Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 149
Filter
Add more filters

Publication year range
1.
J Cell Mol Med ; 24(8): 4791-4803, 2020 04.
Article in English | MEDLINE | ID: mdl-32180339

ABSTRACT

Coronary artery bypass grafting (CABG) triggers a systemic inflammatory response that may contribute to adverse outcomes. Dendritic cells (DC) and monocytes are immunoregulatory cells potentially affected by CABG, contributing to an altered immune state. This study investigated changes in DC and monocyte responses in CABG patients at 5 time-points: admission, peri-operative, ICU, day 3 and day 5. Whole blood from 49 CABG patients was used in an ex vivo whole blood culture model to prospectively assess DC and monocyte responses. Lipopolysaccharide (LPS) was added in parallel to model responses to an infectious complication. Co-stimulatory and adhesion molecule expression and intracellular mediator production was measured by flow cytometry. CABG modulated monocyte and DC responses. In addition, DC and monocytes were immunoparalysed, evidenced by failure of co-stimulatory and adhesion molecules (eg HLA-DR), and intracellular mediators (eg IL-6) to respond to LPS stimulation. DC and monocyte modulation was associated with prolonged ICU length of stay and post-operative atrial fibrillation. DC and monocyte cytokine production did not recover by day 5 post-surgery. This study provides evidence that CABG modulates DC and monocyte responses. Using an ex vivo model to assess immune competency of CABG patients may help identify biomarkers to predict adverse outcomes.


Subject(s)
Coronary Artery Bypass/adverse effects , Dendritic Cells/immunology , HLA-DR Antigens/genetics , Interleukin-6/genetics , Monocytes/immunology , Aged , Cell Adhesion Molecules/genetics , Dendritic Cells/drug effects , Female , Gene Expression Regulation/drug effects , HLA-DR Antigens/blood , Humans , Interleukin-6/blood , Lipopolysaccharides/pharmacology , Male , Monocytes/drug effects , Paralysis/blood , Paralysis/immunology , Paralysis/pathology , Thoracic Surgery
2.
Mol Pharm ; 17(11): 4201-4211, 2020 11 02.
Article in English | MEDLINE | ID: mdl-32903002

ABSTRACT

Many autoimmune therapies focus on immune suppression to reduce symptom severity and halt disease progression; however, currently approved treatments lack specificity for the autoantigen and rely on more global immune suppression. Multivalent antigen arrays can disarm pathogenic autoimmune B cell populations that specifically recognize the antigen of interest via their B cell receptor (BCR). Disarmament may be achieved by BCR engagement, cross-linking, and sustained receptor occupancy as a result of multivalent, high avidity BCR binding. To engage and explore this mechanism, a tetramer display of the encephalogenic proteolipid peptide (PLP139-151), referred to as 4-arm PLP139-151, was synthesized by copper-catalyzed azide-alkyne cycloaddition chemistry. Subcutaneous administration of 4-arm PLP139-151 completely ameliorated symptoms of paralysis in a mouse model of multiple sclerosis known as experimental autoimmune encephalomyelitis. Competitive binding of 4-arm PLP139-151 to PLP139-151-specific IgG in the mouse serum demonstrated the enhanced avidity associated with the multivalent array compared to the free peptide. Furthermore, key PLP139-151-reactive B cells were depleted following 4-arm PLP139-151 treatment, resulting in significant reduction of proinflammatory cytokines. Together, these data demonstrate the potential of 4-arm PLP139-151 to silence autoreactive B cell populations and limit the downstream activation of effector cells.


Subject(s)
Autoantigens/administration & dosage , B-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Immune Tolerance , Immunotherapy/methods , Multiple Sclerosis/therapy , Myelin Proteolipid Protein/administration & dosage , Peptide Fragments/administration & dosage , Administration, Topical , Animals , Autoantigens/blood , Autoantigens/immunology , Encephalomyelitis, Autoimmune, Experimental/blood , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Immunoglobulin G/blood , Mice , Multiple Sclerosis/blood , Multiple Sclerosis/immunology , Myelin Proteolipid Protein/blood , Myelin Proteolipid Protein/immunology , Paralysis/blood , Paralysis/immunology , Paralysis/therapy , Peptide Fragments/blood , Peptide Fragments/immunology , Receptors, Antigen, B-Cell/immunology , Treatment Outcome
3.
J Neurovirol ; 24(3): 372-375, 2018 06.
Article in English | MEDLINE | ID: mdl-29322435

ABSTRACT

We report a case of vaccine-associated paralytic poliomyelitis (VAPP) in an immunocompromised patient with acute lymphocytic leukemia who was initially diagnosed with aseptic meningitis. Isolation of Sabin-like type 1 poliovirus from the patient's cerebrospinal fluid made this a case of vaccine-related poliovirus (VRPV) infection. The patient developed paralysis and respiratory distress and deceased a few months after onset of paralysis with respiratory failure. This tragic case report highlights the emergence of VAPP and indicates the importance of timely diagnosis of VRPV infections to improve clinical management of VRPV-infected patients and to prevent the devastating consequences of silent introduction of VRPVs in treatment wards and eventually in the society.


Subject(s)
Immunocompromised Host , Meningitis, Aseptic/diagnosis , Poliomyelitis/diagnosis , Poliovirus Vaccine, Oral/adverse effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Adult , Diagnostic Errors , Fatal Outcome , Humans , Male , Meningitis, Aseptic/immunology , Meningitis, Aseptic/pathology , Meningitis, Aseptic/virology , Paralysis/diagnosis , Paralysis/immunology , Paralysis/pathology , Paralysis/virology , Poliomyelitis/etiology , Poliomyelitis/immunology , Poliomyelitis/virology , Poliovirus Vaccine, Oral/administration & dosage , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/virology , Respiratory Distress Syndrome/diagnosis , Respiratory Distress Syndrome/immunology , Respiratory Distress Syndrome/pathology , Respiratory Distress Syndrome/virology
4.
Spinal Cord ; 56(10): 964-970, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29795171

ABSTRACT

STUDY DESIGN: This was an animal study. OBJECTIVES: Local inflammation is attenuated below high thoracic SCI, where innervation of major lymphoid organs is involved. However, whether inflammatory responses are affected after low thoracic SCI, remains undetermined. The aim of this study was to characterize the influence of low thoracic SCI on carrageenan-induced paw swelling in intact and paralyzed limbs, at acute and subacute stages. SETTING: University and hospital-based research center, Mexico City, Mexico. METHODS: Rats received a severe contusive SCI at T9 spinal level or sham injury. Then, 1 and 15 days after lesion, carrageenan or vehicle was subcutaneously injected in forelimb and hindlimb paws. Paw swelling was measured over a 6-h period using a plethysmometer. RESULTS: Swelling increased progressively reaching the maximum 6 h post-carrageenan injection. Swelling increase in sham-injured rats was approximately 130% and 70% compared with baseline values of forelimbs and hindlimbs, respectively. Paws injected with saline exhibited no measurable swelling. Carrageenan-induced paw swelling 1-day post-SCI was suppressed in both intact and paralyzed limbs. Fifteen days post-injury, the swelling response to carrageenan was completely reestablished in forelimbs, whereas in hindlimbs it remained significantly attenuated compared with sham-injured rats. CONCLUSIONS: SCI at low spinal level affects the induced swelling response in a different way depending on both, the neurological status of challenged regions and the stage of injury. These findings suggest that neurological compromise of the main immunological organs is not a prerequisite for the local swelling response to be affected after injury.


Subject(s)
Inflammation/physiopathology , Spinal Cord Injuries/immunology , Acute Disease , Animals , Carrageenan , Disease Models, Animal , Disease Progression , Female , Forelimb , Hindlimb , Inflammation/pathology , Paralysis/immunology , Paralysis/pathology , Random Allocation , Rats, Sprague-Dawley , Spinal Cord Injuries/pathology , Thoracic Vertebrae , Time Factors
5.
Am J Physiol Cell Physiol ; 313(5): C533-C540, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28855162

ABSTRACT

Transient muscle paralysis engendered by a single injection of botulinum toxin A (BTxA) rapidly induces profound focal bone resorption within the medullary cavity of adjacent bones. While initially conceived as a model of mechanical disuse, osteoclastic resorption in this model is disproportionately severe compared with the modest gait defect that is created. Preliminary studies of bone marrow following muscle paralysis suggested acute upregulation of inflammatory cytokines, including TNF-α and IL-1. We therefore hypothesized that BTxA-induced muscle paralysis would rapidly alter the inflammatory microenvironment and the osteoclastic potential of bone marrow. We tested this hypothesis by defining the time course of inflammatory cell infiltration, osteoinflammatory cytokine expression, and alteration in osteoclastogenic potential in the tibia bone marrow following transient muscle paralysis of the calf muscles. Our findings identified inflammatory cell infiltration within 24 h of muscle paralysis. By 72 h, osteoclast fusion and pro-osteoclastic inflammatory gene expression were upregulated in tibia bone marrow. These alterations coincided with bone marrow becoming permissive to the formation of osteoclasts of greater size and greater nuclei numbers. Taken together, our data are consistent with the thesis that transient calf muscle paralysis induces acute inflammation within the marrow of the adjacent tibia and that these alterations are temporally consistent with a role in mediating muscle paralysis-induced bone resorption.


Subject(s)
Bone Resorption/physiopathology , Inflammation/etiology , Muscle, Skeletal/drug effects , Osteoclasts/pathology , Paralysis/physiopathology , Animals , Bone Marrow/pathology , Bone Resorption/etiology , Botulinum Toxins, Type A/toxicity , Female , Inflammation/physiopathology , Mice , Mice, Inbred C57BL , Neuromuscular Agents/toxicity , Paralysis/chemically induced , Paralysis/immunology , T-Lymphocytes/immunology
6.
Lancet ; 388(10045): 717-27, 2016 Aug 13.
Article in English | MEDLINE | ID: mdl-26948435

ABSTRACT

Guillain-Barré syndrome is the most common and most severe acute paralytic neuropathy, with about 100,000 people developing the disorder every year worldwide. Under the umbrella term of Guillain-Barré syndrome are several recognisable variants with distinct clinical and pathological features. The severe, generalised manifestation of Guillain-Barré syndrome with respiratory failure affects 20-30% of cases. Treatment with intravenous immunoglobulin or plasma exchange is the optimal management approach, alongside supportive care. Understanding of the infectious triggers and immunological and pathological mechanisms has advanced substantially in the past 10 years, and is guiding clinical trials investigating new treatments. Investigators of large, worldwide, collaborative studies of the spectrum of Guillain-Barré syndrome are accruing data for clinical and biological databases to inform the development of outcome predictors and disease biomarkers. Such studies are transforming the clinical and scientific landscape of acute autoimmune neuropathies.


Subject(s)
Axons/immunology , Guillain-Barre Syndrome , Immunoglobulins, Intravenous/therapeutic use , Neural Conduction/immunology , Paralysis/immunology , Plasma Exchange , Axons/pathology , Campylobacter Infections/complications , Campylobacter Infections/immunology , Campylobacter jejuni , Clinical Trials as Topic , Diagnosis, Differential , Disease Management , Electrophysiology , Guillain-Barre Syndrome/complications , Guillain-Barre Syndrome/diagnosis , Guillain-Barre Syndrome/epidemiology , Guillain-Barre Syndrome/immunology , Guillain-Barre Syndrome/physiopathology , Guillain-Barre Syndrome/therapy , Humans , Influenza Vaccines/administration & dosage , Influenza Vaccines/adverse effects , Muscle Weakness/complications , Muscle Weakness/etiology , Paralysis/complications , Plasmapheresis , Prognosis , Respiratory Insufficiency/etiology , Respiratory Insufficiency/prevention & control , Treatment Outcome
7.
Proc Natl Acad Sci U S A ; 110(25): 10306-11, 2013 Jun 18.
Article in English | MEDLINE | ID: mdl-23733958

ABSTRACT

Sensory abnormalities such as numbness and paresthesias are often the earliest symptoms in neuroinflammatory diseases including multiple sclerosis. The increased production of various cytokines occurs in the early stages of neuroinflammation and could have detrimental effects on the central nervous system, thereby contributing to sensory and cognitive deficits. However, it remains unknown whether and when elevation of cytokines causes changes in brain structure and function under inflammatory conditions. To address this question, we used a mouse model for experimental autoimmune encephalomyelitis (EAE) to examine the effect of inflammation and cytokine elevation on synaptic connections in the primary somatosensory cortex. Using in vivo two-photon microscopy, we found that the elimination and formation rates of dendritic spines and axonal boutons increased within 7 d of EAE induction--several days before the onset of paralysis--and continued to rise during the course of the disease. This synaptic instability occurred before T-cell infiltration and microglial activation in the central nervous system and was in conjunction with peripheral, but not central, production of TNF-α. Peripheral administration of a soluble TNF inhibitor prevented abnormal turnover of dendritic spines and axonal boutons in presymptomatic EAE mice. These findings indicate that peripheral production of TNF-α is a key mediator of synaptic instability in the primary somatosensory cortex and may contribute to sensory and cognitive deficits seen in autoimmune diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Somatosensory Cortex/abnormalities , Somatosensory Cortex/immunology , Tumor Necrosis Factor-alpha/blood , Animals , Axons/immunology , Axons/pathology , Dendritic Spines/immunology , Dendritic Spines/pathology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/metabolism , Mice , Mice, Inbred C57BL , Microglia/immunology , Microglia/pathology , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Paralysis/immunology , Paralysis/metabolism , Paralysis/pathology , Presynaptic Terminals/immunology , Presynaptic Terminals/pathology , Somatosensory Cortex/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Necrosis Factor-alpha/immunology
8.
J Virol ; 86(23): 12561-70, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22973027

ABSTRACT

We previously reported that mice lacking alpha/beta and gamma interferon receptors (IFN-α/ßR and -γR) uniformly exhibit paralysis following infection with the dengue virus (DENV) clinical isolate PL046, while only a subset of mice lacking the IFN-γR alone and virtually no mice lacking the IFN-α/ßR alone develop paralysis. Here, using a mouse-passaged variant of PL046, strain S221, we show that in the absence of the IFN-α/ßR, signaling through the IFN-γR confers approximately 140-fold greater resistance against systemic vascular leakage-associated dengue disease and virtually complete protection from dengue-induced paralysis. Viral replication in the spleen was assessed by immunohistochemistry and flow cytometry, which revealed a reduction in the number of infected cells due to IFN-γR signaling by 2 days after infection, coincident with elevated levels of IFN-γ in the spleen and serum. By 4 days after infection, IFN-γR signaling was found to restrict DENV replication systemically. Clearance of DENV, on the other hand, occurred in the absence of IFN-γR, except in the central nervous system (CNS) (brain and spinal cord), where clearance relied on IFN-γ from CD8(+) T cells. These results demonstrate the roles of IFN-γR signaling in protection from initial systemic and subsequent CNS disease following DENV infection and demonstrate the importance of CD8(+) T cells in preventing DENV-induced CNS disease.


Subject(s)
Central Nervous System/immunology , Dengue Virus/physiology , Dengue/immunology , Disease Susceptibility/immunology , Paralysis/immunology , Receptors, Interferon/metabolism , Virus Replication/physiology , Adoptive Transfer , Analysis of Variance , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line , Central Nervous System/virology , Dengue/complications , Dengue/pathology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunohistochemistry , Interferon-gamma/blood , Interferon-gamma/metabolism , Mice , Mice, Mutant Strains , Paralysis/etiology , Real-Time Polymerase Chain Reaction , Receptor, Interferon alpha-beta/genetics , Regression Analysis , Signal Transduction/physiology , Spleen/virology , Interferon gamma Receptor
9.
Appl Environ Microbiol ; 78(11): 3800-5, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22447593

ABSTRACT

The progress of the Global Polio Eradication Initiative is monitored by acute flaccid paralysis (AFP) surveillance supplemented with environmental surveillance in selected areas. To assess the sensitivity of environmental surveillance, stools from (re)vaccinated elderly persons with a low seroprevalence and from wastewater were concurrently collected and analyzed in the Netherlands over a prolonged period of time. A total number of 228 healthy individuals with different levels of immunity were challenged with monovalent oral polio vaccine serotype 1 or 3. Poliovirus concentrations were determined by the titration of fecal suspensions on poliovirus-sensitive L20B cells and of sewage concentrates by L20B monolayer plaque assay. Almost half of the individuals (45%) shed poliovirus on day 3 after challenge, which peaked (57%) on day 8 with an average poliovirus excretion of 1.3 × 10(5) TCID(50) per g of feces and gradually decreased to less than 5% on day 42. The virus concentrations in sewage peaked on days 6 to 8 at approximately 100 PFU per liter, remained high until day 14, and subsequently decreased to less than 10 PFU per liter on day 29. The estimated poliovirus concentration in sewage approximated the measured initial virus excretion in feces, within 1 log(10) variation, resulting in a sensitivity of detection of 100 infected but mostly asymptomatic individuals in tens of thousands of individuals. An additional second peak observed in sewage may indicate secondary transmission missed by enterovirus or AFP surveillance in patients. This enables the detection of circulating poliovirus by environmental surveillance, supporting its feasibility as an early warning system.


Subject(s)
Environmental Monitoring/methods , Paralysis/prevention & control , Poliomyelitis/prevention & control , Poliovirus/immunology , Population Surveillance/methods , Aged , Aged, 80 and over , Animals , Cell Line , Epidemiological Monitoring , Feces/virology , Female , Humans , Male , Mice , Middle Aged , Netherlands/epidemiology , Paralysis/epidemiology , Paralysis/immunology , Paralysis/virology , Poliomyelitis/epidemiology , Poliomyelitis/immunology , Poliomyelitis/virology , Poliovirus/isolation & purification , Poliovirus Vaccine, Oral/administration & dosage , Poliovirus Vaccine, Oral/immunology , Seroepidemiologic Studies , Sewage/virology
10.
Med Microbiol Immunol ; 201(2): 177-87, 2012 May.
Article in English | MEDLINE | ID: mdl-22102098

ABSTRACT

Immunopathogenesis of Campylobacter jejuni-associated Guillain-Barré syndrome (GBS) is not yet well established probably due to lack of experimental model. Therefore, we studied the Th1/Th2 immune response and pathological changes in C. jejuni-induced chicken model for GBS. C. jejuni (5 × 10(9) CFU/ml) and placebo were fed to 30 chickens each. Stools of all birds were negative for C. jejuni by culture and PCR before experiment. The birds were regularly assessed for disease symptoms up to 30 days. Sciatic nerves from all chickens were examined at 5 days intervals by histopathology and immunohistochemistry, and also for the expression of Th1/Th2 cytokines. Twenty-two chickens (73.3%) developed diarrhea after C. jejuni infection; 18 (60.0%) experimental chickens developed GBS-like paralytic neuropathy. Pathology in the sciatic nerves of these chickens included perinodal and/or patchy demyelination, perivascular focal lymphocytic infiltration, myelin swelling and presence of macrophages within the nerve fibers on 10th-20th post-infection day (PID). Cytokines (IFN-γ, IL-1ß, TNF-α, IL-6 and IL-2) were elevated in early phase (5th-15th PID) and TGF-ß2, IL-10 and IL-4 in the recovery phase (25th-30th PID) of the disease. The study provides evidence that C. jejuni infection in the chicken can provide an experimental animal model of GBS.


Subject(s)
Campylobacter Infections/veterinary , Campylobacter jejuni/isolation & purification , Guillain-Barre Syndrome/veterinary , Paralysis/veterinary , Poultry Diseases/microbiology , Th1 Cells/immunology , Th2 Cells/immunology , Animals , Campylobacter Infections/complications , Campylobacter Infections/immunology , Campylobacter Infections/pathology , Campylobacter jejuni/immunology , Campylobacter jejuni/pathogenicity , Chickens , Cytokines/biosynthesis , Disease Models, Animal , Gene Expression Profiling , Guillain-Barre Syndrome/immunology , Guillain-Barre Syndrome/pathology , Histocytochemistry , Lymphocytes/immunology , Microscopy, Electron, Transmission , Paralysis/immunology , Paralysis/pathology , Poultry Diseases/immunology , Poultry Diseases/pathology , Reverse Transcriptase Polymerase Chain Reaction , Sciatic Nerve/pathology
11.
J Immunol ; 184(10): 5859-64, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20382883

ABSTRACT

Relapsing fever borreliosis is a multisystemic infection characterized primarily by bacteremia but can extend to the CNS. The incidence of CNS disease manifestations in humans depends on the infecting relapsing fever Borrelia species. In the murine model of Borrelia hermsii infection we found high incidence of distinct signs of CNS disease that ranged from a flaccid tail to complete paralysis of hind limbs. Infiltration of large number of T cells into the spinal cord of B. hermsii-infected mice and the upregulation of MHC class II and CD80 on infiltrating macrophages and on microglial cells suggested a role for T cell and Ag-presenting cell interactions in this pathogenesis. Indeed, B. hermsii infection did not induce CNS disease manifestations in T cell-deficient mice (TCR-beta x delta(-/-)), although it resulted in bacteremia comparable to wild-type (Wt) level. Moreover, the infiltration of immune cells into the spinal cord of TCR-beta x delta(-/-) mice was reduced and the resident microglial cells were not activated. Histopathological analysis of lumbar sections of the spinal cord confirmed severe inflammation in Wt but not in TCR-beta x delta(-/-) mice. Induction of CNS disease was dependent on the B. hermsii strain as well as on the ability of the host to control bacteremia. Mice that are impaired in controlling B. hermsii, such as CD14(-/-) mice, exhibited more severe CNS disease than Wt mice. This study demonstrates that distinct neurologic disease manifestations develop during relapsing fever and that T cells play a critical role in the induction of neuropathogenesis.


Subject(s)
Borrelia/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Relapsing Fever/immunology , Relapsing Fever/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , Animals , Cell Movement/genetics , Cell Movement/immunology , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , Immunocompetence/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Paralysis/genetics , Paralysis/immunology , Paralysis/pathology , Relapsing Fever/genetics , Spinal Cord/immunology , Spinal Cord/microbiology , Spinal Cord/pathology , T-Lymphocyte Subsets/microbiology
12.
Respir Res ; 12: 147, 2011 Nov 06.
Article in English | MEDLINE | ID: mdl-22054060

ABSTRACT

BACKGROUND: Neonatal mice developed neurological disease and pulmonary dysfunction after an infection with a mouse-adapted human Enterovirus 71 (EV71) strain MP4. However, the hallmark of severe human EV71 infection, pulmonary edema (PE), was not evident. METHODS: To test whether EV71-induced PE required a proinflammatory cytokine response, exogenous pro-inflammatory cytokines were administered to EV71-infected mice during the late stage of infection. RESULTS: After intracranial infection of EV71/MP4, 7-day-old mice developed hind-limb paralysis, pulmonary dysfunction, and emphysema. A transient increase was observed in serum IL-6, IL-10, IL-13, and IFN-γ, but not noradrenaline. At day 3 post infection, treatment with IL-6, IL-13, and IFN-γ provoked mild PE and severe emphysema that were accompanied by pulmonary dysfunction in EV71-infected, but not herpes simplex virus-1 (HSV-1)-infected control mice. Adult mice did not develop PE after an intracerebral microinjection of EV71 into the nucleus tractus solitarii (NTS). While viral antigen accumulated in the ventral medulla and the NTS of intracerebrally injected mice, neuronal loss was observed in the ventral medulla only. CONCLUSIONS: Exogenous IL-6, IL-13, and IFN-γ treatment could induce mild PE and exacerbate pulmonary abnormality of EV71-infected mice. However, other factors such as over-activation of the sympathetic nervous system may also be required for the development of classic PE symptoms.


Subject(s)
Enterovirus A, Human , Enterovirus Infections/pathology , Interferon-gamma/toxicity , Interleukin-13/toxicity , Interleukin-6/toxicity , Lung/pathology , Pulmonary Edema/pathology , Pulmonary Emphysema/pathology , Animals , Animals, Newborn , Chlorocebus aethiops , Enterovirus A, Human/immunology , Enterovirus A, Human/pathogenicity , Enterovirus Infections/blood , Enterovirus Infections/immunology , Humans , Interferon-gamma/blood , Interleukin-13/blood , Interleukin-6/blood , Lung/immunology , Lung/virology , Mice , Mice, Inbred ICR , Paralysis/immunology , Paralysis/pathology , Paralysis/virology , Pulmonary Edema/blood , Pulmonary Edema/immunology , Pulmonary Emphysema/immunology , Pulmonary Emphysema/virology , Vero Cells
13.
Front Immunol ; 12: 648184, 2021.
Article in English | MEDLINE | ID: mdl-34305887

ABSTRACT

Enterovirus 71 (EV71) is a positive single-stranded RNA virus from the enterovirus genus of the Picornaviridae family. Most young children infected with EV71 develop mild symptoms of hand, foot and mouth disease, but some develop severe symptoms with neurological involvement. Limb paralysis from EV71 infection is presumed to arise mainly from dysfunction of motor neurons in the spinal cord. However, EV71 also targets and damages skeletal muscle, which may also contribute to the debilitating symptoms. In this study, we have delineated the impacts of EV71 infection on skeletal muscle using a mouse model. Mouse pups infected with EV71 developed limb paralysis, starting at day 3 post-infection and peaking at day 5-7 post-infection. At later times, mice recovered gradually but not completely. Notably, severe disease was associated with high levels of myositis accompanied by muscle calcification and persistent motor end plate abnormalities. Interestingly, macrophages exhibited a dynamic change in phenotype, with inflammatory macrophages (CD45+CD11b+Ly6Chi) appearing in the early stage of infection and anti-inflammatory/restorative macrophages (CD45+CD11b+Ly6Clow/-) appearing in the late stage. The presence of inflammatory macrophages was associated with severe inflammation, while the restorative macrophages were associated with recovery. Altogether, we have demonstrated that EV71 infection causes myositis, muscle calcification and structural defects in motor end plates. Subsequent muscle regeneration is associated with a dynamic change in macrophage phenotype.


Subject(s)
Enterovirus A, Human , Enterovirus Infections/immunology , Macrophages/immunology , Muscle, Skeletal/pathology , Myositis/immunology , Phenotype , Recovery of Function/immunology , Animals , Antigens, Ly/metabolism , CD11b Antigen/metabolism , Calcinosis/immunology , Disease Models, Animal , Enterovirus Infections/virology , Leukocyte Common Antigens/metabolism , Mice , Mice, Inbred C57BL , Paralysis/immunology , Regeneration/immunology
14.
Viruses ; 13(3)2021 02 27.
Article in English | MEDLINE | ID: mdl-33673590

ABSTRACT

This report is an overview of enterovirus (EV) detection in Tunisian polio-suspected paralytic cases (acute flaccid paralysis (AFP) cases), healthy contacts and patients with primary immunodeficiencies (PID) during an 11-year period. A total of 2735 clinical samples were analyzed for EV isolation and type identification, according to the recommended protocols of the World Health Organization. Three poliovirus (PV) serotypes and 28 different nonpolio enteroviruses (NPEVs) were detected. The NPEV detection rate was 4.3%, 2.8% and 12.4% in AFP cases, healthy contacts and PID patients, respectively. The predominant species was EV-B, and the circulation of viruses from species EV-A was noted since 2011. All PVs detected were of Sabin origin. The PV detection rate was higher in PID patients compared to AFP cases and contacts (6.8%, 1.5% and 1.3% respectively). PV2 was not detected since 2015. Using nucleotide sequencing of the entire VP1 region, 61 strains were characterized as Sabin-like. Among them, six strains of types 1 and 3 PV were identified as pre-vaccine-derived polioviruses (VDPVs). Five type 2 PV, four strains belonging to type 1 PV and two strains belonging to type 3 PV, were classified as iVDPVs. The data presented provide a comprehensive picture of EVs circulating in Tunisia over an 11-year period, reveal changes in their epidemiology as compared to previous studies and highlight the need to set up a warning system to avoid unnoticed PVs.


Subject(s)
Enterovirus Infections/epidemiology , Enterovirus Infections/virology , Enterovirus/genetics , Poliomyelitis/epidemiology , Poliomyelitis/virology , Enterovirus/immunology , Enterovirus Infections/immunology , Humans , Molecular Epidemiology/methods , Paralysis/immunology , Paralysis/virology , Phylogeny , Poliomyelitis/immunology , Poliovirus/genetics , Poliovirus/immunology , Poliovirus Vaccine, Oral/immunology , Tunisia/epidemiology
15.
J Exp Med ; 152(3): 507-20, 1980 Sep 01.
Article in English | MEDLINE | ID: mdl-6774045

ABSTRACT

Mice immunized with acetylcholine receptor (AChR) purified from Torpedo californica form anti-AChR antibodies and often develop muscular weakness and flaccid paralysis closely resembling the human disease myasthenia gravis. This condition, termed experimental myasthenia gravis (EMG), is strain dependent in that the frequency of paralysis is much greater in some strains than in others. Differences in the frequency of EMG might result from differences in the immune system or the neuromuscular junction. In these studies, we have identified two loci, the major histocompatibility complex (H-2) region on chromosome 17 and the region that contains the structural genes for the constant region of immunoglobulin heavy chains (IgCH region) on chromosome 12, which significantly effect the probability with which a mouse immunized with T. californica AChR can be expected to become paralyzed. One genotype (H-2b, Ig-1b) correlated with high susceptibility to EMG in four strains with three dissimilar backgrounds. These studies demonstrate that susceptibility to EMG is a heritable trait determined by at least two distinct loci that are linked to regions of the mouse genome that regulate immune responsiveness.


Subject(s)
Genes, MHC Class II , H-2 Antigens/genetics , Immunoglobulin Constant Regions/genetics , Immunoglobulins/genetics , Myasthenia Gravis/immunology , Receptors, Cholinergic/immunology , Receptors, Nicotinic/immunology , Animals , Disease Models, Animal , Fishes , Immunization , Mice , Muscle Hypotonia/immunology , Myasthenia Gravis/physiopathology , Paralysis/immunology
16.
J Exp Med ; 162(6): 2107-24, 1985 Dec 01.
Article in English | MEDLINE | ID: mdl-2415664

ABSTRACT

Class II-restricted T cell clones specific for myelin basic protein (MBP) have been generated from PL/J and (PL/J X SJL/J)F1 [((PLSJ)F1] mice following sensitization to rat MBP. Of 17 T cell clones generated from (PLSJ)F1 mice, 5 are I-Au(A alpha uA beta u) restricted, one is restricted to I-As(A alpha sA beta s), 10 are restricted to hybrid I-A(u X s)F1 (A alpha sA beta u) determinants, and one clone is restricted to hybrid I-E(u X s) (E alpha uE beta s) molecules. Thus, of 16 I-A-restricted T cell clones generated from (PLSJ)F1 mice, only one is I-As-restricted, reflecting a lack of priming to MBP in association with I-As. T cell clones restricted to I-Au and to I-E (E alpha u E beta s) molecules recognize mouse (self) MBP. Furthermore, only the five T cell clones restricted to I-Au molecules recognize a determinant in common with mouse (self) MBP within the encephalitogenic N-terminal peptide. Three such I-Au restricted T cell clones, derived independently, cause paralysis in 100% of (PL/J X SJL/J)F1 mice tested. Acute, chronic unremitting, and chronic relapsing paralysis are all induced following injection of these clones. Administration of greater numbers of cloned T cells causes acute and fatal experimental allergic encephalomyelitis, while administration of lower numbers of cloned T cells is associated with chronic unremitting and relapsing paralysis. Paralysis induced with T cell clones shares many clinical, immunologic, and histologic aspects with human demyelinating diseases such as multiple sclerosis. Histopathology reveals perivascular lymphocytic infiltration, demyelination, and remyelination. These studies demonstrate the utility of T cell clones for analyzing the association between class II major histocompatibility complex molecules and disease susceptibility.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Epitopes/immunology , Myelin Basic Protein/immunology , T-Lymphocytes/immunology , Animals , Antigen-Presenting Cells/immunology , Autoimmune Diseases/immunology , Clone Cells/classification , Clone Cells/immunology , Crosses, Genetic , Female , H-2 Antigens/genetics , H-2 Antigens/immunology , Histocompatibility Antigens/genetics , Histocompatibility Antigens/immunology , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Paralysis/immunology , Peptide Fragments/immunology , T-Lymphocytes/classification , T-Lymphocytes/transplantation
17.
J Vasc Surg ; 52(2): 435-43, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20541344

ABSTRACT

INTRODUCTION: Hypothermia is widely used to mediate ischemia-reperfusion injury associated with repair of the thoracoabdominal aorta. Experiments were designed in a murine model of thoracic aortic ischemia-reperfusion (TAR) to evaluate the effect of moderate systemic hypothermia on neurologic function, spinal cord morphology, and indices of inflammation in critical organs. METHODS: C57BL/6 mice were subjected to TAR under hypothermic (34 degrees C) or normothermic (38 degrees C) conditions, followed by 24 or 48 hours of normothermic reperfusion. Neurologic functions were assessed during reperfusion. Spinal cords were examined at 24 and 48 hours after reperfusion, and the degree of injury qualified by counting the number of viable motor neurons within the anterior horns. Keratinocyte chemokine, interleukin-6, and myeloperoxidase levels were measured from lung, liver, and kidney at 24 and 48 hours. RESULTS: Normothermic TAR resulted in a dense neurologic deficit in all mice throughout the reperfusion period. Mice subjected to TAR under hypothermic conditions had transient, mild neurologic deficit during the initial periods of reperfusion. Between 24 and 48 hours, delayed paralysis developed in half of these mice, whereas the other half remained neurologically intact. Spinal cord histology showed a graded degree of injury that correlated with neurologic function. There was no correlation between markers of inflammation in various organs and neurologic outcomes following TAR. CONCLUSION: Systemic moderate hypothermia was protective against immediate paralysis after TAR in all cases and was associated with delayed paralysis in 50% of mice. This study suggests that delayed-onset paralysis may be the result of a local insult, rather than a systemic inflammatory event, precipitating spinal cord injury.


Subject(s)
Aorta, Thoracic/surgery , Hypothermia, Induced , Paralysis/prevention & control , Reperfusion Injury/prevention & control , Spinal Cord Ischemia/prevention & control , Animals , Anterior Horn Cells/pathology , Anterior Horn Cells/physiopathology , Cell Survival , Chemokines/metabolism , Disease Models, Animal , Inflammation Mediators/metabolism , Interleukin-6/metabolism , Kidney/immunology , Liver/immunology , Lung/immunology , Male , Mice , Mice, Inbred C57BL , Neurologic Examination , Paralysis/immunology , Paralysis/pathology , Paralysis/physiopathology , Peroxidase/metabolism , Reperfusion Injury/immunology , Reperfusion Injury/pathology , Reperfusion Injury/physiopathology , Severity of Illness Index , Spinal Cord Ischemia/immunology , Spinal Cord Ischemia/pathology , Spinal Cord Ischemia/physiopathology , Time Factors
18.
Neuroimmunomodulation ; 17(4): 270-8, 2010.
Article in English | MEDLINE | ID: mdl-20203533

ABSTRACT

LINGO-1 (leucine-rich repeat and Ig domain-containing, Nogo receptor-interacting protein) is an important component of the NgR receptor complex involved in RhoA activation and axon regeneration. The authors report on passive immunization with LINGO-1 polyclonal antiserum, a therapeutic approach to overcome NgR-mediated growth inhibition after spinal cord injury (SCI). The intrathecally administered high-titer rabbit-derived antiserum can be detected around the injury site within a wide time window; it blocks LINGO-1 in vivo with high molecular specificity. In this animal model, passive immunization with LINGO-1 antiserum significantly decreased RhoA activation and increased neuronal survival. Adult rats immunized in this manner show recovery of certain hindlimb motor functions after dorsal hemisection of the spinal cord. Thus, passive immunotherapy with LINGO-1 polyclonal antiserum may represent a promising repair strategy following acute SCI.


Subject(s)
Cytoprotection/drug effects , Immunization, Passive/methods , Membrane Proteins/antagonists & inhibitors , Nerve Tissue Proteins/antagonists & inhibitors , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , Animals , Cell Survival/drug effects , Cell Survival/immunology , Cytoprotection/immunology , Disease Models, Animal , Female , Immune Sera/immunology , Immune Sera/pharmacology , Injections, Spinal , Membrane Proteins/immunology , Nerve Degeneration/drug therapy , Nerve Degeneration/immunology , Nerve Degeneration/physiopathology , Nerve Tissue Proteins/immunology , Paralysis/drug therapy , Paralysis/immunology , Paralysis/physiopathology , Rats , Rats, Sprague-Dawley , Recovery of Function/immunology , Spinal Cord Injuries/immunology , Spinal Cord Injuries/physiopathology , Treatment Outcome , rhoA GTP-Binding Protein/drug effects , rhoA GTP-Binding Protein/metabolism
19.
Front Immunol ; 11: 589380, 2020.
Article in English | MEDLINE | ID: mdl-33178221

ABSTRACT

Severe COVID-19 patients show various immunological abnormalities including T-cell reduction and cytokine release syndrome, which can be fatal and is a major concern of the pandemic. However, it is poorly understood how T-cell dysregulation can contribute to the pathogenesis of severe COVID-19. Here we show single cell-level mechanisms for T-cell dysregulation in severe COVID-19, demonstrating new pathogenetic mechanisms of T-cell activation and differentiation underlying severe COVID-19. By in silico sorting CD4+ T-cells from a single cell RNA-seq dataset, we found that CD4+ T-cells were highly activated and showed unique differentiation pathways in the lung of severe COVID-19 patients. Notably, those T-cells in severe COVID-19 patients highly expressed immunoregulatory receptors and CD25, whilst repressing the expression of FOXP3. Furthermore, we show that CD25+ hyperactivated T-cells differentiate into multiple helper T-cell lineages, showing multifaceted effector T-cells with Th1 and Th2 characteristics. Lastly, we show that CD25-expressing hyperactivated T-cells produce the protease Furin, which facilitates the viral entry of SARS-CoV-2. Collectively, CD4+ T-cells from severe COVID-19 patients are hyperactivated and FOXP3-mediated negative feedback mechanisms are impaired in the lung, which may promote immunopathology. Therefore, our study proposes a new model of T-cell hyperactivation and paralysis that drives immunopathology in severe COVID-19.


Subject(s)
COVID-19/immunology , Lymphocyte Activation/immunology , Paralysis/immunology , SARS-CoV-2/immunology , Severity of Illness Index , Single-Cell Analysis/methods , T-Lymphocytes, Regulatory/immunology , COVID-19/virology , Databases, Genetic , Forkhead Transcription Factors/metabolism , Furin/metabolism , Humans , Interleukin-2 Receptor alpha Subunit/metabolism , RNA-Seq , Receptors, Antigen, T-Cell/metabolism , Transcriptome , Virus Internalization
20.
Science ; 181(4103): 945-7, 1973 Sep 07.
Article in English | MEDLINE | ID: mdl-4354228

ABSTRACT

A neurogenic paralysis of the lower limb can be induced and serially transmitted in mice by a nontransforming type C virus strain that originated in an embryo of a wild mouse. The virus exerted a neurotropic effect on the anterior horn neurons.


Subject(s)
Oncogenic Viruses/isolation & purification , Paralysis/etiology , RNA Viruses/isolation & purification , Animals , Antigens, Viral/analysis , Brain/immunology , Cells, Cultured , Complement Fixation Tests , Embryo, Mammalian/microbiology , Hindlimb , Lymphoma/etiology , Mice , Muscles/immunology , Paralysis/immunology , Retroviridae/growth & development , Retroviridae/immunology , Retroviridae/isolation & purification , Spinal Cord/immunology , Spleen/immunology
SELECTION OF CITATIONS
SEARCH DETAIL