Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Fish Shellfish Immunol ; 100: 179-185, 2020 May.
Article in English | MEDLINE | ID: mdl-32126245

ABSTRACT

In this study, a new continuous muscle cell line, LYCMS (large yellow croaker muscle cell line), derived from the muscle tissue of larva of large yellow croaker (Larimichthys crocea) was developed with modified DMEM/F12 medium at 27 °C. The muscle cell line could be passaged at different ratios for different growth rates. Karyotype analysis showed that a large proportion of LYCMS cells had 48 chromosomes. The proliferation of LYCMS cell line could be affected by mammalian growth factors such as human basic fibroblast growth factor (b-FGF), epidermal growth factor (EGF), and hepatocyte growth factor (HGF). GFP expression experiments indicated that the LYCMS cell line could be used for exogenous genes' expression. Different virus response-related genes tested in this study showed diverse change types in expression before and after (0-24 h) polycytidylic acid (poly I: C) challenge of LYCMS cells. This is the first study of virus response signaling pathways of large yellow croaker based on the muscle cell line. The results showed that compared with the in vivo experiments, the use of the LYCMS cell line for immune research is more convenient, efficient, and rapid. By using this model, we demonstrated that MDA5-IPS1-TRAF6-NFκB-cytokines, MDA5-IPS1-TRAF3-IRF3-interferon or TLR22-TRIF-IRF3-interferon, TLR8-MyD88-NFκB-cytokines, and TLR3-TRIF-IRF3-interferon pathways were able to response to poly I: C challenge in the muscle cell line of large yellow croaker.


Subject(s)
Fish Proteins/genetics , Immunity, Innate/genetics , Muscle Cells/drug effects , Muscles/cytology , Perciformes/immunology , Poly I-C/pharmacology , Animals , Cell Line , Fish Proteins/immunology , Gene Expression Profiling , Karyotype , Larva/anatomy & histology , Larva/cytology , Muscle Cells/immunology , Signal Transduction , Tissue Culture Techniques , Viruses
2.
Proc Natl Acad Sci U S A ; 117(6): 2751-2760, 2020 02 11.
Article in English | MEDLINE | ID: mdl-31980524

ABSTRACT

Obesity is associated with a chronic state of low-grade inflammation and progressive tissue infiltration by immune cells and increased expression of inflammatory cytokines. It is established that interleukin 6 (IL6) regulates multiple aspects of metabolism, including glucose disposal, lipolysis, oxidative metabolism, and energy expenditure. IL6 is secreted by many tissues, but the role of individual cell types is unclear. We tested the role of specific cells using a mouse model with conditional expression of the Il6 gene. We found that IL6 derived from adipocytes increased, while IL6 derived from myeloid cells and muscle suppressed, macrophage infiltration of adipose tissue. These opposite actions were associated with a switch of IL6 signaling from a canonical mode (myeloid cells) to a noncanonical trans-signaling mode (adipocytes and muscle) with increased expression of the ADAM10/17 metalloprotease that promotes trans-signaling by the soluble IL6 receptor α. Collectively, these data demonstrate that the source of IL6 production plays a major role in the physiological regulation of metabolism.


Subject(s)
Adipose Tissue/immunology , Interleukin-6/immunology , Obesity/immunology , ADAM10 Protein/genetics , ADAM10 Protein/immunology , ADAM17 Protein/genetics , ADAM17 Protein/immunology , Adipocytes/immunology , Animals , Female , Humans , Interleukin-6/genetics , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Muscle Cells/immunology , Myeloid Cells/immunology , Obesity/genetics , Species Specificity
3.
Immunity ; 51(5): 899-914.e7, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31732166

ABSTRACT

Myocardial infarction, stroke, and sepsis trigger systemic inflammation and organism-wide complications that are difficult to manage. Here, we examined the contribution of macrophages residing in vital organs to the systemic response after these injuries. We generated a comprehensive catalog of changes in macrophage number, origin, and gene expression in the heart, brain, liver, kidney, and lung of mice with myocardial infarction, stroke, or sepsis. Predominantly fueled by heightened local proliferation, tissue macrophage numbers increased systemically. Macrophages in the same organ responded similarly to different injuries by altering expression of tissue-specific gene sets. Preceding myocardial infarction improved survival of subsequent pneumonia due to enhanced bacterial clearance, which was caused by IFNÉ£ priming of alveolar macrophages. Conversely, EGF receptor signaling in macrophages exacerbated inflammatory lung injury. Our data suggest that local injury activates macrophages in remote organs and that targeting macrophages could improve resilience against systemic complications following myocardial infarction, stroke, and sepsis.


Subject(s)
Disease Susceptibility , Macrophages/immunology , Macrophages/metabolism , Animals , Biomarkers , Cell Count , Disease Susceptibility/immunology , ErbB Receptors/metabolism , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Ischemia/etiology , Ischemia/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mice , Muscle Cells/immunology , Muscle Cells/metabolism , Myocardial Infarction/etiology , Myocardial Infarction/metabolism , Organ Specificity/genetics , Organ Specificity/immunology , Pneumonia/etiology , Pneumonia/metabolism , Pneumonia/pathology
4.
Parasitol Res ; 117(5): 1647-1655, 2018 May.
Article in English | MEDLINE | ID: mdl-29550999

ABSTRACT

Megaesophagus is one of the major manifestations of the chronic phase of Chagas disease. Its primary symptom is generally dysphagia due to disturbance in the lower esophageal sphincter. Microscopically, the affected organ presents denervation, which has been considered as consequence of an inflammatory process that begins at the acute phase and persists in the chronic phase. Inflammatory infiltrates are composed of lymphocytes, macrophages, natural killer cells, mast cells, and eosinophils. In this study, we evaluated the immunoreactivity of nerve growth factor (NGF), and of its receptor tropomyosin receptor kinase A (TrkA), molecules that are well known for having a relevant role in neuroimmune communication in the gastrointestinal tract. Esophageal samples obtained via autopsy or surgery procedures from six noninfected individuals, six infected individuals without megaesophagus, and six infected individuals with megaesophagus were analyzed. Infected individuals without megaesophagus presented increased numbers of NGF immunoreactive (IR) mast cells and increased areas of TrkA-IR epithelial cells and inner muscle cells. Infected individuals with megaesophagus showed increased numbers of NGF-IR eosinophils and mast cells, TrkA-IR eosinophils and mast cells, increased area of NGF-IR epithelial cells, and increased areas of TrkA-IR epithelials cells and inner muscle cells. The data presented here point to the participation of NGF and its TrkA receptor in the pathology of chagasic megaesophagus.


Subject(s)
Chagas Disease/pathology , Esophageal Achalasia/pathology , Nerve Growth Factor/immunology , Receptor, trkA/immunology , Trypanosoma cruzi/pathogenicity , Cell Count , Chagas Disease/parasitology , Eosinophils/immunology , Esophageal Achalasia/parasitology , Esophagus/parasitology , Esophagus/pathology , Female , Humans , Macrophages/immunology , Male , Mast Cells/immunology , Middle Aged , Muscle Cells/immunology , Neurons/metabolism , Parasite Load , Protein Kinases , Tropomyosin/metabolism , Trypanosoma cruzi/isolation & purification
5.
Sci Transl Med ; 10(424)2018 01 17.
Article in English | MEDLINE | ID: mdl-29343625

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) is a major cause of morbidity and mortality worldwide and is a frequent cause of skin and soft tissue infections (SSTIs). Lymphedema-fluid accumulation in tissue caused by impaired lymphatic vessel function-is a strong risk factor for SSTIs. SSTIs also frequently recur in patients and sometimes lead to acquired lymphedema. However, the mechanism of how SSTIs can be both the consequence and the cause of lymphatic vessel dysfunction is not known. Intravital imaging in mice revealed an acute reduction in both lymphatic vessel contractility and lymph flow after localized MRSA infection. Moreover, chronic lymphatic impairment is observed long after MRSA is cleared and inflammation is resolved. Associated with decreased collecting lymphatic vessel function was the loss and disorganization of lymphatic muscle cells (LMCs), which are critical for lymphatic contraction. In vitro, incubation with MRSA-conditioned supernatant led to LMC death. Proteomic analysis identified several accessory gene regulator (agr)-controlled MRSA exotoxins that contribute to LMC death. Infection with agr mutant MRSA resulted in sustained lymphatic function compared to animals infected with wild-type MRSA. Our findings suggest that agr is a promising target to preserve lymphatic vessel function and promote immunity during SSTIs.


Subject(s)
Lymphatic Vessels/immunology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Animals , Cell Survival/physiology , Cells, Cultured , Female , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Inflammation/immunology , Male , Mice, Inbred C57BL , Microbial Sensitivity Tests , Muscle Cells/immunology
6.
Infect Immun ; 86(1)2018 01.
Article in English | MEDLINE | ID: mdl-28993459

ABSTRACT

The histozoic myxozoan parasite Kudoa thyrsites causes postmortem myoliquefaction and is responsible for economic losses to salmon aquaculture in the Pacific Northwest. Despite its importance, little is known about the host-parasite relationship, including the host response to infection. The present work sought to characterize the immune response in Atlantic salmon during infection, recovery, and reexposure to K. thyrsites After exposure to infective seawater, infected and uninfected smolts were sampled three times over 4,275 degree-days. Histological analysis revealed infection severity decreased over time in exposed fish, while in controls there was no evidence of infection. Following a secondary exposure of all fish, severity of infection in the controls was similar to that measured in exposed fish at the first sampling time but was significantly reduced in reexposed fish, suggesting the acquisition of protective immunity. Using immunohistochemistry, we detected a population of MHIIß+ cells in infected muscle that followed a pattern of abundance concordant with parasite prevalence. Infiltration of these cells into infected myocytes preceded destruction of the plasmodium and dissemination of myxospores. Dual labeling indicated a majority of these cells were CD83+/MHIIß+ Using reverse transcription-quantitative PCR, we detected significant induction of cellular effectors, including macrophage/dendritic cells (mhii/cd83/mcsf), B cells (igm/igt), and cytotoxic T cells (cd8/nkl), in the musculature of infected fish. These data support a role for cellular effectors such as antigen-presenting cells (monocyte/macrophage and dendritic cells) along with B and T cells in the acquired protective immune response of Atlantic salmon against K. thyrsites.


Subject(s)
Adaptive Immunity/immunology , Antigen-Presenting Cells/immunology , Antigens, CD/immunology , Immunoglobulins/immunology , Membrane Glycoproteins/immunology , Myxozoa/immunology , Salmo salar/immunology , Salmo salar/parasitology , Salmon/immunology , Salmon/parasitology , Animals , Antigen-Presenting Cells/parasitology , Aquaculture/methods , B-Lymphocytes/immunology , B-Lymphocytes/parasitology , Dendritic Cells/immunology , Dendritic Cells/parasitology , Fish Diseases/immunology , Fish Diseases/parasitology , Host-Parasite Interactions/immunology , Macrophages/immunology , Macrophages/parasitology , Muscle Cells/immunology , Muscle Cells/parasitology , Muscle, Skeletal/immunology , Muscle, Skeletal/parasitology , Parasitic Diseases, Animal/immunology , Parasitic Diseases, Animal/parasitology , T-Lymphocytes/immunology , T-Lymphocytes/parasitology , CD83 Antigen
7.
Sci Rep ; 7(1): 8580, 2017 08 17.
Article in English | MEDLINE | ID: mdl-28819261

ABSTRACT

Modified Vaccinia virus Ankara (MVA) is a promising vaccine vector with an excellent safety profile. However, despite extensive pre-clinical and clinical testing, surprisingly little is known about the cellular tropism of MVA, especially in relevant animal species. Here, we performed in vitro, ex vivo and in vivo experiments with recombinant MVA expressing green fluorescent protein (rMVA-GFP). In both human peripheral blood mononuclear cells and mouse lung explants, rMVA-GFP predominantly infected antigen presenting cells. Subsequent in vivo experiments performed in mice, ferrets and non-human primates indicated that preferential targeting of dendritic cells and alveolar macrophages was observed after respiratory administration, although subtle differences were observed between the respective animal species. Following intramuscular injection, rMVA-GFP was detected in interdigitating cells between myocytes, but also in myocytes themselves. These data are important in advancing our understanding of the basis for the immunogenicity of MVA-based vaccines and aid rational vaccine design and delivery strategies.


Subject(s)
Antigen-Presenting Cells/immunology , Leukocytes, Mononuclear/immunology , Vaccinia virus/immunology , Viral Vaccines/immunology , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/virology , Ferrets , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Macaca fascicularis , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/virology , Mice , Microscopy, Confocal , Muscle Cells/immunology , Muscle Cells/metabolism , Muscle Cells/virology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Vaccinia virus/genetics , Vaccinia virus/physiology
8.
Curr Opin Immunol ; 38: 94-100, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26741528

ABSTRACT

Alarmins are a heterogeneous group of endogenous molecules that signal cellular damage when sensed extracellularly. Heme is an endogenous molecule that acts as a prosthetic group of hemoproteins, such as hemoglobin and myoglobin. When released from damaged red blood cells or muscle cells, oxidized hemoglobin and myoglobin release their prosthetic heme groups, respectively. This generates labile heme, which is sensed by pattern recognition receptors (PRR) expressed by innate immune cells and possibly regulatory T cells (TREG). The ensuing adaptive response, which alerts for the occurrence of red blood cell or muscle cell damage, regulates the pathologic outcome of hemolysis or rhabdomyolysis, respectively. In conclusion, we propose that labile heme is an alarmin.


Subject(s)
Alarmins/immunology , Gene Expression Regulation/immunology , Heme/immunology , Immunity, Innate , Reactive Oxygen Species/immunology , Adaptive Immunity , Alarmins/metabolism , Animals , Endothelial Cells/cytology , Endothelial Cells/immunology , Erythrocytes/chemistry , Erythrocytes/immunology , Heme/metabolism , Humans , Macrophages/cytology , Macrophages/immunology , Muscle Cells/chemistry , Muscle Cells/immunology , Neutrophils/cytology , Neutrophils/immunology , Reactive Oxygen Species/metabolism , Receptors, Pattern Recognition/genetics , Receptors, Pattern Recognition/immunology , Signal Transduction
9.
Cardiovasc Diabetol ; 14: 56, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25986700

ABSTRACT

BACKGROUND: Endotoxin (i.e. LPS) administration induces a robust inflammatory response with accompanying cardiovascular dysfunction and insulin resistance. Overabundance of nitric oxide (NO) contributes to the vascular dysfunction. However, inflammation itself also induces insulin resistance in skeletal muscle. We sought to investigate whether the cardiovascular dysfunction induced by increased NO availability without inflammatory stress can promote insulin resistance. Additionally, we examined the role of inducible nitric oxide synthase (iNOS or NOS2), the source of the increase in NO availability, in modulating LPS-induced decrease in insulin-stimulated muscle glucose uptake (MGU). METHODS: The impact of NO donor infusion on insulin-stimulated whole-body and muscle glucose uptake (hyperinsulinemic-euglycemic clamps), and the cardiovascular system was assessed in chronically catheterized, conscious mice wild-type (WT) mice. The impact of LPS on insulin action and the cardiovascular system were assessed in WT and global iNOS knockout (KO) mice. Tissue blood flow and cardiac function were assessed using microspheres and echocardiography, respectively. Insulin signaling activity, and gene expression of pro-inflammatory markers were also measured. RESULTS: NO donor infusion decreased mean arterial blood pressure, whole-body glucose requirements, and MGU in the absence of changes in skeletal muscle blood flow. LPS lowered mean arterial blood pressure and glucose requirements in WT mice, but not in iNOS KO mice. Lastly, despite an intact inflammatory response, iNOS KO mice were protected from LPS-mediated deficits in cardiac output. LPS impaired MGU in vivo, regardless of the presence of iNOS. However, ex vivo, insulin action in muscle obtained from LPS treated iNOS KO animals was protected. CONCLUSION: Nitric oxide excess and LPS impairs glycemic control by diminishing MGU. LPS impairs MGU by both the direct effect of inflammation on the myocyte, as well as by the indirect NO-driven cardiovascular dysfunction.


Subject(s)
Endothelium-Dependent Relaxing Factors/pharmacology , Glucose/metabolism , Heart/drug effects , Insulin Resistance , Lipopolysaccharides/pharmacology , Muscle, Skeletal/drug effects , Nitric Oxide Synthase Type II/genetics , Nitric Oxide/pharmacology , Animals , Arterial Pressure/drug effects , Cardiac Output/drug effects , Chemokine CCL2/genetics , Echocardiography , Gene Expression , Glucose Clamp Technique , Inflammation , Interleukin-6/genetics , Mice , Mice, Knockout , Microspheres , Muscle Cells/drug effects , Muscle Cells/immunology , Muscle Cells/metabolism , Muscle, Skeletal/immunology , Muscle, Skeletal/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Regional Blood Flow/drug effects , Serpin E2/genetics , Tumor Necrosis Factor-alpha/genetics
10.
Am J Physiol Gastrointest Liver Physiol ; 308(10): G852-62, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25747352

ABSTRACT

Myocytes are nonhemopoietic in origin and functionally essential in generating gastrointestinal motility. In endotoxemia, a rapid-onset nonhemopoietic mechanism potently triggers early ileus in a Toll-like receptor 4 (TLR4)/myeloid differentiation primary response gene 88 (MyD88)-dependent manner. Moreover, synergistically with hemopoietic cells, nonhemopoietic cells escalate late ileus via an IL-6 receptor-dependent inflammation-driven pathway. We therefore specifically investigated the role of myocytes in TLR4-triggered inflammation and ileus. TLR4(+/+), TLR4(-/-), bmTLR4(+/+)/TLR4(-/-) chimera, SM22-Cre(-/-)TLR4(flox/flox), and selective myocyte TLR4-deficient (SM22-Cre(+/-)TLR4(flox/flox)) mice were injected intraperitoneally with purified lipopolysaccharide. SM22-driven Cre recombinase activity was selectively detected in cardiac, gastrointestinal, skeletal, and vascular myocytes, of small-sized vessels in a two-color fluorescent Cre reporter mouse. In contrast to nonhemopoietic TLR4 deficiency, deletion of myocyte TLR4 signaling prevented neither endotoxin-induced suppression of spontaneous jejunal contractility in vitro nor early ileus in vivo at 6 h. Circulating plasma colony-stimulating factor 3 was greatly elevated during endotoxemia, independent of myocyte TLR4 signaling or time. TLR4 activation of myocytes contributed significantly to an early enteric IL-6 mRNA induction and systemic IL-6 release, as well as to a late increase in circulating chemokine (C-X-C motif) ligand 1 (CXCL1) and IL-17. Consequently, inhibition of myocyte TLR4 signaling allowed functional recovery of motility by preventing inflammation-driven late ileus at 24 h. Direct TLR4 activation of myocytes is not responsible for nonhemopoietic-mediated early ileus. However, myocytes are proinflammatory cells that potently drive enteric and systemic inflammation, subsequently fueling late mediator-triggered ileus. Specifically, the myocyte TLR4-dependent inflammatory signature of elevated plasma IL-6, CXCL1, and IL-17 is strongly associated with late rodent ileus.


Subject(s)
Chemokines/immunology , Ileitis/immunology , Ileitis/pathology , Ileus/immunology , Ileus/pathology , Muscle Cells/immunology , Toll-Like Receptor 4/immunology , Animals , Ileitis/chemically induced , Immunologic Factors/immunology , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Muscle Cells/pathology
11.
Trends Endocrinol Metab ; 25(9): 444-51, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24997879

ABSTRACT

Adiponectin, one of the most abundant adipose-derived hormones, has variable actions in many tissues and organs. Although principally known for its insulin-sensitizing activity, recent data also highlight its homeostatic function, which is mediated both by direct actions on metabolic cells and indirectly through immunomodulatory effects on immune cells. Here we review the multifaceted immunometabolic actions of adiponectin and attempt to unify some of the contradictory reports on adiponectin function in inflammatory processes. We propose that a holistic understanding of adiponectin function can be garnered only from understanding its actions both on the immune system and on metabolism.


Subject(s)
Adiponectin/metabolism , Energy Metabolism , Homeostasis , Immunologic Factors/metabolism , Models, Biological , Animals , Humans , Insulin Resistance , Kupffer Cells/immunology , Kupffer Cells/metabolism , Macrophages/immunology , Macrophages/metabolism , Muscle Cells/immunology , Muscle Cells/metabolism , Receptors, Adiponectin/agonists , Receptors, Adiponectin/metabolism , Signal Transduction , Synovial Membrane/immunology , Synovial Membrane/metabolism
12.
Cell Death Dis ; 5: e1002, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24434510

ABSTRACT

Adaptive responses of skeletal muscle regulate the nuclear shuttling of the sarcomeric protein Ankrd2 that can transduce different stimuli into specific adaptations by interacting with both structural and regulatory proteins. In a genome-wide expression study on Ankrd2-knockout or -overexpressing primary proliferating or differentiating myoblasts, we found an inverse correlation between Ankrd2 levels and the expression of proinflammatory genes and identified Ankrd2 as a potent repressor of inflammatory responses through direct interaction with the NF-κB repressor subunit p50. In particular, we identified Gsk3ß as a novel direct target of the p50/Ankrd2 repressosome dimer and found that the recruitment of p50 by Ankrd2 is dependent on Akt2-mediated phosphorylation of Ankrd2 upon oxidative stress during myogenic differentiation. Surprisingly, the absence of Ankrd2 in slow muscle negatively affected the expression of cytokines and key calcineurin-dependent genes associated with the slow-twitch muscle program. Thus, our findings support a model in which alterations in Ankrd2 protein and phosphorylation levels modulate the balance between physiological and pathological inflammatory responses in muscle.


Subject(s)
Cell Differentiation , Muscle Cells/cytology , Muscle Proteins/immunology , Muscle, Skeletal/cytology , NF-kappa B/immunology , Nuclear Proteins/immunology , Repressor Proteins/immunology , Animals , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Cells/immunology , Muscle Proteins/genetics , Muscle, Skeletal/immunology , NF-kappa B/genetics , Nuclear Proteins/genetics , Protein Binding , Repressor Proteins/genetics
13.
Vet Parasitol ; 194(2-4): 150-4, 2013 May 20.
Article in English | MEDLINE | ID: mdl-23462254

ABSTRACT

The nurse cell (NC), formed from muscle cells upon infection with the parasitic nematode Trichinella spp. constitutes a confined habitat for muscle larvae of encapsulating species. Signaling pathway-directed analysis of microarray data allowed identification of the stage of NC cell cycle arrest as being of G1-like type, accompanied by cellular senescence. In accord with the specificity of senescent cellular systems, up-regulation of pro-inflammatory molecules was also found within the NC preparations. Potential immune-related activities associated with NCs as inferred from the aforementioned analysis, are reviewed herein. Transcriptional data suggest that the NC which harbors the larvae may exhibit the following immune-related functions: (i) production of complement components, (ii) antigen presentation and phagocytosis, (iii) pro-inflammatory cytokine secretion, (iv) oxidative stress generation and (v) eicosanoid synthesis.


Subject(s)
Antigens, Helminth/immunology , Inflammation/immunology , Muscle Cells/immunology , Signal Transduction/immunology , Trichinella/immunology , Trichinellosis/immunology , Animals , Antigen Presentation , Cytokines/metabolism , Eicosanoids/metabolism , Gene Expression Regulation/immunology , Larva , Muscle Cells/metabolism , Muscle Cells/parasitology , Phagocytosis , Phenotype , Respiratory Burst , Trichinellosis/parasitology
14.
Nat Immunol ; 14(1): 1-5, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23238748

ABSTRACT

Antibody gene transfer, which involves the delivery of genes that encode potent, broadly neutralizing antibodies to human immunodeficiency virus (HIV), is a promising new strategy for preventing HIV infection. A satellite symposium at the AIDS Vaccine 2012 conference brought together many of the groups working in this field.


Subject(s)
AIDS Vaccines/genetics , Antibodies, Neutralizing/genetics , Antibodies, Viral/genetics , Gene Transfer Techniques , HIV Infections/prevention & control , HIV/immunology , Animals , CD4 Immunoadhesins/genetics , CD4 Immunoadhesins/immunology , Clinical Trials as Topic , Dependovirus , Disease Models, Animal , Genetic Engineering , Genetic Vectors/genetics , Humans , Muscle Cells/immunology , United States
15.
J Immunol Methods ; 385(1-2): 35-44, 2012 Nov 30.
Article in English | MEDLINE | ID: mdl-22910000

ABSTRACT

High titer autoantibodies, which are often associated with specific clinical phenotypes, are useful diagnostically and prognostically in systemic autoimmune diseases. In several autoimmune rheumatic diseases (e.g. myositis and Sjogren's syndrome), 20-40% of patients are autoantibody negative as assessed by conventional assays. The recent discovery of new specificities (e.g., anti-MDA5) in a subset of these autoantibody-negative subjects demonstrates that additional specificities await identification. In this manuscript, we describe a rapid multidimensional method to identify new autoantigens. A central foundation of this rapid approach is the use of an antigen source in which a pathogenic pathway active in the disease is recapitulated. Additionally, the method involves a modified serological proteome analysis strategy which allows confirmation that the correct gel plug has been removed prior to sending for sequencing. Lastly, the approach uses multiple sources of information to enable rapid triangulation and identification of protein candidates. Possible permutations and underlying principles of this triangulation strategy are elaborated to demonstrate the broad utility of this approach for antigen discovery.


Subject(s)
Autoantigens/immunology , Oligonucleotide Array Sequence Analysis/methods , Proteome/immunology , Proteomics/methods , Autoantigens/genetics , Autoantigens/metabolism , Cells, Cultured , Electrophoresis, Gel, Two-Dimensional , HeLa Cells , Humans , Immunoblotting , Interferon-alpha/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Muscle Cells/drug effects , Muscle Cells/immunology , Muscle Cells/metabolism , Muscles/immunology , Muscles/metabolism , Muscles/pathology , Myosin Light Chains/genetics , Myosin Light Chains/immunology , Myosin Light Chains/metabolism , Myositis/blood , Myositis/immunology , Proteome/genetics , Proteome/metabolism , Reproducibility of Results , Tandem Mass Spectrometry
16.
J Immunol ; 188(10): 4876-84, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22491251

ABSTRACT

PD-1, a member of the CD28 family of immune regulatory molecules, is expressed on activated T cells, interacts with its ligands, PD-L1/B7-H1 and PD-L2/B7-DC, on other cells, and delivers inhibitory signals to the T cell. We studied the role of this pathway in modulating autoreactive T cell responses in two models of myocarditis. In a CD8(+) T cell-mediated adoptive transfer model, we found that compared with Pd1(+/+) CD8(+) T cells, Pd1(-/-) CD8(+) T cells cause enhanced disease, with increased inflammatory infiltrate, particularly rich in neutrophils. Additionally, we show enhanced proliferation in vivo and enhanced cytotoxic activity of PD-1-deficient T lymphocytes against myocardial endothelial cells in vitro. In experimental autoimmune myocarditis, a disease model dependent on CD4(+) T cells, we show that mice lacking PD-1 develop enhanced disease compared with wild-type mice. PD-1-deficient mice displayed increased inflammation, enhanced serum markers of myocardial damage, and an increased infiltration of inflammatory cells, including CD8(+) T cells. Together, these studies show that PD-1 plays an important role in limiting T cell responses in the heart.


Subject(s)
Muscle Cells/immunology , Muscle Cells/pathology , Myocarditis/immunology , Myocarditis/pathology , Programmed Cell Death 1 Receptor/physiology , T-Lymphocyte Subsets/immunology , Amino Acid Sequence , Animals , Cell Movement/genetics , Cell Movement/immunology , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Female , Inflammation/genetics , Inflammation/pathology , Inflammation/prevention & control , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Myocarditis/genetics , Programmed Cell Death 1 Receptor/deficiency , Programmed Cell Death 1 Receptor/genetics , T-Lymphocyte Subsets/pathology
17.
Curr Rheumatol Rep ; 14(3): 252-6, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22350607

ABSTRACT

Since the identification of interleukin (IL)-17 as a T-cell-derived cytokine 15 years ago, the contribution of the T-helper type 17 (Th17) pathway in inducing and maintaining chronic inflammation has been well-established, particularly in rheumatoid arthritis. In addition to the main Th1 profile first suggested to contribute to inflammatory myopathies, the presence in inflamed muscle tissue of myositis of IL-17-producing cells, in association with activated dendritic cells, suggests a local activation of the IL-23-Th17 pathway. IL-17 can act on muscle cells together with proinflammatory cytokines produced by monocytes and innate immunity to amplify the immune response that could lead to muscle destruction. Evidence for activation of the Th17 pathway in myositis lesions and in vitro effects of IL-17 on muscle cells suggest IL-17 as a therapeutic target. Inhibitors of IL-17 have been tested in other inflammatory conditions, but the position of IL-17 inhibition in the treatment of inflammatory myopathies remains to be defined.


Subject(s)
Interleukin-17/immunology , Myositis/immunology , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Humans , Immunologic Factors/therapeutic use , Interleukin-17/antagonists & inhibitors , Molecular Targeted Therapy/methods , Muscle Cells/immunology , Myositis/drug therapy , Th17 Cells/immunology
18.
Eur J Cell Biol ; 91(2): 139-49, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22176919

ABSTRACT

Inflammatory myopathies (IMs) are systemic diseases characterized by a T helper (Th) 1 type inflammatory response and cell infiltrates within skeletal muscles. The mainstay of treatment is drugs aimed at suppressing the immune system - corticosteroids and immunosuppressants. About 25% of patients are non-responders. Skeletal muscle cells seem actively involved in the immune-inflammatory response and not only a target; understanding the molecular bases of IMs might help drug development strategies. Within muscles the interaction between the chemokine interferon (IFN)γ inducible 10 kDa protein, CXCL10 or IP-10, and its specific receptor CXCR3, present on Th1 type infiltrating cells, likely plays a pivotal role, potentially offering the opportunity for therapeutic intervention. We aimed to clarify the involvement of human skeletal muscle cells in inflammatory processes in terms of CXCL10 secretion, to elucidate the engaged molecular mechanism(s) and, finally, to evaluate muscular cell responses, if any, to some immunosuppressants routinely used in IM treatment, such as methylprednisolone, methotrexate, cyclosporin A and Infliximab. We first isolated and characterized human fetal skeletal muscle cells (Hfsmc), which expressed the specific lineage markers and showed the competence to react in the context of an in vitro alloresponse. CXCL10 protein secretion by Hfsmc was similarly induced by the inflammatory cytokines interferon (IFN)γ and tumor necrosis factor (TNF)α, above undetectable control levels, through the activation of Stat1 and NF-kB pathways, respectively; CXCL10 secretion was significantly magnified by cytokine combination, and this synergy was associated to a significant up-regulation of TNFαRII; cytokine-induced CXCL10 secretion was considerably affected only by Infliximab. Our data suggested that human skeletal muscle cells might actively self-promote muscular inflammation by eliciting CXCL10 secretion, which is known to amplify Th1 cell tissue infiltration in vivo. In conclusion, we sustain that pharmacological targeting of CXCL10 within muscular cells might contribute to keep in control pro-Th1 polarization of the immune/inflammatory response.


Subject(s)
Chemokine CXCL10/metabolism , Muscle Cells/metabolism , Muscle, Skeletal/metabolism , Myositis/metabolism , Receptors, CXCR3/metabolism , Antibodies, Monoclonal/pharmacology , Cells, Cultured , Chemokine CXCL10/immunology , Cyclosporine/pharmacology , Fetus , Humans , Immunosuppressive Agents/pharmacology , Infliximab , Interferon-gamma/pharmacology , Lymphocyte Activation , Methylprednisolone/pharmacology , Muscle Cells/drug effects , Muscle Cells/immunology , Muscle, Skeletal/drug effects , Muscle, Skeletal/immunology , Myositis/immunology , NF-kappa B/metabolism , Receptors, CXCR3/immunology , STAT1 Transcription Factor/metabolism , Signal Transduction , Th1 Cells/immunology , Tumor Necrosis Factor-alpha/pharmacology
19.
BMC Dev Biol ; 11: 61, 2011 Oct 17.
Article in English | MEDLINE | ID: mdl-22004330

ABSTRACT

BACKGROUND: Determining the type and source of cells involved in regenerative processes has been one of the most important goals of researchers in the field of regeneration biology. We have previously used several cellular markers to characterize the cells involved in the regeneration of the intestine in the sea cucumber Holothuria glaberrima. RESULTS: We have now obtained a monoclonal antibody that labels the mesothelium; the outer layer of the gut wall composed of peritoneocytes and myocytes. Using this antibody we studied the role of this tissue layer in the early stages of intestinal regeneration. We have now shown that the mesothelial cells of the mesentery, specifically the muscle component, undergo dedifferentiation from very early on in the regeneration process. Cell proliferation, on the other hand, increases much later, and mainly takes place in the mesothelium or coelomic epithelium of the regenerating intestinal rudiment. Moreover, we have found that the formation of the intestinal rudiment involves a novel regenerative mechanism where epithelial cells ingress into the connective tissue and acquire mesenchymal phenotypes. CONCLUSIONS: Our results strongly suggest that the dedifferentiating mesothelium provides the initial source of cells for the formation of the intestinal rudiment. At later stages, cell proliferation supplies additional cells necessary for the increase in size of the regenerate. Our data also shows that the mechanism of epithelial to mesenchymal transition provides many of the connective tissue cells found in the regenerating intestine. These results present some new and important information as to the cellular basis of organ regeneration and in particular to the process of regeneration of visceral organs.


Subject(s)
Cell Dedifferentiation/physiology , Epithelial-Mesenchymal Transition/physiology , Holothuria/physiology , Intestinal Mucosa/metabolism , Regeneration/physiology , Animals , Antibodies, Monoclonal , Cell Proliferation , Epithelium/immunology , Holothuria/cytology , Intestines/growth & development , Mesentery/cytology , Mesentery/physiology , Muscle Cells/immunology , Regeneration/genetics
20.
J Vis Exp ; (52)2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21673633

ABSTRACT

Various techniques of cardiac tissue engineering have been pursued in the past decades including scaffolding strategies using either native or bioartificial scaffold materials, entrapment of cardiac myocytes in hydrogels such as fibrin or collagen and stacking of myocyte monolayers. These concepts aim at restoration of compromised cardiac function (e.g. after myocardial infarction) or as experimental models (e.g. predictive toxicology and substance screening or disease modelling). Precise monitoring of cell survival after implantation of engineered heart tissue (EHT) has now become possible using in-vivo bioluminescence imaging (BLI) techniques. Here we describe the generation of fibrin-based EHT from a transgenic rat strain with ubiquitous expression of firefly luciferase (ROSA/luciferase-LEW Tg; ). Implantation is performed into the greater omentum of different rat strains to assess immune responses of the recipient organism following EHT implantation. Comparison of results generated by BLI and the Enzyme Linked Immuno Spot Technique (ELISPOT) confirm the usability of BLI for the assessment of immune responses.


Subject(s)
Heart Transplantation/methods , Luminescent Measurements/methods , Tissue Engineering/methods , Tissue Transplantation/methods , Animals , Fibrin/chemistry , Luciferases, Firefly/biosynthesis , Luciferases, Firefly/genetics , Muscle Cells/immunology , Muscle Cells/metabolism , Muscle Cells/physiology , Muscle Cells/transplantation , Myocardium/cytology , Myocardium/immunology , Myocardium/metabolism , Rats , Rats, Transgenic
SELECTION OF CITATIONS
SEARCH DETAIL
...