Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Hepatol Commun ; 4(11): 1591-1609, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33163831

RESUMO

Patients with nonalcoholic fatty liver disease/steatohepatitis (NAFLD/NASH) commonly develop atherosclerosis through a mechanism that is not well delineated. These diseases are associated with steatosis, inflammation, oxidative stress, and fibrosis. The role of insulin resistance in their pathogenesis remains controversial. Albumin (Alb)Cre+ Cc1flox ( fl ) /fl mice with the liver-specific null deletion of the carcinoembryonic antigen-related cell adhesion molecule 1 (Ceacam1; alias Cc1) gene display hyperinsulinemia resulting from impaired insulin clearance followed by hepatic insulin resistance, elevated de novo lipogenesis, and ultimately visceral obesity and systemic insulin resistance. We therefore tested whether this mutation causes NAFLD/NASH and atherosclerosis. To this end, mice were propagated on a low-density lipoprotein receptor (Ldlr) -/- background and at 4 months of age were fed a high-cholesterol diet for 2 months. We then assessed the biochemical and histopathologic changes in liver and aortae. Ldlr-/-AlbCre+Cc1fl/fl mice developed chronic hyperinsulinemia with proatherogenic hypercholesterolemia, a robust proinflammatory state associated with visceral obesity, elevated oxidative stress (reduced NO production), and an increase in plasma and tissue endothelin-1 levels. In parallel, they developed NASH (steatohepatitis, apoptosis, and fibrosis) and atherosclerotic plaque lesions. Mechanistically, hyperinsulinemia caused down-regulation of the insulin receptor followed by inactivation of the insulin receptor substrate 1-protein kinase B-endothelial NO synthase pathway in aortae, lowering the NO level. This also limited CEACAM1 phosphorylation and its sequestration of Shc-transforming protein (Shc), activating the Shc-mitogen-activated protein kinase-nuclear factor kappa B pathway and stimulating endothelin-1 production. Thus, in the presence of proatherogenic dyslipidemia, hyperinsulinemia and hepatic insulin resistance driven by liver-specific deletion of Ceacam1 caused metabolic and vascular alterations reminiscent of NASH and atherosclerosis. Conclusion: Altered CEACAM1-dependent hepatic insulin clearance pathways constitute a molecular link between NASH and atherosclerosis.

2.
J Neuroendocrinol ; 32(11): e12854, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32350959

RESUMO

Growth hormone (GH) excess in bovine (b)GH transgenic mice has been shown to alter white adipose tissue (WAT) immune cell populations. The present study aimed to evaluate the effects of GH resistance on WAT immune cell populations using GH receptor knockout (GHR-/- ) mice. Eight- and 24-month-old, male GHR-/- and wild-type mice were used. Body composition and tissue weights were determined, and systemic inflammation was assessed by measuring serum cytokine levels. The stromal vascular fraction (SVF) was isolated from three distinct WAT depots, and immune cell populations were quantified using flow cytometry. GHR-/- mice at both ages had decreased body weight but were obese. Although no significant changes were observed in serum levels of the measured cytokines, SVF cell alterations were seen and differed from depot to depot. Total SVF cells were decreased in epidydimal (Epi) depots, whereas SVF cells per gram adipose tissue weight were increased in mesenteric (Mes) depots of GHR-/- mice relative to controls. T cells and T helper cells were increased in Mes at 8 months old, whereas cytotoxic T cells were decreased in subcutaneous (SubQ) at 24 months old. Other cells were unchanged at both ages measured. The present study demonstrates that removal of GH action results in modest and depot-specific changes to several immune cell populations in WAT of intra-abdominal depots (Epi and Mes), which are somewhat surprising results because the SubQ has the largest change in size, whereas the Mes has no size change. Taken together with previous results from bovine GH transgenic mice, these data suggest that GH induces changes in the immune cell population of WAT in a depot-specific manner. Notably, GHR-/- mice appear to be protected from age-related WAT inflammation and immune cell infiltration despite obesity.


Assuntos
Tecido Adiposo Branco/patologia , Proteínas de Transporte/genética , Inflamação/genética , Inflamação/patologia , Obesidade/genética , Obesidade/patologia , Gordura Abdominal/imunologia , Gordura Abdominal/patologia , Tecido Adiposo Branco/imunologia , Envelhecimento , Animais , Composição Corporal , Citocinas/sangue , Epididimo/patologia , Hormônio do Crescimento/genética , Hormônio do Crescimento/metabolismo , Imunidade Celular/genética , Imunidade Celular/imunologia , Inflamação/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Obesidade/imunologia , Tamanho do Órgão , Linfócitos T/imunologia , Linfócitos T Auxiliares-Indutores/imunologia
3.
Adv Exp Med Biol ; 1223: 81-97, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32030686

RESUMO

The involvement of inflammation in cancer progression is well-established. The immune system can play both tumor-promoting and -suppressive roles, and efforts to harness the immune system to help fight tumor growth are at the forefront of research. Of particular importance is the inflammatory profile at the site of the tumor, with respect to both the leukocyte population numbers, the phenotype of these cells, as well as the contribution of the tumor cells themselves. In this regard, the pro-inflammatory effects of pattern recognition receptor expression and activation in the tumor microenvironment have emerged as a relevant issue both for therapy and to understand tumor development.Pattern recognition receptors (PRRs) were originally recognized as components of immune cells, particularly innate immune cells, as detectors of pathogens. PRR signaling in immune cells activates them, inducing robust antimicrobial responses. In particular, toll-like receptors (TLRs) constitute a family of membrane-bound PRRs which can recognize pathogen-associated molecular patterns (PAMPs) carried by bacteria, virus, and fungi. In addition, PRRs can recognize products generated by stressed cells or damaged tissues, namely damage-associated molecular patterns or DAMPS. Taking into account the role of the immune system in fighting tumors together with the presence of immune cells in the microenvironment of different types of tumors, strategies to activate immune cells via PRR ligands have been envisioned as an anticancer therapeutic approach.In the last decades, it has been determined that PRRs are present and functional on nonimmune cells and that their activation in these cells contributes to the inflammation in the tumor microenvironment. Both tumor-promoting and antitumor effects have been observed when tumor cell PRRs are activated. This argues against nonspecific activation of PRR ligands in the tumor microenvironment as a therapeutic approach. Therefore, the use of PRR ligands for anticancer therapy might benefit from strategies that specifically deliver these ligands to immune cells, thus avoiding tumor cells in some settings. This review focuses on these aspects of TLR signaling in the tumor microenvironment.


Assuntos
Neoplasias/imunologia , Neoplasias/metabolismo , Transdução de Sinais , Receptores Toll-Like/metabolismo , Microambiente Tumoral , Humanos , Receptores de Reconhecimento de Padrão/imunologia , Receptores de Reconhecimento de Padrão/metabolismo , Transdução de Sinais/imunologia , Receptores Toll-Like/imunologia , Microambiente Tumoral/imunologia
4.
Immunobiology ; 225(1): 151854, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31753553

RESUMO

Dendritic cells (DCs) are immune cells that surveil the organism for infections or malignancies and activate specific T lymphocytes initiating specific immune responses. Contrariwise, DCs have been show to participate in the development of diseases, among them some types of cancer by inducing angiogenesis or immunosuppression. The ultimate fate of DC functions regarding their role in disease or health is prompted by signals from the microenvironment. We have previously shown that the interaction of DCs with various extracellular matrix components modifies the immune properties and angiogenic potential of these cells. The objective of the current studies was to investigate the angiogenic and immune profile of murine myeloid DCs upon interaction with laminin environments, with a particular emphasis on ovarian cancer. Our results show that murine ovarian tumors produce several types of laminins, as determined by PCR analysis, and also that tumor-associated DCs, both from ascites or solid tumors express adhesion molecules capable of interacting with these molecules as determined by flow cytometry and PCR analysis. Further, we established that DCs cultured on laminin upregulate both AKT and MEK signaling pathways, and that long-term culture on laminin surfaces decreases the immunological capacities of these cells when compared to the same cells cultured on synthetic substrates. In addition, we observed that tumor conditioned media was able to modify the metabolic status of these cells, and also reprogram the development of DCs from bone marrow precursors towards the generation of myeloid-derived suppressor cells. Overall, these studies demonstrate that the interaction between soluble factors and extracellular matrix components of the ovarian cancer microenvironment shape the biology of DCs and thus help them become co-conspirators of tumor growth.


Assuntos
Células Dendríticas/fisiologia , Matriz Extracelular/metabolismo , Laminina/metabolismo , Células Mieloides/fisiologia , Neoplasias Ovarianas/imunologia , Animais , Antígenos de Neoplasias/imunologia , Carcinogênese , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Laminina/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neovascularização Patológica , Microambiente Tumoral
5.
Int Rev Immunol ; 38(4): 157-171, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31286783

RESUMO

Obesity predisposes the affected individuals to several metabolic, inflammatory, cardiovascular and malignant pathologies and is a top risk factor for premature mortality. It is now well known that inflammation has a major causative role in obesity-associated disease development and that obesity favors the establishment of a pro-inflammatory milieu at the level of adipose microenvironment. These inflammatory signals result in a disruption of normal cellular-crosstalk between adipose and non-adipose components leading to an altered metabolic and immunological status and a dysfunctional phenotype. Abnormal secretion of adipokines - small adipose-derived signaling molecules - can further assist in the inflammatory processes to offset the adipose tissue towards a dysfunctional state. Although adipokines have been recognized as the link between obesity and pathogenesis, studies are needed to fully understand their mechanism of action and underscore their therapeutic value. Here, we have reviewed obesity-induced metabolic and immunological changes at the level of vasculature and emphasize on the importance of adipokines, particularly leptin, vaspin and visfatin, for their therapeutic relevance.


Assuntos
Adipocinas/metabolismo , Células Endoteliais/imunologia , Mediadores da Inflamação/metabolismo , Obesidade/imunologia , Tecido Adiposo/imunologia , Tecido Adiposo/patologia , Células Endoteliais/patologia , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Obesidade/metabolismo , Transdução de Sinais
6.
Cancer Cell ; 35(6): 885-900.e10, 2019 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-31185212

RESUMO

We investigated the role of chemokines in regulating T cell accumulation in solid tumors. CCL5 and CXCL9 overexpression was associated with CD8+ T cell infiltration in solid tumors. T cell infiltration required tumor cell-derived CCL5 and was amplified by IFN-γ-inducible, myeloid cell-secreted CXCL9. CCL5 and CXCL9 coexpression revealed immunoreactive tumors with prolonged survival and response to checkpoint blockade. Loss of CCL5 expression in human tumors was associated with epigenetic silencing through DNA methylation. Reduction of CCL5 expression caused tumor-infiltrating lymphocyte (TIL) desertification, whereas forced CCL5 expression prevented Cxcl9 expression and TILs loss, and attenuated tumor growth in mice through IFN-γ. The cooperation between tumor-derived CCL5 and IFN-γ-inducible CXCR3 ligands secreted by myeloid cells is key for orchestrating T cell infiltration in immunoreactive and immunoresponsive tumors.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Quimiotaxia de Leucócito , Citocinas/metabolismo , Células Dendríticas/metabolismo , Ativação Linfocitária , Linfócitos do Interstício Tumoral/metabolismo , Macrófagos/metabolismo , Neoplasias Ovarianas/metabolismo , Animais , Antineoplásicos Imunológicos/farmacologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Quimiocina CCL5/genética , Quimiocina CCL5/imunologia , Quimiocina CCL5/metabolismo , Quimiocina CXCL9/genética , Quimiocina CXCL9/imunologia , Quimiocina CXCL9/metabolismo , Quimiotaxia de Leucócito/efeitos dos fármacos , Técnicas de Cocultura , Citocinas/genética , Citocinas/imunologia , Metilação de DNA , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Epigênese Genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Imunoterapia/métodos , Interferon gama/genética , Interferon gama/imunologia , Interferon gama/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos Endogâmicos C57BL , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/terapia , Comunicação Parácrina , Receptores CXCR3/genética , Receptores CXCR3/imunologia , Receptores CXCR3/metabolismo , Transdução de Sinais
7.
Future Virol ; 13(9): 671-680, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30416535

RESUMO

Oncolytic viruses (OVs) do more than simply infect and kill host cells. The accepted mechanism of action for OVs consists of a primary lytic phase and a subsequent antitumor and antiviral immune response. However, not all cells are subject to the direct effects of OV therapy, and it is becoming clear that OVs can also impact uninfected cells in the periphery. This review discusses the effects of OVs on uninfected neighboring cells, so-called bystander effects, and implications for OV therapies alone or in combination with other standard of care chemotherapy.

8.
Immunobiology ; 223(6-7): 466-476, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29331323

RESUMO

Malignant cells are not the only components of a tumor mass since other cells (e.g., fibroblasts, infiltrating leukocytes and endothelial cells) are also part of it. In combination with the extracellular matrix, all these cells constitute the tumor microenvironment. In the last decade the role of the tumor microenvironment in cancer progression has gained increased attention and prompted efforts directed to abrogate its deleterious effects on anti-cancer therapies. The immune system can detect and attack tumor cells, and tumor-infiltrating lymphocytes (particularly CD8 T cells) have been associated with improved survival or better response to therapies in colorectal, melanoma, breast, prostate and ovarian cancer patients among others. Contrariwise, tumor-associated myeloid cells (myeloid-derived suppressor cells [MDSCs], dendritic cells [DCs], macrophages) or lymphoid cells such as regulatory T cells can stimulate tumor growth via inhibition of immune responses against the tumor or by participating in tumor neoangiogenesis. Herewith we analyzed the chemokine profile of mouse breast tumors regarding their capacity to generate factors capable of attracting and sequestering DCs to their midst. Chemoattractants from tumors were investigated by molecular biology and immunological techniques and tumor infiltrating DCs were investigated for matched chemokine receptors. In addition, we investigated the inflammatory response of breast cancer cells, a major component of the tumor microenvironment, to double-stranded RNA stimulation. By using molecular biology techniques such as qualitative and quantitative PCR, PCR arrays, and immunological techniques (ELISA, cytokine immunoarrays) we examined the effects of dsRNA treatment on the cytokine secretion profiles of mouse and human breast cancer cells and non-transformed cells. We were able to determine that tumors generate chemokines that are able to interact with receptors present on the surface of tumor infiltrating DCs. We observed that PRR signaling is able to modify the production of chemokines by breast tumor cells and normal breast cells, thereby constituting a possible player in shaping the profile of the leukocyte population in the TME.


Assuntos
Neoplasias da Mama/imunologia , Quimiocinas/metabolismo , Inflamação/imunologia , Animais , Movimento Celular , Quimiocinas/genética , DNA/imunologia , Feminino , Humanos , Mediadores da Inflamação/imunologia , Células MCF-7 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Receptores de Reconhecimento de Padrão/metabolismo
9.
Oncotarget ; 9(94): 36666-36683, 2018 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-30613350

RESUMO

Inflammation and cancer are inter-related, and both pro- and anti-tumorigenic effects are possible in different contexts, highlighting the importance of characterizing specific inflammatory pathways in distinct tumor types. Malignant cells and non-cancerous cells such as fibroblasts, infiltrating leukocytes (i.e., dendritic cells [DC], macrophages, or lymphocytes) and endothelial cells, in combination with the extracellular matrix, constitute the tumor microenvironment (TME). In the last decades, the role of the TME in cancer progression has gained increased attention and efforts directed at abrogating its deleterious effects on anti-cancer therapies have been ongoing. In this context, we investigated the potential of mouse and human ovarian cancer cells to produce inflammatory factors in response to pathogen recognition receptor (PRR) signaling, which might help to shape the biology of the TME. We determined that mouse ovarian tumors generate chemokines that are able to interact with receptors harbored by tumor-associated DCs. We also found that dsRNA triggers significant pro-inflammatory cytokine up-regulation in both human and mouse ovarian tumor cell lines, and that several PRR can simultaneously contribute to the stimulated inflammatory response displayed by these cells. Thus, dsRNA-activated PRRs may not only constitute potentially relevant drug targets for therapies aiming to prevent inflammation associated with leukocyte recruitment, or as co-adjuvants of therapeutic treatments, but also might have a role in development of nascent tumors, for example via activation of cancer cells by microbial molecules associated to pathogens, or with those appearing in circulation due to dysbiosis.

10.
Psychosom Med ; 79(3): 306-310, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28002383

RESUMO

OBJECTIVE: It has been proposed that the inflammatory cytokine system is regulated through the vagus nerve, where vagal activation inhibits release of inflammatory cytokines and, therefore, inflammation. Thus, loss of vagal activation (i.e., reduced high-frequency heart rate variability [HF-HRV]) should result in greater inflammation. Evidence to date for this relationship has relied on animal models and resting states in humans. The present study used a psychosocial stressor to test whether stress-induced decreases in HF-HRV predict increases in circulating inflammatory markers. METHODS: Thirty healthy young women completed a speech stressor. HF-HRV was assessed before and during the stressor while circulating plasma interleukin 6, tumor necrosis factor α, and C-reactive protein were assessed before and 1 hour after the stressor. RESULTS: Consistent with the neural reflex for immunity, greater reductions in HF-HRV during the stressor were associated with greater increases in tumor necrosis factor α (ß = -0.29 to -0.47) and interleukin 6 (ß = -0.40 to -0.68) but not C-reactive protein (ß = 0.10 to 0.29) 1 hour after the stressor. CONCLUSIONS: These findings expand on the current literature by showing that changes in HF-HRV predict and precede changes in circulating inflammatory cytokines in humans and may have implications for treatment of inflammatory diseases.


Assuntos
Frequência Cardíaca/fisiologia , Inflamação/sangue , Sistema Nervoso Parassimpático/fisiopatologia , Estresse Psicológico/sangue , Estresse Psicológico/fisiopatologia , Adulto , Proteína C-Reativa/metabolismo , Feminino , Humanos , Interleucina-6/sangue , Fator de Necrose Tumoral alfa/sangue , Adulto Jovem
11.
Immunol Invest ; 45(8): 712-720, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27420920

RESUMO

In the last 15 years, it has become apparent that ovarian cancer is recognized by the immune system, taking into account that T cell infiltration can be associated with increased overall survival. Several studies indicate that a correct combination of cluster of differentiation 8 and cluster of differentiation 4 T cells is key to fight tumor progression and that the presence of regulatory T cells (Tregs) infiltrating ovarian solid tumors (or present in ascites) is deleterious. Several markers that characterize Tregs include glucocorticoid-induced tumor necrosis factor receptor, cytotoxic T lymphocyte antigen-4, and forkhead box protein 3 (Foxp3). Research has shown that Tregs can infiltrate cancerous tissue and contribute to tumor growth by secreting immunosuppressive cytokines such as transforming growth factor beta and interleukin (IL)-10. Importantly, these cells might hamper the efficacy of immunotherapeutic approaches, thus strategies involving depletion or regulation of this population have been proposed and tested in experimental models. In this Minireview, we will discuss the relevance of Tregs in ovarian cancer and the experimental approaches destined to impair their immunosuppressive effects.


Assuntos
Regulação Neoplásica da Expressão Gênica , Imunoterapia/métodos , Neoplasias Ovarianas/terapia , Linfócitos T Citotóxicos/patologia , Linfócitos T Reguladores/patologia , Antígeno CTLA-4/genética , Antígeno CTLA-4/imunologia , Movimento Celular , Feminino , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Humanos , Tolerância Imunológica , Interleucina-10/genética , Interleucina-10/imunologia , Ativação Linfocitária , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/mortalidade , Neoplasias Ovarianas/patologia , Transdução de Sinais , Análise de Sobrevida , Linfócitos T Citotóxicos/imunologia , Linfócitos T Reguladores/imunologia , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/imunologia
12.
Brain Behav Immun ; 52: 27-31, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26526647

RESUMO

Past work has linked negative repetitive thought (worry, rumination) about stressors to sustained stress responses. Less is known about the effects of neutral types of repetitive thought (e.g., reflection) on physiological stress responses. The present study examined whether greater trait reflection was associated with a lower inflammatory response to an acute psychosocial stressor. Thirty-four healthy undergraduate women completed a speech stressor, and plasma interleukin-6 (IL-6), C-reactive protein, and tumor necrosis factor-α levels were assessed before and after the stressor. Higher levels of reflection predicted lower IL-6 responses 1h after the stressor. Stressor appraisal was not a significant mediator. These preliminary findings stand in contrast to existing evidence that other forms of repetitive thought like worry and rumination may exacerbate or prolong the inflammatory stress response and indicate that reflection is a notable factor worth considering when examining the relationship between stress, inflammation, and health.


Assuntos
Interleucina-6/sangue , Estresse Psicológico/sangue , Estresse Psicológico/psicologia , Adolescente , Adulto , Ansiedade/sangue , Ansiedade/psicologia , Proteína C-Reativa/metabolismo , Cognição/fisiologia , Citocinas/sangue , Feminino , Humanos , Hidrocortisona/sangue , Inflamação/sangue , Fator de Necrose Tumoral alfa/sangue , Adulto Jovem
13.
Semin Cancer Biol ; 35 Suppl: S276-S304, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26590477

RESUMO

Targeted therapies and the consequent adoption of "personalized" oncology have achieved notable successes in some cancers; however, significant problems remain with this approach. Many targeted therapies are highly toxic, costs are extremely high, and most patients experience relapse after a few disease-free months. Relapses arise from genetic heterogeneity in tumors, which harbor therapy-resistant immortalized cells that have adopted alternate and compensatory pathways (i.e., pathways that are not reliant upon the same mechanisms as those which have been targeted). To address these limitations, an international task force of 180 scientists was assembled to explore the concept of a low-toxicity "broad-spectrum" therapeutic approach that could simultaneously target many key pathways and mechanisms. Using cancer hallmark phenotypes and the tumor microenvironment to account for the various aspects of relevant cancer biology, interdisciplinary teams reviewed each hallmark area and nominated a wide range of high-priority targets (74 in total) that could be modified to improve patient outcomes. For these targets, corresponding low-toxicity therapeutic approaches were then suggested, many of which were phytochemicals. Proposed actions on each target and all of the approaches were further reviewed for known effects on other hallmark areas and the tumor microenvironment. Potential contrary or procarcinogenic effects were found for 3.9% of the relationships between targets and hallmarks, and mixed evidence of complementary and contrary relationships was found for 7.1%. Approximately 67% of the relationships revealed potentially complementary effects, and the remainder had no known relationship. Among the approaches, 1.1% had contrary, 2.8% had mixed and 62.1% had complementary relationships. These results suggest that a broad-spectrum approach should be feasible from a safety standpoint. This novel approach has potential to be relatively inexpensive, it should help us address stages and types of cancer that lack conventional treatment, and it may reduce relapse risks. A proposed agenda for future research is offered.


Assuntos
Heterogeneidade Genética , Terapia de Alvo Molecular , Neoplasias/terapia , Medicina de Precisão , Antineoplásicos Fitogênicos/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Neoplasias/genética , Neoplasias/patologia , Neoplasias/prevenção & controle , Transdução de Sinais , Microambiente Tumoral/genética
14.
J Endocrinol ; 226(1): 13-23, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25943560

RESUMO

GH influences adipocyte differentiation, but both stimulatory and inhibitory effects have been described. Adipose tissue-derived mesenchymal stem cells (AT-MSCs) are multipotent and are able to differentiate into adipocytes, among other cells. Canonical Wnt/ß-catenin signaling activation impairs adipogenesis. The aim of the present study was to elucidate the role of GH on AT-MSC adipogenesis using cells isolated from male GH receptor knockout (GHRKO), bovine GH transgenic (bGH) mice, and wild-type littermate control (WT) mice. AT-MSCs from subcutaneous (sc), epididiymal (epi), and mesenteric (mes) AT depots were identified and isolated by flow cytometry (Pdgfrα+ Sca1+ Cd45- Ter119- cells). Their in vitro adipogenic differentiation capacity was determined by cell morphology and real-time RT-PCR. Using identical in vitro conditions, adipogenic differentiation of AT-MSCs was only achieved in the sc depot, and not in epi and mes depots. Notably, we observed an increased differentiation in cells isolated from sc-GHRKO and an impaired differentiation of sc-bGH cells as compared to sc-WT cells. Axin2, a marker of Wnt/ß-catenin activation, was increased in mature sc-bGH adipocytes, which suggests that activation of this pathway may be responsible for the decreased adipogenesis. Thus, the present study demonstrates that (i) adipose tissue in mice has a well-defined population of Pdgfrα+ Sca1+ MSCs; (ii) the differentiation capacity of AT-MSCs varies from depot to depot regardless of GH genotype; (iii) the lack of GH action increases adipogenesis in the sc depot; and iv) activation of the Wnt/ß-catenin pathway might mediate the GH effect on AT-MSCs. Taken together, the present results suggest that GH diminishes fat mass in part by altering adipogenesis of MSCs.


Assuntos
Adipogenia/fisiologia , Tecido Adiposo Branco/citologia , Hormônio do Crescimento/fisiologia , Células-Tronco Mesenquimais/citologia , Adipócitos/citologia , Animais , Antígenos Ly/metabolismo , Bovinos , Diferenciação Celular , Hormônio do Crescimento/genética , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptores da Somatotropina/deficiência , Receptores da Somatotropina/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo
16.
Semin Cancer Biol ; 35 Suppl: S199-S223, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25865775

RESUMO

Cancer arises in the context of an in vivo tumor microenvironment. This microenvironment is both a cause and consequence of tumorigenesis. Tumor and host cells co-evolve dynamically through indirect and direct cellular interactions, eliciting multiscale effects on many biological programs, including cellular proliferation, growth, and metabolism, as well as angiogenesis and hypoxia and innate and adaptive immunity. Here we highlight specific biological processes that could be exploited as targets for the prevention and therapy of cancer. Specifically, we describe how inhibition of targets such as cholesterol synthesis and metabolites, reactive oxygen species and hypoxia, macrophage activation and conversion, indoleamine 2,3-dioxygenase regulation of dendritic cells, vascular endothelial growth factor regulation of angiogenesis, fibrosis inhibition, endoglin, and Janus kinase signaling emerge as examples of important potential nexuses in the regulation of tumorigenesis and the tumor microenvironment that can be targeted. We have also identified therapeutic agents as approaches, in particular natural products such as berberine, resveratrol, onionin A, epigallocatechin gallate, genistein, curcumin, naringenin, desoxyrhapontigenin, piperine, and zerumbone, that may warrant further investigation to target the tumor microenvironment for the treatment and/or prevention of cancer.


Assuntos
Carcinogênese/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Microambiente Tumoral/genética , Antineoplásicos/uso terapêutico , Carcinogênese/genética , Proliferação de Células/efeitos dos fármacos , Humanos , Terapia de Alvo Molecular , Neoplasias/genética , Neoplasias/prevenção & controle , Neovascularização Patológica/genética , Neovascularização Patológica/prevenção & controle , Transdução de Sinais , Microambiente Tumoral/efeitos dos fármacos
17.
Am J Physiol Cell Physiol ; 308(1): C68-78, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25339657

RESUMO

Adhesion of circulating tumor cells to vascular endothelium is mediated by specialized molecules that are functional under shear forces exerted by hematogenous flow. Endothelial E-selectin binding to glycoforms of CD44 mediates shear-resistant cell adhesion in numerous physiological and pathological conditions. However, this pathway is poorly understood in breast cancer and is the focus of the present investigation. All breast cancer cell lines used in this study strongly expressed CD44. In particular, BT-20 cells expressed CD44s and multiple CD44v isoforms, whereas MDA-MB-231 cells predominantly expressed CD44s but weakly expressed CD44v isoforms. CD44 expressed by BT-20, but not MDA-MB-231, cells possessed E-selectin ligand activity as detected by Western blotting and antigen capture assays. Importantly, CD44 expressed by intact BT-20 cells were functional E-selectin ligands, regulating cell rolling and adhesion under physiological flow conditions, as found by shRNA-targeted silencing of CD44. Antigen capture assays strongly suggest greater shear-resistant E-selectin ligand activity of BT-20 cell CD44v isoforms than CD44s. Surprisingly, CD44 was not recognized by the HECA-452 MAb, which detects sialofucosylated epitopes traditionally expressed by selectin ligands, suggesting that BT-20 cells express a novel glycoform of CD44v as an E-selectin ligand. The activity of this glycoform was predominantly attributed to N-linked glycans. Furthermore, expression of CD44v as an E-selectin ligand correlated with high levels of fucosyltransferase-3 and -6 and epithelial, rather than mesenchymal, cell phenotype. Together, these data demonstrate that expression of CD44 as a functional E-selectin ligand may be important in breast cancer metastasis.


Assuntos
Neoplasias da Mama/metabolismo , Adesão Celular , Selectina E/metabolismo , Receptores de Hialuronatos/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Células CHO , Linhagem Celular Tumoral , Movimento Celular , Cricetulus , Selectina E/genética , Células Endoteliais/metabolismo , Transição Epitelial-Mesenquimal , Feminino , Fucosiltransferases/metabolismo , Glicosilação , Humanos , Receptores de Hialuronatos/genética , Ligantes , Metástase Neoplásica , Fenótipo , Isoformas de Proteínas , Interferência de RNA , Fluxo Sanguíneo Regional , Transfecção
18.
Endocrinology ; 156(2): 453-61, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25422874

RESUMO

Group B coxsackieviruses (CVBs) are involved in triggering some cases of type 1 diabetes mellitus (T1DM). However, the molecular mechanism(s) responsible for this remain elusive. Toll-like receptor 3 (TLR3), a receptor that recognizes viral double-stranded RNA, is hypothesized to play a role in virus-induced T1DM, although this hypothesis is yet to be substantiated. The objective of this study was to directly investigate the role of TLR3 in CVB-triggered T1DM in nonobese diabetic (NOD) mice, a mouse model of human T1DM that is widely used to study both spontaneous autoimmune and viral-induced T1DM. As such, we infected female wild-type (TLR3(+/+)) and TLR3 knockout (TLR3(-/-)) NOD mice with CVB4 and compared the incidence of diabetes in CVB4-infected mice with that of uninfected counterparts. We also evaluated the islets of uninfected and CVB4-infected wild-type and TLR3 knockout NOD mice by immunohistochemistry and insulitis scoring. TLR3 knockout mice were markedly protected from CVB4-induced diabetes compared with CVB4-infected wild-type mice. CVB4-induced T-lymphocyte-mediated insulitis was also significantly less severe in TLR3 knockout mice compared with wild-type mice. No differences in insulitis were observed between uninfected animals, either wild-type or TLR3 knockout mice. These data demonstrate for the first time that TLR3 is 1) critical for CVB4-induced T1DM, and 2) modulates CVB4-induced insulitis in genetically prone NOD mice.


Assuntos
Infecções por Coxsackievirus/complicações , Diabetes Mellitus Tipo 1/virologia , Receptor 3 Toll-Like/metabolismo , Animais , Infecções por Coxsackievirus/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Enterovirus Humano B/isolamento & purificação , Feminino , Camundongos Endogâmicos NOD , Camundongos Knockout , Pâncreas/virologia , Distribuição Aleatória
19.
Endocrinology ; 156(5): 1794-803, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25521584

RESUMO

White adipose tissue (WAT) is composed of mature adipocytes and a stromal vascular fraction (SVF), which contains a variety of cells, including immune cells that vary among the different WAT depots. Growth hormone (GH) impacts immune function and adiposity in an adipose depot-specific manner. However, its effects on WAT immune cell populations remain unstudied. Bovine GH transgenic (bGH) mice are commonly used to study the in vivo effects of GH. These giant mice have an excess of GH action, impaired glucose metabolism, decreased adiposity, increased lean mass, and a shortened lifespan. Therefore, the purpose of this study was to characterize the WAT depot-specific differences in immune cell populations in the presence of excess GH in vivo. Three WAT depots were assessed: inguinal (sc), epididymal (EPI), and mesenteric (MES). Subcutaneous and MES bGH WAT depots showed a significantly higher number of total SVF cells, yet only MES bGH WAT had higher leukocyte counts compared with control samples. By means of flow cytometry analysis of the SVF, we detected greater macrophage and regulatory T-cell infiltration in sc and MES bGH WAT depots compared with controls. However, no differences were observed in the EPI WAT depot. RNA-sequencing confirmed significant alterations in pathways related to T-cell infiltration and activation in the sc depot with fewer significant changes in the EPI bGH WAT depot. These findings collectively point to a previously unrecognized role for GH in influencing the distribution of WAT immune cell populations in a depot-specific manner.


Assuntos
Tecido Adiposo Branco/imunologia , Hormônio do Crescimento/genética , Macrófagos/imunologia , Células Estromais/citologia , Linfócitos T Reguladores/imunologia , Tecido Adiposo Branco/citologia , Adiposidade/imunologia , Animais , Bovinos , Hormônio do Crescimento/imunologia , Gordura Intra-Abdominal/citologia , Gordura Intra-Abdominal/imunologia , Contagem de Leucócitos , Macrófagos/citologia , Masculino , Camundongos , Camundongos Transgênicos , Gordura Subcutânea/citologia , Gordura Subcutânea/imunologia
20.
Front Immunol ; 5: 341, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25101083

RESUMO

In the last decade, it has become apparent that toll-like receptor (TLR) signaling can play an important role in ovarian cancer (OC) progression. Interestingly, TLR activation in immune cells can help activate an anti-tumor response, while TLR signaling in tumor cells themselves is often associated with cancer-promoting inflammation. For example, it has been shown that TLR activation in dendritic cells can result in more effective antigen presentation to T cells, thereby favoring tumor eradication. However, aberrant TLR expression in OC cells is associated with more aggressive disease (likely due to recruitment of pro-tumoral leukocytes to the tumor site) and has also been implicated in resistance to mainstream chemotherapy. The delicate balance of TLR activation in the tumor microenvironment in different cell types altogether help shape the inflammatory profile and outcome of tumor growth or regression. With further studies, specific activation or repression of TLRs may be harnessed to offer novel immunotherapies or adjuvants to traditional chemotherapy for some OC patients. Herewith, we review recent literature on basic and translational research concerning therapeutic targeting of TLR pathways for the treatment of OC.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...