Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 43(6): 114288, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38814782

RESUMEN

Lipids have emerged as potent regulators of immune cell function. In the skin, adipocyte lipolysis increases the local pool of free fatty acids and is essential for coordinating early macrophage inflammation following injury. Here, we investigate G-protein-coupled receptor 84 (GPR84), a medium-chain fatty acid (MCFA) receptor, for its potential to propagate pro-inflammatory signaling after skin injury. GPR84 signaling was identified as a key component of regulating myeloid cell numbers and subsequent tissue repair through in vivo administration of a pharmacological antagonist and the MCFA decanoic acid. We found that impaired injury-induced dermal adipocyte lipolysis is a hallmark of diabetes, and lipidomic analysis demonstrated that MCFAs are significantly reduced in diabetic murine wounds. Furthermore, local administration of decanoic acid rescued myeloid cell numbers and tissue repair during diabetic wound healing. Thus, GPR84 is a readily targetable lipid signaling pathway for manipulating injury-induced tissue inflammation with beneficial effects on acute diabetic healing.


Asunto(s)
Diabetes Mellitus Experimental , Inflamación , Receptores Acoplados a Proteínas G , Piel , Cicatrización de Heridas , Animales , Receptores Acoplados a Proteínas G/metabolismo , Piel/patología , Piel/metabolismo , Piel/lesiones , Ratones , Inflamación/patología , Inflamación/metabolismo , Cicatrización de Heridas/efectos de los fármacos , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Ratones Endogámicos C57BL , Masculino , Transducción de Señal , Ácidos Decanoicos/farmacología , Lipólisis/efectos de los fármacos , Humanos , Adipocitos/metabolismo , Células Mieloides/metabolismo
2.
J Exp Clin Cancer Res ; 43(1): 60, 2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38414061

RESUMEN

BACKGROUND: Cancer cells can overexpress CD47, an innate immune checkpoint that prevents phagocytosis upon interaction with signal regulatory protein alpha (SIRPα) expressed in macrophages and other myeloid cells. Several clinical trials have reported that CD47 blockade reduces tumor growth in hematological malignancies. However, CD47 blockade has shown modest results in solid tumors, including melanoma. Our group has demonstrated that histone deacetylase 6 inhibitors (HDAC6is) have immunomodulatory properties, such as controlling macrophage phenotype and inflammatory properties. However, the molecular and cellular mechanisms controlling these processes are not fully understood. In this study, we evaluated the role of HDAC6 in regulating the CD47/SIRPα axis and phagocytosis in macrophages. METHODS: We tested the role of HDAC6is, especially Nexturastat A, in regulating macrophage phenotype and phagocytic function using bone marrow-derived macrophages and macrophage cell lines. The modulation of the CD47/SIRPα axis and phagocytosis by HDAC6is was investigated using murine and human melanoma cell lines and macrophages. Phagocytosis was evaluated via coculture assays of macrophages and melanoma cells by flow cytometry and immunofluorescence. Lastly, to evaluate the antitumor activity of Nexturastat A in combination with anti-CD47 or anti-SIRPα antibodies, we performed in vivo studies using the SM1 and/or B16F10 melanoma mouse models. RESULTS: We observed that HDAC6is enhanced the phenotype of antitumoral M1 macrophages while decreasing the protumoral M2 phenotype. In addition, HDAC6 inhibition diminished the expression of SIRPα, increased the expression of other pro-phagocytic signals in macrophages, and downregulated CD47 expression in mouse and human melanoma cells. This regulatory role on the CD47/SIRPα axis translated into enhanced antitumoral phagocytic capacity of macrophages treated with Nexturastat A and anti-CD47. We also observed that the systemic administration of HDAC6i enhanced the in vivo antitumor activity of anti-CD47 blockade in melanoma by modulating macrophage and natural killer cells in the tumor microenvironment. However, Nexturastat A did not enhance the antitumor activity of anti-SIRPα despite its modulation of macrophage populations in the SM1 tumor microenvironment. CONCLUSIONS: Our results demonstrate the critical regulatory role of HDAC6 in phagocytosis and innate immunity for the first time, further underscoring the use of these inhibitors to potentiate CD47 immune checkpoint blockade therapeutic strategies.


Asunto(s)
Ácidos Hidroxámicos , Melanoma , Neoplasias , Compuestos de Fenilurea , Humanos , Ratones , Animales , Antígeno CD47/metabolismo , Fagocitosis , Inmunoterapia/métodos , Neoplasias/patología , Microambiente Tumoral , Histona Desacetilasa 6
3.
Mol Cancer Ther ; 22(12): 1376-1389, 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-37586844

RESUMEN

Radiotherapy is a curative cancer treatment modality that imparts damage to cellular DNA, induces immunogenic cell death, and activates antitumor immunity. Despite the radiotherapy-induced direct antitumor effect seen within the treated volume, accumulating evidence indicates activation of innate antitumor immunity. Acute proinflammatory responses mediated by anticancer M1 macrophages are observed in the immediate aftermath following radiotherapy. However, after a few days, these M1 macrophages are converted to anti-inflammatory and pro-cancer M2 phenotype, leading to cancer resistance and underlying potential tumor relapse. Histone deacetylase 6 (HDAC6) plays a crucial role in regulating macrophage polarization and innate immune responses. Here, we report targeting HDAC6 function with a novel selective inhibitor (SP-2-225) as a potential therapeutic candidate for combination therapy with radiotherapy. This resulted in decreased tumor growth and enhanced M1/M2 ratio of infiltrating macrophages within tumors. These observations support the use of selective HDAC6 inhibitors to improve antitumor immune responses and prevent tumor relapse after radiotherapy.


Asunto(s)
Neoplasias , Humanos , Histona Desacetilasa 6 , Neoplasias/tratamiento farmacológico , Neoplasias/radioterapia , Macrófagos , Inmunidad Innata , Recurrencia
4.
J Immunother Cancer ; 10(11)2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36343976

RESUMEN

BACKGROUND: Novel therapies are urgently needed for ovarian cancer (OC), the fifth deadliest cancer in women. Preclinical work has shown that DNA methyltransferase inhibitors (DNMTis) can reverse the immunosuppressive tumor microenvironment in OC. Inhibiting DNA methyltransferases activate transcription of double-stranded (ds)RNA, including transposable elements. These dsRNAs activate sensors in the cytoplasm and trigger type I interferon (IFN) signaling, recruiting host immune cells to kill the tumor cells. Adenosine deaminase 1 (ADAR1) is induced by IFN signaling and edits mammalian dsRNA with an A-to-I nucleotide change, which is read as an A-to-G change in sequencing data. These edited dsRNAs cannot be sensed by dsRNA sensors, and thus ADAR1 inhibits the type I IFN response in a negative feedback loop. We hypothesized that decreasing ADAR1 editing would enhance the DNMTi-induced immune response. METHODS: Human OC cell lines were treated in vitro with DNMTi and then RNA-sequenced to measure RNA editing. Adar1 was stably knocked down in ID8 Trp53-/- mouse OC cells. Control cells (shGFP) or shAdar1 cells were tested with mock or DNMTi treatment. Tumor-infiltrating immune cells were immunophenotyped using flow cytometry and cell culture supernatants were analyzed for secreted chemokines/cytokines. Mice were injected with syngeneic shAdar1 ID8 Trp53-/- cells and treated with tetrahydrouridine/DNMTi while given anti-interferon alpha and beta receptor 1, anti-CD8, or anti-NK1.1 antibodies every 3 days. RESULTS: We show that ADAR1 edits transposable elements in human OC cell lines after DNMTi treatment in vitro. Combining ADAR1 knockdown with DNMTi significantly increases pro-inflammatory cytokine/chemokine production and sensitivity to IFN-ß compared with either perturbation alone. Furthermore, DNMTi treatment and Adar1 loss reduces tumor burden and prolongs survival in an immunocompetent mouse model of OC. Combining Adar1 loss and DNMTi elicited the most robust antitumor response and transformed the immune microenvironment with increased recruitment and activation of CD8+ T cells. CONCLUSION: In summary, we showed that the survival benefit from DNMTi plus ADAR1 inhibition is dependent on type I IFN signaling. Thus, epigenetically inducing transposable element transcription combined with inhibition of RNA editing is a novel therapeutic strategy to reverse immune evasion in OC, a disease that does not respond to current immunotherapies.


Asunto(s)
Neoplasias Ováricas , Edición de ARN , Femenino , Humanos , Animales , Ratones , Microambiente Tumoral , Metilación de ADN , Elementos Transponibles de ADN , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , ARN Bicatenario/uso terapéutico , Carcinoma Epitelial de Ovario/genética , Neoplasias Ováricas/terapia , Neoplasias Ováricas/tratamiento farmacológico , Citocinas/metabolismo , Mamíferos/genética , Mamíferos/metabolismo
6.
Oncologist ; 26(12): e2136-e2142, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34427023

RESUMEN

LESSONS LEARNED: Entinostat at the selected dose levels in combination with a standard dose of enzalutamide showed a promising safety profile in this small phase I study BACKGROUND: Entinostat inhibits prostate cancer (PCa) growth and suppresses Treg cell function in vitro and in vivo. METHODS: This was a phase I study to explore the safety and preliminary efficacy of entinostat (3 and 5 mg orally per week) in combination with enzalutamide in castration resistant PCa (CRPC). The study was carried out in an open-label two-cohort design. Patients who had developed disease progression on or were eligible for enzalutamide were enrolled in the study. The safety profile of the combination therapy, Prostate specific antigen (PSA) levels, the pharmacokinetics of enzalutamide after entinostat administration, peripheral T-cell subtype (including Treg quantitation), and mononuclear cell (PBMC) histone H3 acetylation were analyzed. RESULTS: Six patients with metastatic CRPC were enrolled. There was no noticeable increment of fatigue related to entinostat. Toxicities possibly or probably related to entinostat or the combination therapy included grade 3 anemia 1/6 (17%), grade 2 white blood cell (WBC) decrease 1/6 (17%), and other self-limiting grade 1 adverse events (AEs). Median duration of treatment with entinostat was 18 weeks. Entinostat did not affect the steady plasma concentration of enzalutamide. Increased PBMC histone H3 acetylation was observed in blood samples. No evident T-cell subtype changes were detected, including in Treg quantitation. CONCLUSION: Entinostat 5 mg weekly in combination with enzalutamide showed an acceptable safety profile in this small phase I study. A planned phase II part of the trial was terminated because of sponsor withdrawal.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración , Benzamidas , Humanos , Leucocitos Mononucleares , Masculino , Nitrilos , Feniltiohidantoína , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Piridinas
7.
Int J Mol Sci ; 22(4)2021 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-33669239

RESUMEN

Irregular inflammatory responses are a major contributor to tissue dysfunction and inefficient repair. Skin has proven to be a powerful model to study mechanisms that regulate inflammation. In particular, skin wound healing is dependent on a rapid, robust immune response and subsequent dampening of inflammatory signaling. While injury-induced inflammation has historically been attributed to keratinocytes and immune cells, a vast body of evidence supports the ability of non-immune cells to coordinate inflammation in numerous tissues and diseases. In this review, we concentrate on the active participation of tissue-resident adipocytes and fibroblasts in pro-inflammatory signaling after injury, and how altered cellular communication from these cells can contribute to irregular inflammation associated with aberrant wound healing. Furthering our understanding of how tissue-resident mesenchymal cells contribute to inflammation will likely reveal new targets that can be manipulated to regulate inflammation and repair.


Asunto(s)
Adipocitos Blancos/inmunología , Dermis/citología , Dermis/lesiones , Fibroblastos/inmunología , Cicatrización de Heridas/inmunología , Envejecimiento/inmunología , Envejecimiento/metabolismo , Animales , Comunicación Celular/inmunología , Polaridad Celular/inmunología , Citocinas/metabolismo , Diabetes Mellitus/inmunología , Diabetes Mellitus/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Células Madre Mesenquimatosas/metabolismo , Transducción de Señal/inmunología
8.
J Med Chem ; 63(18): 10246-10262, 2020 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-32815366

RESUMEN

Selective inhibition of histone deacetylase 6 (HDAC6) is being recognized as a therapeutic approach for cancers. In this study, we designed a new HDAC6 inhibitor, named Suprastat, using in silico simulations. X-ray crystallography and molecular dynamics simulations provide strong evidence to support the notion that the aminomethyl and hydroxyl groups in the capping group of Suprastat establish significant hydrogen bond interactions, either direct or water-mediated, with residues D460, N530, and S531, which play a vital role in regulating the deacetylase function of the enzyme and which are absent in other isoforms. In vitro characterization of Suprastat demonstrates subnanomolar HDAC6 inhibitory potency and a hundred- to a thousand-fold HDAC6 selectivity over the other HDAC isoforms. In vivo studies reveal that a combination of Suprastat and anti-PD1 immunotherapy enhances antitumor immune response, mediated by a decrease of protumoral M2 macrophages and increased infiltration of antitumor CD8+ effector and memory T-cells.


Asunto(s)
Histona Desacetilasa 6/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/uso terapéutico , Ácidos Hidroxámicos/uso terapéutico , Factores Inmunológicos/uso terapéutico , Melanoma/tratamiento farmacológico , Compuestos de Fenilurea/uso terapéutico , Animales , Línea Celular Tumoral , Cristalografía por Rayos X , Diseño de Fármacos , Femenino , Histona Desacetilasa 6/metabolismo , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/metabolismo , Humanos , Enlace de Hidrógeno , Ácidos Hidroxámicos/síntesis química , Ácidos Hidroxámicos/metabolismo , Factores Inmunológicos/síntesis química , Factores Inmunológicos/metabolismo , Inmunoterapia , Melanoma/terapia , Ratones Endogámicos C57BL , Microsomas Hepáticos/metabolismo , Simulación de Dinámica Molecular , Compuestos de Fenilurea/síntesis química , Compuestos de Fenilurea/metabolismo , Unión Proteica , Ratas
9.
Cancer Res ; 80(17): 3649-3662, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32605998

RESUMEN

Despite the outstanding clinical results of immune checkpoint blockade (ICB) in melanoma and other cancers, clinical trials in breast cancer have reported low responses to these therapies. Current efforts are now focused on improving the treatment efficacy of ICB in breast cancer using new combination designs such as molecularly targeted agents, including histone deacetylase inhibitors (HDACi). These epigenetic drugs have been widely described as potent cytotoxic agents for cancer cells. In this work, we report new noncanonical regulatory properties of ultra-selective HDAC6i over the expression and function of epithelial-mesenchymal transition pathways and the invasiveness potential of breast cancer. These unexplored roles position HDAC6i as attractive options to potentiate ongoing immunotherapeutic approaches. These new functional activities of HDAC6i involved regulation of the E-cadherin/STAT3 axis. Pretreatment of tumors with HDAC6i induced critical changes in the tumor microenvironment, resulting in improved effectiveness of ICB and preventing dissemination of cancer cells to secondary niches. Our results demonstrate for the first time that HDAC6i can both improve ICB antitumor immune responses and diminish the invasiveness of breast cancer with minimal cytotoxic effects, thus departing from the cytotoxicity-centric paradigm previously assigned to HDACi. SIGNIFICANCE: Ultraselective HDAC6 inhibitors can reduce tumor growth and invasiveness of breast cancer by noncanonical mechanisms unrelated to the previously cytotoxic properties attributed to HDAC inhibitors.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/fisiología , Histona Desacetilasa 6/metabolismo , Inhibidores de Puntos de Control Inmunológico/farmacología , Neoplasias Mamarias Experimentales/patología , Neoplasias de la Mama Triple Negativas/patología , Animales , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/fisiología , Femenino , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Invasividad Neoplásica/patología , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/metabolismo
10.
Am J Cancer Res ; 9(10): 2194-2208, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31720082

RESUMEN

Previous studies showed that intratumoral 27-Hydroxycholesterol (27-HC), a metabolite of cholesterol, promotes growth, invasion and migration of breast cancer cells and that tumor-associated macrophages (TAMs) in breast cancers are closely related to tumor growth and metastatic progression. However, the relationship between 27-HC and TAMs in breast cancer remains unclear. In the present study, we observed that CYP27A1, the 27-HC synthesizing enzyme, was expressed in a much higher level in THP1 monocytes and THP1-derived macrophages than in breast cancer cells, and the promoter of CYP7B1, the degrading enzyme for 27-HC, was highly methylated in breast tumor cells. In addition, THP-1 monocytes and murine bone marrow cells were differentiated toward M2 type macrophages after being co-cultured with breast cancer cells or being exposed to exosomes derived from breast cancer cells. M2 type macrophages produced higher amounts of 27-HC than M0 and M1 type macrophages. 27-HC not only stimulated ER+ cancer cell proliferation as reported, but also promoted the recruitment of CCR2- and CCR5-expressing monocytes by inducing macrophages to express multiple chemokines including CCL2, CCL3 and CCL4. Taken together, our data demonstrate that the hypermethylation of CYP7B1 and recruitment of monocytes likely contribute to the accumulation of 27-Hydroxycholesterol in breast cancer and that the interaction of 27-HC with macrophages further promote the development of breast cancer.

12.
J Med Chem ; 62(18): 8557-8577, 2019 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-31414801

RESUMEN

Isoxazole is a five-membered heterocycle that is widely used in drug discovery endeavors. Here, we report the design, synthesis, and structural and biological characterization of SS-208, a novel HDAC6-selective inhibitor containing the isoxazole-3-hydroxamate moiety as a zinc-binding group as well as a hydrophobic linker. A crystal structure of the Danio rerio HDAC6/SS-208 complex reveals a bidentate coordination of the active-site zinc ion that differs from the preferred monodentate coordination observed for HDAC6 complexes with phenylhydroxamate-based inhibitors. While SS-208 has minimal effects on the viability of murine SM1 melanoma cells in vitro, it significantly reduced in vivo tumor growth in a murine SM1 syngeneic melanoma mouse model. These findings suggest that the antitumor activity of SS-208 is mainly mediated by immune-related antitumor activity as evidenced by the increased infiltration of CD8+ and NK+ T cells and the enhanced ratio of M1 and M2 macrophages in the tumor microenvironment.


Asunto(s)
Histona Desacetilasa 6/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Isoxazoles/farmacología , Melanoma/tratamiento farmacológico , Animales , Linfocitos T CD8-positivos/citología , Dominio Catalítico , Línea Celular Tumoral , Descubrimiento de Drogas , Inhibidores de Histona Desacetilasas/química , Humanos , Ácidos Hidroxámicos/química , Isoxazoles/química , Macrófagos/citología , Ratones , Microsomas/química , Células T Asesinas Naturales/citología , Trasplante Isogénico , Pez Cebra , Zinc/química
13.
Methods Mol Biol ; 1983: 279-307, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31087305

RESUMEN

HDACs, originally described as histone modifiers, have recently been demonstrated to modify a variety of other proteins that are involved in diverse cellular processes unrelated to the chromatin environment. This includes deacetylation of nonhistone targets involved in multiple signaling pathways. In this regard, a considerable number of reports have analyzed the role of nonspecific inhibition of HDACs through pan-HDACi in cancer as well as processes of immune regulation. However, with pan-HDACi there is a lack of understanding about the exact contribution of inhibition of each individual HDAC, which makes the rational design of improved drug candidates extremely difficult. Additionally, current approaches using nonselective HDACi in the clinic have critical limitations, including pan-HDACi which elicit poor activity in solid tumors and cardiac toxicity, class I HDACi which activate multiple apoptotic pathways, limiting its use for longer periods of time, and class I-HDAC6i that evidenced a number of adverse effects in initial clinical trials. Therefore, there is a growing interest in the identification of more selective HDACi, and the subsequent development of accurate functional tests to identify the effectiveness and selectivity of these inhibitors. In this chapter, we are describing some selected methodologies to identify the individual activities of HDACs. In addition, we present specific methods to identify enzymatic and nonenzymatic molecular targets of HDACs.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Histona Desacetilasas/metabolismo , Neoplasias/etiología , Neoplasias/metabolismo , Acetilación , Animales , Apoptosis/genética , Biomarcadores , Supervivencia Celular/genética , Transformación Celular Neoplásica/genética , Inmunoprecipitación de Cromatina , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Histonas/metabolismo , Humanos , Inmunohistoquímica , Ratones , Neoplasias/patología
14.
Sci Rep ; 9(1): 6136, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30992475

RESUMEN

Histone deacetylases (HDACs) are involved in diverse cellular regulatory mechanisms including non-canonical functions outside the chromatin environment. Several publications have demonstrated that selective HDAC inhibitors (HDACi) can influence tumor immunogenicity and the functional activity of specific immune cells. In particular, the selective inhibition of HDAC6 has been reported to decrease tumor growth in several malignancies. However, there is still no clarity about the cellular components mediating this effect. In this study, we evaluated the HDAC6i Nexturastat A as a priming agent to facilitate the transition of the tumor microenvironment from "cold" to "hot", and potentially augment immune check-point blockade therapies. This combination modality demonstrated to significantly reduce tumor growth in syngeneic melanoma tumor models. Additionally, we observed a complete neutralization of the up-regulation of PD-L1 and other immunosuppressive pathways induced by the treatment with anti-PD-1 blockade. This combination also showed profound changes in the tumor microenvironment such as enhanced infiltration of immune cells, increased central and effector T cell memory, and a significant reduction of pro-tumorigenic M2 macrophages. The evaluation of individual components of the tumor microenvironment suggested that the in vivo anti-tumor activity of HDAC6i is mediated by its effect on tumor cells and tumor-associated macrophages, and not directly over T cells. Overall, our results indicate that selective HDAC6i could be used as immunological priming agents to sensitize immunologically "cold" tumors and subsequently improve ongoing immune check-point blockade therapies.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Histona Desacetilasa 6/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/uso terapéutico , Macrófagos/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Antiinflamatorios/uso terapéutico , Antígeno B7-H1/inmunología , Histona Desacetilasa 6/inmunología , Tolerancia Inmunológica/efectos de los fármacos , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Microambiente Tumoral/efectos de los fármacos
15.
Am J Respir Cell Mol Biol ; 58(5): 614-624, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29115856

RESUMEN

One of the early events in the progression of LPS-mediated acute lung injury in mice is the disruption of the pulmonary endothelial barrier resulting in lung edema. However, the molecular mechanisms by which the endothelial barrier becomes compromised remain unresolved. The SRY (sex-determining region on the Y chromosome)-related high-mobility group box (Sox) group F family member, SOX18, is a barrier-protective protein through its ability to increase the expression of the tight junction protein CLDN5. Thus, the purpose of this study was to determine if downregulation of the SOX18-CLDN5 axis plays a role in the pulmonary endothelial barrier disruption associated with LPS exposure. Our data indicate that both SOX18 and CLDN5 expression is decreased in two models of in vivo LPS exposure (intraperitoneal, intratracheal). A similar downregulation was observed in cultured human lung microvascular endothelial cells (HLMVECs) exposed to LPS. SOX18 overexpression in HLMVECs or in the mouse lung attenuated the LPS-mediated vascular barrier disruption. Conversely, reduced CLDN5 expression (siRNA) reduced the HLMVEC barrier-protective effects of SOX18 overexpression. The mechanism by which LPS decreases SOX18 expression was identified as transcriptional repression through binding of NF-κB (p65) to a SOX18 promoter sequence located between -1,082 and -1,073 bp with peroxynitrite contributing to LPS-mediated NF-κB activation. We conclude that NF-κB-dependent decreases in the SOX18-CLDN5 axis are essentially involved in the disruption of human endothelial cell barrier integrity associated with LPS-mediated acute lung injury.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Permeabilidad Capilar , Células Endoteliales/metabolismo , Lipopolisacáridos , Pulmón/irrigación sanguínea , FN-kappa B/metabolismo , Edema Pulmonar/metabolismo , Factores de Transcripción SOXF/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Animales , Sitios de Unión , Células Cultivadas , Claudina-5/genética , Claudina-5/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Células Endoteliales/patología , Humanos , Masculino , Ratones Endogámicos C57BL , FN-kappa B/genética , Ácido Peroxinitroso/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Edema Pulmonar/inducido químicamente , Edema Pulmonar/genética , Edema Pulmonar/patología , Factores de Transcripción SOXF/genética , Transducción de Señal , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo
16.
Adv Exp Med Biol ; 967: 105-137, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29047084

RESUMEN

The generation of reactive oxygen species (ROS) plays an important role for the maintenance of cellular processes and functions in the body. However, the excessive generation of oxygen radicals under pathological conditions such as acute lung injury (ALI) and its most severe form acute respiratory distress syndrome (ARDS) leads to increased endothelial permeability. Within this hallmark of ALI and ARDS, vascular microvessels lose their junctional integrity and show increased myosin contractions that promote the migration of polymorphonuclear leukocytes (PMNs) and the transition of solutes and fluids in the alveolar lumen. These processes all have a redox component, and this chapter focuses on the role played by ROS during the development of ALI/ARDS. We discuss the origins of ROS within the cell, cellular defense mechanisms against oxidative damage, the role of ROS in the development of endothelial permeability, and potential therapies targeted at oxidative stress.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Síndrome de Dificultad Respiratoria/metabolismo , Lesión Pulmonar Aguda/fisiopatología , Animales , Antioxidantes/metabolismo , Permeabilidad Capilar , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Humanos , Modelos Biológicos , Oxidación-Reducción , Estrés Oxidativo , Síndrome de Dificultad Respiratoria/fisiopatología , Transducción de Señal
17.
Cancer Immunol Res ; 5(4): 330-344, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28264810

RESUMEN

Triple-negative breast cancer (TNBC) cells are modulated in reaction to tumor-infiltrating lymphocytes. However, their specific responses to this immune pressure are unknown. In order to address this question, we first used mRNA sequencing to compare the immunophenotype of the TNBC cell line MDA-MB-231 and the luminal breast cancer cell line MCF7 after both were cocultured with activated human T cells. Despite similarities in the cytokine-induced immune signatures of the two cell lines, MDA-MD-231 cells were able to transcribe more IDO1 than MCF7 cells. The two cell lines had similar upstream JAK/STAT1 signaling and IDO1 mRNA stability. However, using a series of breast cancer cell lines, IFNγ stimulated IDO1 protein expression and enzymatic activity only in ER-, not ER+, cell lines. Treatment with 5-aza-deoxycytidine reversed the suppression of IDO1 expression in MCF7 cells, suggesting that DNA methylation was potentially involved in IDO1 induction. By analyzing several breast cancer datasets, we discovered subtype-specific mRNA and promoter methylation differences in IDO1, with TNBC/basal subtypes exhibiting lower methylation/higher expression and ER+/luminal subtypes exhibiting higher methylation/lower expression. We confirmed this trend of IDO1 methylation by bisulfite pyrosequencing breast cancer cell lines and an independent cohort of primary breast tumors. Taken together, these findings suggest that IDO1 promoter methylation regulates anti-immune responses in breast cancer subtypes and could be used as a predictive biomarker for IDO1 inhibitor-based immunotherapy. Cancer Immunol Res; 5(4); 330-44. ©2017 AACR.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Metilación de ADN , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Regiones Promotoras Genéticas , Linfocitos T/inmunología , Linfocitos T/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Línea Celular Tumoral , Citocinas/metabolismo , Activación Enzimática , Femenino , Regulación Neoplásica de la Expresión Génica , Genes Reporteros , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Factor 1 Regulador del Interferón/metabolismo , Quinasas Janus/metabolismo , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Estabilidad Proteica , Estabilidad del ARN , ARN Mensajero/genética , Factor de Transcripción STAT1/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/mortalidad
18.
Free Radic Biol Med ; 102: 217-228, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27838434

RESUMEN

The molecular mechanisms by which the endothelial barrier becomes compromised during lipopolysaccharide (LPS) mediated acute lung injury (ALI) are still unresolved. We have previously reported that the disruption of the endothelial barrier is due, at least in part, to the uncoupling of endothelial nitric oxide synthase (eNOS) and increased peroxynitrite-mediated nitration of RhoA. The purpose of this study was to elucidate the molecular mechanisms by which LPS induces eNOS uncoupling during ALI. Exposure of pulmonary endothelial cells (PAEC) to LPS increased pp60Src activity and this correlated with an increase in nitric oxide (NO) production, but also an increase in NOS derived superoxide, peroxynitrite formation and 3-nitrotyrosine (3-NT) levels. These effects could be simulated by the over-expression of a constitutively active pp60Src (Y527FSrc) mutant and attenuated by over-expression of dominant negative pp60Src mutant or reducing pp60Src expression. LPS induces both RhoA nitration and endothelial barrier disruption and these events were attenuated when pp60Src expression was reduced. Endothelial NOS uncoupling correlated with an increase in the levels of asymmetric dimethylarginine (ADMA) in both LPS exposed and Y527FSrc over-expressing PAEC. The effects in PAEC were also recapitulated when we transiently over-expressed Y527FSrc in the mouse lung. Finally, we found that the pp60-Src-mediated decrease in DDAH activity was mediated by the phosphorylation of DDAH II at Y207 and that a Y207F mutant DDAH II was resistant to pp60Src-mediated inhibition. We conclude that pp60Src can directly inhibit DDAH II and this is involved in the increased ADMA levels that enhance eNOS uncoupling during the development of ALI.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Amidohidrolasas/genética , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/genética , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/patología , Amidohidrolasas/metabolismo , Animales , Arginina/análogos & derivados , Arginina/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/patología , Regulación de la Expresión Génica , Lipopolisacáridos/toxicidad , Ratones , Mitocondrias/metabolismo , Mitocondrias/patología , Mutación , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ácido Peroxinitroso/biosíntesis , Fosforilación , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Superóxidos/metabolismo
19.
J Genet Genomics ; 42(7): 355-71, 2015 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-26233891

RESUMEN

Glioblastoma (GBM) is the most common and most aggressive primary brain tumor in adults. The existence of a small population of stem-like tumor cells that efficiently propagate tumors and resist cytotoxic therapy is one proposed mechanism leading to the resilient behavior of tumor cells and poor prognosis. In this study, we performed an in-depth analysis of the DNA methylation landscape in GBM-derived cancer stem cells (GSCs). Parallel comparisons of primary tumors and GSC lines derived from these tumors with normal controls (a neural stem cell (NSC) line and normal brain tissue) identified groups of hyper- and hypomethylated genes that display a trend of either increasing or decreasing methylation levels in the order of controls, primary GBMs, and their counterpart GSC lines, respectively. Interestingly, concurrent promoter hypermethylation and gene body hypomethylation were observed in a subset of genes including MGMT, AJAP1 and PTPRN2. These unique DNA methylation signatures were also found in primary GBM-derived xenograft tumors indicating that they are not tissue culture-related epigenetic changes. Integration of GSC-specific epigenetic signatures with gene expression analysis further identified candidate tumor suppressor genes that are frequently down-regulated in GBMs such as SPINT2, NEFM and PENK. Forced re-expression of SPINT2 reduced glioma cell proliferative capacity, anchorage independent growth, cell motility, and tumor sphere formation in vitro. The results from this study demonstrate that GSCs possess unique epigenetic signatures that may play important roles in the pathogenesis of GBM.


Asunto(s)
Glioblastoma/genética , Células Madre Neoplásicas/metabolismo , Moléculas de Adhesión Celular/genética , Línea Celular Tumoral , Metilación de ADN/genética , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Epigénesis Genética/genética , Humanos , Glicoproteínas de Membrana/genética , Proteínas de Neurofilamentos/genética , Regiones Promotoras Genéticas/genética , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores/genética , Proteínas Supresoras de Tumor/genética
20.
Oncotarget ; 6(16): 14632-45, 2015 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-25999352

RESUMEN

Chronic lymphocytic leukemia (CLL), the most common adult leukemia, remains incurable despite advancements in treatment regimens over the past decade. Several expression profile studies have been pursued to better understand CLL pathogenesis. However, these large-scale studies only provide information at the transcriptional level. To better comprehend the differential protein changes that take place in CLL, we performed a reverse-phase protein array (RPPA) analysis using 167 different antibodies on B-cell lysates from 18 CLL patients and 6 normal donors. From our analysis, we discovered an enrichment of protein alterations involved with mRNA translation, specifically upregulation of the translation initiator eIF4G and phosphorylation of the cap-dependent translation inhibitor 4E-BP1 at serine 65. Interestingly, 4E-BP1 phosphorylation occurred independently of AKT phosphorylation, suggesting a disconnect between PI3K/AKT pathway activation and 4E-BP1 phosphorylation. Based on these results, we treated primary CLL samples with NVP-BEZ235, a PI3K/mTOR dual inhibitor, and compared its apoptotic-inducing potential against the BTK inhibitor Ibrutinib and the PI3Kδ inhibitor Idelalisib. We demonstrated that treatment with NVP-BEZ235 caused greater apoptosis, greater apoptotic cleavage of eIF4G, and greater dephosphorylation of 4E-BP1 in primary CLL cells. Taken together, these results highlight the potential dependence of eIF4G overexpression and 4E-BP1 phosphorylation in CLL survival.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Factor 4G Eucariótico de Iniciación/metabolismo , Leucemia Linfocítica Crónica de Células B/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Análisis por Matrices de Proteínas/métodos , Serina/metabolismo , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Humanos , Leucemia Linfocítica Crónica de Células B/mortalidad , Proteínas Nucleares/genética , Fenotipo , Fosforilación , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...