Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Cancer Res Commun ; 4(4): 1063-1081, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38506672

RESUMEN

Intestinal chronic inflammation is associated with microbial dysbiosis and accumulation of various immune cells including myeloid-derived suppressor cells (MDSC), which profoundly impact the immune microenvironment, perturb homeostasis and increase the risk to develop colitis-associated colorectal cancer (CAC). However, the specific MDSCs-dysbiotic microbiota interactions and their collective impact on CAC development remain poorly understood. In this study, using a murine model of CAC, we demonstrate that CAC-bearing mice exhibit significantly elevated levels of highly immunosuppressive MDSCs, accompanied by microbiota alterations. Both MDSCs and bacteria that infiltrate the colon tissue and developing tumors can be found in close proximity, suggesting intricate MDSC-microbiota cross-talk within the tumor microenvironment. To investigate this phenomenon, we employed antibiotic treatment to disrupt MDSC-microbiota interactions. This intervention yielded a remarkable reduction in intestinal inflammation, decreased MDSC levels, and alleviated immunosuppression, all of which were associated with a significant reduction in tumor burden. Furthermore, we underscore the causative role of dysbiotic microbiota in the predisposition toward tumor development, highlighting their potential as biomarkers for predicting tumor load. We shed light on the intimate MDSCs-microbiota cross-talk, revealing how bacteria enhance MDSC suppressive features and activities, inhibit their differentiation into mature beneficial myeloid cells, and redirect some toward M2 macrophage phenotype. Collectively, this study uncovers the role of MDSC-bacteria cross-talk in impairing immune responses and promoting tumor growth, providing new insights into potential therapeutic strategies for CAC. SIGNIFICANCE: MDSCs-dysbiotic bacteria interactions in the intestine play a crucial role in intensifying immunosuppression within the CAC microenvironment, ultimately facilitating tumor growth, highlighting potential therapeutic targets for improving the treatment outcomes of CAC.


Asunto(s)
Neoplasias Asociadas a Colitis , Microbioma Gastrointestinal , Células Supresoras de Origen Mieloide , Neoplasias , Animales , Ratones , Inflamación , Microambiente Tumoral
2.
Curr Protoc ; 2(10): e561, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36214619

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are heterogenous populations of immature myeloid cells that can be divided into two main subpopulations, polymorphonuclear (PMN) MDSCs and monocytic (M) MDSCs. These cells accumulate during chronic inflammation and induce immunosuppression evident in an array of pathologies such as cancer, inflammatory bowel disease, and infectious and autoimmune diseases. Herein, we describe methods to isolate and characterize MDSCs from various murine tissue, as well as to phenotype blood-derived MDSCs from patients. The protocols describe methods for isolation of total MDSCs and their subpopulations, for characterization, and for evaluation of their distribution within tissue, as well as for assessing their maturation stage by flow cytometry, immunofluorescence analyses, and Giemsa staining. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Single-cell suspension generation from different tissue Alternate Protocol 1: Single-cell suspension generation from subcutaneous melanoma tumors Basic Protocol 2: Characterization of MDSC phenotype Basic Protocol 3: Cell separation using magnetic beads: Separating pan-MDSCs or PMN-MDSC and M-MDSC subpopulations Alternate Protocol 2: Staining and preparing MDSCs for sorting Support Protocol: PMN-MDSC and M-MDSC gating strategy in mouse Basic Protocol 4: Immunofluorescence analysis of MDSCs Basic Protocol 5: Handling human blood samples and characterizing human MDSCs Alternate Protocol 3: Flow cytometry staining of thawed human whole blood samples.


Asunto(s)
Células Supresoras de Origen Mieloide , Animales , Citometría de Flujo , Humanos , Ratones , Monocitos , Células Mieloides , Fenotipo
3.
Curr Protoc ; 2(10): e557, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36282094

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are heterogenous populations of immature myeloid cells that can be divided into two main subpopulations, polymorphonuclear (PMN) MDSCs and monocytic (M) MDSCs. These cells accumulate during chronic inflammation, characterizing an array of pathologies such as cancer, inflammatory bowel disease, and infectious and autoimmune diseases, and induce immunosuppression. The suppressive effects of MDSCs on the immune system are studied mainly when focusing on their features, functions, and impact on target cells such as T cells, natural killer cells, and B cells, among others. Herein, we describe methods for the analysis of MDSC immunosuppressive features and functions, measuring different mediators that contribute to their activities and how they impact on T cell function. The protocols described are a continuation to those in a companion Current Protocols article by Reuven et al. (2022), which uses a generated single-cell suspension and isolated cells to test their activity. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Evaluating MDSC suppressive features Alternate Protocol 1: Dichlorofluorescein diacetate-based reactive oxygen species detection Support Protocol 1: Detection of nitric oxide secretion Support Protocol 2: Measurement of arginase activity Basic Protocol 2: Evaluating MDSC suppressive function Alternate Protocol 2: In vitro effects of MDSCs on expression of T cell receptor complex during activation Support Protocol 3: Effect of MDSCs on interferon γ production Basic Protocol 3: Effect of MDSCs on T cell proliferation Basic Protocol 4: Effect of MDSCs on T cell cytotoxic activity Alternate Protocol 3: In vivo cytotoxicity assay Basic Protocol 5: Analysis of MDSC differentiation.


Asunto(s)
Células Supresoras de Origen Mieloide , Especies Reactivas de Oxígeno/metabolismo , Arginasa/metabolismo , Interferón gamma/metabolismo , Óxido Nítrico/metabolismo , Terapia de Inmunosupresión , Receptores de Antígenos de Linfocitos T/metabolismo
4.
Curr Protoc ; 2(10): e558, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36239438

RESUMEN

Myeloid-derived suppressor cells (MDSCs) represent a heterogeneous population of immature myeloid cells known to play a role in perpetuating a wide range of pathologies, such as chronic infections, autoimmune diseases, and cancer. MDSCs were first identified in mice by the markers CD11b+ Gr1+ , and later, based on their morphology, they were classified into two subsets: polymorphonuclear MDSCs, identified by the markers CD11b+ Ly6G+ Ly6CLow , and monocytic MDSCs, detected as being CD11b+ Ly6G- Ly6CHi . MDSCs are studied as immunosuppressive cells in various diseases characterized by chronic inflammation and are associated with disease causes/triggers such as pathogens, autoantigens, and cancer. Therefore, different diseases may diversely affect MDSC metabolism, migration, and differentiation, thus influencing the generated MDSC functional features and ensuing suppressive environment. In order to study MDSCs in a pathology-free environment, we established and calibrated a highly reproducible mouse model that results in the development of chronic inflammation, which is the major cause of MDSC accumulation and immune suppression. The model presented can be used to study MDSC phenotypes, functional diversity, and plasticity. It also permits study of MDSC migration from the bone marrow to peripheral lymphatic and non-lymphatic organs and MDSC crosstalk with extrinsic factors, both in vivo and ex vivo. Furthermore, this model can serve as a platform to assess the effects of anti-MDSC modalities. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol: Repetitive M.tb immunizations for the induction of chronic inflammation Alternate Protocol 1: Creating a lower grade of inflammation by changing the site of immunization Alternate Protocol 2: In vivo evaluation of immune status Support Protocol 1: Preparation of reconstituted M.tb aliquots and M.tb-IFA emulsions for each of the three injections Support Protocol 2: Preparation of an ovalbumin lentiviral expression vector Support Protocol 3: Fluorescence titering assay for the lentiviral expression vector Support Protocol 4: Spleen excision, tissue dissociation, and preparation of a single-cell suspension Support Protocol 5: Labeling of splenocytes with CFSE proliferation dye.


Asunto(s)
Células Supresoras de Origen Mieloide , Neoplasias , Animales , Autoantígenos/metabolismo , Modelos Animales de Enfermedad , Inflamación , Ratones , Células Supresoras de Origen Mieloide/metabolismo , Neoplasias/metabolismo , Ovalbúmina/metabolismo
5.
Bone Res ; 10(1): 36, 2022 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-35396510

RESUMEN

Elevated osteoclast (OC) activity is a major contributor to inflammatory bone loss (IBL) during chronic inflammatory diseases. However, the specific OC precursors (OCPs) responding to inflammatory cues and the underlying mechanisms leading to IBL are poorly understood. We identified two distinct OCP subsets: Ly6ChiCD11bhi inflammatory OCPs (iOCPs) induced during chronic inflammation, and homeostatic Ly6ChiCD11blo OCPs (hOCPs) which remained unchanged. Functional and proteomic characterization revealed that while iOCPs were rare and displayed low osteoclastogenic potential under normal conditions, they expanded during chronic inflammation and generated OCs with enhanced activity. In contrast, hOCPs were abundant and manifested high osteoclastogenic potential under normal conditions but generated OCs with low activity and were unresponsive to the inflammatory environment. Osteoclasts derived from iOCPs expressed higher levels of resorptive and metabolic proteins than those generated from hOCPs, highlighting that different osteoclast populations are formed by distinct precursors. We further identified the TNF-α and S100A8/A9 proteins as key regulators that control the iOCP response during chronic inflammation. Furthermore, we demonstrated that the response of iOCPs but not that of hOCPs was abrogated in tnf-α-/- mice, in correlation with attenuated IBL. Our findings suggest a central role for iOCPs in IBL induction. iOCPs can serve as potential biomarkers for IBL detection and possibly as new therapeutic targets to combat IBL in a wide range of inflammatory conditions.

6.
Cell Discov ; 7(1): 89, 2021 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-34580278

RESUMEN

SARS-CoV-2 outbreak has been declared by World Health Organization as a worldwide pandemic. However, there are many unknowns about the antigen-specific T-cell-mediated immune responses to SARS-CoV-2 infection. Here, we present both single-cell TCR-seq and RNA-seq to analyze the dynamics of TCR repertoire and immune metabolic functions of blood T cells collected from recently discharged COVID-19 patients. We found that while the diversity of TCR repertoire was increased in discharged patients, it returned to basal level ~1 week after becoming virus-free. The dynamics of T cell repertoire correlated with a profound shift of gene signatures from antiviral response to metabolism adaptation. We also demonstrated that the top expanded T cell clones (~10% of total T cells) display the key anti-viral features in CD8+ T cells, confirming a critical role of antigen-specific T cells in fighting against SARS-CoV-2. Our work provides a basis for further analysis of adaptive immunity in COVID-19 patients, and also has implications in developing a T-cell-based vaccine for SARS-CoV-2.

7.
Cell Immunol ; 365: 104361, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33984533

RESUMEN

The mucosal immune system plays a vital role in protecting the host from the external environment. Its major challenge is to balance immune responses against harmful and harmless agents and serve as a 'homeostatic gate keeper'. Myeloid derived suppressor cells (MDSCs) are a heterogeneous population of undifferentiated cells that are characterized by an immunoregulatory and immunosuppressive phenotype. Herein we postulate that MDSCs may be involved in shaping immune responses related to mucosal immunity, due to their immunomodulatory and tissue remodeling functions. Until recently, MDSCs were investigated mainly in cancerous diseases, where they induce and contribute to an immunosuppressive and inflammatory environment that favors tumor development. However, it is now becoming clear that MDSCs participate in non-cancerous conditions such as chronic infections, autoimmune diseases, pregnancy, aging processes and immune tolerance to commensal microbiota at mucosal sites. Since MDSCs are found in the periphery only in small numbers under normal conditions, their role is highlighted during pathologies characterized by acute or chronic inflammation, when they accumulate and become activated. In this review, we describe several aspects of the current knowledge characterizing MDSCs and their involvement in the regulation of the mucosal epithelial barrier, their crosstalk with commensal microbiota and pathogenic microorganisms, and their complex interactions with a variety of surrounding regulatory and effector immune cells. Finally, we discuss the beneficial and harmful outcomes of the MDSC regulatory functions in diseases affecting mucosal tissues. We wish to illuminate the pivotal role of MDSCs in mucosal immunity, the limitations in our understanding of all the players and the intricate challenges stemming from the complex interactions of MDSCs with their environment.


Asunto(s)
Epitelio/inmunología , Inflamación/inmunología , Microbiota/inmunología , Células Supresoras de Origen Mieloide/inmunología , Animales , Homeostasis , Interacciones Huésped-Parásitos , Humanos , Inmunidad Mucosa , Inmunomodulación
8.
J Clin Invest ; 131(10)2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-33848267

RESUMEN

Stimulation of TAM (TYRO3, AXL, and MERTK) receptor tyrosine kinases promotes tumor progression through numerous cellular mechanisms. TAM cognate ligands GAS6 and PROS1 (for TYRO3 and MERTK) are secreted by host immune cells, an interaction which may support tumor progression. Here, we revealed an unexpected antimetastatic role for myeloid-derived PROS1: suppressing metastatic potential in lung and breast tumor models. Pros1 deletion in myeloid cells led to increased lung metastasis, independent of primary tumor infiltration. PROS1-cKO bone marrow-derived macrophages (BMDMs) led to elevated TNF-α, IL-6, Nos2, and IL-10 via modulation of the Socs3/NF-κB pathway. Conditioned medium from cKO BMDMs enhanced EMT, ERK, AKT, and STAT3 activation within tumor cells and promoted IL-10-dependent invasion and survival. Macrophages isolated from metastatic lungs modulated T cell proliferation and function, as well as expression of costimulatory molecules on DCs in a PROS1-dependent manner. Inhibition of MERTK kinase activity blocked PROS1-mediated suppression of TNF-α and IL-6 but not IL-10. Overall, using lung and breast cancer models, we identified the PROS1/MERTK axis within BMDMs as a potent regulator of adaptive immune responses with a potential to suppress metastatic seeding and revealed IL-10 regulation by PROS1 to deviate from that of TNF-α and IL-6.


Asunto(s)
Proteínas de Unión al Calcio/inmunología , Interleucina-10/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Mamarias Experimentales/inmunología , Proteínas de Neoplasias/inmunología , Macrófagos Asociados a Tumores/inmunología , Animales , Proteínas de Unión al Calcio/genética , Femenino , Interleucina-10/genética , Interleucina-6/genética , Interleucina-6/inmunología , Neoplasias Pulmonares/genética , Neoplasias Mamarias Experimentales/genética , Ratones , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
9.
Curr Opin Immunol ; 51: 154-161, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29614426

RESUMEN

Myeloid derived suppressor cells (MDSCs) are immature myeloid cells characterized by diverse phenotypes and functions. They impair effector functions of immune cells and promote tumor growth, angiogenesis, and tissue damage. In pathologies characterized by chronic inflammation, MDSCs are arrested in their immature state and migrate from the bone marrow to the periphery and to the site of inflammation, where they mediate immunosuppression. When reaching new environments, which exhibit a different array of cytokines, chemokines, and pro-inflammatory mediators, MDSCs sense and adapt to the altered micro-environment by virtue of acquiring different suppressive features/functions that involve changing their cell fate, surface receptors, metabolism and intracellular as well as secreted molecules. This review summarizes some of the latest publications highlighting various layers of MDSC plasticity in relation to different pathologies. We discuss treatments capitalizing on MDSC plasticity aimed at combating MDSCs or manipulating their suppressive activity for improved therapy.


Asunto(s)
Biodiversidad , Plasticidad de la Célula , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Animales , Biomarcadores , Plasticidad de la Célula/inmunología , Terapia Combinada , Exosomas/metabolismo , Humanos , Redes y Vías Metabólicas , Neoplasias/etiología , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia , Fenotipo , Pronóstico , Transducción de Señal
10.
Cytometry B Clin Cytom ; 94(2): 211-218, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28188679

RESUMEN

BACKGROUND: Quantitative flow cytometry (QFCM) can be an important element within the developing toolbox for clinical diagnostics which relies on precise and rapid tests that provide a conclusive answer for physicians. The FC technology combines all of these features. Until recently, this imperative discipline was based on qualitative assessments of cell populations. However, due to the enormous advancement in FC technology, which allows the quantification of a number of antigens on cell surface and within the cells by units of median fluorescence intensity (MFI), this method becomes meaningful and fits the clinical needs. METHODS: On the basis of our experience in the field of quantitative FC, we wish to highlight some of the key concerns related to this methodology and suggest possible solutions for achieving uniform and standardized QFCM tests based on MFI. RESULTS: Several parameters are responsible for inter and intra laboratory variations. The standardization of quantitative FC relies on three major components; Samples and reagents handling, FC maintenance and data analysis. The use of specialized beads as a part of the routine calibration process lowers inter-test variability between different operators and different FC instruments. Similarly, the use of agreed biological controls contributes significantly to lowering test variability. CONCLUSIONS: The field of QFCM displays a significant part in the diagnostic clinical toolbox. We believe that the recommendations described herein can improve significantly the stability and accuracy of this method, thus assuring a more standardized cell analyses. © 2017 International Clinical Cytometry Society.


Asunto(s)
Citometría de Flujo/métodos , Antígenos/química , Calibración , Estudios de Evaluación como Asunto , Fluorescencia , Humanos , Indicadores y Reactivos/química , Estándares de Referencia
11.
Cancer Immunol Immunother ; 66(8): 1089-1101, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28674756

RESUMEN

Chronic inflammation arising in a diverse range of non-cancerous and cancerous diseases, dysregulates immunity and exposes patients to a variety of complications. These include immunosuppression, tissue damage, cardiovascular diseases and more. In cancer, chronic inflammation and related immunosuppression can directly support tumor growth and dramatically reduce the efficacies of traditional treatments, as well as novel immune-based therapies, which require a functional immune system. Nowadays, none of the immune biomarkers, regularly used by clinicians can sense a developing chronic inflammation, thus complications can only be detected upon their appearance. This review focuses on the necessity for such immune status biomarkers, which could predict complications prior to their appearance. Herein we bring examples for the use of cellular and molecular biomarkers in diagnosis, prognosis and follow-up of patients suffering from various cancers, for prediction of response to immune-based anti-cancer therapy and for prediction of cardiovascular disease in type 2 diabetes patients. Monitoring such biomarkers is expected to have a major clinical impact in addition to unraveling of the entangled complexity underlying dysregulated immunity in chronic inflammation. Thus, newly discovered biomarkers and those that are under investigation are projected to open a new era towards combating the silent damage induced by chronic inflammation.


Asunto(s)
Biomarcadores/metabolismo , Sistema Inmunológico , Mediadores de Inflamación/metabolismo , Inflamación/inmunología , Neoplasias/inmunología , Animales , Detección Precoz del Cáncer , Humanos , Tolerancia Inmunológica , Pruebas Inmunológicas , Inflamación/diagnóstico , Neoplasias/diagnóstico
12.
Cancer Immunol Immunother ; 65(7): 857-67, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27225641

RESUMEN

Chronic inflammation, typical of various diseases including cancer, is a "silent bomb within the body," leading to complications that are only evident in most cases upon their appearance, when disease is already deteriorated. Chronic inflammation is associated with accumulation of myeloid-derived suppressor cells (MDSCs), which lead to immunosuppression. MDSCs have numerous harmful effects as they support tumor initiation, tumor growth and spreading, which in turn, perpetuate the inflammatory and suppressive conditions, thus preventing anticancer responses. As the concept of the immune system combating many types of tumors was revived in recent years, immunotherapy has dramatically changed the view of cancer treatment, and numerous novel therapies have been developed and approved by the FDA. However, cumulative clinical data point at very limited success rates. It is most likely that the developing chronic inflammation and MDSC-induced immunosuppression interfere with responses to such treatments and hence are major obstacles in achieving higher response rates to immune-based therapies. Moreover, chemotherapies were shown to have adverse immunoregulatory effects, enhancing or decreasing MDSC levels and activity, thus affecting treatment success. Therefore, therapeutic manipulations of chronic inflammation and MDSCs during cancer development are likely to enhance efficacy of immune- and chemo-based treatments, switching chronic pro-cancer inflammatory environments to an anticancerous milieu. Based on the functional relevance of immune networking in tumors, it is critical to merge monitoring immune system biomarkers into the traditional patient's categorization and treatment regimens. This will provide new tools for clinical practice, allowing appropriate management of cancer patients toward a better-personalized medicine.


Asunto(s)
Inmunoterapia/métodos , Células Supresoras de Origen Mieloide/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Animales , Humanos , Células Supresoras de Origen Mieloide/patología , Neoplasias/patología , Microambiente Tumoral/inmunología
13.
Clin Cancer Res ; 22(23): 5661-5672, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27178742

RESUMEN

PURPOSE: High levels of circulating myeloid-derived suppressor cells (MDSCs) in various cancer types, including melanoma, were shown to correlate with poor survival. We investigated whether frequencies of circulating CD33+CD11b+HLA-DR- MDSCs could be used as immune system monitoring biomarkers to predict response and survival of patients with stage IV melanoma treated with anti-CTLA4 (ipilimumab) therapy. EXPERIMENTAL DESIGN: Peripheral blood samples from 56 patients and 50 healthy donors (HDs) were analyzed for CD33+CD11b+HLA-DR- MDSC percentage, NO-, and hROS levels by flow cytometry. We determined whether MDSC levels and suppressive features detected before anti-CTLA4 therapy correlate with the patients' response and overall survival (OS). RESULTS: Patients with melanoma had significantly higher levels of circulating CD33+CD11b+HLA-DR- MDSCs with suppressive phenotype when compared with HDs. Low levels of MDSCs before CTLA-4 therapy correlated with an objective clinical response, long-term survival, increased CD247 expression in T cells, and an improved clinical status. No predictive impact was observed for lactate dehydrogenase (LDH). Kaplan-Meier and log-rank tests performed on the 56 patients showed that the presence of more than 55.5% of circulating CD33+CD11b+ out of the HLA-DR- cells, were associated with significant short OS (P < 0.003), a median of 6.5 months, in comparison with the group showing lower MDSC frequencies, with a median survival of 15.6 months. CONCLUSIONS: Our study suggests the use of CD33+CD11b+HLA-DR- cells as a predictive and prognostic biomarker in patients with stage IV melanoma treated with anti-CTLA4 therapy. This monitoring system may aid in the development of combinatorial modalities, targeting the suppressive environment in conjunction with iplimumab, toward facilitating better disease outcomes. Clin Cancer Res; 22(23); 5661-72. ©2016 AACR.


Asunto(s)
Antígeno CD11b/sangre , Antígenos HLA-DR/sangre , Ipilimumab/uso terapéutico , Melanoma/sangre , Melanoma/tratamiento farmacológico , Células Mieloides/metabolismo , Lectina 3 Similar a Ig de Unión al Ácido Siálico/sangre , Biomarcadores de Tumor/sangre , Antígeno CTLA-4/sangre , Femenino , Humanos , Masculino , Melanoma/patología , Persona de Mediana Edad , Células Mieloides/efectos de los fármacos , Células Mieloides/patología , Estadificación de Neoplasias/métodos , Pronóstico , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Linfocitos T/patología
14.
Cell Rep ; 14(6): 1462-1476, 2016 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-26854224

RESUMEN

Factors linking inflammation and cancer are of great interest. We now report that the chromatin-targeting E3 ubiquitin ligase RNF20/RNF40, driving histone H2B monoubiquitylation (H2Bub1), modulates inflammation and inflammation-associated cancer in mice and humans. Downregulation of RNF20 and H2Bub1 favors recruitment of p65-containing nuclear factor κB (NF-κB) dimers over repressive p50 homodimers and decreases the heterochromatin mark H3K9me3 on a subset of NF-κB target genes to augment their transcription. Concordantly, RNF20(+/-) mice are predisposed to acute and chronic colonic inflammation and inflammation-associated colorectal cancer, with excessive myeloid-derived suppressor cells (MDSCs) that may quench antitumoral T cell activity. Notably, colons of human ulcerative colitis patients, as well as colorectal tumors, reveal downregulation of RNF20/RNF40 and H2Bub1 in both epithelium and stroma, supporting the clinical relevance of our tissue culture and mouse model findings.


Asunto(s)
Colitis Ulcerosa/genética , Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica , Histonas/genética , FN-kappa B/genética , Ubiquitina-Proteína Ligasas/genética , Animales , Cromatina/química , Cromatina/inmunología , Colitis Ulcerosa/complicaciones , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/patología , Colon/inmunología , Colon/patología , Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Histonas/inmunología , Humanos , Inflamación , Masculino , Ratones , Ratones Transgénicos , Células Mieloides/inmunología , Células Mieloides/patología , FN-kappa B/inmunología , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/patología , Transcripción Genética , Ubiquitina-Proteína Ligasas/inmunología , Ubiquitinación
15.
J Immunol ; 196(1): 156-67, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26608909

RESUMEN

Chronic inflammation is associated with immunosuppression and downregulated expression of the TCR CD247. In searching for new biomarkers that could validate the impaired host immune status under chronic inflammatory conditions, we discovered that sorting nexin 9 (SNX9), a protein that participates in early stages of clathrin-mediated endocytosis, is downregulated as well under such conditions. SNX9 expression was affected earlier than CD247 by the generated harmful environment, suggesting that it is a potential marker sensing the generated immunosuppressive condition. We found that myeloid-derived suppressor cells, which are elevated in the course of chronic inflammation, are responsible for the observed SNX9 reduced expression. Moreover, SNX9 downregulation is reversible, as its expression levels return to normal and immune functions are restored when the inflammatory response and/or myeloid-derived suppressor cells are neutralized. SNX9 downregulation was detected in numerous mouse models for pathologies characterized by chronic inflammation such as chronic infection (Leishmania donovani), cancer (melanoma and colorectal carcinoma), and an autoimmune disease (rheumatoid arthritis). Interestingly, reduced levels of SNX9 were also observed in blood samples from colorectal cancer patients, emphasizing the feasibility of its use as a diagnostic and prognostic biomarker sensing the host's immune status and inflammatory stage. Our new discovery of SNX9 as being regulated by chronic inflammation and its association with immunosuppression, in addition to the CD247 regulation under such conditions, show the global impact of chronic inflammation and the generated immune environment on different cellular pathways in a diverse spectrum of diseases.


Asunto(s)
Complejo CD3/biosíntesis , Huésped Inmunocomprometido/inmunología , Inflamación/inmunología , Células Mieloides/inmunología , Nexinas de Clasificación/biosíntesis , Animales , Artritis Reumatoide/inmunología , Biomarcadores de Tumor/sangre , Proliferación Celular , Células Cultivadas , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Inflamación/patología , Leishmania donovani/inmunología , Leishmaniasis Visceral/inmunología , Leishmaniasis Visceral/parasitología , Masculino , Melanoma/diagnóstico , Melanoma/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Nexinas de Clasificación/sangre , Células TH1/inmunología , Células Th2/inmunología
16.
Front Immunol ; 6: 523, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26528286

RESUMEN

Cancer development is dependent on intrinsic cellular changes as well as inflammatory factors in the tumor macro and microenvironment. The inflammatory milieu nourishes the tumor and contributes to cancer progression. Numerous studies, including ours, have demonstrated that the tumor microenvironment is immunosuppressive, impairing the anticancer immune responses. Chronic inflammation was identified as the key process responsible for this immunosuppression via induction of immature myeloid-derived suppressor cells (MDSCs). Upon a prolonged immune response, MDSCs are polarized toward immunosuppressive cells meant to control the exacerbated immune response. In cancer, the chronic inflammatory response renders the MDSCs harmful. Polarized MDSCs suppress T-cells and natural killer cells, as well as antigen-presenting cells, abrogating the beneficial immune response. These changes in the immunological milieu could also lead to high frequency of mutations, enhanced cancer cell stemness, and angiogenesis, directly supporting tumor initiation, growth, and spreading. The presence of MDSCs in cancer poses a serious obstacle in a variety of immune-based therapies, which rely on the stimulation of antitumor immune responses. Cumulative data, including our own, suggest that the selection of an appropriate and effective anticancer therapy must take into consideration the host's immune status as well as tumor-related parameters. Merging biomarkers for immune monitoring into the traditional patient's categorization and follow-up can provide new predictive and diagnostic tools to the clinical practice. Chronic inflammation and MDSCs could serve as novel targets for therapeutic interventions, which can be combined with conventional cancer treatments such as chemotherapy, radiotherapy, and cancer cell-targeted and immune-based therapies. Intervention in environmental and tumor-specific inflammatory mechanisms will allow better clinical management of cancer toward more efficient treatment.

17.
Diabetes Care ; 38(1): 113-8, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25368105

RESUMEN

OBJECTIVE: We have previously shown that chronic inflammation results in immunosuppression associated with CD247 downregulation in T lymphocytes. Type 2 diabetes mellitus (T2DM) is known to be associated with chronic inflammation. We therefore sought to examine CD247 expression levels in patients with T2DM and to assess whether it can serve as a diagnostic and prognostic biomarker for disease complications and outcomes. RESEARCH DESIGN AND METHODS: Peripheral blood samples from 75 T2DM patients and 40 healthy control subjects were collected and analyzed for the expression level of CD247 in T lymphocytes. Subjects with T2DM underwent a medical interview with physical examination and were followed for an additional average of 19.2 ± 0.9 months to determine the occurrence of major adverse disease end points. The relationship between the level of CD247 expression and disease status at the time of blood draw and the ability of the marker to identify future complications was evaluated. RESULTS: We observed a significant reduction in CD247 expression levels in T lymphocytes of T2DM patients when compared with healthy volunteers. CD247 downregulation was associated with disease severity, complications, and the occurrence of future cardiovascular events, suggesting its potential use not only as a diagnostic but also as a prognostic biomarker. CONCLUSIONS: Our results suggest the use of CD247 as a biomarker in diabetic patients for evaluating the state of chronic inflammation that contributes to morbidity and mortality in this disease and for the prediction of future cardiovascular events.


Asunto(s)
Biomarcadores/metabolismo , Complejo CD3/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Progresión de la Enfermedad , Inflamación/diagnóstico , Linfocitos T/metabolismo , Adulto , Anciano , Complejo CD3/genética , Enfermedades Cardiovasculares/complicaciones , Enfermedades Cardiovasculares/genética , Estudios de Casos y Controles , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/genética , Regulación hacia Abajo , Femenino , Voluntarios Sanos , Humanos , Inflamación/complicaciones , Inflamación/genética , Masculino , Persona de Mediana Edad
18.
Proc Natl Acad Sci U S A ; 111(49): 17582-7, 2014 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-25422452

RESUMEN

The inflamed tumor microenvironment plays a critical role in tumorigenesis. However, the mechanisms through which immune cells, particularly macrophages, promote tumorigenesis have only been partially elucidated, and the full scope of signaling pathways supplying macrophages with protumorigenic phenotypes still remain largely unknown. Here we report that germ-line absence of c-Jun N-terminal phosphorylation at serines 63 and 73 impedes inflammation-associated hepatocarcinogenesis, yet deleting c-Jun only in hepatocytes does not inhibit hepatocellular carcinoma (HCC) formation. Moreover, in human HCC-bearing livers, c-Jun phosphorylation is found in inflammatory cells, whereas it is mostly absent from malignant hepatocytes. Interestingly, macrophages in livers of mice with chronic hepatitis gradually switch their phenotype along the course of disease. Macrophage phenotype and density are dictated by c-Jun phosphorylation, in vitro and in vivo. Transition of macrophage phenotype, from antitumorigenic to protumorigenic, occurs before tumorigenesis, resulting in the production of various chemokines, including chemokine (C-C motif) ligand 17 (CCL17) and CCL22. Such signals, emanating from the liver microenvironment, direct the recruitment of regulatory T cells, which are known to facilitate HCC growth. Our findings identify c-Jun phosphorylation as a key mediator of macrophage education and point to the recruitment of immunosuppressive regulatory T cells as a possible protumorigenic mechanism.


Asunto(s)
Macrófagos/citología , Macrófagos/inmunología , Proteínas Proto-Oncogénicas c-jun/metabolismo , Animales , Apoptosis , Carcinoma Hepatocelular/metabolismo , Proliferación Celular , Quimiocinas/metabolismo , Regulación Neoplásica de la Expresión Génica , Hepatitis/metabolismo , Hepatocitos/citología , Humanos , Inmunidad Innata , Inflamación , Hígado/metabolismo , Neoplasias Hepáticas/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Fenotipo , Fosforilación , Pronóstico , Estructura Terciaria de Proteína , Microambiente Tumoral
19.
Cancer Res ; 74(21): 6022-35, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25209187

RESUMEN

Colorectal cancer is associated with chronic inflammation and immunosuppression mediated by myeloid-derived suppressor cells (MDSC). Although chemotherapy reduces tumor burden at early stages, it tends to have limited effect on a progressive disease, possibly due to adverse effects on the immune system in dictating disease outcome. Here, we show that patients with advanced colorectal cancer display enhanced MDSC levels and reduced CD247 expression and that some conventional colorectal cancer chemotherapy supports the immunosuppressive tumor microenvironment. A FOLFOX combined therapy reduced immunosuppression, whereas a FOLFIRI combined therapy enhanced immunosuppression. Mechanistic studies in a colorectal cancer mouse model revealed that FOLFIRI-like therapy including the drugs CPT11 and 5-fluorouracil (5FU) damaged host immunocompetence in a manner that limits treatment outcomes. CPT11 blocked MDSC apoptosis and myeloid cell differentiation, increasing MDSC immunosuppressive features and mouse mortality. In contrast, 5FU promoted immune recovery and tumor regression. Thus, CPT11 exhibited detrimental immunoregulatory effects that offset 5FU benefits when administered in combination. Our results highlight the importance of developing therapeutic regimens that can target both the immune system and tumor towards improved personalized treatments for colorectal cancer.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Fluorouracilo/administración & dosificación , Células Mieloides/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Apoptosis/efectos de los fármacos , Camptotecina/administración & dosificación , Camptotecina/análogos & derivados , Diferenciación Celular/efectos de los fármacos , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Humanos , Leucovorina/administración & dosificación , Ratones , Células Mieloides/inmunología , Compuestos Organoplatinos/administración & dosificación , Oxaliplatino , Resultado del Tratamiento , Microambiente Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Eur J Immunol ; 44(1): 58-68, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24185712

RESUMEN

TCR-mediated activation induces receptor microclusters that evolve to a defined immune synapse (IS). Many studies showed that actin polymerization and remodeling, which create a scaffold critical to IS formation and stabilization, are TCR mediated. However, the mechanisms controlling simultaneous TCR and actin dynamic rearrangement in the IS are yet not fully understood. Herein, we identify two novel TCR ζ-chain motifs, mediating the TCR's direct interaction with actin and inducing actin bundling. While T cells expressing the ζ-chain mutated in these motifs lack cytoskeleton (actin) associated (cska)-TCRs, they express normal levels of non-cska and surface TCRs as cells expressing wild-type ζ-chain. However, such mutant cells are unable to display activation-dependent TCR clustering, IS formation, expression of CD25/CD69 activation markers, or produce/secrete cytokine, effects also seen in the corresponding APCs. We are the first to show a direct TCR-actin linkage, providing the missing gap linking between TCR-mediated Ag recognition, specific cytoskeleton orientation toward the T-cell-APC interacting pole and long-lived IS maintenance.


Asunto(s)
Citoesqueleto/metabolismo , Sinapsis Inmunológicas/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Actinas/metabolismo , Secuencias de Aminoácidos/genética , Animales , Células Cultivadas , Citocinas/metabolismo , Femenino , Sinapsis Inmunológicas/inmunología , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Mutantes , Mutación/genética , Agregación de Receptores/genética , Receptores de Antígenos de Linfocitos T/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA