Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Rep Med ; 5(1): 101378, 2024 01 16.
Article in English | MEDLINE | ID: mdl-38232704

ABSTRACT

A study by Régnier et al. shows that Flt3-ligand (FL) levels program two distinct mechanisms of anti-tumor immunity. Low FL levels allow recruitment of T effectors and T cell-mediated responses whereas high FL levels support recruitment of classical dendritic cells (cDC) and natural killer (NK) cells and NK-mediated anti-tumor responses.


Subject(s)
Killer Cells, Natural , Neoplasms , Humans , T-Lymphocytes , Neoplasms/drug therapy , Lymphocyte Activation
2.
Front Immunol ; 14: 1197364, 2023.
Article in English | MEDLINE | ID: mdl-37342323

ABSTRACT

During the past decade, there has been a revolution in cancer therapeutics by the emergence of antibody-based immunotherapies that modulate immune responses against tumors. These therapies have offered treatment options to patients who are no longer responding to classic anti-cancer therapies. By blocking inhibitory signals mediated by surface receptors that are naturally upregulated during activation of antigen-presenting cells (APC) and T cells, predominantly PD-1 and its ligand PD-L1, as well as CTLA-4, such blocking agents have revolutionized cancer treatment. However, breaking these inhibitory signals cannot be selectively targeted to the tumor microenvironment (TME). Since the physiologic role of these inhibitory receptors, known as immune checkpoints (IC) is to maintain peripheral tolerance by preventing the activation of autoreactive immune cells, IC inhibitors (ICI) induce multiple types of immune-related adverse effects (irAEs). These irAEs, together with the natural properties of ICs as gatekeepers of self-tolerance, have precluded the use of ICI in patients with pre-existing autoimmune diseases (ADs). However, currently accumulating data indicates that ICI might be safely administered to such patients. In this review, we discuss mechanisms of well established and newly recognized irAEs and evolving knowledge from the application of ICI therapies in patients with cancer and pre-existing ADs.


Subject(s)
Antineoplastic Agents, Immunological , Autoimmune Diseases , Drug-Related Side Effects and Adverse Reactions , Neoplasms , Humans , Antineoplastic Agents, Immunological/therapeutic use , Neoplasms/drug therapy , Neoplasms/pathology , Immunotherapy/adverse effects , Antibodies, Monoclonal/therapeutic use , Drug-Related Side Effects and Adverse Reactions/etiology , Autoimmune Diseases/etiology , Tumor Microenvironment
3.
Cancer Immunol Res ; 11(5): 629-645, 2023 05 03.
Article in English | MEDLINE | ID: mdl-36881002

ABSTRACT

The composition of the tumor immune microenvironment (TIME) is considered a key determinant of patients' response to immunotherapy. The mechanisms underlying TIME formation and development over time are poorly understood. Glioblastoma (GBM) is a lethal primary brain cancer for which there are no curative treatments. GBMs are immunologically heterogeneous and impervious to checkpoint blockade immunotherapies. Utilizing clinically relevant genetic mouse models of GBM, we identified distinct immune landscapes associated with expression of EGFR wild-type and mutant EGFRvIII cancer driver mutations. Over time, accumulation of polymorphonuclear myeloid-derived suppressor cells (PMN-MDSC) was more pronounced in EGFRvIII-driven GBMs and was correlated with resistance to PD-1 and CTLA-4 combination checkpoint blockade immunotherapy. We determined that GBM-secreted CXCL1/2/3 and PMN-MDSC-expressed CXCR2 formed an axis regulating output of PMN-MDSCs from the bone marrow leading to systemic increase in these cells in the spleen and GBM tumor-draining lymph nodes. Pharmacologic targeting of this axis induced a systemic decrease in the numbers of PMN-MDSC, facilitated responses to PD-1 and CTLA-4 combination checkpoint blocking immunotherapy, and prolonged survival in mice bearing EGFRvIII-driven GBM. Our results uncover a relationship between cancer driver mutations, TIME composition, and sensitivity to checkpoint blockade in GBM and support the stratification of patients with GBM for checkpoint blockade therapy based on integrated genotypic and immunologic profiles.


Subject(s)
Brain Neoplasms , Glioblastoma , Myeloid-Derived Suppressor Cells , Animals , Mice , Glioblastoma/therapy , Glioblastoma/drug therapy , CTLA-4 Antigen/genetics , CTLA-4 Antigen/metabolism , Programmed Cell Death 1 Receptor , Cell Line, Tumor , Immunotherapy , Mutation , Tumor Microenvironment/genetics , Brain Neoplasms/genetics , Brain Neoplasms/therapy
4.
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
5.
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
6.
Bone ; 158: 116354, 2022 05.
Article in English | MEDLINE | ID: mdl-35150909

ABSTRACT

INTRODUCTION: GnRH-analogs induce bone loss. We aimed to investigate the effects of goserelin-induced menstrual cessation (MC) and subsequent menstrual restoration (MR) on bone metabolism (BM). METHODS: In this prospective cohort study, premenopausal women (PMW) with histologically verified endometriosis (n = 21) received goserelin monthly for 6 months (6 m) resulting in MC and were followed up for another 6 m after MR (12 m). Age- and BMI-matched healthy PMW (n = 20) served as controls for bone mineral density (BMD) measurements. The primary endpoint was changes in lumbar spine (LS)-BMD at 6 m and 12 m; Secondary endpoints were changes in femoral neck (FN)-BMD, bone turnover markers (P1NP and CΤx), sclerostin, and expression of bone-related circulating microRNAs (miRNAs) at 6 m and 12 m. RESULTS: Goserelin-induced MC reduced LS- and FN-BMD at 6 m (both p < 0.001). From 6 m to 12 m, LS-BMD increased (p < 0.001) but remained below baseline values (p = 0.012), whereas FN-BMD remained stable (p = 1.000). CTx and P1NP levels increased at 6 m (both p < 0.001) and decreased at 12 m (p < 0.001 and p = 0.013, respectively), while CTx (p = 1.000) alone and not P1NP (p = 0.020) returned to baseline. Sclerostin levels did not change. Relative expression of miRNAs targeting RUNX 2 and beta-catenin was significantly downregulated at 6 m compared to baseline (p < 0.001), while the expression of miRNAs targeting osteoblast and osteoclast function at both directions demonstrated a robust increase (up to 400fold) at 12 m (p < 0.001). CONCLUSIONS: Six months of goserelin-induced MC lead to significant bone loss associated with increased bone turnover and changes in the expression of bone-related miRNAs, changes that are only partially reversed at 6 m after MR.


Subject(s)
Endometriosis , MicroRNAs , Biomarkers , Bone Density , Bone Remodeling , Endometriosis/drug therapy , Female , Humans , Menstrual Cycle , MicroRNAs/genetics , Prospective Studies
7.
Gut ; 71(7): 1399-1411, 2022 07.
Article in English | MEDLINE | ID: mdl-34548339

ABSTRACT

OBJECTIVE: Tissue-resident memory T cells (TRM) are vital immune sentinels that provide protective immunity. While hepatic CD8+ TRM have been well described, little is known about the location, phenotype and function of CD4+ TRM. DESIGN: We used multiparametric flow cytometry, histological assessment and novel human tissue coculture systems to interrogate the ex vivo phenotype, function and generation of the intrahepatic CD4+ T-cell compartment. We also used leukocytes isolated from human leukocyte antigen (HLA)-disparate liver allografts to assess long-term retention. RESULTS: Hepatic CD4+ T cells were delineated into three distinct populations based on CD69 expression: CD69-, CD69INT and CD69HI. CD69HICD4+ cells were identified as tissue-resident CD4+ T cells on the basis of their exclusion from the circulation, phenotypical profile (CXCR6+CD49a+S1PR1-PD-1+) and long-term persistence within the pool of donor-derived leukcoocytes in HLA-disparate liver allografts. CD69HICD4+ T cells produced robust type 1 polyfunctional cytokine responses on stimulation. Conversely, CD69INTCD4+ T cells represented a more heterogenous population containing cells with a more activated phenotype, a distinct chemokine receptor profile (CX3CR1+CXCR3+CXCR1+) and a bias towards interleukin-4 production. While CD69INTCD4+ T cells could be found in the circulation and lymph nodes, these cells also formed part of the long-term resident pool, persisting in HLA-mismatched allografts. Notably, frequencies of CD69INTCD4+ T cells correlated with necroinflammatory scores in chronic hepatitis B infection. Finally, we demonstrated that interaction with hepatic epithelia was sufficient to generate CD69INTCD4+ T cells, while additional signals from the liver microenvironment were required to generate liver-resident CD69HICD4+ T cells. CONCLUSIONS: High and intermediate CD69 expressions mark human hepatic CD4+ TRM and a novel functionally distinct recirculating population, respectively, both shaped by the liver microenvironment to achieve diverse immunosurveillance.


Subject(s)
CD4-Positive T-Lymphocytes , Liver , CD8-Positive T-Lymphocytes , Cytokines/immunology , Humans , Immunologic Memory , Liver/immunology , Monitoring, Immunologic
8.
Viruses ; 13(6)2021 05 28.
Article in English | MEDLINE | ID: mdl-34071712

ABSTRACT

Alphavirus non-structural proteins 1-4 (nsP1, nsP2, nsP3, and nsP4) are known to be crucial for alphavirus RNA replication and translation. To date, nsP3 has been demonstrated to mediate many virus-host protein-protein interactions in several fundamental alphavirus mechanisms, particularly during the early stages of replication. However, the molecular pathways and proteins networks underlying these mechanisms remain poorly described. This is due to the low genetic sequence homology of the nsP3 protein among the alphavirus species, especially at its 3' C-terminal domain, the hypervariable domain (HVD). Moreover, the nsP3 HVD is almost or completely intrinsically disordered and has a poor ability to form secondary structures. Evolution in the nsP3 HVD region allows the alphavirus to adapt to vertebrate and insect hosts. This review focuses on the putative roles and functions of indel, repetition, and duplication events that have occurred in the alphavirus nsP3 HVD, including characterization of the differences and their implications for specificity in the context of virus-host interactions in fundamental alphavirus mechanisms, which have thus directly facilitated the evolution, adaptation, viability, and re-emergence of these viruses.


Subject(s)
Alphavirus/genetics , Evolution, Molecular , Host-Pathogen Interactions , Viral Nonstructural Proteins/genetics , Alphavirus/metabolism , Animals , Binding Sites , Cell Line , Humans , Mice , Viral Nonstructural Proteins/metabolism , Virus Replication
9.
Methods Mol Biol ; 1591: 195-214, 2017.
Article in English | MEDLINE | ID: mdl-28349484

ABSTRACT

The liver is the largest internal organ and filters around 3 pints of blood per minute. This continuous flux of blood should not be confused with rapid egress of lymphocytes through the liver; this organ has intricate corridors of specialized sinusoidal spaces, ensuring that immune cells decelerate to shear flow rates, and providing ample opportunities to interact with parenchymal cells. Migration has been intricately linked to T cell function; it is therefore important to study liver T cell biology into context within the liver microenvironment. Here we discuss the highly organized architecture of liver-resident cells (sinusoidal endothelia, Kupffer cells, stellate cells/myofibroblasts, and biliary and hepatic epithelia) and showcase basic, multicellular, and complex systems to model T cell migration through the human liver microenvironment in vitro and ex vivo.


Subject(s)
Cell Movement/immunology , Liver/immunology , Parenchymal Tissue/immunology , T-Lymphocytes/immunology , Cellular Microenvironment/immunology , Endothelium/immunology , Epithelial Cells/immunology , Hepatic Stellate Cells/immunology , Humans , Kupffer Cells/immunology , Myofibroblasts/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...