Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 185(11): 1924-1942.e23, 2022 05 26.
Article in English | MEDLINE | ID: mdl-35525247

ABSTRACT

For many solid malignancies, lymph node (LN) involvement represents a harbinger of distant metastatic disease and, therefore, an important prognostic factor. Beyond its utility as a biomarker, whether and how LN metastasis plays an active role in shaping distant metastasis remains an open question. Here, we develop a syngeneic melanoma mouse model of LN metastasis to investigate how tumors spread to LNs and whether LN colonization influences metastasis to distant tissues. We show that an epigenetically instilled tumor-intrinsic interferon response program confers enhanced LN metastatic potential by enabling the evasion of NK cells and promoting LN colonization. LN metastases resist T cell-mediated cytotoxicity, induce antigen-specific regulatory T cells, and generate tumor-specific immune tolerance that subsequently facilitates distant tumor colonization. These effects extend to human cancers and other murine cancer models, implicating a conserved systemic mechanism by which malignancies spread to distant organs.


Subject(s)
Lymph Nodes , Melanoma , Animals , Immune Tolerance , Immunotherapy , Lymphatic Metastasis/pathology , Melanoma/pathology , Mice
2.
Immunity ; 57(6): 1289-1305.e9, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38772366

ABSTRACT

Adipose tissue group 2 innate lymphoid cells (ILC2s) help maintain metabolic homeostasis by sustaining type 2 immunity and promoting adipose beiging. Although impairment of the ILC2 compartment contributes to obesity-associated insulin resistance, the underlying mechanisms have not been elucidated. Here, we found that ILC2s in obese mice and humans exhibited impaired liver kinase B1 (LKB1) activation. Genetic ablation of LKB1 disrupted ILC2 mitochondrial metabolism and suppressed ILC2 responses, resulting in exacerbated insulin resistance. Mechanistically, LKB1 deficiency induced aberrant PD-1 expression through activation of NFAT, which in turn enhanced mitophagy by suppressing Bcl-xL expression. Blockade of PD-1 restored the normal functions of ILC2s and reversed obesity-induced insulin resistance in mice. Collectively, these data present the LKB1-PD-1 axis as a promising therapeutic target for the treatment of metabolic disease.


Subject(s)
Adipose Tissue , Homeostasis , Insulin Resistance , Lymphocytes , Mitochondria , Obesity , Programmed Cell Death 1 Receptor , Protein Serine-Threonine Kinases , Animals , Insulin Resistance/immunology , Programmed Cell Death 1 Receptor/metabolism , Mice , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Mitochondria/metabolism , Humans , Adipose Tissue/metabolism , Adipose Tissue/immunology , Obesity/immunology , Obesity/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , AMP-Activated Protein Kinases/metabolism , Mice, Inbred C57BL , Mice, Knockout , Immunity, Innate , Male , Mitophagy/immunology , AMP-Activated Protein Kinase Kinases
3.
Cell ; 168(3): 487-502.e15, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28111070

ABSTRACT

Immune responses involve coordination across cell types and tissues. However, studies in cancer immunotherapy have focused heavily on local immune responses in the tumor microenvironment. To investigate immune activity more broadly, we performed an organism-wide study in genetically engineered cancer models using mass cytometry. We analyzed immune responses in several tissues after immunotherapy by developing intuitive models for visualizing single-cell data with statistical inference. Immune activation was evident in the tumor and systemically shortly after effective therapy was administered. However, during tumor rejection, only peripheral immune cells sustained their proliferation. This systemic response was coordinated across tissues and required for tumor eradication in several immunotherapy models. An emergent population of peripheral CD4 T cells conferred protection against new tumors and was significantly expanded in patients responding to immunotherapy. These studies demonstrate the critical impact of systemic immune responses that drive tumor rejection.


Subject(s)
Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocyte Subsets/immunology , Animals , B7-H1 Antigen/antagonists & inhibitors , Bone Marrow/immunology , Cell Proliferation , Disease Models, Animal , Female , Humans , Immune Tolerance , Killer Cells, Natural/immunology , Lymphocyte Activation , Lymphoid Tissue/immunology , Male , Melanoma/immunology , Melanoma/therapy , Mice, Inbred BALB C , Mice, Inbred C57BL , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/therapy , Tumor Microenvironment
4.
Nature ; 626(7999): 635-642, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38297127

ABSTRACT

Type 2 diabetes mellitus is a major risk factor for hepatocellular carcinoma (HCC). Changes in extracellular matrix (ECM) mechanics contribute to cancer development1,2, and increased stiffness is known to promote HCC progression in cirrhotic conditions3,4. Type 2 diabetes mellitus is characterized by an accumulation of advanced glycation end-products (AGEs) in the ECM; however, how this affects HCC in non-cirrhotic conditions is unclear. Here we find that, in patients and animal models, AGEs promote changes in collagen architecture and enhance ECM viscoelasticity, with greater viscous dissipation and faster stress relaxation, but not changes in stiffness. High AGEs and viscoelasticity combined with oncogenic ß-catenin signalling promote HCC induction, whereas inhibiting AGE production, reconstituting the AGE clearance receptor AGER1 or breaking AGE-mediated collagen cross-links reduces viscoelasticity and HCC growth. Matrix analysis and computational modelling demonstrate that lower interconnectivity of AGE-bundled collagen matrix, marked by shorter fibre length and greater heterogeneity, enhances viscoelasticity. Mechanistically, animal studies and 3D cell cultures show that enhanced viscoelasticity promotes HCC cell proliferation and invasion through an integrin-ß1-tensin-1-YAP mechanotransductive pathway. These results reveal that AGE-mediated structural changes enhance ECM viscoelasticity, and that viscoelasticity can promote cancer progression in vivo, independent of stiffness.


Subject(s)
Carcinoma, Hepatocellular , Disease Progression , Elasticity , Extracellular Matrix , Liver Cirrhosis , Liver Neoplasms , Animals , Humans , beta Catenin/metabolism , Carcinoma, Hepatocellular/complications , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Collagen/chemistry , Collagen/metabolism , Computer Simulation , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Extracellular Matrix/metabolism , Glycation End Products, Advanced/metabolism , Integrin beta1/metabolism , Liver Neoplasms/complications , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Neoplasm Invasiveness , Viscosity , YAP-Signaling Proteins/metabolism , Liver Cirrhosis/complications , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology
5.
Immunity ; 47(6): 1037-1050.e6, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29221729

ABSTRACT

Given the limited efficacy of clinical approaches that rely on ex vivo generated dendritic cells (DCs), it is imperative to design strategies that harness specialized DC subsets in situ. This requires delineating the expression of surface markers by DC subsets among individuals and tissues. Here, we performed a multiparametric phenotypic characterization and unbiased analysis of human DC subsets in blood, tonsil, spleen, and skin. We uncovered previously unreported phenotypic heterogeneity of human cDC2s among individuals, including variable expression of functional receptors such as CD172a. We found marked differences in DC subsets localized in blood and lymphoid tissues versus skin, and a striking absence of the newly discovered Axl+ DCs in the skin. Finally, we evaluated the capacity of anti-receptor monoclonal antibodies to deliver vaccine components to skin DC subsets. These results offer a promising path for developing DC subset-specific immunotherapies that cannot be provided by transcriptomic analysis alone.


Subject(s)
Antigens, Differentiation/immunology , Biological Variation, Individual , Dendritic Cells/immunology , Phenotype , Proto-Oncogene Proteins/immunology , Receptor Protein-Tyrosine Kinases/immunology , Receptors, Immunologic/immunology , Skin/immunology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacokinetics , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation/genetics , Biomarkers/analysis , Cancer Vaccines/administration & dosage , Cancer Vaccines/biosynthesis , Cytophotometry/methods , Dendritic Cells/cytology , Female , Gene Expression , Humans , Immunophenotyping , Immunotherapy , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Organ Specificity , Palatine Tonsil/cytology , Palatine Tonsil/immunology , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/deficiency , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Immunologic/genetics , Skin/cytology , Spleen/cytology , Spleen/immunology , Axl Receptor Tyrosine Kinase
6.
Nat Methods ; 19(6): 759-769, 2022 06.
Article in English | MEDLINE | ID: mdl-35654951

ABSTRACT

Advances in multiplexed in situ imaging are revealing important insights in spatial biology. However, cell type identification remains a major challenge in imaging analysis, with most existing methods involving substantial manual assessment and subjective decisions for thousands of cells. We developed an unsupervised machine learning algorithm, CELESTA, which identifies the cell type of each cell, individually, using the cell's marker expression profile and, when needed, its spatial information. We demonstrate the performance of CELESTA on multiplexed immunofluorescence images of colorectal cancer and head and neck squamous cell carcinoma (HNSCC). Using the cell types identified by CELESTA, we identify tissue architecture associated with lymph node metastasis in HNSCC, and validate our findings in an independent cohort. By coupling our spatial analysis with single-cell RNA-sequencing data on proximal sections of the same specimens, we identify cell-cell crosstalk associated with lymph node metastasis, demonstrating the power of CELESTA to facilitate identification of clinically relevant interactions.


Subject(s)
Head and Neck Neoplasms , Cohort Studies , Humans , Lymphatic Metastasis , Squamous Cell Carcinoma of Head and Neck
7.
Immunity ; 45(3): 641-655, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27590114

ABSTRACT

Although all-trans-retinoic acid (atRA) is a key regulator of intestinal immunity, its role in colorectal cancer (CRC) is unknown. We found that mice with colitis-associated CRC had a marked deficiency in colonic atRA due to alterations in atRA metabolism mediated by microbiota-induced intestinal inflammation. Human ulcerative colitis (UC), UC-associated CRC, and sporadic CRC specimens have similar alterations in atRA metabolic enzymes, consistent with reduced colonic atRA. Inhibition of atRA signaling promoted tumorigenesis, whereas atRA supplementation reduced tumor burden. The benefit of atRA treatment was mediated by cytotoxic CD8(+) T cells, which were activated due to MHCI upregulation on tumor cells. Consistent with these findings, increased colonic expression of the atRA-catabolizing enzyme, CYP26A1, correlated with reduced frequencies of tumoral cytotoxic CD8(+) T cells and with worse disease prognosis in human CRC. These results reveal a mechanism by which microbiota drive colon carcinogenesis and highlight atRA metabolism as a therapeutic target for CRC.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Colorectal Neoplasms/immunology , Microbiota/immunology , Tretinoin/metabolism , Animals , CD8-Positive T-Lymphocytes/metabolism , Carcinogenesis/immunology , Colon/immunology , Colon/metabolism , Colorectal Neoplasms/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Retinoic Acid 4-Hydroxylase/metabolism , Signal Transduction/immunology , Up-Regulation/immunology
8.
Nature ; 567(7746): 56-60, 2019 03.
Article in English | MEDLINE | ID: mdl-30814731

ABSTRACT

The cytokine interferon-γ (IFNγ) is a central coordinator of innate and adaptive immunity, but its highly pleiotropic actions have diminished its prospects for use as an immunotherapeutic agent. Here, we took a structure-based approach to decoupling IFNγ pleiotropy. We engineered an affinity-enhanced variant of the ligand-binding chain of the IFNγ receptor IFNγR1, which enabled us to determine the crystal structure of the complete hexameric (2:2:2) IFNγ-IFNγR1-IFNγR2 signalling complex at 3.25 Å resolution. The structure reveals the mechanism underlying deficits in IFNγ responsiveness in mycobacterial disease syndrome resulting from a T168N mutation in IFNγR2, which impairs assembly of the full signalling complex. The topology of the hexameric complex offers a blueprint for engineering IFNγ variants to tune IFNγ receptor signalling output. Unexpectedly, we found that several partial IFNγ agonists exhibited biased gene-expression profiles. These biased agonists retained the ability to induce upregulation of major histocompatibility complex class I antigen expression, but exhibited impaired induction of programmed death-ligand 1 expression in a wide range of human cancer cell lines, offering a route to decoupling immunostimulatory and immunosuppressive functions of IFNγ for therapeutic applications.


Subject(s)
Drug Design , Interferon-gamma/agonists , Interferon-gamma/immunology , Receptors, Interferon/chemistry , Receptors, Interferon/metabolism , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/genetics , Cell Line, Tumor , Drug Partial Agonism , Gene Expression Profiling , Gene Expression Regulation/drug effects , Histocompatibility Antigens Class I/biosynthesis , Histocompatibility Antigens Class I/genetics , Humans , Interferon-gamma/chemistry , Interferon-gamma/genetics , Ligands , Models, Molecular , Mutation , Mycobacterium Infections/genetics , Mycobacterium Infections/immunology , Protein Stability , Receptors, Interferon/genetics , Signal Transduction , Structure-Activity Relationship , Interferon gamma Receptor
9.
Int J Mol Sci ; 24(1)2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36614196

ABSTRACT

Large numbers of neutrophils infiltrate tumors and comprise a notable component of the inflammatory tumor microenvironment. While it is established that tumor cells exhibit the Warburg effect for energy production, the contribution of the neutrophil metabolic state to tumorigenesis is unknown. Here, we investigated whether neutrophil infiltration and metabolic status promotes tumor progression in an orthotopic mouse model of pancreatic ductal adenocarcinoma (PDAC). We observed a large increase in the proportion of neutrophils in the blood and tumor upon orthotopic transplantation. Intriguingly, these tumor-infiltrating neutrophils up-regulated glycolytic factors and hypoxia-inducible factor 1-alpha (HIF-1α) expression compared to neutrophils from the bone marrow and blood of the same mouse. This enhanced glycolytic signature was also observed in human PDAC tissue samples. Strikingly, neutrophil-specific deletion of HIF-1α (HIF-1αΔNφ) significantly reduced tumor burden and improved overall survival in orthotopic transplanted mice, by converting the pro-tumorigenic neutrophil phenotype to an anti-tumorigenic phenotype. This outcome was associated with elevated reactive oxygen species production and activated natural killer cells and CD8+ cytotoxic T cells compared to littermate control mice. These data suggest a role for HIF-1α in neutrophil metabolism, which could be exploited as a target for metabolic modulation in cancer.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Animals , Mice , Neutrophils/metabolism , Cell Line, Tumor , Mice, Knockout , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/pathology , Carcinogenesis , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Tumor Microenvironment/genetics , Pancreatic Neoplasms
10.
J Neuroinflammation ; 18(1): 199, 2021 Sep 12.
Article in English | MEDLINE | ID: mdl-34511110

ABSTRACT

BACKGROUND: Patients with chronic inflammatory disorders such as inflammatory bowel disease frequently experience neurological complications including epilepsy, depression, attention deficit disorders, migraines, and dementia. However, the mechanistic basis for these associations is unknown. Given that many patients are unresponsive to existing medications or experience debilitating side effects, novel therapeutics that target the underlying pathophysiology of these conditions are urgently needed. METHODS: Because intestinal disorders such as inflammatory bowel disease are robustly associated with neurological symptoms, we used three different mouse models of colitis to investigate the impact of peripheral inflammatory disease on the brain. We assessed neuronal hyperexcitability, which is associated with many neurological symptoms, by measuring seizure threshold in healthy and colitic mice. We profiled the neuroinflammatory phenotype of colitic mice and used depletion and neutralization assays to identify the specific mediators responsible for colitis-induced neuronal hyperexcitability. To determine whether our findings in murine models overlapped with a human phenotype, we performed gene expression profiling, pathway analysis, and deconvolution on microarray data from hyperexcitable human brain tissue from patients with epilepsy. RESULTS: We observed that murine colitis induces neuroinflammation characterized by increased pro-inflammatory cytokine production, decreased tight junction protein expression, and infiltration of monocytes and neutrophils into the brain. We also observed sustained neuronal hyperexcitability in colitic mice. Colitis-induced neuronal hyperexcitability was ameliorated by neutrophil depletion or TNFα blockade. Gene expression profiling of hyperexcitable brain tissue resected from patients with epilepsy also revealed a remarkably similar pathology to that seen in the brains of colitic mice, including neutrophil infiltration and high TNFα expression. CONCLUSIONS: Our results reveal neutrophils and TNFα as central regulators of neuronal hyperexcitability of diverse etiology. Thus, there is a strong rationale for evaluating anti-inflammatory agents, including clinically approved TNFα inhibitors, for the treatment of neurological and psychiatric symptoms present in, and potentially independent of, a diagnosed inflammatory disorder.


Subject(s)
Colitis , Epilepsy , Animals , Brain/metabolism , Colitis/chemically induced , Disease Models, Animal , Epilepsy/complications , Humans , Mice , Neurons , Neutrophils/metabolism , Tumor Necrosis Factor-alpha/metabolism
11.
Nature ; 521(7550): 99-104, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25924063

ABSTRACT

Whereas cancers grow within host tissues and evade host immunity through immune-editing and immunosuppression, tumours are rarely transmissible between individuals. Much like transplanted allogeneic organs, allogeneic tumours are reliably rejected by host T cells, even when the tumour and host share the same major histocompatibility complex alleles, the most potent determinants of transplant rejection. How such tumour-eradicating immunity is initiated remains unknown, although elucidating this process could provide the basis for inducing similar responses against naturally arising tumours. Here we find that allogeneic tumour rejection is initiated in mice by naturally occurring tumour-binding IgG antibodies, which enable dendritic cells (DCs) to internalize tumour antigens and subsequently activate tumour-reactive T cells. We exploited this mechanism to treat autologous and autochthonous tumours successfully. Either systemic administration of DCs loaded with allogeneic-IgG-coated tumour cells or intratumoral injection of allogeneic IgG in combination with DC stimuli induced potent T-cell-mediated antitumour immune responses, resulting in tumour eradication in mouse models of melanoma, pancreas, lung and breast cancer. Moreover, this strategy led to eradication of distant tumours and metastases, as well as the injected primary tumours. To assess the clinical relevance of these findings, we studied antibodies and cells from patients with lung cancer. T cells from these patients responded vigorously to autologous tumour antigens after culture with allogeneic-IgG-loaded DCs, recapitulating our findings in mice. These results reveal that tumour-binding allogeneic IgG can induce powerful antitumour immunity that can be exploited for cancer immunotherapy.


Subject(s)
Antibodies, Neoplasm/immunology , Antigens, Neoplasm/immunology , Dendritic Cells/immunology , Immunoglobulin G/immunology , Neoplasms/immunology , T-Lymphocytes/immunology , Animals , Antibodies, Neoplasm/administration & dosage , CD40 Antigens/metabolism , Disease Models, Animal , Female , Immunoglobulin G/administration & dosage , Isoantibodies/administration & dosage , Isoantibodies/immunology , Lymphocyte Activation/immunology , Male , Mice , Neoplasm Metastasis , Neoplasm Transplantation/immunology , Neoplasms/pathology , Receptors, IgG/immunology , Tumor Necrosis Factor-alpha/immunology
12.
Int J Mol Sci ; 22(12)2021 Jun 14.
Article in English | MEDLINE | ID: mdl-34198548

ABSTRACT

Inflammation in the tumor microenvironment has been shown to promote disease progression in pancreatic ductal adenocarcinoma (PDAC); however, the role of macrophage metabolism in promoting inflammation is unclear. Using an orthotopic mouse model of PDAC, we demonstrate that macrophages from tumor-bearing mice exhibit elevated glycolysis. Macrophage-specific deletion of Glucose Transporter 1 (GLUT1) significantly reduced tumor burden, which was accompanied by increased Natural Killer and CD8+ T cell activity and suppression of the NLRP3-IL1ß inflammasome axis. Administration of mice with a GLUT1-specific inhibitor reduced tumor burden, comparable with gemcitabine, the current standard-of-care. In addition, we observe that intra-tumoral macrophages from human PDAC patients exhibit a pronounced glycolytic signature, which reliably predicts poor survival. Our data support a key role for macrophage metabolism in tumor immunity, which could be exploited to improve patient outcomes.


Subject(s)
Adenocarcinoma/pathology , Carcinoma, Pancreatic Ductal/pathology , Cytoprotection , Glycolysis , Macrophages/metabolism , Pancreatic Neoplasms/pathology , Adenocarcinoma/immunology , Animals , Carcinoma, Pancreatic Ductal/immunology , Cell Proliferation/drug effects , Cytoprotection/drug effects , Drug Resistance, Neoplasm/drug effects , Glucose Transporter Type 1/metabolism , Glycolysis/drug effects , Humans , Hydroxybenzoates/pharmacology , Inflammation/pathology , Interleukin-1beta/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Macrophages/drug effects , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pancreatic Neoplasms/immunology , Survival Analysis , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Tumor Burden/drug effects , Pancreatic Neoplasms
13.
BMC Bioinformatics ; 21(1): 346, 2020 Aug 10.
Article in English | MEDLINE | ID: mdl-32778050

ABSTRACT

BACKGROUND: While technological advances have made it possible to profile the immune system at high resolution, translating high-throughput data into knowledge of immune mechanisms has been challenged by the complexity of the interactions underlying immune processes. Tools to explore the immune network are critical for better understanding the multi-layered processes that underlie immune function and dysfunction, but require a standardized network map of immune interactions. To facilitate this we have developed ImmunoGlobe, a manually curated intercellular immune interaction network extracted from Janeway's Immunobiology textbook. RESULTS: ImmunoGlobe is the first graphical representation of the immune interactome, and is comprised of 253 immune system components and 1112 unique immune interactions with detailed functional and characteristic annotations. Analysis of this network shows that it recapitulates known features of the human immune system and can be used uncover novel multi-step immune pathways, examine species-specific differences in immune processes, and predict the response of immune cells to stimuli. ImmunoGlobe is publicly available through a user-friendly interface at www.immunoglobe.org and can be downloaded as a computable graph and network table. CONCLUSION: While the fields of proteomics and genomics have long benefited from network analysis tools, no such tool yet exists for immunology. ImmunoGlobe provides a ground truth immune interaction network upon which such tools can be built. These tools will allow us to predict the outcome of complex immune interactions, providing mechanistic insight that allows us to precisely modulate immune responses in health and disease.


Subject(s)
Cell Communication , Data Curation , Extracellular Space/metabolism , Immune System/metabolism , Protein Interaction Maps , Software , Systems Biology , Animals , Humans , Mice , Models, Immunological
14.
Nature ; 565(7741): 573-574, 2019 01.
Article in English | MEDLINE | ID: mdl-30683936
15.
Proc Natl Acad Sci U S A ; 114(8): 1988-1993, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28167780

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are known mainly for their secretion of type I IFN upon viral encounter. We describe a CD2hiCD5+CD81+ pDC subset, distinguished by prominent dendrites and a mature phenotype, in human blood, bone marrow, and tonsil, which can be generated from CD34+ progenitors. These CD2hiCD5+CD81+ cells express classical pDC markers, as well as the toll-like receptors that enable conventional pDCs to respond to viral infection. However, their gene expression profile is distinct, and they produce little or no type I IFN upon stimulation with CpG oligonucleotides, likely due to their diminished expression of IFN regulatory factor 7. A similar population of CD5+CD81+ pDCs is present in mice and also does not produce type I IFN after CpG stimulation. In contrast to conventional CD5-CD81- pDCs, human CD5+CD81+ pDCs are potent stimulators of B-cell activation and antibody production and strong inducers of T-cell proliferation and Treg formation. These findings reveal the presence of a discrete pDC population that does not produce type I IFN and yet mediates important immune functions previously attributed to all pDCs.


Subject(s)
B-Lymphocytes/physiology , Cell Differentiation , Cell Proliferation/physiology , Dendritic Cells/physiology , Lymphocyte Activation , T-Lymphocytes/physiology , Animals , CD2 Antigens/metabolism , CD5 Antigens/metabolism , Cell Separation , Flow Cytometry , Humans , Interferon Type I/metabolism , Male , Mice , Mice, Inbred C57BL , Oligodeoxyribonucleotides/immunology , Tetraspanin 28/metabolism , Toll-Like Receptors/metabolism
16.
J Autoimmun ; 105: 102290, 2019 12.
Article in English | MEDLINE | ID: mdl-31202617

ABSTRACT

The critical role of sphingosine-1-phosphate (S1P) signaling in lymphocyte trafficking is well recognized, however, the contribution of myeloid cell-S1P signaling in neuroimmunity is less well understood. We previously reported that C57BL/6J mice harboring phosphorylation defective S1P receptor 1 (S1P1) (with mutated serines in the carboxyl terminus, leading to impaired receptor internalization) [S1P1(S5A)] developed severe, TH17-dominant experimental autoimmune encephalomyelitis. In this study, we demonstrate that S1P1-mediated TH17 polarization is not an intrinsic T cell effect, but dependent on sustained S1P1 signaling in myeloid cells. First, utilizing the S1P1(S5A) mice in the EAE model, we observed that S1P1 activated and enhanced antigen presentation function in myeloid cells. Second, sequential phosphorylation of STAT3 occurred in dendritic cells, monocytes, and macrophages/microglia during neuroinflammation. Third, we show that pro-inflammatory (CD45hiCD11b+Ly6Chi) monocytes contribute to TH17 differentiation and neuroinflammation by regulating IL-6 expression. Finally, results from experiments utilizing myeloid cell-specific S1P1 overexpression (S1pr1f/stop/f:LysMCre) mice demonstrate that myeloid cell S1P1 directly contributes to severity of neuroinflammation. These findings reveal the critical contribution of myeloid-S1P1 signaling in CNS autoimmunity.


Subject(s)
Autoimmunity/immunology , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Inflammation/immunology , Myeloid Cells/immunology , Sphingosine-1-Phosphate Receptors/immunology , Animals , Central Nervous System/metabolism , Central Nervous System/pathology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism , Myeloid Cells/metabolism , STAT3 Transcription Factor/immunology , STAT3 Transcription Factor/metabolism , Signal Transduction/immunology , Sphingosine-1-Phosphate Receptors/genetics , Sphingosine-1-Phosphate Receptors/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism
17.
Blood ; 129(12): 1718-1728, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28096089

ABSTRACT

The combination of total lymphoid irradiation and anti-T-cell antibodies safely induces immune tolerance to combined hematopoietic cell and organ allografts in humans. Our mouse model required host natural killer T (NKT) cells to induce tolerance. Because NKT cells normally depend on signals from CD8+ dendritic cells (DCs) for their activation, we used the mouse model to test the hypothesis that, after lymphoid irradiation, host CD8+ DCs play a requisite role in tolerance induction through interactions with NKT cells. Selective deficiency of either CD8+ DCs or NKT cells abrogated chimerism and organ graft acceptance. After radiation, the CD8+ DCs increased expression of surface molecules required for NKT and apoptotic cell interactions and developed suppressive immune functions, including production of indoleamine 2,3-deoxygenase. Injection of naive mice with apoptotic spleen cells generated by irradiation led to DC changes similar to those induced by lymphoid radiation, suggesting that apoptotic body ingestion by CD8+ DCs initiates tolerance induction. Tolerogenic CD8+ DCs induced the development of tolerogenic NKT cells with a marked T helper 2 cell bias that, in turn, regulated the differentiation of the DCs and suppressed rejection of the transplants. Thus, reciprocal interactions between CD8+ DCs and invariant NKT cells are required for tolerance induction in this system that was translated into a successful clinical protocol.


Subject(s)
Dendritic Cells/immunology , Graft Rejection/prevention & control , Immune Tolerance , Natural Killer T-Cells/immunology , Animals , Bone Marrow Transplantation , CD8-Positive T-Lymphocytes/immunology , Cell Communication/immunology , Graft Rejection/immunology , Heart Transplantation , Mice
18.
Rapid Commun Mass Spectrom ; 32(6): 480-488, 2018 Mar 30.
Article in English | MEDLINE | ID: mdl-29334584

ABSTRACT

RATIONALE: A novel benzimidazole compound ZLN005 was previously identified as a transcriptional activator of peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) in certain metabolic tissues. Upregulation of PGC-1α by ZLN005 has been shown to have a beneficial effect in a diabetic mouse model and in a coronary artery disease model in vitro. ZLN005 could also have therapeutic potential in neurodegenerative diseases involving down-regulation of PGC-1α. Given the phenotypic efficacy of ZLN005 in several animal models of human disease, its metabolic profile was investigated to guide the development of novel therapeutics using ZLN005 as the lead compound. METHODS: ZLN005 was incubated with both rat and human liver microsomes and S9 fractions to identify in vitro metabolites. Urine from rats dosed with ZLN005 was used to identify in vivo metabolites. Extracted metabolites were analyzed by liquid chromatography/tandem mass spectrometry (LC/MS/MS) using a hybrid linear ion trap triple quadrupole mass spectrometer in full scan, enhanced product ion scan, neutral loss scan and precursor scan modes. Metabolites in plasma and brain of ZLN005-treated rats were also profiled using multiple reaction monitoring. RESULTS: Identified in vitro transformations of ZLN005 include mono- and dihydroxylation, further oxidation to carboxylic acids, and mono-O-glucuronide and sulfate conjugation to hydroxy ZLN005 as well as glutathione conjugation. Identified in vivo metabolites are mainly glucuronide and sulfate conjugates of dihydroxyl, carboxyl, and hydroxy acid of the parent compound. The parent compound as well as several major phase I metabolites were found in rat plasma and brain. CONCLUSIONS: Using both in vitro and in vivo methods, we elucidated the metabolic pathway of ZLN005. Phase I metabolites with hydroxylation and carboxylation, as well as phase II metabolites with glucuronide, sulfate and glutathione conjugation, were identified.

19.
Angew Chem Int Ed Engl ; 57(12): 3137-3142, 2018 03 12.
Article in English | MEDLINE | ID: mdl-29370452

ABSTRACT

The C-type lectins dectin-1 and dectin-2 contribute to innate immunity against microbial pathogens by recognizing their foreign glycan structures. These receptors are promising targets for vaccine development and cancer immunotherapy. However, currently available agonists are heterogeneous glycoconjugates and polysaccharides from natural sources. Herein, we designed and synthesized the first chemically defined ligands for dectin-1 and dectin-2. They comprised glycopolypeptides bearing mono-, di-, and trisaccharides and were built through polymerization of glycosylated N-carboxyanhydrides. Through this approach, we achieved glycopolypeptides with high molecular weights and low dispersities. We identified structures that elicit a pro-inflammatory response through dectin-1 or dectin-2 in antigen-presenting cells. With their native proteinaceous backbones and natural glycosidic linkages, these agonists are attractive for translational applications.


Subject(s)
Anhydrides/metabolism , Antigen-Presenting Cells/metabolism , Glycopeptides/metabolism , Lectins, C-Type/metabolism , Anhydrides/chemistry , Cells, Cultured , Glycopeptides/chemistry , Humans , Lectins, C-Type/chemistry , Ligands , Molecular Structure , Polymerization
20.
Eur J Immunol ; 45(2): 612-23, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25352488

ABSTRACT

IgG autoantibodies, including antibodies to double-stranded DNA (dsDNA), are pathogenic in systemic lupus erythematosus (SLE), but the mechanisms controlling their production are not understood. To assess the role of invariant natural killer T (iNKT) cells in this process, we studied 44 lupus patients. We took advantage of the propensity of PBMCs from patients with active disease to spontaneously secrete IgG in vitro. Despite the rarity of iNKT cells in lupus blood (0.002-0.05% of CD3-positive T cells), antibody blockade of the conserved iNKT TCR or its ligand, CD1d, or selective depletion of iNKT cells, inhibited spontaneous secretion of total IgG and anti-dsDNA IgG by lupus PBMCs. Addition of anti-iNKT or anti-CD1d antibody to PBMC cultures also reduced the frequency of plasma cells, suggesting that lupus iNKT cells induce B-cell maturation. Like fresh iNKT cells, expanded iNKT-cell lines from lupus patients, but not healthy subjects, induced autologous B cells to secrete antibodies, including IgG anti-dsDNA. This activity was inhibited by anti-CD40L antibody, as well as anti-CD1d antibody, confirming a role for CD40L-CD40 and TCR-CD1d interactions in lupus iNKT-cell-mediated help. These results reveal a critical role for iNKT cells in B-cell maturation and autoantibody production in patients with lupus.


Subject(s)
Antibodies, Antinuclear/biosynthesis , Antigens, CD1d/immunology , Immunoglobulin G/biosynthesis , Lupus Erythematosus, Systemic/immunology , Natural Killer T-Cells/immunology , Receptors, Antigen, T-Cell/immunology , Adult , Antibodies, Neutralizing/pharmacology , Antigens, CD1d/genetics , CD40 Ligand/antagonists & inhibitors , CD40 Ligand/genetics , CD40 Ligand/immunology , Cell Differentiation , Cell Separation , Female , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/pathology , Lymphocyte Activation , Lymphocyte Depletion , Natural Killer T-Cells/pathology , Plasma Cells/immunology , Plasma Cells/pathology , Primary Cell Culture , Receptors, Antigen, T-Cell/antagonists & inhibitors , Receptors, Antigen, T-Cell/genetics
SELECTION OF CITATIONS
SEARCH DETAIL