Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 133(6): 994-1005, 2008 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-18555776

RESUMEN

The effects of primary tumors on the host systemic environment and resulting contributions of the host to tumor growth are poorly understood. Here, we find that human breast carcinomas instigate the growth of otherwise-indolent tumor cells, micrometastases, and human tumor surgical specimens located at distant anatomical sites. This systemic instigation is accompanied by incorporation of bone-marrow cells (BMCs) into the stroma of the distant, once-indolent tumors. We find that BMCs of hosts bearing instigating tumors are functionally activated prior to their mobilization; hence, when coinjected with indolent cells, these activated BMCs mimic the systemic effects imparted by instigating tumors. Secretion of osteopontin by instigating tumors is necessary for BMC activation and the subsequent outgrowth of the distant otherwise-indolent tumors. These results reveal that outgrowth of indolent tumors can be governed on a systemic level by endocrine factors released by certain instigating tumors, and hold important experimental and therapeutic implications.


Asunto(s)
Adenocarcinoma/metabolismo , Células de la Médula Ósea/citología , Neoplasias de la Mama/metabolismo , Metástasis de la Neoplasia , Osteopontina/metabolismo , Animales , Células de la Médula Ósea/metabolismo , División Celular , Línea Celular Tumoral , Movimiento Celular , Neoplasias del Colon/metabolismo , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Trasplante Heterólogo
2.
Cell Immunol ; 361: 104285, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33484943

RESUMEN

Myeloid derived suppressor cells (MDSCs) are a diverse collection of immune cells that suppress anti-tumor immune responses. Decreasing MDSCs accumulation in the tumor microenvironment could improve the anti-tumor immune response and improve immunotherapy. Here, we examine the impact of physiologically relevant thermal treatments on the accumulation of MDSCs in tumors in mice. We found that different temperature-based protocols, including 1) weekly whole-body hyperthermia, 2) housing mice at their thermoneutral temperature (TT, ~30 °C), and 3) housing mice at a subthermoneutral temperature (ST,~22 °C) while providing a localized heat source, each resulted in a reduction in MDSC accumulation and improved tumor growth control compared to control mice housed at ST, which is the standard, mandated housing temperature for laboratory mice. Additionally, we found that low dose ß-adrenergic receptor blocker (propranolol) therapy reduced MDSC accumulation and improved tumor growth control to a similar degree as the models that relieved cold stress. These results show that thermal treatments can decrease MDSC accumulation and tumor growth comparable to propranolol therapy.


Asunto(s)
Calor/uso terapéutico , Células Supresoras de Origen Mieloide/inmunología , Neoplasias/inmunología , Antagonistas Adrenérgicos beta/farmacología , Animales , Línea Celular Tumoral , Femenino , Respuesta al Choque Térmico/fisiología , Calefacción/métodos , Hipertermia Inducida/métodos , Inmunoterapia/métodos , Masculino , Ratones , Ratones Endogámicos BALB C , Células Supresoras de Origen Mieloide/metabolismo , Células Supresoras de Origen Mieloide/fisiología , Microambiente Tumoral/inmunología
3.
J Immunol ; 202(3): 631-636, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30670578

RESUMEN

Mice are the preeminent research organism in which to model human diseases and study the involvement of the immune response. Rapidly accumulating evidence indicates a significant involvement of stress hormones in cancer progression, resistance to therapies, and suppression of immune responses. As a result, there has been a concerted effort to model human stress in mice. In this article, we discuss recent literature showing how mice in research facilities are chronically stressed at baseline because of environmental factors. Focusing on housing temperature, we suggest that the stress of cool housing temperatures contributes to the impact of other imposed experimental stressors and therefore has a confounding effect on mouse stress models. Furthermore, we propose that manipulation of housing temperature is a useful approach for studying the impact of chronic stress on disease and the immune response and for testing therapeutic methods of reducing the negative effects of chronic stress.


Asunto(s)
Frío , Vivienda para Animales , Neoplasias/inmunología , Estrés Fisiológico/inmunología , Animales , Modelos Animales de Enfermedad , Ratones
4.
Br J Cancer ; 120(2): 207-217, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30518816

RESUMEN

BACKGROUND: Advanced cancer causes necrosis and releases damage-associated molecular patterns (DAMPs). Mitochondrial DAMPs activate neutrophils, including generation of neutrophil extracellular traps (NETs), which are injurious, thrombogenic, and implicated in metastasis. We hypothesised that extracellular mitochondrial DNA (mtDNA) in ascites from patients with epithelial ovarian cancer (EOC) would correlate with worse outcomes. METHODS: Banked ascites supernatants from patients with newly diagnosed advanced EOC were analysed for mtDNA, neutrophil elastase, and activation of healthy donor neutrophils and platelets. TCGA was mined for expression of SELP and ELANE. RESULTS: The highest quartile of ascites mtDNA correlated with reduced progression-free survival (PFS) and a higher likelihood of disease progression within 12-months following primary surgery (n = 68, log-rank, p = 0.0178). NETs were detected in resected tumours. Ascites supernatants chemoattracted neutrophils, induced NETs, and activated platelets. Ascites exposure rendered neutrophils suppressive, based on abrogation of ex vivo stimulated T cell proliferation. Increased SELP mRNA expression correlated with worse overall survival (n = 302, Cox model, p = 0.02). CONCLUSION: In this single-centre retrospective analysis, ascites mtDNA correlated with worse PFS in advanced EOC. Mitochondrial and other DAMPs in ascites may activate neutrophil and platelet responses that facilitate metastasis and obstruct anti-tumour immunity. These pathways are potential prognostic markers and therapeutic targets.


Asunto(s)
Alarminas/genética , Carcinoma Epitelial de Ovario/genética , ADN Mitocondrial/genética , Trampas Extracelulares/genética , Anciano , Ascitis/genética , Ascitis/patología , Plaquetas/metabolismo , Carcinoma Epitelial de Ovario/patología , Trampas Extracelulares/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Elastasa de Leucocito/genética , Persona de Mediana Edad , Metástasis de la Neoplasia , Estadificación de Neoplasias , Neutrófilos/metabolismo , Neutrófilos/patología , Supervivencia sin Progresión , Microambiente Tumoral/genética
5.
Cancer Immunol Immunother ; 68(1): 11-22, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30229289

RESUMEN

Primary and secondary lymphoid organs are heavily innervated by the autonomic nervous system. Norepinephrine, the primary neurotransmitter secreted by post-ganglionic sympathetic neurons, binds to and activates ß-adrenergic receptors expressed on the surface of immune cells and regulates the functions of these cells. While it is known that both activated and memory CD8+ T-cells primarily express the ß2-adrenergic receptor (ß2-AR) and that signaling through this receptor can inhibit CD8+ T-cell effector function, the mechanism(s) underlying this suppression is not understood. Under normal activation conditions, T-cells increase glucose uptake and undergo metabolic reprogramming. In this study, we show that treatment of murine CD8+ T-cells with the pan ß-AR agonist isoproterenol (ISO) was associated with a reduced expression of glucose transporter 1 following activation, as well as decreased glucose uptake and glycolysis compared to CD8+ T-cells activated in the absence of ISO. The effect of ISO was specifically dependent upon ß2-AR, since it was not seen in adrb2-/- CD8+ T-cells and was blocked by the ß-AR antagonist propranolol. In addition, we found that mitochondrial function in CD8+ T-cells was also impaired by ß2-AR signaling. This study demonstrates that one mechanism by which ß2-AR signaling can inhibit CD8+ T-cell activation is by suppressing the required metabolic reprogramming events which accompany activation of these immune cells and thus reveals a new mechanism by which adrenergic stress can suppress the effector activity of immune cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Activación de Linfocitos/inmunología , Receptores Adrenérgicos beta 2/inmunología , Transducción de Señal/inmunología , Agonistas Adrenérgicos beta/farmacología , Antagonistas Adrenérgicos beta/farmacología , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Células Cultivadas , Femenino , Glucosa/inmunología , Glucosa/metabolismo , Tolerancia Inmunológica/efectos de los fármacos , Tolerancia Inmunológica/inmunología , Isoproterenol/farmacología , Activación de Linfocitos/efectos de los fármacos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Propranolol/farmacología , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal/efectos de los fármacos
6.
Int J Hyperthermia ; 36(sup1): 83-89, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31795833

RESUMEN

There is substantial research being conducted on the relationships between the gut microbiome, the immune response and health and disease. Environmental temperature and heat stress are known to modify the gut microbiome. Changes in core temperature have been linked, in multiple phyla, to altered microbiome composition and function. This raises the question of whether local/regional or whole body thermal therapies which target tumors in the abdomen, peritoneal cavity, or pelvis influence the gut microbiome. To date, there is little information on whether thermal therapy exerts positive or negative effects on the microbiome. This is an intriguing question since there is growing interest in the immunological impact of various thermal therapies. The goal of this brief review is to highlight research on how environmental conditions, particularly temperature (internal as well as external temperatures) influences the gut microbiome. Given the potential for temperature shifts to modulate gut microbe function and composition, it is likely that various forms of thermal therapy, including hyperthermic intraperitoneal chemotherapy (HIPEC), deep regional, and whole body hyperthermia influence the microbiome in ways that are currently not appreciated. More research is needed to determine whether thermal therapy induced changes in the microbiome occur, and whether they are beneficial or detrimental to the host. Currently, although approaches to microbiome modification such as dietary intervention, fecal transfer, probiotics and prebiotics are being developed, the potential of temperature manipulation has, as yet, not been explored. Therefore, new research could reveal whether perturbations of the microbiome composition that have negative health consequences (dysbiosis) could be an important target for treatment by thermal medicine.


Asunto(s)
Temperatura Corporal/fisiología , Microbioma Gastrointestinal/fisiología , Hipertermia Inducida/métodos , Animales , Humanos , Hipertermia Inducida/efectos adversos , Temperatura
7.
Int J Hyperthermia ; 34(2): 135-143, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29498310

RESUMEN

Stress in patients and pre-clinical research animals plays a critical role in disease progression Activation of the sympathetic nervous system (SNS) by stress results in secretion of the catecholamines epinephrine (Epi) and norepinephrine (NE) from the adrenal gland and sympathetic nerve endings. Adrenergic receptors for catecholamines are present on immune cells and their activity is affected by stress and the accompanying changes in levels of these neurotransmitters. In this short review, we discuss how this adrenergic stress impacts two categories of immune responses, infections and autoimmune diseases. Catecholamines signal primarily through the ß2-adrenergic receptors present on innate and adaptive immune cells which are critical in responding to infections caused by pathogens. In general, this adrenergic input, particularly chronic stimulation, suppresses lymphocytes and allows infections to progress. On the other hand, insufficient adrenergic control of immune responses allows progression of several autoimmune diseases.


Asunto(s)
Autoinmunidad/inmunología , Enfermedades Transmisibles/inmunología , Sistema Nervioso Simpático/fisiopatología , Humanos
8.
Adv Exp Med Biol ; 1036: 173-189, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29275472

RESUMEN

In the last 10-15 years, there has been a recognition that the catecholamines (norepinephrine, NE, and epinephrine, Epi) released by the sympathetic nervous system under stressful conditions promote tumor growth through a variety of mechanisms. Tumors recruit autonomic nerves during their development and NE is then released locally in the tumor microenvironment (TME). Acting through adrenergic receptors present on a variety of cells in the TME, NE and Epi induce proliferation, resistance to apoptosis, epithelial to mesenchymal transition, metastasis of tumor cells, angiogenesis, and inflammation in the TME. These pre-clinical studies have been conducted in mouse models whose care and housing parameters are outlined in "The Guide for the Care and Use of Laboratory Animals [1]. In particular, the Guide mandates that mice be housed at standardized sub-thermoneutral temperatures; however, this causes a state of chronic cold-stress and elevated levels of NE. Although mice are able to maintain a normal body temperature when kept at these cool temperatures, it is becoming clear that this cold-stress is sufficient to activate physiological changes which affect experimental outcomes. We find that when mice are housed under standard, sub-thermoneutral temperatures (~22 °C, ST), tumor growth is significantly greater than when mice are housed at thermoneutrality (~30 °C TT). We also find that the anti-tumor immune response is suppressed at ST and this immunosuppression can be reversed by housing mice at TT or by administration of propranolol (a ß-adrenergic receptor antagonist) to mice housed at ST. Furthermore, at ST tumors are more resistant to therapy and can also be sensitized to cytotoxic therapies by housing mice at TT or by treating mice with propranolol. The implications of these observations are particularly relevant to the way in which experiments conducted in preclinical models are interpreted and the findings implemented in the clinic. It may be that the disappointing failure of many new therapies to fulfill their promise in the clinic is related to an incomplete preclinical assessment in mouse models. Further, an expanded understanding of the efficacy of a therapy alone or in combination obtained by testing under a wider range of conditions would better predict how patients, who are under various levels of stress, might respond in a clinical setting. This may be particularly important to consider since we now appreciate that long term outcome of many therapies depends on eliciting an immune response.It is clear that the outcome of metabolic experiments, immunological investigations and therapeutic efficacy testing in tumors of mice housed at ST is restricted and expanding these experiments to include results obtained at TT may provide us with valuable information that would otherwise be overlooked.


Asunto(s)
Neoplasias Experimentales , Animales , Enfermedad Crónica , Humanos , Ratones , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Sistema Nervioso/inmunología , Sistema Nervioso/patología , Estrés Psicológico/inmunología , Estrés Psicológico/patología , Estrés Psicológico/terapia
9.
Proc Natl Acad Sci U S A ; 110(50): 20176-81, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24248371

RESUMEN

We show here that fundamental aspects of antitumor immunity in mice are significantly influenced by ambient housing temperature. Standard housing temperature for laboratory mice in research facilities is mandated to be between 20-26 °C; however, these subthermoneutral temperatures cause mild chronic cold stress, activating thermogenesis to maintain normal body temperature. When stress is alleviated by housing at thermoneutral ambient temperature (30-31 °C), we observe a striking reduction in tumor formation, growth rate and metastasis. This improved control of tumor growth is dependent upon the adaptive immune system. We observe significantly increased numbers of antigen-specific CD8(+) T lymphocytes and CD8(+) T cells with an activated phenotype in the tumor microenvironment at thermoneutrality. At the same time there is a significant reduction in numbers of immunosuppressive MDSCs and regulatory T lymphocytes. Notably, in temperature preference studies, tumor-bearing mice select a higher ambient temperature than non-tumor-bearing mice, suggesting that tumor-bearing mice experience a greater degree of cold-stress. Overall, our data raise the hypothesis that suppression of antitumor immunity is an outcome of cold stress-induced thermogenesis. Therefore, the common approach of studying immunity against tumors in mice housed only at standard room temperature may be limiting our understanding of the full potential of the antitumor immune response.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vivienda para Animales/normas , Inmunoterapia/métodos , Neoplasias/inmunología , Estrés Fisiológico/inmunología , Temperatura , Análisis de Varianza , Animales , Recuento de Células Sanguíneas , Línea Celular Tumoral , Femenino , Inmunohistoquímica , Modelos Lineales , Ratones , Ratones Endogámicos BALB C
10.
Cancer Immunol Immunother ; 63(11): 1115-28, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25307152

RESUMEN

Long conserved mechanisms maintain homeostasis in living creatures in response to a variety of stresses. However, continuous exposure to stress can result in unabated production of stress hormones, especially catecholamines, which can have detrimental health effects. While the long-term effects of chronic stress have well-known physiological consequences, recent discoveries have revealed that stress may affect therapeutic efficacy in cancer. Growing epidemiological evidence reveals strong correlations between progression-free and long-term survival and ß-blocker usage in cancer patients. In this review, we summarize the current understanding of how the catecholamines, epinephrine and norepinephrine, affect cancer cell survival and tumor progression. We also highlight new data exploring the potential contributions of stress to immunosuppression in the tumor microenvironment and the implications of these findings for the efficacy of immunotherapies.


Asunto(s)
Terapia de Inmunosupresión/métodos , Inmunosupresores/uso terapéutico , Neoplasias/patología , Neoplasias/psicología , Microambiente Tumoral , Animales , Ansiedad/complicaciones , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Epinefrina/metabolismo , Miedo , Homeostasis , Humanos , Tolerancia Inmunológica , Ratones , Norepinefrina/metabolismo , Receptores Adrenérgicos/metabolismo , Estrés Psicológico , Sistema Nervioso Simpático
11.
J Transl Med ; 11: 110, 2013 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-23639003

RESUMEN

BACKGROUND: Studies of primary patient tumor xenografts grown in immunodeficient mice have shown that these tumors histologically and genetically closely resemble the original tumors. These patient xenograft models are becoming widely used for therapeutic efficacy studies. Because many therapies are directed at tumor stromal components and because the tumor microenvironment also is known to influence the response of a tumor to therapy, it is important to understand the nature of the stroma and, in particular, the vascular supply of patient xenografts. METHODS: Patient tumor xenografts were established by implanting undisrupted pieces of patient tumors in SCID mice. For this study, formalin fixed, paraffin embedded specimens from several types of solid tumors were selected and, using species-specific antibodies which react with formalin fixed antigens, we analyzed the species origin of the stroma and blood vessels that supported tumor growth in these models. Additionally, we investigated the kinetics of the vascularization process in a colon tumor and a mesothelioma xenograft. In mice bearing a head and neck xenograft, a perfusion study was performed to compare the functionality of the human and mouse tumor vessels. RESULTS: In patient tumors which successfully engrafted, the human stroma and vessels which were engrafted as part of the original tumor did not survive and were no longer detectable at the time of first passage (15-25 weeks). Uniformly, the stroma and vessels supporting the growth of these tumors were of murine origin. The results of the kinetic studies showed that the loss of the human vessels and vascularization by host vessels occurred more rapidly in a colon tumor (by 3 weeks) than in a mesothelioma (by 9 weeks). Finally, the perfusion studies revealed that while mouse vessels in the periphery of the tumor were perfused, those in the central regions were rarely perfused. No vessels of human origin were detected in this model. CONCLUSIONS: In the tumors we investigated, we found no evidence that the human stromal cells and vessels contained in the original implant either survived or contributed in any substantive way to the growth of these xenografts.


Asunto(s)
Trasplante de Neoplasias , Neoplasias/patología , Neovascularización Patológica/patología , Adulto , Anciano , Animales , Neoplasias del Colon/patología , Femenino , Humanos , Masculino , Mesotelioma/patología , Ratones , Ratones SCID , Persona de Mediana Edad , Neoplasias/terapia , Neovascularización Patológica/terapia , Células Tumorales Cultivadas , Microambiente Tumoral
12.
Biochimie ; 210: 71-81, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36693616

RESUMEN

Preclinical mouse models are widely used for studying mechanisms of disease and responses to therapeutics, however there is concern about the lack of experimental reproducibility and failure to predict translational success. The gut microbiome has emerged as a regulator of metabolism and immunological processes in health and disease. The gut microbiome of mice differs by supplier and this affects experimental outcomes. We have previously reported that the mandated, mildly cool housing temperature for research mice (22°-26 °C) induces chronic adrenergic stress which suppresses anti-tumor immunity and promotes tumor growth compared to thermoneutral housing (30 °C). Therefore, we wondered how housing temperature affects the microbiome. Here, we demonstrate that the gut microbiome of BALB/c mice is easily modulated by a few degrees difference in temperature. Our results reveal significant differences between the gut microbiome of mice housed at 22°-23 °C vs. 30 °C. Although the genera vary, we consistently observed an enrichment of members of the family Lachnospiraceae when mice are housed at 22°-23 °C. These findings demonstrate that adrenergic stress and need for increased energy harvest to support thermogenesis, in addition to other factors such as diet, modulates the gut microbiome and this could be one mechanism by which housing temperature affects experimental outcomes. Additionally, tumor growth in mice housed at 30 °C also increases the proportion of Lachnospiraceae. The idea that stress can alter the gut microbiome and cause differences in experimental outcomes is applicable to mouse studies in general and is a variable that has significant potential to affect experimental reproducibility.


Asunto(s)
Microbioma Gastrointestinal , Neoplasias , Animales , Ratones , Temperatura , Reproducibilidad de los Resultados , Vivienda para Animales , Adrenérgicos
13.
J Hepatol ; 56(1): 176-83, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21835141

RESUMEN

BACKGROUND & AIMS: The mammalian target of rapamycin (mTOR) plays a pivotal role in hepatocellular carcinoma (HCC). Previous studies indicated that inhibition of mTORC1 enhanced histone deacetylase inhibitors (HDACis)-mediated anti-tumor activity, accompanied with feedback activation of AKT. Therefore, dual targeting of mTORC1/C2 should be more efficient in suppressing AKT activity and in enhancing the anti-tumor activity of HDACi in HCC. METHODS: The interactions between mTOR kinase inhibitors (mTORKis) (i.e., Pp242, AZD8055, OSI027) and HDACis (i.e., SAHA, LBH589) were examined in vitro using HCC cell lines and in vivo using patient-derived primary HCC xenografts on SCID mice. RESULTS: mTORKis significantly enhanced HDACi-induced apoptosis in HCC cells. The inhibition of both mTORC1/2 not only efficiently blocked mTORC1 signaling, but also abrogated AKT-feedback activation caused by selective mTORC1 inhibition. The co-treatment of mTORKi and HDACi further inhibited AKT signaling and upregulated Bim. Dysfunction of mTORC2 by shRNA significantly lowered the threshold of HDACi-induced cytotoxicity by abrogating AKT activation. Knockdown of AKT1 sensitized Pp242/HDACi-induced apoptosis and ectopic expression of constitutively active AKT1 abrogated the combination-induced cytotoxicity, indicating AKT plays a vital role in the combination-induced effects. Knockdown of Bim prevented Pp242/HDACis-induced cytotoxicity in HCC. Lastly, in vivo studies indicated that the combination of AZD8055 and SAHA almost completely inhibited tumor-growth, without obvious adverse effects, by abrogating AKT and upregulating Bim; while either agent alone shows only 30% inhibition in primary HCC xenografts. CONCLUSIONS: Our findings suggest that a combining-regimen of mTORKi and HDACi may be an effective therapeutic strategy for HCC.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/administración & dosificación , Neoplasias Hepáticas/tratamiento farmacológico , Proteínas/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Animales , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Secuencia de Bases , Proteína 11 Similar a Bcl2 , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Sinergismo Farmacológico , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Diana Mecanicista del Complejo 1 de la Rapamicina , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones SCID , Complejos Multiproteicos , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Animals (Basel) ; 12(3)2022 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-35158694

RESUMEN

Mice are the most common animal used to study disease, but there are real concerns about the reproducibility of many of these experiments. This review discusses how several different sources of chronic stress can directly impact experimental outcomes. Mandated housing conditions induce an underappreciated level of chronic stress but are not usually considered or reported as part of the experimental design. Since chronic stress plays a critical role in the development and progression of many somatic diseases including cancer, obesity, and auto-immune diseases, this baseline stress can directly affect outcomes of such experiments. To study the role of stress in both physical and psychiatric diseases, there has been a proliferation of protocols for imposing chronic stress on mice. For somatic diseases, biomarkers can be used to compare the models with the disease in patients, but to evaluate the validity of psychiatric models, behavioral tests are carried out to assess changes in behavior and these tests may themselves cause an underappreciated degree of additional stress. Therefore, it is important for animal welfare to reduce baseline stress and select the most humane protocols for inducing and assessing chronic stress to obtain the most reliable outcomes.

15.
Cancer Immunol Res ; 9(6): 651-664, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33762351

RESUMEN

Metabolic dysfunction and exhaustion in tumor-infiltrating T cells have been linked to ineffectual antitumor immunity and the failure of immune checkpoint inhibitor therapy. We report here that chronic stress plays a previously unrecognized role in regulating the state of T cells in the tumor microenvironment (TME). Using two mouse tumor models, we found that blocking chronic adrenergic stress signaling using the pan ß-blocker propranolol or by using mice lacking the ß2-adrenergic receptor (ß2-AR) results in reduced tumor growth rates with significantly fewer tumor-infiltrating T cells that express markers of exhaustion, with a concomitant increase in progenitor exhausted T cells. We also report that blocking ß-AR signaling in mice increases glycolysis and oxidative phosphorylation in tumor-infiltrating lymphocytes (TIL), which associated with increased expression of the costimulatory molecule CD28 and increased antitumor effector functions, including increased cytokine production. Using T cells from Nur77-GFP reporter mice to monitor T-cell activation, we observed that stress-induced ß-AR signaling suppresses T-cell receptor (TCR) signaling. Together, these data suggest that chronic stress-induced adrenergic receptor signaling serves as a "checkpoint" of immune responses and contributes to immunosuppression in the TME by promoting T-cell metabolic dysfunction and exhaustion. These results also support the possibility that chronic stress, which unfortunately is increased in many patients with cancer following their diagnoses, could be exerting a major negative influence on the outcome of therapies that depend upon the status of TILs and support the use of strategies to reduce stress or ß-AR signaling in combination with immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Experimentales/inmunología , Receptores Adrenérgicos beta 2/inmunología , Microambiente Tumoral/inmunología , Antagonistas Adrenérgicos beta/farmacología , Animales , Línea Celular Tumoral , Respuesta al Choque por Frío , Femenino , Inmunoterapia/métodos , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Fenotipo , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal/inmunología
16.
Int J Hyperthermia ; 26(4): 366-75, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20210603

RESUMEN

PURPOSE: We employed a grp170-secreting tumour cell system to determine whether tumour cells engineered to secrete grp170 generate an antitumour-specific immune response. Further, we examine the possibility that secreted grp170 can bind to and co-transport out of tumour cells full-length tumour antigens that may play a role in the anti-tumour immune response. MATERIALS AND METHODS: Wild type Colon-26 and Colon-26 engineered to secrete grp170 were subcutaneously inoculated into BALB/c mice. Tumour growth was monitored, and variations in immunoregulatory mechanisms were evaluated using immunohistochemistry, lymphocyte depletion, ELISpot assays, and Western blot analysis. RESULTS: Immunisation of animals with grp170-secreting tumour cells results in rejection of the tumour by induction of antigen-specific, CD8-dependent immune responses. The secreted grp170 is able to deliver full-length tumour antigens to the tumour microenvironment, thus making them available for uptake by antigen presenting cells (APCs) to initiate tumour-specific immune responses. CONCLUSIONS: These data parallel our studies showing that hsp110 or grp170 are able to chaperone full-length proteins, and when complexed with protein antigens and used as vaccines, these complexes elicit immune responses in vivo against the protein antigens. This cell-based approach has the potential to be utilised as a tumour-specific vaccine in tumours of various histological origins.


Asunto(s)
Inmunidad Adaptativa/inmunología , Antígenos de Neoplasias/metabolismo , Glicoproteínas/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Chaperonas Moleculares/metabolismo , Neoplasias/inmunología , Neoplasias/metabolismo , Animales , Antígenos de Neoplasias/inmunología , Antígenos Virales de Tumores/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Femenino , Glicoproteínas/genética , Antígenos H-2/metabolismo , Proteínas HSP70 de Choque Térmico/genética , Interferón gamma/metabolismo , Luciferasas/metabolismo , Activación de Linfocitos/inmunología , Depleción Linfocítica , Ratones , Ratones Endogámicos BALB C , Chaperonas Moleculares/genética , Neoplasias/patología , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica/inmunología , Señales de Clasificación de Proteína/genética , Bazo/citología , Bazo/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T Citotóxicos/inmunología , Transfección , Proteínas del Envoltorio Viral/inmunología , Proteínas del Envoltorio Viral/metabolismo
17.
Nat Commun ; 11(1): 1821, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32286326

RESUMEN

The abscopal effect following ionizing radiation therapy (RT) is considered to be a rare event. This effect does occur more frequently when combined with other therapies, including immunotherapy. Here we demonstrate that the frequency of abscopal events following RT alone is highly dependent upon the degree of adrenergic stress in the tumor-bearing host. Using a combination of physiologic, pharmacologic and genetic strategies, we observe improvements in the control of both irradiated and non-irradiated distant tumors, including metastatic tumors, when adrenergic stress or signaling through ß-adrenergic receptor is reduced. Further, we observe cellular and molecular evidence of improved, antigen-specific, anti-tumor immune responses which also depend upon T cell egress from draining lymph nodes. These data suggest that blockade of ß2 adrenergic stress signaling could be a useful, safe, and feasible strategy to improve efficacy in cancer patients undergoing radiation therapy.


Asunto(s)
Adrenérgicos/farmacología , Inmunidad , Neoplasias/inmunología , Neoplasias/radioterapia , Radiación Ionizante , Estrés Fisiológico , Antagonistas Adrenérgicos beta/farmacología , Animales , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ganglios Linfáticos/patología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias/genética , Receptores Adrenérgicos beta 2/metabolismo , Transducción de Señal/efectos de los fármacos
18.
Biochem Biophys Res Commun ; 386(2): 305-10, 2009 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-19523437

RESUMEN

Lung tumor xenografts grown in immunocompromised mice provide a renewable source of tumor tissue for research and a means to study individualized response to chemotherapy. Critical to this utility is verification that the xenograft cells retain core phenotypic characteristics of the original tumor. We compared eight non-small cell lung carcinomas with their corresponding xenografts grown in mice with severe combined immunodeficiency by way of histology, immunohistochemistry, and microRNA expression profiling. Six of the eight xenografts closely resembled their original tumor by light microscopy. The xenografts also largely retained key immunophenotypic features. With expression profiling of human microRNAs, however, xenografts clustered separately from the original tumors. While this may be partly due to contamination by non-neoplastic human and mouse stroma, the results suggest that miRNA expression may be altered in xenografts and that this possibility should be further evaluated.


Asunto(s)
Neoplasias Pulmonares/genética , MicroARNs/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Perfilación de la Expresión Génica , Ratones , Ratones SCID
19.
Clin Cancer Res ; 25(8): 2363-2365, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30670491

RESUMEN

Depression induces secretion of neuropeptide Y from prostate cancer cells, which, in turn, recruits myeloid-derived suppressor cells (MDSC) to the tumor; tumor cells and MDSCs secrete IL6, which activates STAT3 within cancer cells. Prostate cancer samples from depressed patients reveal a similar phenotype, suggesting new treatment strategies based upon blockade of ß2-adrenergic receptors and/or neuropeptide Y.See related article by Cheng et al., p. 2621.


Asunto(s)
Células Supresoras de Origen Mieloide/inmunología , Neoplasias de la Próstata/genética , Depresión , Humanos , Masculino , Factor de Transcripción STAT3/genética , Transducción de Señal
20.
J Clin Invest ; 129(12): 5537-5552, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31566578

RESUMEN

Catecholamines released by sympathetic nerves can activate adrenergic receptors present on nearly every cell type, including myeloid-derived suppressor cells (MDSCs). Using in vitro systems, murine tumor models in wild-type and genetically modified (ß2-AR-/-) mice, and adoptive transfer approaches, we found that the degree of ß2-AR signaling significantly influences MDSC frequency and survival in tumors and other tissues. It also modulates their expression of immunosuppressive molecules such as arginase-I and PD-L1 and alters their ability to suppress the proliferation of T cells. The regulatory functions of ß2-AR signaling in MDSCs were also found to be dependent upon STAT3 phosphorylation. Moreover, we observed that the ß2-AR-mediated increase in MDSC survival is dependent upon Fas-FasL interactions, and this is consistent with gene expression analyses, which reveal a greater expression of apoptosis-related genes in ß2-AR-/- MDSCs. Our data reveal the potential of ß2-AR signaling to increase the generation of MDSCs from both murine and human peripheral blood cells and that the immunosuppressive function of MDSCs can be mitigated by treatment with ß-AR antagonists, or enhanced by ß-AR agonists. This strongly supports the possibility that reducing stress-induced activation of ß2-ARs could help to overcome immune suppression and enhance the efficacy of immunotherapy and other cancer therapies.


Asunto(s)
Tolerancia Inmunológica , Células Supresoras de Origen Mieloide/inmunología , Receptores Adrenérgicos beta 2/inmunología , Animales , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células Supresoras de Origen Mieloide/fisiología , Neoplasias/irrigación sanguínea , Fosforilación , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA