpopulation of hepatic Tregs that are primed to respond to ICI agents and mediate resistance.


Asunto(s)
Neoplasias Hepáticas , Linfocitos T Reguladores , Animales , Ratones , Interleucina-2 , Integrina beta1 , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología
2.
J Immunol ; 211(7): 1099-1107, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37624046

RESUMEN

The gut microbiome is an important modulator of the host immune system. In this study, we found that altering the gut microbiome by oral vancomycin increases liver invariant NKT (iNKT) cell function. Enhanced iNKT cytokine production and activation marker expression were observed in vancomycin-treated mice following both Ag-specific and Ag-independent in vivo iNKT stimulations, with a more prominent effect in the liver than in the spleen. Fecal transplantation studies demonstrated that the iNKT functional regulation is mediated by altering the gut microbiome but uncoupled from the modulation of iNKT cell population size. Interestingly, when stimulated in vitro, iNKT cells from vancomycin-treated mice did not show increased activation, suggesting an indirect regulation. iNKT cells expressed high levels of IL-18 receptor, and vancomycin increased the expression of IL-18 in the liver. Blocking IL-18 by neutralizing Ab or using genetically deficient mice attenuated the enhanced iNKT activation. Liver macrophages were identified as a major source of IL-18. General macrophage depletion by clodronate abolished this iNKT activation. Using anti-CSF-1R depletion or LyzCrexCSF-1RLsL-DTR mice identified CSF-1R+ macrophages as a critical modulator of iNKT function. Vancomycin treatment had no effect on iNKT cell function in vivo in IL-18 knockout macrophage reconstituted mice. Together, our results demonstrate that the gut microbiome controls liver iNKT function via regulating CSF-1R+ macrophages to produce IL-18.


Asunto(s)
Microbioma Gastrointestinal , Ratones , Animales , Interleucina-18 , Vancomicina/farmacología , Macrófagos , Hígado , Ratones Noqueados , Proteínas Tirosina Quinasas Receptoras
3.
Cancer Cell ; 40(10): 1173-1189.e6, 2022 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-36220073

RESUMEN

Cancer immunotherapy often depends on recognition of peptide epitopes by cytotoxic T lymphocytes (CTLs). The tumor microenvironment (TME) is enriched for peroxynitrite (PNT), a potent oxidant produced by infiltrating myeloid cells and some tumor cells. We demonstrate that PNT alters the profile of MHC class I bound peptides presented on tumor cells. Only CTLs specific for PNT-resistant peptides have a strong antitumor effect in vivo, whereas CTLs specific for PNT-sensitive peptides are not effective. Therapeutic targeting of PNT in mice reduces resistance of tumor cells to CTLs. Melanoma patients with low PNT activity in their tumors demonstrate a better clinical response to immunotherapy than patients with high PNT activity. Our data suggest that intratumoral PNT activity should be considered for the design of neoantigen-based therapy and also may be an important immunotherapeutic target.


Asunto(s)
Melanoma , Microambiente Tumoral , Animales , Antígenos de Neoplasias/metabolismo , Epítopos , Antígenos de Histocompatibilidad Clase I/metabolismo , Inmunoterapia , Melanoma/metabolismo , Ratones , Oxidantes/metabolismo , Péptidos , Ácido Peroxinitroso/metabolismo , Linfocitos T Citotóxicos
4.
Cancer Cell ; 40(9): 986-998.e5, 2022 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-36055226

RESUMEN

Platelets, the often-overlooked component of the immune system, have been shown to promote tumor growth. Non-alcoholic fatty liver disease (NAFLD) is a common disease in the Western world and rising risk for hepatocellular carcinoma (HCC). Unexpectedly, we observed that platelets can inhibit the growth of established HCC in NAFLD mice. Through pharmacological inhibition and genetic depletion of P2Y12 as well as in vivo transfusion of wild-type (WT) or CD40L-/- platelets, we demonstrate that the anti-tumor function of platelets is mediated through P2Y12-dependent CD40L release, which leads to CD8+ T cell activation by the CD40 receptor. Unlike P2Y12 inhibition, blocking platelets with aspirin does not prevent platelet CD40L release nor accelerate HCC in NAFLD mice. Similar findings were observed in liver metastasis models. All together, our study reveals a complex role of platelets in tumor regulation. Anti-platelet treatment without inhibiting CD40L release could be considered for liver cancer patients with NAFLD.


Asunto(s)
Plaquetas/inmunología , Carcinoma Hepatocelular , Neoplasias Hepáticas , Enfermedad del Hígado Graso no Alcohólico , Receptores Purinérgicos P2Y12/metabolismo , Animales , Ligando de CD40/genética , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Ratones , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/genética
5.
Cancer Immunol Res ; 9(9): 1024-1034, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34193462

RESUMEN

Mucosal-associated invariant T (MAIT) cells are MR1-restricted innate-like T cells that recognize non-peptide antigens including riboflavin derivates. Although in vitro-activated MAIT cells show antitumor activity, the in vivo role of MAIT cells in cancer is still unclear. Here, we have shown that MAIT cells have antitumor function in vivo when activated by a combination of the synthetic riboflavin synthesis pathway-derived antigen 5-OP-RU [5-(2-oxopropylideneamino)-6-D-ribitylaminouracil] and the Toll-like receptor 9 (TLR9) agonist CpG. Coadministration of 5-OP-RU and CpG induced strong systemic in vivo expansion and activation of MAIT cells with high CD69 expression, pronounced effector memory phenotype, and upregulated levels of effector molecules including IFNγ, granzyme B, and perforin. Activated and expanded MAITs induced a potent and broad antitumor immune response in murine models of liver metastasis and hepatocellular carcinoma, lung metastasis, and subcutaneous tumors in two different mouse strains. Such tumor inhibition was absent in MAIT-deficient Mr1 -/- mice. CRISPR/Cas9-mediated MR1 knockout in tumor cells did not affect efficacy of this MAIT-directed immunotherapy, pointing toward an indirect mechanism of action. Our findings suggest that MAIT cells are an attractive target for cancer immunotherapy.See related Spotlight by Lantz, p. 996.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Activación de Linfocitos/inmunología , Antígenos de Histocompatibilidad Menor/metabolismo , Células T Invariantes Asociadas a Mucosa/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Animales , Antígenos CD , Antígenos de Diferenciación de Linfocitos T , Sistemas CRISPR-Cas , Línea Celular Tumoral , Femenino , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Lectinas Tipo C , Masculino , Ratones , Antígenos de Histocompatibilidad Menor/genética , Células T Invariantes Asociadas a Mucosa/metabolismo , Neoplasias/metabolismo , Ribitol/administración & dosificación , Ribitol/análogos & derivados , Riboflavina/biosíntesis , Riboflavina/química , Riboflavina/farmacología , Uracilo/administración & dosificación , Uracilo/análogos & derivados
6.
Cell Mol Gastroenterol Hepatol ; 12(3): 1166-1178, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34033968

RESUMEN

BACKGROUND & AIMS: Intrahepatic cholangiocarcinoma (iCCA) accounts for a fraction of primary liver cancers but has a 5-year survival rate of only 10%. Immune checkpoint inhibitors are effective in treating many solid cancers, but immune checkpoint inhibitor monotherapy has no clear benefit in iCCA. Mitogen-activated kinase (MEK) inhibitors, such as trametinib, have shown promising results in preclinical studies for iCCA by inhibiting cell proliferation and modifying the tumor microenvironment. This study aimed to show the potential benefit of combining trametinib with anti-programmed cell death protein 1 (PD-1) therapy in different iCCA mouse models. METHODS: Here, we assessed the in vitro cytotoxicity of trametinib in mouse (SB1 and LD-1) and human (EGI-1) cholangiocarcinoma cell lines. We examined the efficacy of single-agent trametinib, anti-PD-1, and a combination of both in subcutaneous, orthotopic, and plasmid-induced iCCA mouse models. Flow cytometry analysis was used to elucidate changes in the tumor immune microenvironment upon treatment. Whole-exome sequencing (WES) was performed on the SB1 tumor cell line to correlate this preclinical model with iCCAs in patients. RESULTS: Trametinib reduced tumor cell growth of SB1, LD-1, and EGI-1 tumor cells in vitro. Trametinib treatment led to up-regulation of major histocompatibility complex (MHC-I) and programmed cell death ligand 1 (PD-L-1) (programmed cell death ligand 1) on tumor cells in vitro. The combination of trametinib and anti-PD-1 reduced tumor burden in several iCCA tumor models and improved survival in SB1 tumor-bearing mice compared with either agent alone. Immunoprofiling of tumor-bearing mice showed an increase of hepatic effector memory CD8+ and CD4+ T cells, as well as an increased degranulation of CD8+ T cells, indicating enhanced cytotoxicity. WES and somatic mutational analysis showed no mutations of KRAS, BRAF, and ERK in SB1 tumor cells, and showed a similar genetic signature of SB1 found in a cohort of patients with iCCA. CONCLUSIONS: Altogether, our study shows that trametinib improves the immunogenicity of tumor cells by up-regulating MHC-I surface expression. The combination with anti-PD-1 results in optimal treatment efficacy for iCCA. WES of SB1 cells suggests that KRAS wild-type iCCAs also respond to this combination therapy.


Asunto(s)
Neoplasias de los Conductos Biliares/tratamiento farmacológico , Colangiocarcinoma/tratamiento farmacológico , Inhibidores de Puntos de Control Inmunológico/administración & dosificación , Piridonas/administración & dosificación , Pirimidinonas/administración & dosificación , Animales , Neoplasias de los Conductos Biliares/metabolismo , Neoplasias de los Conductos Biliares/mortalidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colangiocarcinoma/metabolismo , Colangiocarcinoma/mortalidad , Sinergismo Farmacológico , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Ratones , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Piridonas/farmacología , Pirimidinonas/farmacología , Secuenciación del Exoma , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cell Immunol ; 361: 104295, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33508529

RESUMEN

Liver cancer is a leading cause of cancer deaths worldwide. Hepatocellular carcinoma (~75-85%) and cholangiocarcinoma (~10-15%) account for the majority of primary liver malignancies. Patients with primary liver cancer are often diagnosed with unresectable diseases and do not respond well to current therapies. The liver is also a common site of metastasis. Liver metastasis is difficult to treat, and the prognosis is poor. Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells with immunosuppressive activity. MDSCs are an important component of the tumor microenvironment and promote tumor progression through various mechanisms. MDSCs expand in both liver cancer patients and mouse liver cancer models. Importantly, MDSCs correlate with poor clinical outcomes for liver cancer patients. The tumor-promoting functions of MDSCs have also been shown in mouse liver cancer models. All these studies suggest that targeting MDSCs can potentially benefit liver cancer treatment. This review summarizes the current findings of MDSC regulation in liver cancer and related disease conditions.


Asunto(s)
Carcinógenos , Carcinoma Hepatocelular/inmunología , Neoplasias Hepáticas/inmunología , Células Supresoras de Origen Mieloide/inmunología , Microambiente Tumoral/inmunología , Animales , Carcinoma Hepatocelular/patología , Modelos Animales de Enfermedad , Humanos , Hígado/inmunología , Hígado/patología , Neoplasias Hepáticas/patología , Células Mieloides/inmunología
8.
Cancer Discov ; 11(5): 1248-1267, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33323397

RESUMEN

Gut dysbiosis is commonly observed in patients with cirrhosis and chronic gastrointestinal disorders; however, its effect on antitumor immunity in the liver is largely unknown. Here we studied how the gut microbiome affects antitumor immunity in cholangiocarcinoma. Primary sclerosing cholangitis (PSC) or colitis, two known risk factors for cholangiocarcinoma which promote tumor development in mice, caused an accumulation of CXCR2+ polymorphonuclear myeloid-derived suppressor cells (PMN-MDSC). A decrease in gut barrier function observed in mice with PSC and colitis allowed gut-derived bacteria and lipopolysaccharide to appear in the liver and induced CXCL1 expression in hepatocytes through a TLR4-dependent mechanism and an accumulation of CXCR2+ PMN-MDSCs. In contrast, neomycin treatment blocked CXCL1 expression and PMN-MDSC accumulation and inhibited tumor growth even in the absence of liver disease or colitis. Our study demonstrates that the gut microbiome controls hepatocytes to form an immunosuppressive environment by increasing PMN-MDSCs to promote liver cancer. SIGNIFICANCE: MDSCs have been shown to be induced by tumors and suppress antitumor immunity. Here we show that the gut microbiome can control accumulation of MDSCs in the liver in the context of a benign liver disease or colitis.See related commentary by Chagani and Kwong, p. 1014.This article is highlighted in the In This Issue feature, p. 995.


Asunto(s)
Colangiocarcinoma/patología , Bacterias Gramnegativas/fisiología , Hepatocitos/fisiología , Neoplasias Hepáticas/patología , Células Supresoras de Origen Mieloide/fisiología , Animales , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Humanos , Ratones
9.
J Hepatol ; 74(5): 1145-1154, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33276030

RESUMEN

BACKGROUND & AIMS: While cholangiocarcinomas (CCAs) commonly express programmed cell death 1 (PD-1) and its ligand (PD-L1), they respond poorly to immune checkpoint inhibitors (ICIs). We aimed to determine whether stimulating antigen-presenting cells, including macrophages and dendritic cells, using a CD40 agonist could improve this response. METHODS: We compared treatment responses in subcutaneous, orthotopic, and 2 plasmid-based murine intrahepatic CCA (iCCA) models. Mice were treated for 4 weeks with weekly IgG control, a CD40 agonistic antibody, anti-PD-1, or the combination of both (anti-CD40/PD-1). Flow cytometric (FACS) analysis of lymphocytes and myeloid cell populations (including activation status) was performed. We used dendritic cell knockout mice, and macrophage, CD4+ and CD8+ T cell depletion models to identify effector cells. Anti-CD40/PD-1 was combined with chemotherapy (gemcitabine/cisplatin) to test for improved therapeutic efficacy. RESULTS: In all 4 models, anti-PD-1 alone was minimally efficacious. Mice exhibited a moderate response to CD40 agonist monotherapy. Combination anti-CD40/PD-1 therapy led to a significantly greater reduction in tumor burden. FACS demonstrated increased number and activation of CD4+ and CD8+ T cells, natural killer cells, and myeloid cells in tumor and non-tumor liver tissue of tumor-bearing mice treated with anti-CD40/PD-1. Depletion of macrophages, dendritic cells, CD4+ T cells, or CD8+ T cells abrogated treatment efficacy. Combining anti-CD40/PD-1 with gemcitabine/cisplatin resulted in a significant survival benefit compared to gemcitabine/cisplatin alone. CONCLUSION: CD40-mediated activation of macrophages and dendritic cells in iCCA significantly enhances response to anti-PD-1 therapy. This regimen may enhance the efficacy of first-line chemotherapy. LAY SUMMARY: Checkpoint inhibition, a common form of immune therapy, is generally ineffective for the treatment of cholangiocarcinoma. These tumors suppress the infiltration and function of surrounding immune cells. Stimulating immune cells such as macrophages and dendritic cells via the CD40 receptor activates downstream immune cells and enhances the response to checkpoint inhibitors.


Asunto(s)
Antígenos CD40/agonistas , Colangiocarcinoma , Inhibidores de Puntos de Control Inmunológico/farmacología , Neoplasias Hepáticas , Activación de Macrófagos/inmunología , Microambiente Tumoral , Animales , Antimetabolitos Antineoplásicos/farmacología , Línea Celular Tumoral , Colangiocarcinoma/inmunología , Colangiocarcinoma/patología , Cisplatino/farmacología , Células Dendríticas/inmunología , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Sensibilidad Colateral al uso de Fármacos , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Factores Activadores de Macrófagos/inmunología , Ratones , Ratones Noqueados , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Gemcitabina
10.
Gastroenterology ; 160(1): 331-345.e6, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33010248

RESUMEN

BACKGROUND & AIMS: Nonalcoholic steatohepatitis causes loss of hepatic CD4+ T cells and promotes tumor growth. The liver is the most common site of distant metastases from a variety of malignancies, many of which respond to immunotherapy. We investigated the effects of steatohepatitis on the efficacy of immunotherapeutic agents against liver tumors in mice. METHODS: Steatohepatitis was induced by feeding C57BL/6NCrl or BALB/c AnNCr mice a methionine and choline-deficient diet or a choline-deficient l-amino acid-defined diet. Mice were given intrahepatic or subcutaneous injections of B16 melanoma and CT26 colon cancer cells, followed by intravenous injections of M30-RNA vaccine (M30) or intraperitoneal injections of an antibody against OX40 (aOX40) on days 3, 7, and 10 after injection of the tumor cells. We measured tumor growth and analyzed immune cells in tumor tissues by flow cytometry. Mice were given N-acetylcysteine to prevent loss of CD4+ T cells from liver. RESULTS: Administration of M30 and aOX40 inhibited growth of tumors from intrahepatic injections of B16 or CT26 cells in mice on regular diet. However, M30 and/or aOX40 did not slow growth of liver tumors from B16 or CT26 cells in mice with diet-induced steatohepatitis (methionine and choline-deficient diet or choline-deficient l-amino acid-defined diet). Steatohepatitis did not affect the ability of M30 to slow growth of subcutaneous B16 tumors. In mice with steatohepatitis given N-acetylcysteine, which prevents loss of CD4+ T cells, M30 and aOX40 were able slow growth of hepatic tumors. Flow cytometry analysis of liver tumors revealed reduced CD4+ T cells and effector memory cells in mice with vs without steatohepatitis. CONCLUSIONS: Steatohepatitis reduces the abilities of immunotherapeutic agents, such as M30 and aOX40, to inhibit tumor liver growth by reducing tumor infiltration by CD4+ T cells and effector memory cells. N-acetylcysteine restores T-cell numbers in tumors and increases the ability of M30 and aOX40 to slow tumor growth in mice.


Asunto(s)
Inmunoterapia , Neoplasias Hepáticas/etiología , Neoplasias Hepáticas/terapia , Melanoma/terapia , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Linfocitos T/fisiología , Animales , Modelos Animales de Enfermedad , Neoplasias Hepáticas/patología , Melanoma/etiología , Melanoma/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/patología
11.
J Hepatol ; 70(3): 449-457, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30414862

RESUMEN

BACKGROUND & AIMS: Cytokine-induced killer (CIK) cell-based immunotherapy is effective as an adjuvant therapy in early stage hepatocellular carcinoma (HCC) but lacks efficacy in advanced HCC. We aimed to investigate immune suppressor mechanisms in HCC, focusing on the role of myeloid-derived suppressor cells (MDSCs) in response to CIK therapy. METHODS: MDSCs were quantified by flow cytometry and quantitative real-time PCR. Cytokines were detected by cytokine array. A lactate dehydrogenase cytotoxicity assay was performed in the presence or absence of MDSCs to study CIK function against HCC cells in vitro. An FDA-approved PDE5 inhibitor, tadalafil, was used to target MDSCs in vitro and in vivo. Two different murine HCC cell lines were tested in subcutaneous and orthotopic tumor models in C57BL/6 and BALB/c mice. The antitumor effects of human CIKs and MDSCs were also tested in vitro. RESULTS: Adoptive cell transfer of CIKs into tumor-bearing mice induced inflammatory mediators (e.g., CX3CL1, IL-13) in the tumor microenvironment and an increase of tumor-infiltrating MDSCs, leading to impaired antitumor activity in 2 different HCC models. MDSCs efficiently suppressed the cytotoxic activity of CIKs in vitro. In contrast, treatment with a PDE5 inhibitor reversed the MDSC suppressor function via ARG1 and iNOS blockade and systemic treatment with a PDE5 inhibitor prevented MDSC accumulation in the tumor microenvironment upon CIK cell therapy and increased its antitumor efficacy. Similar results were observed when human CIKs were tested in vitro in the presence of CD14+HLA-DR-/low MDSCs. Treatment of MDSCs with a PDE5 inhibitor suppressed MDSC suppressor function and enhanced CIK activity against human HCC cell lines in vitro. CONCLUSION: Our results suggest that targeting MDSCs is an efficient strategy to enhance the antitumor efficacy of CIKs for the treatment of patients with HCC. LAY SUMMARY: Cytokine-induced killer cells are a mixture of immune cells given to eliminate cancer cells. However, not all patients respond to this treatment. Herein, we show in 2 different liver cancer models that myeloid-derived suppressor cells are increased in response to cytokine-induced killer cell therapy. Targeting these myeloid-derived suppressor cells may provide an additional therapeutic benefit alongside cytokine-induced killer cell therapy.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Células Asesinas Inducidas por Citocinas/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Células Supresoras de Origen Mieloide/metabolismo , Inhibidores de Fosfodiesterasa 5/uso terapéutico , Tadalafilo/uso terapéutico , Traslado Adoptivo/métodos , Animales , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Terapia Combinada/métodos , Células Asesinas Inducidas por Citocinas/inmunología , Citocinas/metabolismo , Femenino , Humanos , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células Supresoras de Origen Mieloide/efectos de los fármacos , Inhibidores de Fosfodiesterasa 5/farmacología , Transducción de Señal/efectos de los fármacos , Tadalafilo/farmacología , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Cell Res ; 28(11): 1103-1117, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30297869

RESUMEN

Androgen deprivation therapy (ADT) is a main treatment for prostate cancer (PCa) but the disease often recurs and becomes castration-resistant in nearly all patients. Recent data implicate the involvement of immune cells in the development of this castration-resistant prostate cancer (CRPC). In particular, T cells have been found to be expanded in both PCa patients and mouse models shortly after androgen deprivation. However, whether or which of the T cell subtypes play an important role during the development of CRPC is unknown. Here we identified a novel population of CD4lowHLA-G+ T cells that undergo significant expansion in PCa patients after ADT. In mouse PCa models, a similar CD4low T cell population expands during the early stages of CRPC onset. These cells are identified as IL-4-expressing TH17 cells, and are shown to be associated with CRPC onset in patients and essential for the development of CRPC in mouse models. Mechanistically, CD4lowHLA-G+ T cells drive androgen-independent growth of prostate cancer cells by modulating the activity and migration of CD11blowF4/80hi macrophages. Furthermore, following androgen deprivation, elevated PGE2-EP2 signaling inhibited the expression of CD4 in thymocytes, and subsequently induced the polarization of CD4low naïve T cells towards the IL-4-expressing TH17 phenotype via up-regulation of IL23R. Therapeutically, inactivating PGE2 signaling with celecoxib at a time when CD4lowHLA-G+ T cells appeared, but not immediately following androgen deprivation, dramatically suppressed the onset of CRPC. Collectively, our results indicate that an unusual population of CD4lowHLA-G+ T cells is essential for the development of CRPC and point to a new therapeutic avenue of combining ADT with PGE2 inhibition for the treatment of prostate cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Linfocitos T CD4-Positivos/inmunología , Celecoxib/uso terapéutico , Antígenos HLA-G/inmunología , Neoplasias de la Próstata Resistentes a la Castración/inmunología , Neoplasias de la Próstata Resistentes a la Castración/terapia , Animales , Línea Celular Tumoral , Masculino , Ratones , Ratones Endogámicos C57BL
13.
Cancer Immunol Immunother ; 67(8): 1305-1315, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29959458

RESUMEN

Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related death worldwide. Immune checkpoint blockade with anti-CTLA-4 and anti-PD-1 antibodies has shown promising results in the treatment of patients with advanced HCC. The anti-PD-1 antibody, nivolumab, is now approved for patients who have had progressive disease on the current standard of care. However, a subset of patients with advanced HCC treated with immune checkpoint inhibitors failed to respond to therapy. Here, we provide evidence of adaptive resistance to immune checkpoint inhibitors through upregulation of indoleamine 2,3-dioxygenase (IDO) in HCC. Anti-CTLA-4 treatment promoted an induction of IDO1 in resistant HCC tumors but not in tumors sensitive to immune checkpoint blockade. Using both subcutaneous and hepatic orthotopic models, we found that the addition of an IDO inhibitor increases the efficacy of treatment in HCC resistant tumors with high IDO induction. Furthermore, in vivo neutralizing studies demonstrated that the IDO induction by immune checkpoint blockade was dependent on IFN-γ. Similar findings were observed with anti-PD-1 therapy. These results provide evidence that IDO may play a role in adaptive resistance to immune checkpoint inhibitors in patients with HCC. Therefore, inhibiting IDO in combination with immune checkpoint inhibitors may add therapeutic benefit in tumors which overexpress IDO and should be considered for clinical evaluation in HCC.


Asunto(s)
Carcinoma Hepatocelular/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Neoplasias Hepáticas/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Antígeno CTLA-4/inmunología , Línea Celular Tumoral , Células Hep G2 , Humanos , Interferón gamma/inmunología , Ratones , Nivolumab , Receptor de Muerte Celular Programada 1/inmunología
14.
Science ; 360(6391)2018 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-29798856

RESUMEN

Primary liver tumors and liver metastasis currently represent the leading cause of cancer-related death. Commensal bacteria are important regulators of antitumor immunity, and although the liver is exposed to gut bacteria, their role in antitumor surveillance of liver tumors is poorly understood. We found that altering commensal gut bacteria in mice induced a liver-selective antitumor effect, with an increase of hepatic CXCR6+ natural killer T (NKT) cells and heightened interferon-γ production upon antigen stimulation. In vivo functional studies showed that NKT cells mediated liver-selective tumor inhibition. NKT cell accumulation was regulated by CXCL16 expression of liver sinusoidal endothelial cells, which was controlled by gut microbiome-mediated primary-to-secondary bile acid conversion. Our study suggests a link between gut bacteria-controlled bile acid metabolism and liver antitumor immunosurveillance.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Microbioma Gastrointestinal/inmunología , Vigilancia Inmunológica , Neoplasias Hepáticas/inmunología , Hígado/metabolismo , Células T Asesinas Naturales/inmunología , Animales , Quimiocina CXCL16/metabolismo , Clostridium/metabolismo , Humanos , Hígado/inmunología , Hígado/patología , Neoplasias Hepáticas/patología , Depleción Linfocítica , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Receptores CXCR6/metabolismo
15.
Cell Death Dis ; 9(6): 620, 2018 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-29795111

RESUMEN

Hepatocellular carcinoma (HCC) is a common cause of cancer-related death worldwide. As obesity and diabetes become more prevalent, the contribution of non-alcoholic fatty liver disease (NAFLD) to HCC is rising. Recently, we reported intrahepatic CD4+ T cells are critical for anti-tumor surveillance in NAFLD. Lipid accumulation in the liver is the hallmark of NAFLD, which may perturb T cell function. We sought to investigate how the lipid-rich liver environment influences CD4+ T cells by focusing on carnitine palmitoyltransferase (CPT) family members, which control the mitochondrial ß-oxidation of fatty acids and act as key molecules in lipid catabolism. Linoleic acid (C18:2) co-localized within the mitochondria along with a corresponding increase in CPT gene upregulation. This CPT upregulation can be recapitulated by feeding mice with a high-C18:2 diet or the NAFLD promoting methionine-choline-deficient (MCD) diet. Using an agonist and antagonist, the induction of CPT genes was found to be mediated by peroxisome proliferator-activated receptor alpha (PPAR-α). CPT gene upregulation increased mitochondrial reactive oxygen species (ROS) and led to cell apoptosis. In vivo, using liver-specific inducible MYC transgenic mice fed MCD diet, blocking CPT with the pharmacological inhibitor perhexiline decreased apoptosis of intrahepatic CD4+ T cells and inhibited HCC tumor formation. These results provide useful information for potentially targeting the CPT family to rescue intrahepatic CD4+ T cells and to aid immunotherapy for NAFLD-promoted HCC.


Asunto(s)
Apoptosis/genética , Linfocitos T CD4-Positivos/patología , Carcinogénesis/patología , Carcinoma Hepatocelular/genética , Carnitina O-Palmitoiltransferasa/genética , Ácido Linoleico/farmacología , Neoplasias Hepáticas/genética , Regulación hacia Arriba/genética , Células 3T3 , Animales , Apoptosis/efectos de los fármacos , Linfocitos T CD4-Positivos/efectos de los fármacos , Carcinogénesis/genética , Carcinoma Hepatocelular/patología , Carnitina O-Palmitoiltransferasa/antagonistas & inhibidores , Carnitina O-Palmitoiltransferasa/metabolismo , Inhibidores Enzimáticos/farmacología , Humanos , Ácido Linoleico/química , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Modelos Biológicos , Enfermedad del Hígado Graso no Alcohólico/genética , Enfermedad del Hígado Graso no Alcohólico/patología , PPAR alfa/metabolismo , Perhexilina/farmacología , Especies Reactivas de Oxígeno/metabolismo
16.
FEBS J ; 285(4): 752-762, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28857485

RESUMEN

Hepatocellular carcinoma (HCC) is the sixth most frequent neoplasm and the second leading cause of cancer-related deaths worldwide. Nonalcoholic fatty liver disease (NAFLD), a common disorder in obese people, has been identified as an important risk factor for HCC. Following the increasing prevalence of obesity, it is expected that the contribution of NAFLD to HCC's incidence worldwide will grow. Recently, a number of studies have been published, which help us better understand cellular and molecular mechanisms of how NAFLD promotes hepatocarcinogensis. Inflammatory cytokines, ER stress and circadian dysregulation, which mediate hepatocyte injury and NAFLD progression, have been identified to promote malignant transformation of hepatocytes. Besides these 'intrinsic' effects, lipid dysregulation dramatically affects the liver local microenvironment. The reshaped immune environment has also been found to contribute to the NAFLD-mediated hepatocarcinogenesis. This review explores recent findings of both 'intrinsic' effects on hepatocytes and the role of the local environment in NAFLD-promoted HCC development.


Asunto(s)
Carcinoma Hepatocelular/patología , Hepatocitos/patología , Neoplasias Hepáticas/patología , Enfermedad del Hígado Graso no Alcohólico/patología , Animales , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/metabolismo , Hepatocitos/inmunología , Hepatocitos/metabolismo , Humanos , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/metabolismo , Enfermedad del Hígado Graso no Alcohólico/inmunología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Factores de Riesgo , Microambiente Tumoral/inmunología
17.
J Exp Bot ; 68(3): 513-526, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28082511

RESUMEN

Lipid molecules are key structural components of plant male reproductive organs, such as the anther and pollen. Although advances have been made in the understanding of acyl lipids in plant reproduction, the metabolic pathways of other lipid compounds, particularly glycerolipids, are not fully understood. Here we report that an endoplasmic reticulum-localized enzyme, Glycerol-3-Phosphate Acyltransferase 3 (OsGPAT3), plays an indispensable role in anther development and pollen formation in rice. OsGPAT3 is preferentially expressed in the tapetum and microspores of the anther. Compared with wild-type plants, the osgpat3 mutant displays smaller, pale yellow anthers with defective anther cuticle, degenerated pollen with defective exine, and abnormal tapetum development and degeneration. Anthers of the osgpat3 mutant have dramatic reductions of all aliphatic lipid contents. The defective cuticle and pollen phenotype coincide well with the down-regulation of sets of genes involved in lipid metabolism and regulation of anther development. Taking these findings together, this work reveals the indispensable role of a monocot-specific glycerol-3-phosphate acyltransferase in male reproduction in rice.


Asunto(s)
Glicerol-3-Fosfato O-Aciltransferasa/genética , Oryza/fisiología , Proteínas de Plantas/genética , Secuencia de Aminoácidos , Regulación hacia Abajo , Fertilidad , Flores/genética , Flores/crecimiento & desarrollo , Flores/fisiología , Glicerol-3-Fosfato O-Aciltransferasa/química , Glicerol-3-Fosfato O-Aciltransferasa/metabolismo , Oryza/genética , Oryza/crecimiento & desarrollo , Filogenia , Proteínas de Plantas/química , Proteínas de Plantas/metabolismo
18.
Cancer Res ; 75(20): 4312-21, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26249173

RESUMEN

The tumor-promoting chemokine CCL5 has been implicated in malignant transformation of breast epithelial cells, with studies to date focusing mainly on basal-type breast cancers. In this study, we investigated the consequences of CCL5 deletion in the MMTV-PyMT transgenic mouse model of luminal breast cancer. In this model, primary tumor burden and pulmonary metastases were reduced significantly in CCL5-deficient subjects, an effect found to be associated with a deficit of Th2 (IL4⁺CD4⁺ T) cells. Mechanistic investigations revealed that CCL5 activates CCR3, a highly expressed chemokine receptor on CD4⁺ T cells, and also boosts Gfi1 expression to promote the differentiation of Th2 cells, which enhance the prometastatic activity of tumor-associated myeloid cells. Clinically, polarization toward this immunosuppressive Th2 phenotype was also evident in patients with advanced luminal breast cancer. Thus, our findings showed that CCL5/CCR3 signaling promotes metastasis by inducing Th2 polarization of CD4⁺ T cells, with implications for prognosis and immunotherapy of luminal breast cancer.


Asunto(s)
Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Quimiocina CCL5/metabolismo , Células Th2/inmunología , Células Th2/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Estudios de Casos y Controles , Diferenciación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Quimiocina CCL5/sangre , Quimiocina CCL5/deficiencia , Quimiocina CCL5/genética , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Eliminación de Gen , Expresión Génica , Humanos , Interleucina-4/genética , Interleucina-4/metabolismo , Neoplasias Pulmonares/secundario , Ratones , Ratones Noqueados , Células Mieloides/inmunología , Células Mieloides/metabolismo , Metástasis de la Neoplasia , Receptores CCR3/genética , Receptores CCR3/metabolismo , Transducción de Señal , Esferoides Celulares , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Células Th2/citología , Factores de Transcripción/genética , Células Tumorales Cultivadas
19.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 21(5): 1142-7, 2013 Oct.
Artículo en Chino | MEDLINE | ID: mdl-24156422

RESUMEN

The aim of this study was to investigate the clinical characteristics and prognosis of acute erythroleukemia (AEL, AML-M6). The clinical features and results of morphologic, immunophenotypic, cytogenetic and molecular biologic detections were retrospectively analyzed in 13 cases of AEL from 305 acute leukemia patients hospitalized between October 2007 and October 2012. The results showed that the expression of erythroid and non-erythroid cells increased at the same time. The myeloid antigens mainly expressed CD13/CD33/CD117/CD34, while the erythroid antigens expressed Gly and CD71. The karyotypic detection indicated that there were 1 case with normal karyotype, 3 cases with simple karyotypic abnormality and 2 cases with complex karyotypic abnormality, the other cases were not detected. The molecular biological detection found that the poor prognosis gene existed in 5 cases [38.5% (5/13)], including 3 cases with MLL-MLL fusion gene, 1 case with MLL mutation, and 1 cases with NRAS gene mutation, the abnormal genes were not detected in remainder 8 cases. After chemotherapy with decitabine, the complete remission (CR) rate achieved 53.5% (7/13), partial remission (DR) rate achieved 15.4% (2/13). Finally, 8 patients received allo-HSCT, the median overall survival (OS) was 20.7 months, 3 year survival rate was 79%, 3 year disease-free survival rate was 78%. It is concluded that the acute erythroleukemia is a rare subtype of AML, which is transformed from MDS and has harmful genes and poor prognosis. Allo-HSCT and treatment with decitabine may enhance the survival rate of AEL.


Asunto(s)
Leucemia Eritroblástica Aguda/diagnóstico , Leucemia Eritroblástica Aguda/genética , Adolescente , Adulto , Anciano , Niño , Preescolar , Femenino , Humanos , Cariotipificación , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Tasa de Supervivencia , Adulto Joven
20.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 21(5): 1305-8, 2013 Oct.
Artículo en Chino | MEDLINE | ID: mdl-24156455

RESUMEN

The aim of this study was to observe the protective effect of ademetionine 1, 4-butanedisulfonate on liver injury caused by chemotherapy in patients with leukemia. The clinical data of protective effect were analyzed retrospectively from January 2010 to April 2012. A total of 62 acute leukemia patients were divided into A group (27 cases) and B group (35 cases), the polyene phosphatidyl choline combined with ademetionine or combined with compound glycyrrhizin were given in A and B group, respectively. The changes of liver function were observed after 2 weeks, 5 patients in B group suffered from acute liver injury were treated by ademetionine as rescue therapy. Liver function was compared before and after treatment. The results showed that ALT and AST levels were significantly reduced in A group (P < 0.05), none of the patients (0/27) suffered from acute liver injury, but 14.29% (5/35) patients in B group suffered from acute liver injury, and liver function could be recovered by substitution treatment of ademetionine (the median time is 8 days, 5-14 days). It is concluded that the protective and therapeutic effect of ademetionine against liver injury caused by chemotherapy in patients with leukemia is better than that of compound glycyrrhizin.


Asunto(s)
Antineoplásicos/efectos adversos , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Leucemia/tratamiento farmacológico , S-Adenosilmetionina/análogos & derivados , Adulto , Femenino , Ácido Glicirrínico/uso terapéutico , Humanos , Masculino , Persona de Mediana Edad , S-Adenosilmetionina/uso terapéutico , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA