Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
2.
Nature ; 630(8018): 968-975, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38867043

ABSTRACT

Obesity is a leading risk factor for progression and metastasis of many cancers1,2, yet can in some cases enhance survival3-5 and responses to immune checkpoint blockade therapies, including anti-PD-1, which targets PD-1 (encoded by PDCD1), an inhibitory receptor expressed on immune cells6-8. Although obesity promotes chronic inflammation, the role of the immune system in the obesity-cancer connection and immunotherapy remains unclear. It has been shown that in addition to T cells, macrophages can express PD-19-12. Here we found that obesity selectively induced PD-1 expression on tumour-associated macrophages (TAMs). Type I inflammatory cytokines and molecules linked to obesity, including interferon-γ, tumour necrosis factor, leptin, insulin and palmitate, induced macrophage PD-1 expression in an mTORC1- and glycolysis-dependent manner. PD-1 then provided negative feedback to TAMs that suppressed glycolysis, phagocytosis and T cell stimulatory potential. Conversely, PD-1 blockade increased the level of macrophage glycolysis, which was essential for PD-1 inhibition to augment TAM expression of CD86 and major histocompatibility complex I and II molecules and ability to activate T cells. Myeloid-specific PD-1 deficiency slowed tumour growth, enhanced TAM glycolysis and antigen-presentation capability, and led to increased CD8+ T cell activity with a reduced level of markers of exhaustion. These findings show that obesity-associated metabolic signalling and inflammatory cues cause TAMs to induce PD-1 expression, which then drives a TAM-specific feedback mechanism that impairs tumour immune surveillance. This may contribute to increased cancer risk yet improved response to PD-1 immunotherapy in obesity.


Subject(s)
Neoplasms , Obesity , Programmed Cell Death 1 Receptor , Tumor-Associated Macrophages , Animals , Female , Humans , Male , Mice , Antigen Presentation/drug effects , B7-2 Antigen/antagonists & inhibitors , B7-2 Antigen/immunology , B7-2 Antigen/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Glycolysis/drug effects , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Lymphocyte Activation , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mice, Inbred C57BL , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Obesity/immunology , Obesity/metabolism , Phagocytosis/drug effects , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/drug effects
3.
Nat Immunol ; 24(1): 55-68, 2023 01.
Article in English | MEDLINE | ID: mdl-36581713

ABSTRACT

The inhibitory receptor PD-1 suppresses T cell activation by recruiting the phosphatase SHP-2. However, mice with a T-cell-specific deletion of SHP-2 do not have improved antitumor immunity. Here we showed that mice with conditional targeting of SHP-2 in myeloid cells, but not in T cells, had diminished tumor growth. RNA sequencing (RNA-seq) followed by gene set enrichment analysis indicated the presence of polymorphonuclear myeloid-derived suppressor cells and tumor-associated macrophages (TAMs) with enriched gene expression profiles of enhanced differentiation, activation and expression of immunostimulatory molecules. In mice with conditional targeting of PD-1 in myeloid cells, which also displayed diminished tumor growth, TAMs had gene expression profiles enriched for myeloid differentiation, activation and leukocyte-mediated immunity displaying >50% overlap with enriched profiles of SHP-2-deficient TAMs. In bone marrow, GM-CSF induced the phosphorylation of PD-1 and recruitment of PD-1-SHP-2 to the GM-CSF receptor. Deletion of SHP-2 or PD-1 enhanced GM-CSF-mediated phosphorylation of the transcription factors HOXA10 and IRF8, which regulate myeloid differentiation and monocytic-moDC lineage commitment, respectively. Thus, SHP-2 and PD-1-SHP-2 signaling restrained myelocyte differentiation resulting in a myeloid landscape that suppressed antitumor immunity.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor , Neoplasms , Animals , Mice , Cell Differentiation , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Myeloid Cells , Programmed Cell Death 1 Receptor/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Signal Transduction
4.
Semin Cancer Biol ; 86(Pt 2): 187-201, 2022 11.
Article in English | MEDLINE | ID: mdl-35985559

ABSTRACT

During the past decade there has been a revolution in cancer therapeutics by the emergence of antibody-based and cell-based immunotherapies that modulate immune responses against tumors. These new therapies have extended and improved the therapeutic efficacy of chemo-radiotherapy and have offered treatment options to patients who are no longer responding to these classic anti-cancer treatments. Unfortunately, tumor eradication and long-lasting responses are observed in a small fraction of patients, whereas the majority of patients respond only transiently. These outcomes indicate that the maximum potential of immunotherapy has not been reached due to incomplete knowledge of the cellular and molecular mechanisms that guide the development of successful anti-tumor immunity and its failure. In this review, we discuss recent discoveries about the immune cellular composition of the tumor microenvironment (TME) and the role of key signaling mechanisms that compromise the function of immune cells leading to cancer immune escape.


Subject(s)
Neoplasms , Tumor Microenvironment , Humans , Immunotherapy , Neoplasms/pathology , Signal Transduction , Immunologic Factors
5.
Nat Immunol ; 23(8): 1148-1156, 2022 08.
Article in English | MEDLINE | ID: mdl-35879449

ABSTRACT

Long recognized as an evolutionarily ancient cell type involved in tissue homeostasis and immune defense against pathogens, macrophages are being re-discovered as regulators of several diseases, including cancer. Tumor-associated macrophages (TAMs) represent the most abundant innate immune population in the tumor microenvironment (TME). Macrophages are professional phagocytic cells of the hematopoietic system specializing in the detection, phagocytosis and destruction of bacteria and other harmful micro-organisms, apoptotic cells and metabolic byproducts. In contrast to these healthy macrophage functions, TAMs support cancer cell growth and metastasis and mediate immunosuppressive effects on the adaptive immune cells of the TME. Cancer is one of the most potent insults on macrophage physiology, inducing changes that are intimately linked with disease progression. In this Review, we outline hallmarks of TAMs and discuss the emerging mechanisms that contribute to their pathophysiological adaptations and the vulnerabilities that provide attractive targets for therapeutic exploitation in cancer.


Subject(s)
Neoplasms , Tumor Microenvironment , Disease Progression , Humans , Macrophages , Phagocytosis
6.
Nat Immunol ; 23(6): 971-984, 2022 06.
Article in English | MEDLINE | ID: mdl-35624211

ABSTRACT

Glioblastoma (GBM) is an incurable primary malignant brain cancer hallmarked with a substantial protumorigenic immune component. Knowledge of the GBM immune microenvironment during tumor evolution and standard of care treatments is limited. Using single-cell transcriptomics and flow cytometry, we unveiled large-scale comprehensive longitudinal changes in immune cell composition throughout tumor progression in an epidermal growth factor receptor-driven genetic mouse GBM model. We identified subsets of proinflammatory microglia in developing GBMs and anti-inflammatory macrophages and protumorigenic myeloid-derived suppressors cells in end-stage tumors, an evolution that parallels breakdown of the blood-brain barrier and extensive growth of epidermal growth factor receptor+ GBM cells. A similar relationship was found between microglia and macrophages in patient biopsies of low-grade glioma and GBM. Temozolomide decreased the accumulation of myeloid-derived suppressor cells, whereas concomitant temozolomide irradiation increased intratumoral GranzymeB+ CD8+T cells but also increased CD4+ regulatory T cells. These results provide a comprehensive and unbiased immune cellular landscape and its evolutionary changes during GBM progression.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Animals , Brain Neoplasms/metabolism , ErbB Receptors , Glioblastoma/metabolism , Humans , Mice , Sequence Analysis, RNA , Single-Cell Analysis , Temozolomide/therapeutic use , Tumor Microenvironment/genetics
7.
Biomed J ; 45(2): 289-298, 2022 04.
Article in English | MEDLINE | ID: mdl-34601137

ABSTRACT

Leukocytes, the leading players of immune system, are involved in innate and adaptive immune responses. Leukocyte adhesion to endothelial cells during transmigration or to antigen presenting cells during T cell activation, requires integrin activation through a process termed inside-out integrin signaling. In hematopoietic cells, Rap1 and its downstream effector RIAM (Rap1-interacting adaptor molecule) form a cornerstone for inside-out integrin activation. The Rap1/RIAM pathway is involved in signal integration for activation, actin remodeling and cytoskeletal reorganization in T cells, as well as in myeloid cell differentiation and function. RIAM is instrumental for phagocytosis, a process requiring particle recognition, cytoskeletal remodeling and membrane protrusion for engulfment and digestion. In the present review, we discuss the structural and molecular properties of RIAM and the recent discoveries regarding the functional role of the Rap1/RIAM module in hematopoietic cells.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Membrane Proteins/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Cell Adhesion , Endothelial Cells/metabolism , Humans , Integrins/metabolism , Membrane Proteins/metabolism
8.
J Vis Exp ; (177)2021 11 16.
Article in English | MEDLINE | ID: mdl-34866628

ABSTRACT

The respiratory tract is in direct contact with the outside environment and requires a precisely regulated immune system to provide protection while suppressing unwanted reactions to environmental antigens. Lungs host several populations of innate and adaptive immune cells that provide immune surveillance but also mediate protective immune responses. These cells, which keep the healthy pulmonary immune system in balance, also participate in several pathological conditions such as asthma, infections, autoimmune diseases, and cancer. Selective expression of surface and intracellular proteins provides unique immunophenotypic properties to the immune cells of the lung. Consequently, flow cytometry has an instrumental role in the identification of such cell populations during steady-state and pathological conditions. This paper presents a protocol that describes a consistent and reproducible method to identify the immune cells that reside in the lungs of healthy mice under steady-state conditions. However, this protocol can also be used to identify changes in these cell populations in various disease models to help identify disease-specific changes in the lung immune landscape.


Subject(s)
Immunity, Innate , Lung , Animals , Flow Cytometry/methods , Immunophenotyping , Mice
9.
Immunometabolism ; 3(2)2021.
Article in English | MEDLINE | ID: mdl-33927895

ABSTRACT

Metabolism is a common cellular feature. Cancer creates a suppressive microenvironment resulting in inactivation of antigen-specific T cells by metabolic reprogramming. Development of approaches that enhance and sustain physiologic properties of T cell metabolism to prevent T cell inactivation and promote effector function in the tumor microenvironment is an urgent need for improvement of cell-based cancer immunotherapies.

10.
Metabolism ; 114: 154338, 2021 01.
Article in English | MEDLINE | ID: mdl-32791172

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are fatty acid-activated transcription factors of nuclear hormone receptor superfamily that regulate energy metabolism. Currently, three PPAR subtypes have been identified: PPARα, PPARγ, and PPARß/δ. PPARα and PPARδ are highly expressed in oxidative tissues and regulate genes involved in substrate delivery and oxidative phosphorylation (OXPHOS) and regulation of energy homeostasis. In contrast, PPARγ is more important in lipogenesis and lipid synthesis, with highest expression levels in white adipose tissue (WAT). In addition to tissues regulating whole body energy homeostasis, PPARs are expressed in immune cells and have an emerging critical role in immune cell differentiation and fate commitment. In this review, we discuss the actions of PPARs in the function of the innate and the adaptive immune system and their implications in immune-mediated inflammatory conditions.


Subject(s)
Immunity/physiology , Inflammation/metabolism , Lipid Metabolism/physiology , Peroxisome Proliferator-Activated Receptors/metabolism , T-Lymphocytes/metabolism , Animals , Humans
11.
Sci Immunol ; 5(43)2020 01 03.
Article in English | MEDLINE | ID: mdl-31901074

ABSTRACT

PD-1, a T cell checkpoint receptor and target of cancer immunotherapy, is also expressed on myeloid cells. The role of myeloid-specific versus T cell-specific PD-1 ablation on antitumor immunity has remained unclear because most studies have used either PD-1-blocking antibodies or complete PD-1 KO mice. We generated a conditional allele, which allowed myeloid-specific (PD-1f/fLysMcre) or T cell-specific (PD-1f/fCD4cre) targeting of Pdcd1 gene. Compared with T cell-specific PD-1 ablation, myeloid cell-specific PD-1 ablation more effectively decreased tumor growth. We found that granulocyte/macrophage progenitors (GMPs), which accumulate during cancer-driven emergency myelopoiesis and give rise to myeloid-derived suppressor cells (MDSCs), express PD-1. In tumor-bearing PD-1f/fLysMcre but not PD-1f/fCD4cre mice, accumulation of GMP and MDSC was prevented, whereas systemic output of effector myeloid cells was increased. Myeloid cell-specific PD-1 ablation induced an increase of T effector memory cells with improved functionality and mediated antitumor protection despite preserved PD-1 expression in T cells. In PD-1-deficient myeloid progenitors, growth factors driving emergency myelopoiesis induced increased metabolic intermediates of glycolysis, pentose phosphate pathway, and TCA cycle but, most prominently, elevated cholesterol. Because cholesterol is required for differentiation of inflammatory macrophages and DC and promotes antigen-presenting function, our findings indicate that metabolic reprogramming of emergency myelopoiesis and differentiation of effector myeloid cells might be a key mechanism of antitumor immunity mediated by PD-1 blockade.


Subject(s)
Colonic Neoplasms/immunology , Melanoma/immunology , Myeloid Cells/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , Cell Differentiation , Cell Line, Tumor , Female , Male , Mice, Inbred C57BL , Mice, Transgenic , Programmed Cell Death 1 Receptor/genetics
12.
Curr Med Chem ; 25(17): 1954-1967, 2018.
Article in English | MEDLINE | ID: mdl-29189120

ABSTRACT

BACKGROUND: Glioma is a heterogeneous, highly complicated central nervous system (CNS) tumor with uncertain mechanism of initiation and progression, resulting in an unfavorable outcome. An extended network of cytokines is recognized as a major regulator of glioma pathogenesis, either promoting or inhibiting glioma progression based on their type and specificity. Interleukin-8 (IL-8) has been revealed as a critical regulator of CNS function and development with participation in many CNS disorders including gliomas. OBJECTIVE: The aim of the present review is to address the role of IL-8 in glioma pathogenesis focusing on the implicated molecular pathways as well as on its potential targeting for glioma therapy. METHODS AND RESULTS: PubMed-Medline, SCOPUS, and Google Scholar databases were searched for pre-clinical and clinical studies related to IL-8 implication in gliomagenesis and IL-8 targeting strategies for gliomas. Literature data indicate that IL-8 participates in glioma angiogenesis and cell migration and it can serve as a potential biomarker, for early diagnosis, follow-up and response to therapy. CONCLUSION: Several promising approaches that target directly or indirectly IL-8 effects in gliomas are currently in progress while more-in-depth studies are needed to validate its biomarker role and elucidate the underlying molecular mechanisms.


Subject(s)
Glioma/metabolism , Interleukin-8/metabolism , Animals , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor , Carcinogenesis/metabolism , Cell Movement/drug effects , Down-Regulation , Glioma/drug therapy , Humans , Interleukin-8/antagonists & inhibitors , Interleukin-8/genetics , Neovascularization, Pathologic/chemically induced
13.
Oncotarget ; 7(51): 85624-85640, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-27793053

ABSTRACT

Polycomb group proteins regulate chromatin structure and have an important regulatory role on gene expression in various cell types. Two polycomb group complexes (Polycomb repressive complex 1 (PRC1) and 2 (PRC2)) have been identified in mammalian cells. Both PRC1 and PRC2 compact chromatin, and also catalyze histone modifications. PRC1 mediates monoubiquitination of histone H2A, whereas PRC2 catalyzes methylation of histone H3 on lysine 27. These alterations of histones can lead to altered gene expression patterns by regulating chromatin structure. Numerous studies have highlighted the role of the PRC2 catalytic component enhancer of zeste homolog 2 (EZH2) in neoplastic development and progression, and EZH2 mutations have been identified in various malignancies. Through modulating the expression of critical genes, EZH2 is actively involved in fundamental cellular processes such as cell cycle progression, cell proliferation, differentiation and apoptosis. In addition to cancer cells, EZH2 also has a decisive role in the differentiation and function of T effector and T regulatory cells. In this review we summarize the recent progress regarding the role of EZH2 in human malignancies, highlight the molecular mechanisms by which EZH2 aberrations promote the pathogenesis of cancer, and discuss the anti-tumor effects of EZH2 targeting via activating direct anti-cancer mechanisms and anti-tumor immunity.


Subject(s)
DNA Methylation , Enhancer of Zeste Homolog 2 Protein/genetics , Epigenesis, Genetic , Neoplasms/genetics , Tumor Escape , Animals , Antineoplastic Agents/therapeutic use , Chromatin Assembly and Disassembly , DNA Methylation/drug effects , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/immunology , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/immunology , Signal Transduction , Tumor Escape/drug effects
14.
Front Immunol ; 7: 346, 2016.
Article in English | MEDLINE | ID: mdl-27648006

ABSTRACT

[This corrects the article on p. 172 in vol. 7, PMID: 27199994.].

15.
Cytokine ; 71(2): 377-84, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25458967

ABSTRACT

Glioma, a neuroglia originated malignancy, consists of one of the most aggressive primary tumors of the central nervous system with poor prognosis and lack of efficient treatment strategy. Cytokines have been implicated in several stages of glioma progression, participating in tumor onset, growth enhancement, angiogenesis and aggressiveness. Interestingly, cytokines have also the ability to inhibit glioma growth upon specific regulation or interplay with other molecules. This review addresses the dual role of major cytokines implicated in glioma pathology, pointing toward promising therapeutic approaches.


Subject(s)
Brain Neoplasms/physiopathology , Cytokines/metabolism , Gene Expression Regulation, Neoplastic , Glioma/physiopathology , Interleukin-18/metabolism , Animals , Apoptosis , Biomarkers/metabolism , Disease Progression , Epigenesis, Genetic , Gene Expression Regulation , Genetic Therapy/methods , Humans , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Microglia/pathology , Neovascularization, Pathologic , Prognosis , Transforming Growth Factor beta/metabolism
16.
Expert Opin Ther Targets ; 17(12): 1395-403, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24024908

ABSTRACT

INTRODUCTION: Osteoprotegerin (OPG) acts as a soluble decoy receptor for the bone marrow stroma cell-derived and osteoblast-derived receptor activator of nuclear factor-kB ligand (RANKL), thus regulating the RANK-mediated osteoclastogenesis and osteoclast-mediated bone resorption at the metastatic niche of cancer in skeleton. AREAS COVERED: This article discusses the 'key' role of OPG expression during the early events of cancer cell invasion into the bone matrix and the subsequent events underlying the formation of osteoblastic metastasis, a unique event observed in human prostate cancer biology. EXPERT OPINION: Understanding the cellular and molecular events implicated in bone metastasis can facilitate designing new therapeutic strategies for targeting early and/or late events in the metastasis processes. The RANKL/RANK/OPG pathway is a key regulator of pathological bone metabolism in metastatic sites. Targeted manipulation of these molecules may provide sustainable antitumor responses.


Subject(s)
Bone Neoplasms/metabolism , Osteoprotegerin/metabolism , Prostatic Neoplasms/metabolism , Animals , Bone Neoplasms/secondary , Humans , Male , Osteoblasts/metabolism , Prostatic Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...