Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 118
Filter
1.
Eur J Immunol ; 54(5): e2350715, 2024 May.
Article in English | MEDLINE | ID: mdl-38446066

ABSTRACT

Although a role for TLR2 on T cells has been indicated in prior studies, in vivo stimulation of TLR2 on T cells by Mtb and its impact on Mtb infection has not been tested. Furthermore, it is not known if the enhanced susceptibility to Mtb of Tlr2 gene knockout mice is due to its role in macrophages, T cells, or both. To address TLR2 on T cells, we generated Tlr2fl/flxCd4cre/cre mice, which lack expression of TLR2 on both CD4 and CD8 T cells, to study the in vivo role of TLR2 on T cells after aerosol infection with virulent Mtb. Deletion of TLR2 in CD4+ and CD8+ T cells reduces their ability to be co-stimulated by TLR2 ligands for cytokine production. These include both pro- (IFN-γ, TNF-α) and anti-inflammatory cytokines (IL-10). Deletion of TLR2 in T cells affected control of Mtb in the lungs and spleens of infected mice. This suggests that T-cell co-stimulation by mycobacterial TLR2 ligands in vivo contributes to the control of Mtb infection in the lung and spleen.


Subject(s)
CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Mice, Knockout , Mycobacterium tuberculosis , Toll-Like Receptor 2 , Tuberculosis , Animals , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Mice , CD8-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Tuberculosis/microbiology , Mice, Inbred C57BL , Lung/immunology , Lung/microbiology , Spleen/immunology , Interferon-gamma/immunology , Interferon-gamma/metabolism , Lymphocyte Activation/immunology , Cytokines/metabolism , Cytokines/immunology
2.
J Immunol ; 211(9): 1385-1396, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37695687

ABSTRACT

Mycobacterium tuberculosis cell-wall glycolipids such as mannosylated lipoarabinomannan (ManLAM) can inhibit murine CD4+ T cells by blocking TCR signaling. This results in suppression of IL-2 production, reduced T cell proliferation, and induction of CD4+ T cell anergy. This study extended these findings to the interaction between primary human CD4+ T cells and macrophages infected by mycobacteria. Exposure of human CD4+ T cells to ManLAM before activation resulted in loss of polyfunctionality, as measured by IL-2, IFN-γ, and TNF-α expression, and reduced CD25 expression. This was not associated with upregulation of inhibitory receptors CTLA-4, PD-1, TIM-3, and Lag-3. By confocal microscopy and imaging flow cytometry, ManLAM exposure reduced conjugate formation between macrophages and CD4+ T cells. ManLAM colocalized to the immunological synapse (IS) and reduced translocation of lymphocyte-specific protein tyrosine kinase (LCK) to the IS. When CD4+ T cells and Mycobacterium bovis BCG-infected monocytes were cocultured, ManLAM colocalized to CD4+ T cells, which formed fewer conjugates with infected monocytes. These results demonstrate that mycobacterial cell-wall glycolipids such as ManLAM can traffic from infected macrophages to disrupt productive IS formation and inhibit CD4+ T cell activation, contributing to immune evasion by M. tuberculosis.


Subject(s)
Mycobacterium tuberculosis , Humans , CD4-Positive T-Lymphocytes , Glycolipids/metabolism , Immunological Synapses , Interleukin-2/metabolism , Macrophages/microbiology
3.
J Immunol ; 211(5): 767-781, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37486212

ABSTRACT

The gut microbiome and intestinal immune system are engaged in a dynamic interplay that provides myriad benefits to host health. However, the microbiome can also elicit damaging inflammatory responses, and thus establishing harmonious immune-microbiome interactions is essential to maintain homeostasis. Gut microbes actively coordinate the induction of anti-inflammatory responses that establish these mutualistic interactions. Despite this, the microbial pathways that govern this dialogue remain poorly understood. We investigated the mechanisms through which the gut symbiont Bacteroides thetaiotaomicron exerts its immunomodulatory functions on murine- and human-derived cells. Our data reveal that B. thetaiotaomicron stimulates production of the cytokine IL-10 via secreted factors that are packaged into outer membrane vesicles, in a TLR2- and MyD88-dependent manner. Using a transposon mutagenesis-based screen, we identified a key role for the B. thetaiotaomicron-encoded NADH:ubiquinone oxidoreductase (NQR) complex, which regenerates NAD+ during respiration, in this process. Finally, we found that disruption of NQR reduces the capacity of B. thetaiotaomicron to induce IL-10 by impairing biogenesis of outer membrane vesicles. These data identify a microbial pathway with a previously unappreciated role in gut microbe-mediated immunomodulation that may be targeted to manipulate the capacity of the microbiome to shape host immunity.


Subject(s)
Bacteroides thetaiotaomicron , Mice , Humans , Animals , Interleukin-10/metabolism , Mutagenesis , Gastrointestinal Tract , Cytokines/metabolism
4.
Nat Commun ; 13(1): 6648, 2022 11 04.
Article in English | MEDLINE | ID: mdl-36333322

ABSTRACT

The impact of bone cell activation on bacterially-induced osteolysis remains elusive. Here, we show that matrix-embedded osteocytes stimulated with bacterial pathogen-associated molecular patterns (PAMPs) directly drive bone resorption through an MYD88-regulated signaling pathway. Mice lacking MYD88, primarily in osteocytes, protect against osteolysis caused by calvarial injections of bacterial PAMPs and resist alveolar bone resorption induced by oral Porphyromonas gingivalis (Pg) infection. In contrast, mice with targeted MYD88 restoration in osteocytes exhibit osteolysis with inflammatory cell infiltration. In vitro, bacterial PAMPs induce significantly higher expression of the cytokine RANKL in osteocytes than osteoblasts. Mechanistically, activation of the osteocyte MYD88 pathway up-regulates RANKL by increasing binding of the transcription factors CREB and STAT3 to Rankl enhancers and by suppressing K48-ubiquitination of CREB/CREB binding protein and STAT3. Systemic administration of an MYD88 inhibitor prevents jawbone loss in Pg-driven periodontitis. These findings reveal that osteocytes directly regulate inflammatory osteolysis in bone infection, suggesting that MYD88 and downstream RANKL regulators in osteocytes are therapeutic targets for osteolysis in periodontitis and osteomyelitis.


Subject(s)
Alveolar Bone Loss , Osteolysis , Osteomyelitis , Periodontitis , Mice , Animals , Osteocytes/metabolism , Osteolysis/chemically induced , Osteolysis/complications , Osteolysis/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Pathogen-Associated Molecular Pattern Molecules/metabolism , RANK Ligand/metabolism , Porphyromonas gingivalis/metabolism , Periodontitis/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/metabolism , Osteoclasts/metabolism
5.
J Thromb Thrombolysis ; 53(4): 950-953, 2022 May.
Article in English | MEDLINE | ID: mdl-34797472

ABSTRACT

Over the last few years data from our group have indicated that α-synuclein is important in development of immune cells as well as potentially erythrocytes and platelets. The latter is important since this protein may work as negative regulator of granule release. Thus, we sought to begin to understand the structure of this protein in platelets. Flow cytometric analysis of this protein using region-specific (N-terminus, central region and C-terminus) monoclonal antibodies was performed. Antibody to the central region gave the strongest shift among all three antibodies, with the C-terminus having intermediate shift and N-terminus minimal shift. Western blotting using the same antibodies showed similar binding of all antibodies to α-synuclein. These results suggest a similar arrangement of this protein in platelets as seen in neurons. Future studies ought to look at the role that each protein region plays in platelets.


Subject(s)
Blood Platelets , alpha-Synuclein , Antibodies, Monoclonal , Blood Platelets/metabolism , Flow Cytometry , Humans , alpha-Synuclein/analysis , alpha-Synuclein/metabolism
6.
J Alzheimers Dis ; 82(4): 1487-1497, 2021.
Article in English | MEDLINE | ID: mdl-34180415

ABSTRACT

BACKGROUND: Accumulation of iron is a consistent feature of Alzheimer's disease (AD) brains. The underlying cause, however, remains debatable. OBJECTIVE: To explore whether local hepcidin synthesized by brain cells contributes to iron accumulation in AD brains. METHODS: Brain tissue from the cingulate cortex of 33 cases of AD pre-assigned to Braak stage I-VI, 6 cases of non-dementia, and 15 cases of non-AD dementia were analyzed for transcriptional upregulation of hepcidin by RT-qPCR and RT-PCR. Change in the expression of ferritin, ferroportin (Fpn), microglial activation marker Iba1, IL-6, and TGFß2 was determined by western blotting. Total tissue iron was determined by colorimetry. RESULTS: Significant transcriptional upregulation of hepcidin was observed in Braak stage III-VI relative to Braak stage I and II, non-AD dementia, and non-dementia samples. Ferritin was increased in Braak stage V, and a significant increase in tissue iron was evident in Braak stage III-VI. The expression of Iba1 and IL-6 was also increased in Braak stage III-VI relative to Braak stage I and II and non-AD dementia samples. Amyloid-ß plaques were absent in most Braak stage I and II samples, and present in Braak stage III-VI samples with few exceptions. CONCLUSION: These observations suggest that upregulation of brain hepcidin is mediated by IL-6, a known transcriptional activator of hepcidin. The consequent downregulation of Fpn on neuronal and other cells results in accumulation of iron in AD brains. The increase in hepcidin is disease-specific, and increases with disease progression, implicating AD-specific pathology in the accumulation of iron.


Subject(s)
Alzheimer Disease/pathology , Anti-Infective Agents/metabolism , Ferritins/metabolism , Hepcidins/metabolism , Up-Regulation , Aged , Autopsy , Brain/pathology , Female , Humans , Interleukin-6/metabolism , Male , Reverse Transcriptase Polymerase Chain Reaction
7.
Circ Res ; 128(8): 1214-1236, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33856918

ABSTRACT

A pandemic of historic impact, coronavirus disease 2019 (COVID-19) has potential consequences on the cardiovascular health of millions of people who survive infection worldwide. Severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2), the etiologic agent of COVID-19, can infect the heart, vascular tissues, and circulating cells through ACE2 (angiotensin-converting enzyme 2), the host cell receptor for the viral spike protein. Acute cardiac injury is a common extrapulmonary manifestation of COVID-19 with potential chronic consequences. This update provides a review of the clinical manifestations of cardiovascular involvement, potential direct SARS-CoV-2 and indirect immune response mechanisms impacting the cardiovascular system, and implications for the management of patients after recovery from acute COVID-19 infection.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19/virology , Cardiovascular Diseases/virology , Myocytes, Cardiac/virology , SARS-CoV-2/physiology , Virus Internalization , Biomarkers/metabolism , COVID-19/complications , COVID-19/epidemiology , COVID-19/therapy , Cardiomyopathies/virology , Gene Expression , Humans , Immune System/physiology , Myocardium/enzymology , Myocytes, Cardiac/enzymology , Neuropilin-1/metabolism , Platelet Activation , RNA, Messenger/metabolism , Renin-Angiotensin System/physiology , Return to Sport , Risk Factors , SARS-CoV-2/ultrastructure , Spike Glycoprotein, Coronavirus/metabolism , Troponin/metabolism , Ventricular Remodeling , Virus Attachment , Virus Internalization/drug effects
8.
Circ Arrhythm Electrophysiol ; 14(2): e009203, 2021 02.
Article in English | MEDLINE | ID: mdl-33591816

ABSTRACT

Cardiac sarcoidosis is a component of an often multiorgan granulomatous disease of still uncertain cause. It is being recognized with increasing frequency, mainly as the result of heightened awareness and new diagnostic tests, specifically cardiac magnetic resonance imaging and 18F-fluorodeoxyglucose positron emission tomography scans. The purpose of this case-based review is to highlight the potentially life-saving importance of making the early diagnosis of cardiac sarcoidosis using these new tools and to provide a framework for the optimal care of patients with this disease. We will review disease mechanisms as currently understood, associated arrhythmias including conduction abnormalities, and atrial and ventricular tachyarrhythmias, guideline-directed diagnostic criteria, screening of patients with extracardiac sarcoidosis, and the use of pacemakers and defibrillators in this setting. Treatment options, including those related to heart failure, and those which may help clarify disease mechanisms are included.


Subject(s)
Arrhythmias, Cardiac/etiology , Cardiomyopathies/complications , Heart Conduction System/physiopathology , Heart Rate/physiology , Sarcoidosis/complications , Arrhythmias, Cardiac/physiopathology , Humans
9.
Infect Immun ; 89(1)2020 12 15.
Article in English | MEDLINE | ID: mdl-33077627

ABSTRACT

This study investigated responses to Toll-like receptor 2 (TLR2)-driven extracellular signal-related kinase (ERK) signaling in dendritic cells (DCs) versus macrophages. TLR2 signaling was induced with Pam3Cys-Ser-Lys4, and the role of ERK signaling was interrogated pharmacologically with MEK1/2 inhibitor U0126 or genetically with bone marrow-derived macrophages or DCs from Tpl2-/- mice. We assessed cytokine production via enzyme-linked immunosorbent assay (ELISA) or V-Plex, and mRNA levels were assessed via reverse transcriptase quantitative PCR (qRT-PCR). In macrophages, blockade of ERK signaling by pharmacologic or genetic approaches inhibited interleukin 10 (IL-10) expression and increased expression of the p40 subunit shared by IL-12 and IL-23 (IL-12/23p40). In DCs, blockade of ERK signaling similarly inhibited IL-10 expression but decreased IL-12/23p40 expression, which is opposite to the effect of ERK signaling blockade on IL-12/23p40 in macrophages. This difference in IL-12/23p40 regulation correlated with the differential expression of transcription factors cFos and IRF1, which are known to regulate IL-12 family members, including IL-12 and IL-23. Thus, the impact of ERK signaling in response to TLR2 stimulation differs between macrophages and DCs, potentially regulating their distinctive functions in the immune system. ERK-mediated suppression of IL-12/23p40 in macrophages may prevent excessive inflammation and associated tissue damage following TLR2-stimulation, while ERK-mediated induction of IL-12/23p40 in DCs may promote priming of T helper 1 (Th1) responses. A greater understanding of the role that ERK signaling plays in different immune cell types may inform the development of host-directed therapy and optimal adjuvanticity for a number of infectious pathogens.


Subject(s)
Dendritic Cells/metabolism , Interleukin-12/metabolism , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System , Macrophages/metabolism , Proto-Oncogene Proteins/metabolism , Toll-Like Receptor 2/metabolism , Animals , Arginase/genetics , Arginase/metabolism , Dendritic Cells/immunology , Disease Models, Animal , Gene Expression Regulation , Interleukin-10/metabolism , MAP Kinase Kinase Kinases/genetics , Macrophages/immunology , Mice , Mice, Knockout , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Proto-Oncogene Proteins/genetics
10.
JACC Clin Electrophysiol ; 6(8): 1053-1066, 2020 08.
Article in English | MEDLINE | ID: mdl-32819525

ABSTRACT

Coronavirus disease 2019 (COVID-19) has presented substantial challenges to patient care and impacted health care delivery, including cardiac electrophysiology practice throughout the globe. Based upon the undetermined course and regional variability of the pandemic, there is uncertainty as to how and when to resume and deliver electrophysiology services for arrhythmia patients. This joint document from representatives of the Heart Rhythm Society, American Heart Association, and American College of Cardiology seeks to provide guidance for clinicians and institutions reestablishing safe electrophysiological care. To achieve this aim, we address regional and local COVID-19 disease status, the role of viral screening and serologic testing, return-to-work considerations for exposed or infected health care workers, risk stratification and management strategies based on COVID-19 disease burden, institutional preparedness for resumption of elective procedures, patient preparation and communication, prioritization of procedures, and development of outpatient and periprocedural care pathways.


Subject(s)
Arrhythmias, Cardiac/diagnosis , Arrhythmias, Cardiac/therapy , Cardiology , Coronavirus Infections/epidemiology , Delivery of Health Care , Electrophysiologic Techniques, Cardiac , Pneumonia, Viral/epidemiology , Ambulatory Care , American Heart Association , Betacoronavirus , COVID-19 , COVID-19 Testing , Clinical Laboratory Techniques , Coronavirus Infections/diagnosis , Coronavirus Infections/prevention & control , Decision Making, Shared , Health Personnel , Humans , Mass Screening , Organizational Policy , Pandemics/prevention & control , Patient Selection , Personal Protective Equipment/supply & distribution , Pneumonia, Viral/diagnosis , Pneumonia, Viral/prevention & control , Return to Work , Risk Assessment , SARS-CoV-2 , Telemedicine , United States/epidemiology
11.
EBioMedicine ; 58: 102907, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32771682

ABSTRACT

BACKGROUND: SARS-CoV-2 enters cells by binding of its spike protein to angiotensin-converting enzyme 2 (ACE2). Angiotensin-converting enzyme inhibitors (ACEIs) or angiotensin II receptor blockers (ARBs) have been reported to increase ACE2 expression in animal models, and worse outcomes are reported in patients with co-morbidities commonly treated with these agents, leading to controversy during the COVID-19 pandemic over whether these drugs might be helpful or harmful. METHODS: Animal, in vitro and clinical data relevant to the biology of the renin-angiotensin system (RAS), its interaction with the kallikrein-kinin system (KKS) and SARS-CoV-2, and clinical studies were reviewed. FINDINGS AND INTERPRETATION: SARS-CoV-2 hijacks ACE2to invade and damage cells, downregulating ACE2, reducing its protective effects and exacerbating injurious Ang II effects. However, retrospective observational studies do not show higher risk of infection with ACEI or ARB use. Nevertheless, study of the RAS and KKS in the setting of coronaviral infection may yield therapeutic targets.


Subject(s)
Angiotensin Receptor Antagonists/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Coronavirus Infections/drug therapy , Peptidyl-Dipeptidase A/metabolism , Pneumonia, Viral/drug therapy , Angiotensin Receptor Antagonists/pharmacology , Angiotensin-Converting Enzyme 2 , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Betacoronavirus/pathogenicity , COVID-19 , Coronavirus Infections/metabolism , Coronavirus Infections/pathology , Coronavirus Infections/virology , Humans , Kallikrein-Kinin System/drug effects , Pandemics , Peptidyl-Dipeptidase A/genetics , Pneumonia, Viral/metabolism , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , Renin-Angiotensin System/drug effects , SARS-CoV-2
12.
Circ Arrhythm Electrophysiol ; 13(7): e008999, 2020 07.
Article in English | MEDLINE | ID: mdl-32530306

ABSTRACT

Coronavirus disease 2019 (COVID-19) has presented substantial challenges to patient care and impacted healthcare delivery, including cardiac electrophysiology practice throughout the globe. Based upon the undetermined course and regional variability of the pandemic, there is uncertainty as to how and when to resume and deliver electrophysiology services for patients with arrhythmia. This joint document from representatives of the Heart Rhythm Society, American Heart Association, and American College of Cardiology seeks to provide guidance for clinicians and institutions reestablishing safe electrophysiological care. To achieve this aim, we address regional and local COVID-19 disease status, the role of viral screening and serological testing, return-to-work considerations for exposed or infected health care workers, risk stratification and management strategies based on COVID-19 disease burden, institutional preparedness for resumption of elective procedures, patient preparation and communication, prioritization of procedures, and development of outpatient and periprocedural care pathways.


Subject(s)
Arrhythmias, Cardiac/surgery , Coronavirus Infections/epidemiology , Elective Surgical Procedures/statistics & numerical data , Infection Control/organization & administration , Pandemics/statistics & numerical data , Pneumonia, Viral/epidemiology , Practice Guidelines as Topic , American Heart Association , Arrhythmias, Cardiac/diagnostic imaging , Arrhythmias, Cardiac/epidemiology , COVID-19 , COVID-19 Testing , Cardiac Electrophysiology , Cardiac Imaging Techniques , Catheter Ablation/methods , Catheter Ablation/statistics & numerical data , Clinical Laboratory Techniques/methods , Coronavirus Infections/diagnosis , Coronavirus Infections/prevention & control , Elective Surgical Procedures/methods , Female , Humans , Male , Outcome Assessment, Health Care , Pandemics/prevention & control , Pneumonia, Viral/prevention & control , Societies, Medical , United States
13.
Heart Rhythm ; 17(9): e242-e254, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32540298

ABSTRACT

Coronavirus disease 2019 (COVID-19) has presented substantial challenges to patient care and impacted health care delivery, including cardiac electrophysiology practice throughout the globe. Based upon the undetermined course and regional variability of the pandemic, there is uncertainty as to how and when to resume and deliver electrophysiology services for arrhythmia patients. This joint document from representatives of the Heart Rhythm Society, American Heart Association, and American College of Cardiology seeks to provide guidance for clinicians and institutions reestablishing safe electrophysiological care. To achieve this aim, we address regional and local COVID-19 disease status, the role of viral screening and serologic testing, return-to-work considerations for exposed or infected health care workers, risk stratification and management strategies based on COVID-19 disease burden, institutional preparedness for resumption of elective procedures, patient preparation and communication, prioritization of procedures, and development of outpatient and periprocedural care pathways.


Subject(s)
Betacoronavirus , Cardiac Electrophysiology/organization & administration , Coronavirus Infections/prevention & control , Infection Control/organization & administration , Pandemics/prevention & control , Pneumonia, Viral/prevention & control , COVID-19 , Coronavirus Infections/diagnosis , Coronavirus Infections/epidemiology , Humans , Patient Selection , Pneumonia, Viral/diagnosis , Pneumonia, Viral/epidemiology , SARS-CoV-2 , Telemedicine
14.
Am J Clin Pathol ; 154(2): 190-200, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32451533

ABSTRACT

OBJECTIVES: To report methods and findings of 2 autopsies with molecular evaluation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) positive individuals. METHODS: Postmortem examination was completed following Centers for Disease Control and Prevention public guidelines. Numerous formalin-fixed paraffin-embedded (FFPE) tissue types from each case were surveyed for SARS-CoV-2 RNA by quantitative reverse transcription polymerase chain reaction (qRT-PCR). SARS-CoV-2 viral genome was sequenced by next-generation sequencing (NGS) from FFPE lung tissue blocks. RESULTS: Postmortem examinations revealed diffuse alveolar damage, while no viral-associated hepatic, cardiac, or renal damage was observed. Viral RNA was detected in lungs, bronchi, lymph nodes, and spleen in both cases using qRT-PCR method. RNA sequencing using NGS in case 1 revealed mutations most consistent with Western European Clade A2a with ORF1a L3606F mutation. CONCLUSIONS: SARS-CoV-2 testing and viral sequencing can be performed from FFPE tissue. Detection and sequencing of SARS-CoV-2 in combination with morphological findings from postmortem tissue examination can aid in gaining a better understanding of the virus's pathophysiologic effects on human health.


Subject(s)
Betacoronavirus/isolation & purification , Clinical Laboratory Techniques/methods , Coronavirus Infections/diagnosis , Lung/pathology , Molecular Diagnostic Techniques/methods , Pneumonia, Viral/diagnosis , Aged, 80 and over , Autopsy , Betacoronavirus/genetics , COVID-19 , COVID-19 Testing , Coronavirus Infections/pathology , Coronavirus Infections/virology , Fatal Outcome , High-Throughput Nucleotide Sequencing , Humans , Lung/virology , Male , Middle Aged , Pandemics , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , RNA, Viral/analysis , RNA, Viral/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , SARS-CoV-2 , Sequence Analysis, RNA/methods , Tissue Fixation/methods
15.
Eur J Haematol ; 103(2): 99-106, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31107976

ABSTRACT

OBJECTIVES: Type II heparin-induced thrombocytopenia (HIT) is mediated by formation of antibodies to platelet factor 4 (PF4)-heparin complexes. We evaluated anti-PF4-heparin-negative samples for the presence of additional anti-platelet and anti-red blood cell (RBC) antibodies using whole-cell platelet/ RBC ELISAs we developed. METHODS: Seventy-three samples tested for anti-PF4-heparin by ELISA were included: 62 tested negative, 9 tested positive, and 2 had equivocal results. Plasma specimens from healthy donors were used as controls. RESULTS: 100% (9/9) anti-PF4-positive samples had anti-platelet antibodies detected by whole-cell platelet ELISA. 42.2% (27/64) anti-PF4-heparin-negative samples were negative for anti-platelet and anti-RBC antibodies. 32.8% (21/64) negative samples showed reactivity to both platelets and RBC; 12.5% (8/64) negative samples were each reactive with either platelet or RBC ELISA, respectively. Additionally, two samples that tested equivocal by anti-PF4-heparin ELISA had antibodies to both platelets and RBC by whole-cell ELISA. CONCLUSIONS: Our study suggests that patients with thrombocytopenia testing negative for anti-PF4-heparin may still harbor antibodies to platelets. However, additional research is needed to determine the significance of these antibodies. Nevertheless, these findings may encourage clinicians to further investigate patients with possible immune-mediated etiologies of thrombocytopenia and anemia.


Subject(s)
Autoantibodies/immunology , Enzyme-Linked Immunosorbent Assay , Heparin/adverse effects , Thrombocytopenia/diagnosis , Thrombocytopenia/etiology , Adult , Aged , Aged, 80 and over , Autoantibodies/blood , Blood Cell Count , Blood Platelets/immunology , Blood Platelets/metabolism , Case-Control Studies , Comorbidity , Enzyme-Linked Immunosorbent Assay/methods , Erythrocytes/immunology , Erythrocytes/metabolism , Female , Heparin/immunology , Humans , Male , Middle Aged , Platelet Activation/immunology , Platelet Factor 4/immunology , Thrombocytopenia/blood
16.
Nat Commun ; 9(1): 4585, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30389917

ABSTRACT

People living with HIV/AIDS on antiretroviral therapy have increased risk of non-AIDS-defining cancers (NADCs). However, the underlying mechanism for development and progression of certain NADCs remains obscure. Here we show that exosomes released from HIV-infected T cells and those purified from blood of HIV-positive patients stimulate proliferation, migration and invasion of oral/oropharyngeal and lung cancer cells. The HIV transactivation response (TAR) element RNA in HIV-infected T-cell exosomes is responsible for promoting cancer cell proliferation and inducing expression of proto-oncogenes and Toll-like receptor 3 (TLR3)-inducible genes. These effects depend on the loop/bulge region of the molecule. HIV-infected T-cell exosomes rapidly enter recipient cells through epidermal growth factor receptor (EGFR) and stimulate ERK1/2 phosphorylation via the EGFR/TLR3 axis. Thus, our findings indicate that TAR RNA-containing exosomes from HIV-infected T cells promote growth and progression of particular NADCs through activation of the ERK cascade in an EGFR/TLR3-dependent manner.


Subject(s)
Disease Progression , Exosomes/metabolism , HIV Infections/metabolism , HIV Long Terminal Repeat/genetics , HIV-1/physiology , Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , ErbB Receptors/metabolism , Exosomes/ultrastructure , Gene Expression Regulation , HEK293 Cells , HIV Infections/blood , Humans , MAP Kinase Signaling System , Mice, Nude , Phosphorylation , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Toll-Like Receptor 3/metabolism
17.
Blood Cells Mol Dis ; 73: 33-37, 2018 11.
Article in English | MEDLINE | ID: mdl-30195626

ABSTRACT

Effects of α-synuclein deficiency on cellular blood components have not been extensively investigated. This study evaluated ultrastructural changes of leukocytes in α-synuclein knockout (KO) mice using electron microscopy (EM). The following ultrastructural characteristics were quantified in leukocytes: mitochondria, primary granules, specific granules (SG), Golgi apparatus (GA), inclusions, rough-endoplasmic reticulum (RER), smooth-endoplasmic reticulum (SER), and cellular projections (CP). EM showed increased numbers or amounts of SG, inclusions, and SER in KO group (5.3 ±â€¯4.5 in WT vs. 14.1 ±â€¯10.3 in KO, p = 0.02; 0.4 ±â€¯0.9 in WT vs. 3.2 ±â€¯2.8 in KO, p = 0.007; and 7.7 ±â€¯6.7 in WT vs. 17.7 ±â€¯12.2 in KO, p = 0.03, respectively). Although CP number was not significantly different between the two groups (13.4 ±â€¯5.3 in WT vs. 16.3 ±â€¯7.5 in KO, p = 0.32), their size and shapes were altered in KO mice. Notably, findings occurred in the setting of significant lymphopenia. α-Synuclein deficiency leads to changes in size and shape of secretory particles and increases in SER, SG, and inclusions, indicating a potential role for α-synuclein in vesicular trafficking in leukocytes. Further studies are needed to elucidate functions mediated by α-synuclein.


Subject(s)
Leukocytes/ultrastructure , alpha-Synuclein/deficiency , Animals , Cell Shape , Cell Surface Extensions , Mice , Mice, Knockout , Microscopy, Electron , Transport Vesicles , alpha-Synuclein/genetics , alpha-Synuclein/pharmacology
18.
Eur J Haematol ; 2018 Jul 28.
Article in English | MEDLINE | ID: mdl-30055066

ABSTRACT

OBJECTIVES: In platelets, α-synuclein is important in calcium-dependent granule release. Notably, cells release α-synuclein in setting of cell damage or death. Therefore, we investigated α-synuclein levels in plasma of single donor platelet (SDP) units during storage. METHODS: Aliquots were obtained from same SDP units for 7 days from day of donation. Additionally, randomly sampled SDP units at same storage time points were also assayed by enzyme-linked immunosorbent assay. RESULTS: α-Synuclein in SDP plasma increased continuously over time at each assayed time point. Significant increases were measured on day 3 (11.7 ± 9.6 ng/mL, P = 0.025), day 5 (15.3 ± 5.9 ng/mL, P = 0.002), and highest on day 7 (23.7 ± 5.6 ng/mL, P < 0.0001) compared to day 0 (1.1 ± 0.8 ng/mL). Similar significant results were obtained in randomly sampled SDP units at same corresponding time points. Flow cytometry showed that platelets had strong expression of α-synuclein and lacked expression of other synucleins. CONCLUSIONS: Increases of α-synuclein during SDP storage is a steady and continuous process that increases with time. Our findings indicate that α-synuclein may represent a biomarker of platelet biological state during storage. Further research will be needed to determine how α-synuclein increases correlate with platelets' function.

19.
Infect Immun ; 86(10)2018 10.
Article in English | MEDLINE | ID: mdl-30037791

ABSTRACT

Mycobacterium tuberculosis causes persistent infection due to its ability to evade host immune responses. M. tuberculosis induces Toll-like receptor 2 (TLR2) signaling, which influences immune responses to M. tuberculosis TLR2 agonists expressed by M. tuberculosis include lipoproteins (e.g., LprG), the glycolipid phosphatidylinositol mannoside 6 (PIM6), and the lipoglycan lipomannan (LM). Another M. tuberculosis lipoglycan, mannose-capped lipoarabinomannan (ManLAM), lacks TLR2 agonist activity. In contrast, PILAM, from Mycobacterum smegmatis, does have TLR2 agonist activity. Our understanding of how M. tuberculosis lipoproteins and lipoglycans interact with TLR2 is limited, and binding of these molecules to TLR2 has not been measured directly. Here, we directly measured M. tuberculosis lipoprotein and lipoglycan binding to TLR2 and its partner receptor, TLR1. LprG, LAM, and LM were all found to bind to TLR2 in the absence of TLR1, but not to TLR1 in the absence of TLR2. Trimolecular interactions were revealed by binding of TLR2-LprG or TLR2-PIM6 complexes to TLR1, whereas binding of TLR2 to TLR1 was not detected in the absence of the lipoprotein or glycolipid. ManLAM exhibited low affinity for TLR2 in comparison to PILAM, LM, and LprG, which correlated with reduced ability of ManLAM to induce TLR2-mediated extracellular-signal-regulated kinase (ERK) activation and tumor necrosis factor alpha (TNF-α) secretion in macrophages. We provide the first direct affinity measurement and kinetic analysis of M. tuberculosis lipoprotein and lipoglycan binding to TLR2. Our results demonstrate that binding affinity correlates with the functional ability of agonists to induce TLR2 signaling.


Subject(s)
Bacterial Proteins/metabolism , Lipopolysaccharides/metabolism , Lipoproteins/metabolism , Mycobacterium tuberculosis/metabolism , Toll-Like Receptor 2/metabolism , Tuberculosis/metabolism , Animals , Bacterial Proteins/genetics , Female , Humans , Lipopolysaccharides/genetics , Lipoproteins/genetics , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/genetics , Protein Binding , Signal Transduction , Toll-Like Receptor 1/genetics , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 2/genetics , Tuberculosis/genetics , Tuberculosis/microbiology
20.
Acad Med ; 93(2): 150-151, 2018 02.
Article in English | MEDLINE | ID: mdl-29377855

Subject(s)
Physicians
SELECTION OF CITATIONS
SEARCH DETAIL
...