Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 132(9)2022 05 02.
Article in English | MEDLINE | ID: mdl-35316223

ABSTRACT

T cell immunoglobulin mucin domain-containing protein 3 (Tim-3) negatively regulates innate and adaptive immunity in cancer. To identify the mechanisms of Tim-3 in cancer immunity, we evaluated the effects of Tim-3 blockade in human and mouse melanoma. Here, we show that human programmed cell death 1-positive (PD-1+) Tim-3+CD8+ tumor-infiltrating lymphocytes (TILs) upregulate phosphatidylserine (PS), a receptor for Tim-3, and acquire cell surface myeloid markers from antigen-presenting cells (APCs) through transfer of membrane fragments called trogocytosis. Tim-3 blockade acted on Tim-3+ APCs in a PS-dependent fashion to disrupt the trogocytosis of activated tumor antigen-specific CD8+ T cells and PD-1+Tim-3+ CD8+ TILs isolated from patients with melanoma. Tim-3 and PD-1 blockades cooperated to disrupt trogocytosis of CD8+ TILs in 2 melanoma mouse models, decreasing tumor burden and prolonging survival. Deleting Tim-3 in dendritic cells but not in CD8+ T cells impeded the trogocytosis of CD8+ TILs in vivo. Trogocytosed CD8+ T cells presented tumor peptide-major histocompatibility complexes and became the target of fratricide T cell killing, which was reversed by Tim-3 blockade. Our findings have uncovered a mechanism Tim-3 uses to limit antitumor immunity.


Subject(s)
Hepatitis A Virus Cellular Receptor 2/immunology , Melanoma , Animals , CD8-Positive T-Lymphocytes , Hepatitis A Virus Cellular Receptor 2/genetics , Hepatitis A Virus Cellular Receptor 2/metabolism , Humans , Lymphocytes, Tumor-Infiltrating , Melanoma/pathology , Mice , Programmed Cell Death 1 Receptor , Trogocytosis
2.
Nat Med ; 28(3): 545-556, 2022 03.
Article in English | MEDLINE | ID: mdl-35228752

ABSTRACT

Ample evidence indicates that the gut microbiome is a tumor-extrinsic factor associated with antitumor response to anti-programmed cell death protein-1 (PD-1) therapy, but inconsistencies exist between published microbial signatures associated with clinical outcomes. To resolve this, we evaluated a new melanoma cohort, along with four published datasets. Time-to-event analysis showed that baseline microbiota composition was optimally associated with clinical outcome at approximately 1 year after initiation of treatment. Meta-analysis and other bioinformatic analyses of the combined data show that bacteria associated with favorable response are confined within the Actinobacteria phylum and the Lachnospiraceae/Ruminococcaceae families of Firmicutes. Conversely, Gram-negative bacteria were associated with an inflammatory host intestinal gene signature, increased blood neutrophil-to-lymphocyte ratio, and unfavorable outcome. Two microbial signatures, enriched for Lachnospiraceae spp. and Streptococcaceae spp., were associated with favorable and unfavorable clinical response, respectively, and with distinct immune-related adverse effects. Despite between-cohort heterogeneity, optimized all-minus-one supervised learning algorithms trained on batch-corrected microbiome data consistently predicted outcomes to programmed cell death protein-1 therapy in all cohorts. Gut microbial communities (microbiotypes) with nonuniform geographical distribution were associated with favorable and unfavorable outcomes, contributing to discrepancies between cohorts. Our findings shed new light on the complex interaction between the gut microbiome and response to cancer immunotherapy, providing a roadmap for future studies.


Subject(s)
Gastrointestinal Microbiome , Melanoma , Microbiota , Bacteria/genetics , Gastrointestinal Microbiome/genetics , Humans , Immunotherapy/adverse effects , Melanoma/drug therapy
3.
Neurology ; 98(3): e279-e290, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34911748

ABSTRACT

BACKGROUND AND OBJECTIVES: To the best of our knowledge, no comprehensive update of the descriptive epidemiology and trends of ischemic stroke has been released since Global Burden of Disease (GBD) 2017. Thus, our objective was to examine ischemic stroke burden at the global, regional, and national levels in terms of sex, age, and social development index (SDI). METHODS: Data were extracted from the GBD 2019 datasets. Estimated annual percentage changes (EAPCs) were calculated to assess the incidence rate, mortality, and disability-adjusted life-years (DALY) trends of ischemic stroke. Measures were stratified by sex, region, country, age, and SDI. RESULTS: The global age-standardized incidence rate (ASIR) of ischemic stroke decreased from 1990 to 2019, with an EAPC of -0.43 (95% confidence interval [CI] -0.54 to -0.32). High-middle and middle SDI regions had much higher ASIR, age-standardized death rate (ASDR), and age-standardized DALY rates due to ischemic stroke than other SDI regions. Regionally, East Asia had the highest ASIR of ischemic stroke in 2019 and the largest increase in the ASIR from 1990 to 2019. Nationally, Egypt (EAPC 1.40, 95% CI 1.27-1.52) and China (EAPC 1.10, 95% CI 1.00-1.20) had the most pronounced increases in the ASIR of ischemic stroke. Globally, there was an increase in ischemic stroke incidence with increasing age, especially in women 50 to 69 years of age or older. The global ASDR decreased from 1990 to 2019, with an EAPC of -1.63 (95% CI -1.72 to -1.53). The ASDR and age-standardized DALY rates increased most in southern sub-Saharan Africa, eastern sub-Saharan Africa, and southeast Asia. DISCUSSION: The ASIR, ASDR, and age-standardized DALY rates remained high in high-middle and middle SDI regions. East Asia, southern sub-Saharan Africa, eastern sub-Saharan Africa, and Southeast Asia had the greatest burden of ischemic stroke.


Subject(s)
Ischemic Stroke , Aged , China , Female , Global Burden of Disease , Global Health , Humans , Incidence , Middle Aged , Quality-Adjusted Life Years
5.
Front Immunol ; 12: 641188, 2021.
Article in English | MEDLINE | ID: mdl-33828552

ABSTRACT

Precisely controlled lymphocyte migration is critically required for immune surveillance and successful immune responses. Lymphocyte migration is strictly regulated by chemokines and chemokine receptors. Here we show that protein geranylgeranylation, a form of post-translational protein lipid modification, is required for chemokine receptor-proximal signaling. Mature thymocytes deficient for protein geranylgeranylation are impaired for thymus egress. Circulating mature T cells lacking protein geranylgeranylation fail to home to secondary lymphoid organs or to transmigrate in response to chemokines in vitro. Mechanistically, protein geranylgeranylation modifies the γ-subunits of the heterotrimeric small GTPases that are essential for chemokine receptor signaling. In addition, protein geranylgeranylation also promotes the differentiation of IL-17-producing T helper cells while inhibiting the differentiation of Foxp3+ regulatory T cells. Finally, mice with T cell lineage-specific deficiency of protein geranylgeranylation are resistant to experimental autoimmune encephalomyelitis induction. This study elucidated a critical role of protein geranylgeranylation in regulating T lymphocyte migration and function.


Subject(s)
Chemotaxis, Leukocyte/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Protein Prenylation/immunology , Receptors, Chemokine/immunology , Th17 Cells/immunology , Animals , Cell Differentiation/immunology , Mice , Multiple Sclerosis , Signal Transduction/immunology
6.
Science ; 371(6529): 595-602, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33542131

ABSTRACT

Anti-programmed cell death protein 1 (PD-1) therapy provides long-term clinical benefits to patients with advanced melanoma. The composition of the gut microbiota correlates with anti-PD-1 efficacy in preclinical models and cancer patients. To investigate whether resistance to anti-PD-1 can be overcome by changing the gut microbiota, this clinical trial evaluated the safety and efficacy of responder-derived fecal microbiota transplantation (FMT) together with anti-PD-1 in patients with PD-1-refractory melanoma. This combination was well tolerated, provided clinical benefit in 6 of 15 patients, and induced rapid and durable microbiota perturbation. Responders exhibited increased abundance of taxa that were previously shown to be associated with response to anti-PD-1, increased CD8+ T cell activation, and decreased frequency of interleukin-8-expressing myeloid cells. Responders had distinct proteomic and metabolomic signatures, and transkingdom network analyses confirmed that the gut microbiome regulated these changes. Collectively, our findings show that FMT and anti-PD-1 changed the gut microbiome and reprogrammed the tumor microenvironment to overcome resistance to anti-PD-1 in a subset of PD-1 advanced melanoma.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Drug Resistance, Neoplasm , Fecal Microbiota Transplantation , Melanoma/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Skin Neoplasms/therapy , CD8-Positive T-Lymphocytes/immunology , Gastrointestinal Microbiome , Humans , Interleukin-8/immunology , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/immunology , Myeloid Cells/immunology , Tumor Microenvironment/immunology
7.
Semin Immunol ; 49: 101436, 2020 06.
Article in English | MEDLINE | ID: mdl-33288379

ABSTRACT

T cells play a critical role in promoting tumor regression in both experimental models and humans. Yet, T cells that are chronically exposed to tumor antigen during cancer progression can become dysfunctional/exhausted and fail to induce tumor destruction. Such tumor-induced T cell dysfunction may occur via multiple mechanisms. In particular, immune checkpoint inhibitory receptors that are upregulated by tumor-infiltrating lymphocytes in many cancers limit T cell survival and function. Overcoming this inhibitory receptor-mediated T cell dysfunction has been a central focus of recent developments in cancer immunotherapy. Immunotherapies targeting inhibitory receptor pathways such as programmed cell death 1 (PD-1)/programmed death ligand 1 and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), alone or in combination, confer significant clinical benefits in multiple tumor types. However, many patients with cancer do not respond to immune checkpoint blockade, and dual PD-1/CTLA-4 blockade may cause serious adverse events, which limits its indications. Targeting novel non-redundant inhibitory receptor pathways contributing to tumor-induced T cell dysfunction in the tumor microenvironment may prove efficacious and non-toxic. This review presents preclinical and clinical findings supporting the roles of two key pathways-T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) and T cell immunoreceptor with Ig and ITIM domain (TIGIT)/CD226/CD96/CD112R-in cancer immunotherapy.


Subject(s)
Costimulatory and Inhibitory T-Cell Receptors/metabolism , Neoplasms/immunology , Neoplasms/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Biomarkers, Tumor , Humans , Immune Checkpoint Proteins/metabolism , Immunotherapy , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Molecular Targeted Therapy , Neoplasms/pathology , Neoplasms/therapy , Signal Transduction , T-Lymphocyte Subsets/drug effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
8.
Clin Cancer Res ; 26(20): 5520-5533, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32591463

ABSTRACT

PURPOSE: Natural killer (NK) cells play a critical role in tumor immunosurveillance. Multiple activating and inhibitory receptors (IR) regulate NK-cell-mediated tumor control. The IR T-cell immunoglobulin and ITIM domain (TIGIT) and its counter-receptor CD226 exert opposite effects on NK-cell-mediated tumor reactivity. EXPERIMENTAL DESIGN: We evaluated the frequency, phenotype, and functions of NK cells freshly isolated from healthy donors and patients with melanoma with multiparameter flow cytometry. We assessed TIGIT and CD226 cell surface expression and internalization upon binding to CD155. We evaluated the role of IL15 and TIGIT blockade in increasing NK-cell-mediated cytotoxicity in vitro and in two mouse models. RESULTS: NK cells are present at low frequencies in metastatic melanoma, are dysfunctional, and downregulate both TIGIT and CD226 expression. As compared with TIGIT- NK cells, TIGIT+ NK cells exhibit higher cytotoxic capacity and maturation, but paradoxically lower cytotoxicity against CD155+ MHC class I-deficient melanoma cells. Membrane bound CD155 triggers CD226 internalization and degradation, resulting in decreased NK-cell-mediated tumor reactivity. IL15 increases TIGIT and CD226 gene expression by tumor-infiltrating NK cells (TiNKs) and, together with TIGIT blockade, increases NK-cell-mediated melanoma cytotoxicity in vitro and decreases tumor metastasis in two mouse melanoma models. Specific deletion of TIGIT on transferred NK cells enhances the antimetastatic activity of IL15, while CD226 blockade decreases the effects of IL15 and TIGIT blockade. CONCLUSIONS: Our findings support the development of novel combinatorial immunotherapy with IL15 and TIGIT blockade to promote NK-cell-mediated destruction of MHC class I-deficient melanoma, which are refractory to CD8+ T-cell-mediated immunity.See related commentary by Pietra et al., p. 5274.


Subject(s)
Interleukin-15/pharmacology , Killer Cells, Natural/immunology , Melanoma/immunology , Receptors, Immunologic/genetics , Animals , Antigens, Differentiation, T-Lymphocyte/genetics , Antigens, Differentiation, T-Lymphocyte/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Interleukin-15/genetics , Killer Cells, Natural/drug effects , Melanoma/blood , Melanoma/genetics , Melanoma/pathology , Mice , Receptors, Immunologic/antagonists & inhibitors , Receptors, Virus/genetics
9.
Clin Immunol ; 183: 145-157, 2017 10.
Article in English | MEDLINE | ID: mdl-28844702

ABSTRACT

Ficolin-2 is an important serum complement lectin. Here, we describe novel findings indicating that serum ficolin-2 concentrations in multiple tumor patients are significantly lower than those in healthy donors. Administration of exogenous ficolin-2 or ficolin-A (a ficolin-2-like molecule in mouse), with only once, could remarkably inhibit the tumor cells growth in murine tumor models via early macrophages, dendritic cells (DCs) and CD8+ T cells, but not CD4+ T cells. Ficolin-A (FCN-A) knockout (KO) mice exhibits significantly increased tumor cell growth. Ficolin-2 induces macrophage activation, promotes M1 polarization and facilitates proliferation and antigen-specific cytotoxicity of CD8+ T cells. Ficolin-2 binds to Toll-like receptor 4 (TLR4) on macrophages and DCs and promotes their antigen-presenting abilities to CD8+ T cells. Our findings provide a new therapeutic strategy for tumors based on the triggering of immune-mediated antitumor effect by ficolin-2.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Lectins/immunology , Macrophage Activation/immunology , Macrophages/immunology , Neoplasms/blood , Adult , Aged , Animals , Breast Neoplasms/blood , Carcinoma, Hepatocellular/blood , Carcinoma, Lewis Lung , Cell Line, Tumor , Chemokine CCL5/immunology , Colorectal Neoplasms/blood , Dendritic Cells/immunology , Female , Humans , Interleukin-6/immunology , Lectins/blood , Lectins/genetics , Liver Neoplasms/blood , Lung Neoplasms/blood , Male , Mice , Mice, Knockout , Middle Aged , Nitric Oxide/immunology , Toll-Like Receptor 4/immunology , Tumor Necrosis Factor-alpha/immunology , Ficolins
10.
Tuberculosis (Edinb) ; 104: 95-106, 2017 05.
Article in English | MEDLINE | ID: mdl-28454656

ABSTRACT

Macrophages are the primary host target cells of Mycobacterium tuberculosis (M.tb). However, little is known about the changes of membrane glycopatterns of macrophages in response to M. tb infection. Using lectin microarrays we compared the differential expression of glycopatterns of macrophages upon stimulation with the heat-inactivated virulent M.tb H37Rv or attenuate M.tb H37Ra. We found that widespread alteration of macrophage membrane glycopatterns were induced by the heat-inactivated virulent M. tb H37Rv, as shown by the significantly changed binding abilities of 11 lectins (sugar binding proteins) among 40 lectins tested. The binding ability of the lectin ABA to macrophages showed the greatest increase after virulent M. tb H37Rv treatment, which suggests that the expression of N-acetyl-d-lactosamine (ABA binding ligand Galß1-3GalNAc, O-link glycan) is mainly increased on macrophages during virulent M.tb infection. Addition of ABA blocked the attachment/engulfment of M. tb H37Rv, but not H37Ra, to macrophages. Further, increased glycosylated CD44, one of ABA-binding glycoproteins on macrophages, was identified by pull-down assays with ABA-agarose, followed by mass spectrometry and western blotting. ABA directly binds with Galß1-3GalNAc-glycosylated CD44 on macrophage, and inhibits M. tb mannose-capped lipoarabinomannan (ManLAM) binding to glycosylated CD44. Moreover, ABA increases IL-6, but reduces IL-10 production of ManLAM-treated macrophages and inhibits M. tb H37Rv-induced necrosis in macrophages. Our study will help to reveal the mechanism of pathogenicity and virulence of M. tb from a new perspective and provide a potential new diagnostic and therapeutic strategy for tuberculosis based on glycopatterns, ABA and its ligand Galß1-3GalNAc-glycosylated CD44 target molecule on macrophage.


Subject(s)
Glycoproteins/metabolism , Macrophages, Peritoneal/metabolism , Membrane Proteins/metabolism , Mycobacterium tuberculosis/pathogenicity , Polysaccharides/metabolism , Tuberculosis/metabolism , Animals , Biomarkers/metabolism , Disease Models, Animal , Female , Glycosylation , Host-Pathogen Interactions , Humans , Hyaluronan Receptors/metabolism , Macrophages, Peritoneal/microbiology , Mass Spectrometry , Mice, Inbred C57BL , Microarray Analysis , THP-1 Cells , Tuberculosis/microbiology , Virulence
11.
Virol Sin ; 30(4): 249-60, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26220728

ABSTRACT

Human ficolin-2 is an important lectin complement pathway activator that is secreted from liver cells and has been implicated as an anti-infection innate immune molecule. However, the role of ficolin-2 protein and its dynamic changes over the course of and in the prognosis of chronic hepatitis B (CHB) and hepatocellular carcinoma (HCC) remain unclear. In this study, we analyzed ficolin-2 protein expression in a cohort of individuals with CHB infection, HCC and cirrhosis. A sandwich enzyme-linked immunosorbent assay (ELISA) method was used to measure serum ficolin-2 concentrations. Ficolin-2 expression in liver tissues was detected by immunohistochemical staining. Serum ficolin-2 concentrations in CHB patients were significantly higher than in healthy controls and HBV carriers. After 48 weeks of routine amelioration liver function treatment, serum ficolin-2 concentrations decreased and were positively correlated with favorable alanine aminotransferase (ALT), HBV DNA and HBeAg-seroconversion outcomes. Interestingly, we observed much lower expression of serum and intrahepatic ficolin-2 in HCC and cirrhosis compared with healthy controls. Our findings suggest that serum and intrahepatic ficolin-2 levels may be considered one of the indicators for the response of chronic HBV infection, HCC and cirrhosis.


Subject(s)
Carcinoma, Hepatocellular/pathology , Hepatitis B, Chronic/pathology , Lectins/blood , Liver Cirrhosis/pathology , Liver Neoplasms/pathology , Serum/chemistry , Adult , Enzyme-Linked Immunosorbent Assay , Female , Hepatitis B, Chronic/complications , Humans , Immunohistochemistry , Male , Young Adult , Ficolins
12.
Virol Sin ; 29(1): 25-32, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24452543

ABSTRACT

Ficolins are serum complement lectins, with a structure similar to mannose-binding lectin (MBL) and lung surfactant protein (SP)-A and SP-D. Ficolins activate the lectin complement system and play important roles in host innate immunity. Ficolins are members of the collectin family of proteins, which act as pattern recognition receptors (PRRs). They are soluble oligomeric defense proteins with lectin-like activity, and are able to recognize pathogen-associated molecular patterns (PAMPs), which are carbohydrate molecules on the surface of pathogens, and of apoptotic, necrotic, and malignant cells. Upon binding to their specific PAMPs, ficolins may trigger activation of the immune system either (1) by initiating activation of complement via the lectin pathway, (2) by a primitive type of opsonophagocytosis, or (3) by stimulating secretion of the inflammatory cytokines interferon (IFN)-Γ, interleukin (IL)-17, IL-6, and tumor necrosis factor (TNF)-α, and production of nitric oxide (NO) by macrophages, thus limiting the infection and concurrently orchestrating the subsequent adaptive immune response. Recently, a number of reports have shown that dysfunction or abnormal expression of ficolins may play crucial roles in viral and bacterial diseases and in inflammation. This review summarizes the reports on the roles of ficolins in the infectious diseases, and provides insight into ficolins as novel innate immune therapeutic options to treat these diseases.


Subject(s)
Communicable Diseases/immunology , Lectins/immunology , Lectins/metabolism , Receptors, Pattern Recognition/immunology , Receptors, Pattern Recognition/metabolism , Carbohydrate Metabolism , Complement System Proteins/metabolism , Cytokines/metabolism , Humans , Nitric Oxide/metabolism , Phagocytosis , Protein Binding , Ficolins
SELECTION OF CITATIONS
SEARCH DETAIL
...