Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Cell ; 184(6): 1517-1529, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33740452

ABSTRACT

Our immune system has evolved to protect us from pathogens and maintain homeostasis through localization in diverse tissue sites throughout the body. Immune responses are orchestrated by T cells, which direct pathogen clearance at the infection site and establish tissue-resident memory T cells (TRMs) for protection immunity. Here, we discuss how tissue immune responses are influenced by various stressors (e.g., metabolic, environmental, aging) that are rapidly changing due to climate fluctuations and globalization. We propose potential strategies for targeting tissue immunity to mitigate future pathogenic and environmental challenges and areas of investigation that can elucidate mechanisms for adapting and restoring homeostasis.


Subject(s)
Immunity , Organ Specificity/immunology , Aging/immunology , Animals , Environment , Humans , Immune System/physiology , T-Lymphocytes/immunology
2.
Nat Immunol ; 22(1): 25-31, 2021 01.
Article in English | MEDLINE | ID: mdl-33154590

ABSTRACT

Clinical manifestations of COVID-19 caused by the new coronavirus SARS-CoV-2 are associated with age1,2. Adults develop respiratory symptoms, which can progress to acute respiratory distress syndrome (ARDS) in the most severe form, while children are largely spared from respiratory illness but can develop a life-threatening multisystem inflammatory syndrome (MIS-C)3-5. Here, we show distinct antibody responses in children and adults after SARS-CoV-2 infection. Adult COVID-19 cohorts had anti-spike (S) IgG, IgM and IgA antibodies, as well as anti-nucleocapsid (N) IgG antibody, while children with and without MIS-C had reduced breadth of anti-SARS-CoV-2-specific antibodies, predominantly generating IgG antibodies specific for the S protein but not the N protein. Moreover, children with and without MIS-C had reduced neutralizing activity as compared to both adult COVID-19 cohorts, indicating a reduced protective serological response. These results suggest a distinct infection course and immune response in children independent of whether they develop MIS-C, with implications for developing age-targeted strategies for testing and protecting the population.


Subject(s)
Antibodies, Viral/immunology , Antibody Formation/immunology , COVID-19/immunology , Nucleocapsid Proteins/immunology , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Adolescent , Adult , Aged , COVID-19/virology , Child , Child, Preschool , Female , Humans , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Male , Middle Aged , SARS-CoV-2/physiology , Young Adult
3.
Immunity ; 54(4): 797-814.e6, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33765436

ABSTRACT

Immune response dynamics in coronavirus disease 2019 (COVID-19) and their severe manifestations have largely been studied in circulation. Here, we examined the relationship between immune processes in the respiratory tract and circulation through longitudinal phenotypic, transcriptomic, and cytokine profiling of paired airway and blood samples from patients with severe COVID-19 relative to heathy controls. In COVID-19 airways, T cells exhibited activated, tissue-resident, and protective profiles; higher T cell frequencies correlated with survival and younger age. Myeloid cells in COVID-19 airways featured hyperinflammatory signatures, and higher frequencies of these cells correlated with mortality and older age. In COVID-19 blood, aberrant CD163+ monocytes predominated over conventional monocytes, and were found in corresponding airway samples and in damaged alveoli. High levels of myeloid chemoattractants in airways suggest recruitment of these cells through a CCL2-CCR2 chemokine axis. Our findings provide insights into immune processes driving COVID-19 lung pathology with therapeutic implications for targeting inflammation in the respiratory tract.


Subject(s)
COVID-19/immunology , Lung/immunology , Myeloid Cells/immunology , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , COVID-19/blood , COVID-19/mortality , COVID-19/pathology , Cytokines/immunology , Cytokines/metabolism , Humans , Inflammation , Longitudinal Studies , Lung/pathology , Macrophages/immunology , Macrophages/pathology , Middle Aged , Monocytes/immunology , Monocytes/pathology , Myeloid Cells/pathology , SARS-CoV-2 , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Transcriptome , Young Adult
4.
Cancer Immunol Immunother ; 71(8): 1837-1849, 2022 Aug.
Article in English | MEDLINE | ID: mdl-34999916

ABSTRACT

Talimogene Laherparepvec (OncoVEXmGMCSF), an oncolytic virus, immune checkpoint inhibitor anti-programmed cell death protein 1 (anti-PD1), and BRAF inhibition (BRAFi), are all clinically approved for treatment of melanoma patients and are effective through diverse mechanisms of action. Individually, these therapies also have an effect on the tumor immune microenvironment (TIME). Evaluating the combination effect of these three therapies on the TIME can help determine when combination therapy is most appropriate for further study. In this study, we use a transgenic murine melanoma model (Tyr::CreER; BRAFCA/+; PTENflox/flox), to evaluate the TIME in response to combinations of BRAFi, anti-PD1, and OncoVEXmGMCSF. We find that mice treated with the triple combination BRAFi + anti-PD1 + OncoVEXmGMCSF have decreased tumor growth compared to BRAFi alone and prolonged survival compared to control. Flow cytometry shows an increase in percent CD8 + /CD3 + cytotoxic T Lymphocytes (CTLs) and a decrease in percent FOXP3 + /CD4 + T regulatory cells (Tregs) in tumors treated with OncoVEXmGMCSF compared to mice not treated with OncoVEXmGMCSF. Immunogenomic analysis at 30d post-treatment shows an increase in Th1 and interferon-related genes in mice receiving OncoVEXmGMCSF + BRAFi. In summary, treatment with combination BRAFi + anti-PD1 + OncoVEXmGMCSF is more effective than any single treatment in controlling tumor growth, and groups receiving OncoVEXmGMCSF had more tumoral infiltration of CTLs and less intratumoral Tregs in the TIME. This study provides rational basis to combine targeted agents, oncolytic viral therapy, and checkpoint inhibitors in the treatment of melanoma.


Subject(s)
Antineoplastic Agents , Melanoma , Oncolytic Virotherapy , Oncolytic Viruses , Animals , Antineoplastic Agents/therapeutic use , Immunologic Factors/therapeutic use , Immunotherapy , Melanoma/drug therapy , Mice , Proto-Oncogene Proteins B-raf/genetics , Tumor Microenvironment
5.
Blood ; 131(23): 2581-2593, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29666112

ABSTRACT

Macrophages play important roles in recycling iron derived from the clearance of red blood cells (RBCs). They are also a critically important component of host defense, protecting against invading pathogens. However, the effects on macrophage biology of acutely ingesting large numbers of RBCs are not completely understood. To investigate this issue, we used a mouse model of RBC transfusion and clearance, which mimics the clinical setting. In this model, transfusions of refrigerator storage-damaged (ie, "old") RBCs led to increased erythrophagocytosis by splenic red pulp macrophages (RPMs). This robust erythrophagocytosis induced ferroptosis, an iron-dependent form of cell death, in RPMs. This was accompanied by increases in reactive oxygen species and lipid peroxidation in vivo, which were reduced by treatment in vitro with ferrostatin-1, a ferroptosis inhibitor. Old RBC transfusions also induced RPM-dependent chemokine expression by splenic Ly6Chi monocytes, which signaled Ly6Chi monocyte migration from bone marrow to spleen, where these cells subsequently differentiated into RPMs. The combination of cell division among remaining splenic RPMs, along with the influx of bone marrow-derived Ly6Chi monocytes, suggests that, following RPM depletion induced by robust erythrophagocytosis, there is a coordinated effort to restore homeostasis of the RPM population by local self-maintenance and contributions from circulating monocytes. In conclusion, these findings may be clinically relevant to pathological conditions that can arise as a result of increased erythrophagocytosis, such as transfusion-related immunomodulation and impaired host immunity.


Subject(s)
Erythrocyte Transfusion , Erythrocytes/immunology , Macrophages/immunology , Phagocytosis , Animals , Cell Death , Cell Division , Disease Models, Animal , Erythrocyte Transfusion/adverse effects , Erythrocytes/cytology , Lipid Peroxidation , Macrophages/cytology , Mice , Mice, Inbred C57BL , Monocytes/cytology , Monocytes/immunology
6.
Transfusion ; 58(4): 891-895, 2018 04.
Article in English | MEDLINE | ID: mdl-29473178

ABSTRACT

BACKGROUND: Platelets (PLTs) collected and stored in PLT additive solution Intersol (PAS-C) are presumed to reduce recipient exposure to donor plasma components; however, the effects of PAS-C on PLT supernatant composition are poorly defined. Therefore, we compared the total protein concentration, isohemagglutinin titers, and HLA antibodies in supernatants of PAS-C PLTs to plasma PLTs. STUDY DESIGN AND METHODS: Apheresis PLTs from group O blood donors were collected into either 100% donor plasma (n = 50) or a mixture of 65% PAS-C/35% donor plasma (n = 50). Within 12 hours of collection, samples of the PLT supernatant were frozen and stored. PLT supernatants were assayed for total protein concentration and anti-A and anti-B titers and screened for HLA antibodies. Samples positive for HLA antibodies were further tested using single-antigen bead assays to determine antibody strength and specificity. RESULTS: Supernatants of PAS-C PLTs had significantly lower total protein concentration and anti-A and anti-B titers compared to plasma PLTs. There was no significant difference in the number of HLA antibody screen-positive PAS-C (3/50) compared to plasma PLT supernatants (2/50); however, the HLA antibody screen-positive supernatants of PAS-C PLTs had fewer HLA specificities (2) compared to those of the plasma PLTs (18). CONCLUSION: Decreased plasma proteins likely underlie lower rates of allergic and febrile, nonhemolytic transfusion reactions from the infusion of PAS-C PLTs. Decreased anti-A and anti-B titers may prevent hemolysis from minor ABO mismatch. Lower HLA antibody specificities may mitigate transfusion-related acute lung injury.


Subject(s)
Blood Platelets/drug effects , Blood Preservation/methods , Blood Proteins/analysis , Hemagglutinins/blood , Histocompatibility Testing/methods , Isoantibodies/blood , Organ Preservation Solutions/pharmacology , Transfusion-Related Acute Lung Injury/prevention & control , ABO Blood-Group System/immunology , Antibody Affinity , Antibody Specificity , Blood Group Incompatibility/prevention & control , Blood Platelets/chemistry , Blood Platelets/immunology , Epitopes/immunology , Female , HLA Antigens/immunology , Humans , Male , Plasma , Plateletpheresis
7.
Oncoimmunology ; 11(1): 2066767, 2022.
Article in English | MEDLINE | ID: mdl-35558160

ABSTRACT

Patients with pancreatic ductal adenocarcinoma (PDAC) have a grim prognosis despite complete surgical resection and intense systemic therapies. While immunotherapies have been beneficial with many different types of solid tumors, they have almost uniformly failed in the treatment of PDAC. Understanding how therapies affect the tumor immune microenvironment (TIME) can provide insights for the development of strategies to treat PDAC. We used quantitative multiplexed immunofluorescence (qmIF) quantitative spatial analysis (qSA), and immunogenomic (IG) analysis to analyze formalin-fixed paraffin embedded (FFPE) primary tumor specimens from 44 patients with PDAC including 18 treated with neoadjuvant chemoradiation (CRT) and 26 patients receiving no treatment (NT) and compared them with tissues from 40 treatment-naïve melanoma patients. We find that relative to NT tumors, CD3+ T cell infiltration was increased in CRT treated tumors (p = .0006), including increases in CD3+CD8+ cytotoxic T cells (CTLs, p = .0079), CD3+CD4+FOXP3- T helper cells (Th, p = .0010), and CD3+CD4+FOXP3+ regulatory T cells (Tregs, p = .0089) with no difference in CD68+ macrophages. IG analysis from micro-dissected tissues indicated overexpression of genes involved in antigen presentation, T cell activation, and inflammation in CRT treated tumors. Among treated patients, a higher ratio of Tregs to total T cells was associated with shorter survival time (p = .0121). Despite comparable levels of infiltrating T cells in CRT PDACs to melanoma, PDACs displayed distinct spatial profiles with less T cell clustering as defined by nearest neighbor analysis (p < .001). These findings demonstrate that, while CRT can achieve high T cell densities in PDAC compared to melanoma, phenotype and spatial organization of T cells may limit benefit of T cell infiltration in this immunotherapy-resistant tumor.


Subject(s)
Carcinoma, Pancreatic Ductal , Melanoma , Pancreatic Neoplasms , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/therapy , Forkhead Transcription Factors , Humans , Melanoma/therapy , Neoadjuvant Therapy , Pancreatic Neoplasms/therapy , Tumor Microenvironment , Pancreatic Neoplasms
8.
JCI Insight ; 7(11)2022 06 08.
Article in English | MEDLINE | ID: mdl-35446789

ABSTRACT

Respiratory failure in COVID-19 is characterized by widespread disruption of the lung's alveolar gas exchange interface. To elucidate determinants of alveolar lung damage, we performed epithelial and immune cell profiling in lungs from 24 COVID-19 autopsies and 43 uninfected organ donors ages 18-92 years. We found marked loss of type 2 alveolar epithelial (T2AE) cells and increased perialveolar lymphocyte cytotoxicity in all fatal COVID-19 cases, even at early stages before typical patterns of acute lung injury are histologically apparent. In lungs from uninfected organ donors, there was also progressive loss of T2AE cells with increasing age, which may increase susceptibility to COVID-19-mediated lung damage in older individuals. In the fatal COVID-19 cases, macrophage infiltration differed according to the histopathological pattern of lung injury. In cases with acute lung injury, we found accumulation of CD4+ macrophages that expressed distinctly high levels of T cell activation and costimulation genes and strongly correlated with increased extent of alveolar epithelial cell depletion and CD8+ T cell cytotoxicity. Together, our results show that T2AE cell deficiency may underlie age-related COVID-19 risk and initiate alveolar dysfunction shortly after infection, and we define immune cell mediators that may contribute to alveolar injury in distinct pathological stages of fatal COVID-19.


Subject(s)
Acute Lung Injury , COVID-19 , Acute Lung Injury/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Alveolar Epithelial Cells/pathology , Autopsy , Humans , Lung/pathology , Middle Aged , Young Adult
9.
JCI Insight ; 7(17)2022 09 08.
Article in English | MEDLINE | ID: mdl-35925682

ABSTRACT

Checkpoint inhibitors (CPIs) targeting programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) and cytotoxic T lymphocyte antigen 4 (CTLA-4) have revolutionized cancer treatment but can trigger autoimmune complications, including CPI-induced diabetes mellitus (CPI-DM), which occurs preferentially with PD-1 blockade. We found evidence of pancreatic inflammation in patients with CPI-DM with shrinkage of pancreases, increased pancreatic enzymes, and in a case from a patient who died with CPI-DM, peri-islet lymphocytic infiltration. In the NOD mouse model, anti-PD-L1 but not anti-CTLA-4 induced diabetes rapidly. RNA sequencing revealed that cytolytic IFN-γ+CD8+ T cells infiltrated islets with anti-PD-L1. Changes in ß cells were predominantly driven by IFN-γ and TNF-α and included induction of a potentially novel ß cell population with transcriptional changes suggesting dedifferentiation. IFN-γ increased checkpoint ligand expression and activated apoptosis pathways in human ß cells in vitro. Treatment with anti-IFN-γ and anti-TNF-α prevented CPI-DM in anti-PD-L1-treated NOD mice. CPIs targeting the PD-1/PD-L1 pathway resulted in transcriptional changes in ß cells and immune infiltrates that may lead to the development of diabetes. Inhibition of inflammatory cytokines can prevent CPI-DM, suggesting a strategy for clinical application to prevent this complication.


Subject(s)
Diabetes Mellitus , Programmed Cell Death 1 Receptor , Animals , Humans , Inflammation Mediators , Mice , Mice, Inbred NOD , Tumor Necrosis Factor Inhibitors
10.
mSphere ; 6(4): e0057121, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34319126

ABSTRACT

The COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) is ongoing and has shown the community that flexible methods for rapidly identifying and screening candidate antivirals are needed. Assessing virus-neutralizing activity of human serum to monitor population immunity and response to infection and vaccination is key to pandemic control. We developed a virus neutralization platform strategy that relies only on bioinformatic and genetic information of the virus of interest. The platform uses viral envelope glycoprotein cDNAs to set up an assay that mimics multicycle infection but is safe and, therefore, amenable to biosafety level 2 (BSL2) conditions for viruses that require BSL3 facilities (e.g., SARS-CoV-1 and SARS-CoV-2). As a complement to this platform, we present a new cell-based immunofluorescent (CBI) assay that uses SARS-CoV-2 spike protein (S)-expressing cells to accurately measure the neutralization potential of human sera and is readily adaptable to variants of concern. These methods should be useful additions to the tools for assessing antiviral immunity, whether acquired via natural infection or vaccines. IMPORTANCE Assays for rapid biosafety level 2 (BSL2) evaluation of neutralizing properties of antibodies acquired via natural infection or through vaccination is urgently needed. Here, we propose a combinatorial approach in which sera are screened for SARS-CoV-2 spike protein (S) binding using a cell-based immunofluorescent (CBI) assay, and positive samples are further evaluated in a pseudotyped viral multicycle infection-mimicking protocol under BSL2 conditions.


Subject(s)
Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Antiviral Agents/immunology , COVID-19/immunology , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Animals , COVID-19/virology , Cell Line , Chlorocebus aethiops , HEK293 Cells , Humans , Neutralization Tests/methods , Pandemics/prevention & control , Vero Cells
11.
Nat Commun ; 12(1): 2911, 2021 05 18.
Article in English | MEDLINE | ID: mdl-34006859

ABSTRACT

The impact of immune mediators on weight homeostasis remains underdefined. Interrogation of resistance to diet-induced obesity in mice lacking a negative regulator of Toll-like receptor signaling serendipitously uncovered a role for B cell activating factor (BAFF). Here we show that overexpression of BAFF in multiple mouse models associates with protection from weight gain, approximating a log-linear dose response relation to BAFF concentrations. Gene expression analysis of BAFF-stimulated subcutaneous white adipocytes unveils upregulation of lipid metabolism pathways, with BAFF inducing white adipose tissue (WAT) lipolysis. Brown adipose tissue (BAT) from BAFF-overexpressing mice exhibits increased Ucp1 expression and BAFF promotes brown adipocyte respiration and in vivo energy expenditure. A proliferation-inducing ligand (APRIL), a BAFF homolog, similarly modulates WAT and BAT lipid handling. Genetic deletion of both BAFF and APRIL augments diet-induced obesity. Lastly, BAFF/APRIL effects are conserved in human adipocytes and higher BAFF/APRIL levels correlate with greater BMI decrease after bariatric surgery. Together, the BAFF/APRIL axis is a multifaceted immune regulator of weight gain and adipose tissue function.


Subject(s)
B-Cell Activating Factor/genetics , Obesity/genetics , Signal Transduction/genetics , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics , Weight Gain/genetics , Adipocytes/cytology , Adipocytes/metabolism , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/cytology , Adipose Tissue, White/metabolism , Animals , B-Cell Activating Factor/metabolism , Cells, Cultured , Diet, High-Fat/adverse effects , Gene Expression Profiling/methods , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Obesity/etiology , Obesity/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism
12.
Neuron ; 50(1): 127-43, 2006 Apr 06.
Article in English | MEDLINE | ID: mdl-16600861

ABSTRACT

Whereas significant insight exists as to how LTP-related changes can contribute to the formation of long-term memory, little is known about the role of hippocampal LTD-like changes in learning and memory storage. We describe a mouse lacking the transcription factor SRF in the adult forebrain. This mouse could not acquire a hippocampus-based immediate memory for a novel context even across a few minute timespan, which led to a profound but selective deficit in explicit spatial memory. These animals were also impaired in the induction of LTD, including LTD triggered by a cholinergic agonist. Moreover, genes regulating two processes essential for LTD-calcium release from intracellular stores and phosphatase activation-were abnormally expressed in knockouts. These findings suggest that for the hippocampus to form associative spatial memories through LTP-like processes, it must first undergo learning of the context per se through exploration and the learning of familiarity, which requires LTD-like processes.


Subject(s)
Exploratory Behavior/physiology , Learning/physiology , Long-Term Synaptic Depression/physiology , Memory, Short-Term/physiology , Prosencephalon/physiology , Serum Response Factor/physiology , Animals , Animals, Newborn , Behavior, Animal , Blotting, Northern/methods , Carbachol/pharmacology , Cholinergic Agonists/pharmacology , Clathrin Heavy Chains/metabolism , Discrimination, Psychological/physiology , Dose-Response Relationship, Radiation , Early Growth Response Protein 1/metabolism , Electric Stimulation/methods , Enzyme Inhibitors/pharmacology , Gene Expression/genetics , Gene Expression Regulation, Developmental/physiology , Habituation, Psychophysiologic/physiology , Hippocampus/metabolism , Immunohistochemistry/methods , In Situ Hybridization/methods , Indoles/pharmacology , Long-Term Synaptic Depression/genetics , Maze Learning/physiology , Mice , Mice, Knockout , Models, Neurological , Olfactory Bulb/physiology , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Serum Response Factor/deficiency , Time Factors
13.
J Clin Invest ; 117(4): 902-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17364026

ABSTRACT

Monocyte recruitment to sites of inflammation is regulated by members of the chemokine family of chemotactic cytokines. However, the mechanisms that govern the migration of monocytes from bone marrow to blood and from blood to inflamed tissues are not well understood. Here we report that CC chemokine receptor 2 (CCR2) is highly expressed on a subpopulation of blood monocytes whose numbers are markedly decreased in CCR2(-/-) mice. In bone marrow, however, CCR2(-/-) mice had an increased number of monocytes, suggesting that CCR2 is critical for monocyte egress. Intravenous infusion of ex vivo-labeled WT or CCR2(-/-) bone marrow into WT recipient mice demonstrated that CCR2 is necessary for efficient monocyte recruitment from the blood to inflamed tissue. Analysis of mice lacking monocyte chemoattractant protein-1 (MCP-1), MCP-3, MCP-5, or MCP-2 plus MCP-5 revealed that MCP-3 and MCP-1 are the CCR2 agonists most critical for the maintenance of normal blood monocyte counts. These findings provide evidence that CCR2 and MCP-3/MCP-1 are critical for monocyte mobilization and suggest new roles for monocyte chemoattractants in leukocyte homeostasis.


Subject(s)
Bone Marrow Cells/physiology , Inflammation/physiopathology , Monocyte Chemoattractant Proteins/physiology , Monocytes/physiology , Receptors, Chemokine/physiology , Adoptive Transfer , Animals , Blood Cell Count , Bone Marrow Transplantation/physiology , Chemokine CCL7 , Chemokines/blood , Humans , Mice , Mice, Knockout , Monocyte Chemoattractant Proteins/genetics , Monocytes/immunology , Polymorphism, Genetic , Receptors, CCR2 , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics
14.
medRxiv ; 2020 Oct 18.
Article in English | MEDLINE | ID: mdl-33106817

ABSTRACT

Immune responses to respiratory viruses like SARS-CoV-2 originate and function in the lung, yet assessments of human immunity are often limited to blood. Here, we conducted longitudinal, high-dimensional profiling of paired airway and blood samples from patients with severe COVID-19, revealing immune processes in the respiratory tract linked to disease pathogenesis. Survival from severe disease was associated with increased CD4 + T cells and decreased monocyte/macrophage frequencies in the airway, but not in blood. Airway T cells and macrophages exhibited tissue-resident phenotypes and activation signatures, including high level expression and secretion of monocyte chemoattractants CCL2 and CCL3 by airway macrophages. By contrast, monocytes in blood expressed the CCL2-receptor CCR2 and aberrant CD163 + and immature phenotypes. Extensive accumulation of CD163 + monocyte/macrophages within alveolar spaces in COVID-19 lung autopsies suggested recruitment from circulation. Our findings provide evidence that COVID-19 pathogenesis is driven by respiratory immunity, and rationale for site-specific treatment and prevention strategies.

15.
medRxiv ; 2020 Jul 14.
Article in English | MEDLINE | ID: mdl-32699861

ABSTRACT

Clinical manifestations of COVID-19 caused by the novel coronavirus SARS-CoV-2 are associated with age. While children are largely spared from severe respiratory disease, they can present with a SARS-CoV-2-associated multisystem inflammatory syndrome (MIS-C) similar to Kawasaki's disease. Here, we show distinct antibody (Ab) responses in children with MIS-C compared to adults with severe COVID-19 causing acute respiratory distress syndrome (ARDS), and those who recovered from mild disease. There was a reduced breadth and specificity of anti-SARS-CoV-2-specific antibodies in MIS-C patients compared to the COVID patient groups; MIS-C predominantly generated IgG Abs specific for the Spike (S) protein but not for the nucleocapsid (N) protein, while both COVID-19 cohorts had anti-S IgG, IgM and IgA Abs, as well as anti-N IgG Abs. Moreover, MIS-C patients had reduced neutralizing activity compared to COVID-19 cohorts, indicating a reduced protective serological response. These results suggest a distinct infection course and immune response in children and adults who develop severe disease, with implications for optimizing treatments based on symptom and age.

16.
J Clin Invest ; 116(1): 115-24, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16341265

ABSTRACT

The C-C motif chemokine receptor-2 (CCR2) regulates monocyte and macrophage recruitment and is necessary for macrophage-dependent inflammatory responses and the development of atherosclerosis. Although adipose tissue expression and circulating concentrations of CCL2 (also known as MCP1), a high-affinity ligand for CCR2, are elevated in obesity, the role of CCR2 in metabolic disorders, including insulin resistance, hepatic steatosis, and inflammation associated with obesity, has not been studied. To determine what role CCR2 plays in the development of metabolic phenotypes, we studied the effects of Ccr2 genotype on the development of obesity and its associated phenotypes. Genetic deficiency in Ccr2 reduced food intake and attenuated the development of obesity in mice fed a high-fat diet. In obese mice matched for adiposity, Ccr2 deficiency reduced macrophage content and the inflammatory profile of adipose tissue, increased adiponectin expression, ameliorated hepatic steatosis, and improved systemic glucose homeostasis and insulin sensitivity. In mice with established obesity, short-term treatment with a pharmacological antagonist of CCR2 lowered macrophage content of adipose tissue and improved insulin sensitivity without significantly altering body mass or improving hepatic steatosis. These data suggest that CCR2 influences the development of obesity and associated adipose tissue inflammation and systemic insulin resistance and plays a role in the maintenance of adipose tissue macrophages and insulin resistance once obesity and its metabolic consequences are established.


Subject(s)
Dietary Fats , Insulin Resistance , Obesity/physiopathology , Receptors, Chemokine/physiology , Adipose Tissue/physiopathology , Animals , Chemokine CCL2/physiology , Crosses, Genetic , Female , Inflammation/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR2 , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics
17.
Cell Rep ; 29(12): 3916-3932.e5, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31851923

ABSTRACT

Non-recirculating tissue-resident memory T cells (TRMs) are the predominant T cell subset in diverse tissue sites, where they mediate protective immune responses in situ. Here, we reveal a role for TRM in maintaining immune homeostasis in the human pancreas through interactions with resident macrophages and the PD-1/PD-L1 inhibitory pathway. Using tissues obtained from organ donors, we identify that pancreas T cells comprise CD8+PD-1hi TRMs, which are phenotypically, functionally, and transcriptionally distinct compared to TRMs in neighboring jejunum and lymph node sites. Pancreas TRMs cluster with resident macrophages throughout the exocrine areas; TRM effector functions are enhanced by macrophage-derived co-stimulation and attenuated by the PD-1/PD-L1 pathways. Conversely, in samples from chronic pancreatitis, TRMs exhibit reduced PD-1 expression and reduced interactions with macrophages. These findings suggest important roles for PD-1 and TRM-macrophage interactions in controlling tissue homeostasis and immune dysfunctions underlying inflammatory disease, with important implications for PD-1-based immunotherapies.


Subject(s)
B7-H1 Antigen/metabolism , Immunologic Memory/physiology , Pancreas/metabolism , Pancreatitis/immunology , Pancreatitis/metabolism , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocyte Subsets/metabolism , B7-H1 Antigen/genetics , CD58 Antigens/metabolism , Humans , Immunity, Mucosal/genetics , Immunity, Mucosal/physiology , Immunologic Memory/genetics , Macrophages/immunology , Macrophages/metabolism , Pancreas/immunology , Pancreas/pathology , Pancreatitis/genetics , Programmed Cell Death 1 Receptor/genetics , Signal Transduction , T-Lymphocyte Subsets/immunology
18.
Endocrinology ; 149(7): 3549-58, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18403477

ABSTRACT

Obesity is a major risk factor for the development of type 2 diabetes, and both conditions are now recognized to possess significant inflammatory components underlying their pathophysiologies. We tested the hypothesis that the plant polyphenolic compound curcumin, which is known to exert potent antiinflammatory and antioxidant effects, would ameliorate diabetes and inflammation in murine models of insulin-resistant obesity. We found that dietary curcumin admixture ameliorated diabetes in high-fat diet-induced obese and leptin-deficient ob/ob male C57BL/6J mice as determined by glucose and insulin tolerance testing and hemoglobin A1c percentages. Curcumin treatment also significantly reduced macrophage infiltration of white adipose tissue, increased adipose tissue adiponectin production, and decreased hepatic nuclear factor-kappaB activity, hepatomegaly, and markers of hepatic inflammation. We therefore conclude that orally ingested curcumin reverses many of the inflammatory and metabolic derangements associated with obesity and improves glycemic control in mouse models of type 2 diabetes. This or related compounds warrant further investigation as novel adjunctive therapies for type 2 diabetes in man.


Subject(s)
Curcumin/pharmacology , Diabetes Mellitus, Type 2/drug therapy , Inflammation/drug therapy , Obesity/complications , Adiponectin/genetics , Adiponectin/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/administration & dosage , Antioxidants/pharmacology , Blood Glucose/metabolism , Curcumin/administration & dosage , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/etiology , Dietary Supplements , Disease Models, Animal , Gene Expression/drug effects , Immunohistochemistry , Inflammation/etiology , Interleukin-6/genetics , Interleukin-6/metabolism , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , NF-kappa B/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
19.
J Clin Invest ; 112(12): 1796-808, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14679176

ABSTRACT

Obesity alters adipose tissue metabolic and endocrine function and leads to an increased release of fatty acids, hormones, and proinflammatory molecules that contribute to obesity associated complications. To further characterize the changes that occur in adipose tissue with increasing adiposity, we profiled transcript expression in perigonadal adipose tissue from groups of mice in which adiposity varied due to sex, diet, and the obesity-related mutations agouti (Ay) and obese (Lepob). We found that the expression of 1,304 transcripts correlated significantly with body mass. Of the 100 most significantly correlated genes, 30% encoded proteins that are characteristic of macrophages and are positively correlated with body mass. Immunohistochemical analysis of perigonadal, perirenal, mesenteric, and subcutaneous adipose tissue revealed that the percentage of cells expressing the macrophage marker F4/80 (F4/80+) was significantly and positively correlated with both adipocyte size and body mass. Similar relationships were found in human subcutaneous adipose tissue stained for the macrophage antigen CD68. Bone marrow transplant studies and quantitation of macrophage number in adipose tissue from macrophage-deficient (Csf1op/op) mice suggest that these F4/80+ cells are CSF-1 dependent, bone marrow-derived adipose tissue macrophages. Expression analysis of macrophage and nonmacrophage cell populations isolated from adipose tissue demonstrates that adipose tissue macrophages are responsible for almost all adipose tissue TNF-alpha expression and significant amounts of iNOS and IL-6 expression. Adipose tissue macrophage numbers increase in obesity and participate in inflammatory pathways that are activated in adipose tissues of obese individuals.


Subject(s)
Adipose Tissue/metabolism , Macrophages/metabolism , Obesity , Animals , Antigens, CD/biosynthesis , Antigens, Differentiation, Myelomonocytic/biosynthesis , Body Weight , Cell Separation , Female , Flow Cytometry , Humans , Immunohistochemistry , Immunophenotyping , Interleukin-6/metabolism , Leukocyte Common Antigens/biosynthesis , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Obesity/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tumor Necrosis Factor-alpha/metabolism
20.
Arch Pathol Lab Med ; 141(12): 1705-1711, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28849943

ABSTRACT

CONTEXT: - Cytomegalovirus (CMV) can be transmitted by cellular blood products, leading to severe disease in immunosuppressed patients such as neonates and transplant recipients. To mitigate transfusion-transmitted CMV (TT-CMV), "CMV-safe" blood products (leukoreduced and/or CMV-seronegative) are transfused. Attempts to develop practice guidelines for TT-CMV mitigation have been limited by paucity of high-quality clinical trials. OBJECTIVE: - To assess current TT-CMV mitigation strategies across medical institutions for specific at-risk populations. DESIGN: - Supplemental questions regarding TT-CMV and CMV disease mitigation were added to a College of American Pathologists Transfusion Medicine (Comprehensive) Participant Survey in 2015, addressing whether a given institution provided CMV-safe products for 6 at-risk patient populations. RESULTS: - Ninety percent (2712 of 3032) of institutions reported providing universally leukoreduced blood products. Among institutions without universal leukoreduction, 92% (295 of 320) provided leukoreduced products on the basis of clinical criteria. Eighty-three percent (2481 of 3004) of respondents reported having availability of CMV-seronegative products; however, wide variation in policies was reported governing CMV-seronegative product use. Among all respondents, less than 5% reported using CMV prophylaxis and monitoring in high-risk patient groups. Transplant centers reported higher rates of CMV prophylaxis (25% [97 of 394] solid organ) and monitoring (15% [59 of 394] solid organ) for CMV-negative transplant recipients. CONCLUSIONS: - Universal leukoreduction is the primary strategy for mitigating TT-CMV. While most institutions have both CMV-seronegative and leukoreduced blood products available, consensus is lacking on which patients should receive these products. High-quality studies are needed to determine if CMV-seronegative and leukoreduced blood products are needed in high-risk patient populations.


Subject(s)
Blood Transfusion , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Infections/transmission , Transfusion Reaction/prevention & control , Antibodies, Viral/blood , Cytapheresis/methods , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Humans , Infant, Newborn , Pathology, Clinical , Practice Guidelines as Topic , Risk Factors , Societies, Medical , Surveys and Questionnaires , Transplantation/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL