Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 859
Filter
1.
PLoS Pathog ; 20(5): e1012125, 2024 May.
Article in English | MEDLINE | ID: mdl-38696536

ABSTRACT

Major 5'-terminally deleted (5'TD) RNA forms of group-B coxsackievirus (CVB-5'TD) has been associated with myocarditis in both mice and humans. Although it is known that interferon-ß (IFN-ß) signaling is critical for an efficient innate immune response against CVB-induced myocarditis, the link between CVB-5'TD RNA forms and type I IFN signaling in cardiomyocytes remains to be explored. In a mouse model of CVB3/28-induced myocarditis, major early-emerging forms of CVB-5'TD RNA have been characterized as replicative viral populations that impair IFN-ß production in the heart. Synthetic CVB3/28 RNA forms mimicking each of these major 5'TD virus populations were transfected in mice and have been shown to modulate innate immune responses in the heart and to induce myocarditis in mice. Remarkably, transfection of synthetic viral RNA with deletions in the secondary structures of the 5'-terminal CVB3 RNA domain I, modifying stem-loops "b", "c" or "d", were found to impair IFN-ß production in human cardiomyocytes. In addition, the activation of innate immune response by Poly(I:C), was found to restore IFN-ß production and to reduce the burden of CVB-5'TD RNA-forms in cardiac tissues, thereby reducing the mortality rate of infected mice. Overall, our results indicate that major early-emerging CVB3 populations deleted in the domain I of genomic RNA, in the 5' noncoding region, modulate the activation of the type I IFN pathway in cardiomyocytes and induce myocarditis in mice. These findings shed new light on the role of replicative CVB-5'TD RNA forms as key pathophysiological factors in CVB-induced human myocarditis.


Subject(s)
Coxsackievirus Infections , Enterovirus B, Human , Interferon Type I , Myocarditis , Myocytes, Cardiac , RNA, Viral , Myocarditis/virology , Myocarditis/immunology , Myocarditis/genetics , Animals , Myocytes, Cardiac/virology , Myocytes, Cardiac/metabolism , Mice , Enterovirus B, Human/immunology , Coxsackievirus Infections/immunology , Coxsackievirus Infections/virology , Coxsackievirus Infections/genetics , Interferon Type I/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Humans , Immunity, Innate , Signal Transduction , Interferon-beta/metabolism , Interferon-beta/genetics , Interferon-beta/immunology , Male , 5' Untranslated Regions
2.
Hum Antibodies ; 32(2): 61-65, 2024.
Article in English | MEDLINE | ID: mdl-38669524

ABSTRACT

BACKGROUND: The antibody that crosses transplacentally from mother to fetus is very important origin of protective passive immunity against infection neonatal with enterovirus. Important varieties of coxsackievirus B3 (CVB3) are responsible for infections in newborns. The purpose from this study is to investigate in the prevalence of Coxsackie B virus in a sample of Iraqi women with miscarriage and potential role of miscarriage risk. METHODS: Between November 2022 and June 2023, we included 91 parturient women (gestational age: 4-20 weeks) who were between the ages of 15 and 40. Every participant completed a questionnaire, and blood was drawn to assess maternal antibodies against CVB3. RESULTS: The blood seropositive rates were 46 out 91(50.54%), 2 out 46 were IgM positive (4.34%), (8-12 weeks) 23 from 46 (50%) (p-value 0.0294) gestational age more frequent among aborted women that positive for anti-coxsackie B antibody, The 25-35 age group was significantly overrepresented (51/91, 56%) compared to other age groups. CONCLUSION: This investigation posits Coxsackie B virus (CBV) as a possible etiology for miscarriage in the Iraqi female population. Further studies employing larger cohorts and robust methodologies, beyond the current detection technique, are warranted to corroborate these observations and elucidate the potential mechanisms by which CBV might induce miscarriage.


Subject(s)
Abortion, Spontaneous , Antibodies, Viral , Coxsackievirus Infections , Enterovirus B, Human , Humans , Female , Iraq/epidemiology , Adult , Pregnancy , Enterovirus B, Human/immunology , Abortion, Spontaneous/virology , Abortion, Spontaneous/immunology , Abortion, Spontaneous/epidemiology , Coxsackievirus Infections/immunology , Coxsackievirus Infections/virology , Coxsackievirus Infections/epidemiology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Young Adult , Adolescent , Immunoglobulin M/blood , Immunoglobulin M/immunology , Seroepidemiologic Studies , Pregnancy Complications, Infectious/virology , Pregnancy Complications, Infectious/immunology , Pregnancy Complications, Infectious/epidemiology , Prevalence
3.
Immunol Cell Biol ; 101(8): 735-745, 2023 09.
Article in English | MEDLINE | ID: mdl-37253434

ABSTRACT

Coxsackievirus B3 (CVB3)-induced viral myocarditis (VMC) is characterized by immune cell infiltration and myocardial damage. High mobility group box 1 (HMGB1) is a highly conserved nuclear DNA-binding protein that participates in DNA replication, transcriptional regulation, repair response and inflammatory response in different disease models. To investigate the exact function of HMGB1 in CVB3-induced VMC, we crossed Hmgb1-floxed (Hmgb1f/f ) mice with mice carrying a suitable Cre recombinase transgenic strain to achieve conditional inactivation of the Hmgb1 gene in a cardiomyocyte-specific manner and to establish myocarditis. In this study, we found that cardiomyocyte-specific Hmgb1-deficient (Hmgb1f/f TgCre/+ ) mice exhibited exacerbated myocardial injury. Hmgb1-deficient cardiomyocytes may promote early apoptosis via the p53-mediated Bax mitochondrial pathway, as evidenced by the higher localization of p53 protein in the cytosol of Hmgb1-deficient cardiomyocytes upon CVB3 infection. Moreover, cardiomyocyte Hmgb1-deficient mice are more susceptible to cardiac dysfunction after infection. This study provides new insights into HMGB1 in VMC pathogenesis and a strategy for appropriate blocking of HMGB1 in the clinical treatment of VMC.


Subject(s)
Coxsackievirus Infections , Enterovirus B, Human , HMGB1 Protein , Myocarditis , Animals , Mice , Apoptosis/genetics , HMGB1 Protein/metabolism , Mice, Inbred BALB C , Myocarditis/immunology , Myocarditis/pathology , Myocarditis/virology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Tumor Suppressor Protein p53/metabolism , Coxsackievirus Infections/immunology
4.
J Virol ; 96(17): e0123222, 2022 09 14.
Article in English | MEDLINE | ID: mdl-36037480

ABSTRACT

Enteroviruses initiate infection in the gastrointestinal tract, and sex is often a biological variable that impacts pathogenesis. Previous data suggest that sex hormones can influence the intestinal replication of Coxsackievirus B3 (CVB3), an enterovirus in the Picornaviridae family. However, the specific sex hormone(s) that regulates intestinal CVB3 replication is poorly understood. To determine if testosterone promotes intestinal CVB3 replication, we orally inoculated male and female Ifnar-/- mice that were treated with either placebo or testosterone-filled capsules. Following oral inoculation, we found that the testosterone-treated male and female mice shed significantly more CVB3 in their feces than did the placebo-treated mice, indicating that testosterone enhances intestinal replication. Similarly, testosterone enhanced viral dissemination in both sexes, as we observed higher viral loads in peripheral tissues following infection. Further, the testosterone-treated male mice also had a higher mortality rate than did the testosterone-depleted male mice. Finally, we observed that testosterone significantly affected the immune response to CVB3. We found that testosterone broadly increased proinflammatory cytokines and chemokines while decreasing the number of splenic B cells and dendritic cells following CVB3 infection. Moreover, while testosterone did not affect the early CD4 T cell response to CVB3, testosterone reduced the activation of CD8 T cells. These data indicate that testosterone can promote intestinal CVB3 replication and dissemination while also impacting the subsequent viral immune response. IMPORTANCE Biological sex plays a significant role in the outcomes of various infections and diseases. The impact of sex hormones on the intestinal replication and dissemination of Coxsackievirus B3 remains poorly understood. Using an oral inoculation model, we found that testosterone enhances CVB3 shedding and dissemination in male and female mice. Further, testosterone can alter the immune response to CVB3. This work highlights the role of testosterone in CVB3 pathogenesis and suggests that sex hormones can impact the replication and dissemination of enteric viruses.


Subject(s)
Coxsackievirus Infections/immunology , Testosterone/metabolism , Animals , Coxsackievirus Infections/metabolism , Coxsackievirus Infections/virology , Disease Models, Animal , Female , Host-Pathogen Interactions , Male , Mice , Virus Replication
5.
J Virol ; 96(11): e0043522, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35579435

ABSTRACT

Platelet factor 4 (PF4) or the CXC chemokine CXCL4 is the most abundant protein within the α-granules of platelets. Previous studies found that PF4 regulates infections of several viruses, including HIV-1, H1N1, hepatitis C virus (HCV), and dengue virus. Here, we show that PF4 is an inhibitor of enterovirus A71 (EV71) and coxsackievirus A16 (CA16) infections. The secreted form of PF4 from transfected cells or soluble purified PF4 from Escherichia coli, even lacking signal peptide affected secretion, obviously inhibited the propagation of EV71 and CA16. Mechanistically, we demonstrated that PF4 blocked the entry of the virus into the host cells by interactions with VP3 proteins of EV71/CA16 and the interaction with SCARB2 receptor-mediated EV71 and CA16 endocytosis. As expected, the incubation of anti-PF4 antibody with PF4 blocked PF4 inhibition on EV71 and CA16 infections further supported the above conclusion. Importantly, pretreatment of EV71 viruses with PF4 significantly protected the neonatal mice from EV71 lethal challenge and promoted the survival rate of infected mice. PF4 derived from natural platelets by EV71/CA16 activation also presented strong inhibition on EV71 and CA16. In summary, our study identified a new host factor against EV71 and CA16 infections, providing a novel strategy for EV71 and CA16 treatment. IMPORTANCE The virus's life cycle starts with binding to cell surface receptors, resulting in receptor-mediated endocytosis. Targeting the entry of the virus into target cells is an effective strategy to develop a novel drug. EV71 and CA16 are the major pathogens that cause hand, foot, and mouth disease (HFMD) outbreaks worldwide since 2008. However, the treatment of EV71 and CA16 infections is mainly symptomatic because there is no approved drug. Therefore, the underlying pathogenesis of EV71/CA16 and the interaction between host-EV71/CA16 need to be further investigated to develop an inhibitor. Here, we identified PF4 as a potent entry inhibitor of EV71 and CA16 via binding to VP3 proteins of EV71 and CA16 or binding to receptor SCARB2. In the EV71 infection model, PF4 protected mice from EV71 lethal challenge and promoted the survival rate of EV71-infected mice. Our study suggests that PF4 represents a potential candidate host factor for anti-EV71 and CA16 infections.


Subject(s)
Coxsackievirus Infections , Enterovirus Infections , Platelet Factor 4 , Virus Internalization , Animals , Coxsackievirus Infections/immunology , Enterovirus , Enterovirus A, Human , Enterovirus Infections/immunology , Immunologic Factors/metabolism , Mice , Platelet Factor 4/metabolism
6.
Cell Physiol Biochem ; 55(6): 679-703, 2021 Nov 18.
Article in English | MEDLINE | ID: mdl-34791861

ABSTRACT

Viral diseases are a major threat to modern society and the global health system. It is therefore of utter relevance to understand the way viruses affect the host as a basis to find new treatment solutions. The understanding of viral myocarditis (VMC) is incomplete and effective treatment options are lacking. This review will discuss the mechanism, effects, and treatment options of the most frequent myocarditis-causing viruses namely enteroviruses such as Coxsackievirus B3 (CVB3) and Parvovirus B19 (PVB19) on the human heart. Thereby, we focus on: 1. Viral entry: CVB3 use Coxsackievirus-Adenovirus-Receptor (CAR) and Decay Accelerating Factor (DAF) to enter cardiac myocytes while PVB19 use the receptor globoside (Gb4) to enter cardiac endothelial cells. 2. Immune system responses: The innate immune system mediated by activated cardiac toll-like receptors (TLRs) worsen inflammation in CVB3-infected mouse hearts. Different types of cells of the adaptive immune system are recruited to the site of inflammation that have either protective or adverse effects during VMC. 3. Autophagy: CVB3 evades autophagosomal degradation and misuses the autophasomal pathway for viral replication and release. 4. Viral replication sites: CVB3 promotes the formation of double membrane vesicles (DMVs), which it uses as replication sites. PVB19 uses the host cell nucleus as the replication site and uses the host cell DNA replication system. 5. Cell cycle manipulation: CVB3 attenuates the cell cycle at the G1/S phase, which promotes viral transcription and replication. PVB19 exerts cell cycle arrest in the S phase using its viral endonuclease activity. 6. Regulation of apoptosis: Enteroviruses prevent apoptosis during early stages of infection and promote cell death during later stages by using the viral proteases 2A and 3C, and viroporin 2B. PVB19 promotes apoptosis using the non-structural proteins NS1 and the 11 kDa protein. 7. Energy metabolism: Dysregulation of respiratory chain complex expression, activity and ROS production may be altered in CVB3- and PVB19-mediated myocarditis. 8. Ion channel modulation: CVB3-expression was indicated to alter calcium and potassium currents in Xenopus laevis oocytes and rodent cardiomyocytes. The phospholipase 2-like activity of PVB19 may alter several calcium, potassium and sodium channels. By understanding the general pathophysiological mechanisms of well-studied myocarditis-linked viruses, we might be provided with a guideline to handle other less-studied human viruses.


Subject(s)
Coxsackievirus Infections/immunology , Host-Pathogen Interactions/immunology , Myocarditis , Parvoviridae Infections/immunology , Parvovirus B19, Human/physiology , Virus Replication , Coxsackievirus Infections/pathology , Humans , Myocarditis/immunology , Myocarditis/pathology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/pathology , Myocytes, Cardiac/virology , Parvoviridae Infections/pathology , Receptors, Virus/immunology
7.
Front Immunol ; 12: 751341, 2021.
Article in English | MEDLINE | ID: mdl-34804036

ABSTRACT

Seemingly redundant in function, melanoma differentiation-associated protein 5 (MDA5) and toll-like receptor- 3 (TLR3) both sense RNA viruses and induce type I interferon (IFN-I). Herein, we demonstrate that changes in sensing of the same virus by MDA5 and TLR3 can lead to distinct signatures of IFN-α and IFN-ß resulting in different disease outcomes. Specifically, infection with a diabetogenic islet ß cell-tropic strain of coxsackievirus (CB4) results in diabetes protection under reduced MDA5 signaling conditions while reduced TLR3 function retains diabetes susceptibility. Regulating the induction of IFN-I at the site of virus infection creates a local site of interferonopathy leading to loss of T cell regulation and induction of autoimmune diabetes. We have not demonstrated another way to prevent T1D in the NOD mouse, rather we believe this work has provided compounding evidence for a specific control of IFN-I to drive a myriad of responses ranging from virus clearance to onset of autoimmune diabetes.


Subject(s)
Coxsackievirus Infections/immunology , Cytokines/immunology , Diabetes Mellitus, Type 1/immunology , Interferon-Induced Helicase, IFIH1/immunology , Toll-Like Receptor 3/immunology , Animals , Enterovirus B, Human , Female , Interferon-Induced Helicase, IFIH1/genetics , Male , Mice, Inbred NOD , Mice, Transgenic , Toll-Like Receptor 3/genetics
8.
Front Immunol ; 12: 666594, 2021.
Article in English | MEDLINE | ID: mdl-34630378

ABSTRACT

Coxsackievirus B3 (CVB3)-induced viral myocarditis is a common clinical cardiovascular disease without effective available vaccine. In this study, we tried to potentiate the immunoprotection efficacy of our previous CVB3-specific VP1 protein vaccine by introducing a streptococcal protein G-derived, draining lymph nodes (dLNs)-targeting albumin-binding domain (ABD) peptide. We found that compared with the original VP1 vaccine, ABD-fused VP1 (ABD-VP1) vaccine gained the new ability to efficiently bind murine albumin both in vitro and in vivo, possessed a much longer serum half-life in serum and exhibited more abundance in the dLNs after immunization. Accordingly, ABD-VP1 immunization not only significantly facilitated the enrichment and maturation of dendritic cells (DCs), induced higher percentages of IFN-γ+ CD8 + cells in the dLNs, but also robustly promoted VP1-induced T cell proliferation and cytotoxic T lymphocyte (CTL) responses in the spleens. More importantly, ABD-VP1 also elicited higher percentages of protective CD44hi CD62Lhi memory T cells in dLNs and spleens. Consequently, obvious protective effect against viral myocarditis was conferred by ABD-VP1 vaccine compared to the VP1 vaccine, reflected by the less body weight loss, improved cardiac function, alleviated cardiac histomorphological changes and an increased 28-day survival rate. Our results indicated that the ABD might be a promising immune-enhancing regime for vaccine design and development.


Subject(s)
Bacterial Proteins/administration & dosage , Capsid Proteins/administration & dosage , Coxsackievirus Infections/prevention & control , Enterovirus B, Human/immunology , Immunogenicity, Vaccine , Myocarditis/prevention & control , Peptide Fragments/administration & dosage , Serum Albumin/metabolism , Viral Vaccines/administration & dosage , Animals , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Capsid Proteins/immunology , Capsid Proteins/metabolism , Coxsackievirus Infections/immunology , Coxsackievirus Infections/metabolism , Coxsackievirus Infections/virology , Disease Models, Animal , HeLa Cells , Humans , Immunization , Male , Mice, Inbred BALB C , Myocarditis/immunology , Myocarditis/metabolism , Myocarditis/virology , Peptide Fragments/immunology , Peptide Fragments/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Vaccines, Synthetic/metabolism , Viral Vaccines/immunology , Viral Vaccines/metabolism
9.
Viruses ; 13(10)2021 09 24.
Article in English | MEDLINE | ID: mdl-34696348

ABSTRACT

The coxsackievirus B3 strain PD-0 has been proposed as a new oncolytic virus for the treatment of colorectal carcinoma. Here, we generated a cDNA clone of PD-0 and analyzed the virus PD-H, newly generated from this cDNA, in xenografted and syngenic models of colorectal cancer. Replication and cytotoxic assays revealed that PD-H replicated and lysed colorectal carcinoma cell lines in vitro as well as PD-0. Intratumoral injection of PD-H into subcutaneous DLD-1 tumors in nude mice resulted in strong inhibition of tumor growth and significantly prolonged the survival of the animals, but virus-induced systemic infection was observed in one of the six animals. In a syngenic mouse model of subcutaneously growing Colon-26 tumors, intratumoral administration of PD-H led to a significant reduction of tumor growth, the prolongation of animal survival, the prevention of tumor-induced cachexia, and the elevation of CD3+ and dendritic cells in the tumor microenvironment. No virus-induced side effects were observed. After intraperitoneal application, PD-H induced weak pancreatitis and myocarditis in immunocompetent mice. By equipping the virus with target sites of miR-375, which is specifically expressed in the pancreas, organ infections were prevented. Moreover, employment of this virus in a syngenic mouse model of CT-26 peritoneal carcinomatosis resulted in a significant reduction in tumor growth and an increase in animal survival. The results demonstrate that the immune status of the host, the route of virus application, and the engineering of the virus with target sites of suitable microRNAs are crucial for the use of PD-H as an oncolytic virus.


Subject(s)
Coxsackievirus Infections/immunology , Enterovirus/physiology , Oncolytic Viruses/physiology , Animals , CHO Cells , Colorectal Neoplasms , Cricetulus , Enterovirus/classification , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Nude , MicroRNAs , Myocarditis , Neoplasms , Oncolytic Viruses/classification
10.
FASEB J ; 35(11): e21975, 2021 11.
Article in English | MEDLINE | ID: mdl-34618980

ABSTRACT

Autoimmunity contributes to the pathogenesis of viral myocarditis (VMC), which is characterized by the production of anti-heart autoantibodies (AHA) from lymphoid follicles. Recently, the formation of ectopic lymphoid follicles (ELFs) was reported in heart grafts. However, the existence and role of ELFs in myocardial tissues of VMC remain unclear. This study aimed to explore whether and how cardiac ELFs with germinal centers (GCs) could be generated during the development of VMC. We identified the existence of ELFs and explored the underlying mechanism. In a BALB/c mouse model of VMC, the dynamic myocardial infiltrations of lymphocytic aggregates and expressions of associated lymphorganogenic factors were investigated, accompanied by the detection of the production and location of myocardial AHA. The data indicated ELFs formation in myocardial tissues of VMC, and the number of ELFs was in accordance with the severity of VMC. Moreover, the functional ELFs with GCs were capable of facilitating the production of local AHA. Blocking IL-17 or podoplanin (PDPN) could inhibit cardiac ELFs generation, perhaps due to the negative regulation of PDPN neutralization in Th17 cell proliferation and differentiation. The presence of cardiac ELFs and AHA might offer new opportunities for stratification and early identification of VMC patients.


Subject(s)
Coxsackievirus Infections/immunology , Interleukin-17/immunology , Membrane Glycoproteins/immunology , Myocarditis/immunology , Tertiary Lymphoid Structures/immunology , Th17 Cells/immunology , Animals , Cell Differentiation , Cells, Cultured , Male , Mice , Mice, Inbred BALB C , Th17 Cells/cytology
11.
Diabetes ; 70(12): 2871-2878, 2021 12.
Article in English | MEDLINE | ID: mdl-34497136

ABSTRACT

Enteroviruses, including the Coxsackievirus Bs (CVB), have been implicated as causal agents in human type 1 diabetes. Immunization of at-risk individuals with a CVB vaccine provides an attractive strategy for elucidating the role of CVBs in the disease etiology. Previously, we have shown that an inactivated whole-virus vaccine covering all CVB serotypes (CVB1-6) is safe to administer and highly immunogenic in preclinical models, including nonhuman primates. Before initiating clinical trials with this type of vaccine, it was also important to address 1) whether the vaccine itself induces adverse immune reactions, including accelerating diabetes onset in a diabetes-prone host, and 2) whether the vaccine can prevent CVB-induced diabetes in a well-established disease model. Here, we present results from studies in which female NOD mice were left untreated, mock-vaccinated, or vaccinated with CVB1-6 vaccine and monitored for insulitis occurrence or diabetes development. We demonstrate that vaccination induces virus-neutralizing antibodies without altering insulitis scores or the onset of diabetes. We also show that NOD mice vaccinated with a CVB1 vaccine are protected from CVB-induced accelerated disease onset. Taken together, these studies show that CVB vaccines do not alter islet inflammation or accelerate disease progression in an animal model that spontaneously develops autoimmune type 1 diabetes. However, they can prevent CVB-mediated disease progression in the same model.


Subject(s)
Coxsackievirus Infections/prevention & control , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/therapy , Viral Vaccines/therapeutic use , Animals , Antibodies, Neutralizing/therapeutic use , Coxsackievirus Infections/complications , Coxsackievirus Infections/immunology , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Disease Progression , Enterovirus B, Human/immunology , Female , Mice , Mice, Inbred NOD , Vaccination , Viral Vaccines/pharmacology
12.
Front Immunol ; 12: 649520, 2021.
Article in English | MEDLINE | ID: mdl-33968043

ABSTRACT

Rhinovirus C (RV-C) infection is associated with severe asthma exacerbations. Since type 2 inflammation is an important disease mechanism in asthma, we hypothesized that RV-C infection, in contrast to RV-A, preferentially stimulates type 2 inflammation, leading to exacerbated eosinophilic inflammation. To test this, we developed a mouse model of RV-C15 airways disease. RV-C15 was generated from the full-length cDNA clone and grown in HeLa-E8 cells expressing human CDHR3. BALB/c mice were inoculated intranasally with 5 x 106 ePFU RV-C15, RV-A1B or sham. Mice inoculated with RV-C15 showed lung viral titers of 1 x 105 TCID50 units 24 h after infection, with levels declining thereafter. IFN-α, ß, γ and λ2 mRNAs peaked 24-72 hrs post-infection. Immunofluorescence verified colocalization of RV-C15, CDHR3 and acetyl-α-tubulin in mouse ciliated airway epithelial cells. Compared to RV-A1B, mice infected with RV-C15 demonstrated higher bronchoalveolar eosinophils, mRNA expression of IL-5, IL-13, IL-25, Muc5ac and Gob5/Clca, protein production of IL-5, IL-13, IL-25, IL-33 and TSLP, and expansion of type 2 innate lymphoid cells. Analogous results were found in mice treated with house dust mite before infection, including increased airway responsiveness. In contrast to Rorafl/fl littermates, RV-C-infected Rorafl/flIl7rcre mice deficient in ILC2s failed to show eosinophilic inflammation or mRNA expression of IL-13, Muc5ac and Muc5b. We conclude that, compared to RV-A1B, RV-C15 infection induces ILC2-dependent type 2 airway inflammation, providing insight into the mechanism of RV-C-induced asthma exacerbations.


Subject(s)
Asthma/immunology , Coxsackievirus Infections/immunology , Enterovirus/immunology , Eosinophilia/immunology , Lymphocytes/immunology , Animals , Asthma/blood , Asthma/diagnosis , Asthma/virology , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cadherin Related Proteins , Cadherins/genetics , Cadherins/metabolism , Coxsackievirus Infections/blood , Coxsackievirus Infections/complications , Coxsackievirus Infections/virology , Disease Models, Animal , Enterovirus/metabolism , Eosinophilia/blood , Eosinophilia/virology , Eosinophils/immunology , Female , HeLa Cells , Humans , Immunity, Innate , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Symptom Flare Up
13.
Inflammation ; 44(5): 1949-1960, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33961174

ABSTRACT

Viral myocarditis (VMC) is the major cause of sudden death in adolescents. To date, no effective treatment has been identified for VMC. Studies have shown that T helper (Th) cells such as Th1, Th2, Th17, and Th22 cells are involved in the pathogenesis of VMC. However, the role of B cells and their impact on Th cells in VMC is unclear. In this study, we investigated the role of B cells in Th cell differentiation in myocardial damage in an animal model of VMC. C57BL/6 mice were infected with Coxsackievirus B3 (CVB3) intraperitoneally or injected with phosphate-buffered saline as a control condition. At day 7, samples from these mice were analyzed by histology, ELISA, flow cytometry, and gene expression assays. We found that TNF-α-, IL-6-, and IL-17-producing B cell numbers were significantly increased, while IL-4-producing B cell population was significantly reduced in acute VMC. Furthermore, we performed B cell knockout (BKO), SCID, and SCID+B cells reconstitution experiments. We found that BKO alleviated the cardiac damage following CVB3 infection, may hamper the differentiation of Th1 and Th17 cells, may promote the differentiation of Th2 cells, and proved ineffective for the differentiation of Th22 cells. In contrast, SCID+B cells reconstitution experiment exacerbated the cardiac damage. Ex vivo studies further revealed that B cells promote the differentiation of Th1 and Th17 cells and inhibit the differentiation of Th2 cells. Our study shows that B cells are activated and have strong abilities of antigen presentation and producing cytokines in VMC; B cells not only play a pathogenic role in VMC independent of T cells but also promote Th1 and Th17 cell differentiation, and hamper Th2 cell differentiation in VMC.


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation/physiology , Coxsackievirus Infections/immunology , Enterovirus B, Human/immunology , Myocarditis/immunology , Th2 Cells/immunology , Acute Disease , Animals , B-Lymphocytes/metabolism , Cells, Cultured , Coxsackievirus Infections/metabolism , HeLa Cells , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Myocarditis/metabolism , Th2 Cells/metabolism
14.
Int Immunopharmacol ; 95: 107534, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33752081

ABSTRACT

BACKGROUND: The aim of this study was to investigate the mechanism of STAT3 in reducing the inflammatory responses in mice with viral myocarditis (VMC). METHODS: Induce and generate viral myocarditis by using coxsackievirus B3 (CVB3) infected cardiomyocyte-specific STAT3 conditional knockout (STAT3cKO) mice and BALB/c mice. Use RT-PCR and western blot techniques to detect the expression of related cytokines in the uninfected wild-type mice group (Control group), myocarditis wild-type mice group (Model group) and STAT3cKO group, as well as the differentiation of spleen T cells in each group. Eukaryotic expression plasmid pcDNA3-STAT3 can reduce the expression of inflammatory factors the in vitro cultured cardiomyocytes of the STAT3cKO group. RESULTS: RT-PCR showed that compared with the Control group, the expression levels of VMC-related genes (NF-κB, TNF­α, IL-1ß and IL-1) and anti-inflammation-related cytokines (IL-10 and TGF-ß) in the Model group went up (*p < 0.05, **p < 0.01, ***p < 0.001); and also compared with the Control group, the rise in the expression levels of the above VMC-related genes in the STAT3cKO group was particularly significant (***p < 0.001, ****p < 0.0001) but there was no significant difference in the expression of IL-10 and TGF-ß. After 4 weeks, a second RT-PCR showed that the expression of inflammation-related genes in the STAT3cKO group continued to be activated (***p < 0.001, ****p < 0.0001). Western blotting was performed to detect the expression of p65, a key protein of the NF-κB signalling pathway. The results showed that the p65 protein content was increased and the IL-10 protein content was decreased in the STAT3cKO group; the results of the T cell differentiation test showed that the T cell differentiation rate increased in the STAT3cKO group (**p < 0.01). Eukaryotic expression plasmid pcDNA3-STAT3 could reduce the expression of NF-κB, TNF-α, IL-1ß and IL-17 (**p < 0.01). CONCLUSION: The expression of STAT3 gene in VMC could to a certain extent inhibit the NF-κB signalling pathway and reduce the inflammatory responses of VMC.


Subject(s)
Coxsackievirus Infections/immunology , Enterovirus B, Human , Myocarditis/immunology , NF-kappa B/immunology , STAT3 Transcription Factor/immunology , Animals , Cytokines/immunology , Male , Mice, Inbred BALB C , Mice, Knockout , Myocarditis/genetics , Myocardium/immunology , STAT3 Transcription Factor/genetics , Signal Transduction , T-Lymphocytes/immunology
15.
Basic Res Cardiol ; 116(1): 7, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33523326

ABSTRACT

A preclinical model of troponin I-induced myocarditis (AM) revealed a prominent role of the immunoproteasome (ip), the main immune cell-resident proteasome isoform, in heart-directed autoimmunity. Viral infection of the heart is a known trigger of cardiac autoimmunity, with the ip enhancing systemic inflammatory responses after infection with a cardiotropic coxsackievirusB3 (CV). Here, we used ip-deficient A/J-LMP7-/- mice to investigate the role of ip-mediated effects on adaptive immunity in CV-triggered myocarditis and found no alteration of the inflammatory heart tissue damage or cardiac function in comparison to wild-type controls. Aiming to define the impact of the systemic inflammatory storm under the control of ip proteolysis during CV infection, we targeted the ip in A/J mice with the inhibitor ONX 0914 after the first cycle of infection, when systemic inflammation has set in, well before cardiac inflammation. During established acute myocarditis, the ONX 0914 treatment group had the same reduction in cardiac output as the controls, with inflammatory responses in heart tissue being unaffected by the compound. Based on these findings and with regard to the known anti-inflammatory role of ONX 0914 in CV infection, we conclude that the efficacy of ip inhibitors for CV-triggered myocarditis in A/J mice relies on their immunomodulatory effects on the systemic inflammatory reaction.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Coxsackievirus Infections/drug therapy , Inflammation/drug therapy , Myeloid Cells/drug effects , Myocarditis/drug therapy , Myocytes, Cardiac/drug effects , Oligopeptides/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Animals , Cells, Cultured , Coxsackievirus Infections/enzymology , Coxsackievirus Infections/immunology , Disease Models, Animal , Enterovirus B, Human/immunology , Enterovirus B, Human/pathogenicity , Host-Pathogen Interactions , Inflammation/enzymology , Inflammation/immunology , Inflammation/virology , Male , Mice, Knockout , Myeloid Cells/enzymology , Myeloid Cells/immunology , Myeloid Cells/virology , Myocarditis/enzymology , Myocarditis/immunology , Myocarditis/virology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/virology , Proteasome Endopeptidase Complex/genetics , Proteolysis
16.
Virulence ; 12(1): 360-376, 2021 12.
Article in English | MEDLINE | ID: mdl-33380272

ABSTRACT

Abnormalities in CD4+ T cell (Th cell) differentiation play an important role in the pathogenesis of viral myocarditis (VMC). Our previous studies demonstrated that activation of the cholinergic anti-inflammatory pathway (CAP) alleviated the inflammatory response. In addition, we observed that right cervical vagotomy aggravates VMC by inhibiting CAP. However, the vagus nerve's effect on differentiation of CD4+ T cells has not been studied in VMC mice to date. In this study, we investigated the effects of cervical vagotomy and the α7nAChR agonist pnu282987 on CD4+ T cell differentiation in a murine myocarditis model (BALB/c) infected with coxsackievirus B3 (CVB3). Splenic CD4+ T cells from CVB3-induced mice obtained and cultured to investigate the potential mechanism of CD4+ T cell differentiation. Each Th cell subset was analyzed by flow cytometry. Our results showed that right cervical vagotomy increased proportions of Th1 and Th17 cells and decreased proportions of Th2 and Treg cells in the spleen. Vagotomy-induced upregulation of T-bet, Ror-γ, IFN-γ, and IL-17 expression while downregulating the expression of Gata3, Foxp3, and IL-4 in the heart. In addition, we observed upregulated levels of proinflammatory cytokines, aggravated myocardial lesions and cellular infiltration, and worsened cardiac function in VMC mice. Pnu282987 administration reversed these outcomes. Furthermore, vagotomy inhibited JAK2-STAT3 activation and enhanced NF-κB activation in splenic CD4+ T cells. The CD4+ T cell differentiation was related to JAK2-STAT3 and NF-κB signal pathways. In conclusion, vagus nerve modulates the inflammatory response by regulating CD4+ T cell differentiation in response to VMC.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Cell Differentiation/immunology , Coxsackievirus Infections/immunology , Enterovirus B, Human/immunology , Myocarditis/immunology , Myocarditis/virology , Vagus Nerve/immunology , Acute Disease , Animals , CD4-Positive T-Lymphocytes/immunology , Cytokines/immunology , Enterovirus B, Human/classification , Male , Mice , Mice, Inbred BALB C
17.
Nat Rev Cardiol ; 18(3): 169-193, 2021 03.
Article in English | MEDLINE | ID: mdl-33046850

ABSTRACT

Inflammatory cardiomyopathy, characterized by inflammatory cell infiltration into the myocardium and a high risk of deteriorating cardiac function, has a heterogeneous aetiology. Inflammatory cardiomyopathy is predominantly mediated by viral infection, but can also be induced by bacterial, protozoal or fungal infections as well as a wide variety of toxic substances and drugs and systemic immune-mediated diseases. Despite extensive research, inflammatory cardiomyopathy complicated by left ventricular dysfunction, heart failure or arrhythmia is associated with a poor prognosis. At present, the reason why some patients recover without residual myocardial injury whereas others develop dilated cardiomyopathy is unclear. The relative roles of the pathogen, host genomics and environmental factors in disease progression and healing are still under discussion, including which viruses are active inducers and which are only bystanders. As a consequence, treatment strategies are not well established. In this Review, we summarize and evaluate the available evidence on the pathogenesis, diagnosis and treatment of myocarditis and inflammatory cardiomyopathy, with a special focus on virus-induced and virus-associated myocarditis. Furthermore, we identify knowledge gaps, appraise the available experimental models and propose future directions for the field. The current knowledge and open questions regarding the cardiovascular effects associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are also discussed. This Review is the result of scientific cooperation of members of the Heart Failure Association of the ESC, the Heart Failure Society of America and the Japanese Heart Failure Society.


Subject(s)
Cardiomyopathies/physiopathology , Inflammation/physiopathology , Myocarditis/physiopathology , Virus Diseases/physiopathology , Animals , Antiviral Agents/therapeutic use , Autoimmunity/immunology , Biopsy , COVID-19/physiopathology , COVID-19/therapy , Cardiomyopathies/diagnosis , Cardiomyopathies/immunology , Cardiomyopathies/therapy , Cardiomyopathy, Dilated , Coronavirus Infections/immunology , Coronavirus Infections/physiopathology , Coronavirus Infections/therapy , Coxsackievirus Infections/immunology , Coxsackievirus Infections/physiopathology , Coxsackievirus Infections/therapy , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/physiopathology , Cytomegalovirus Infections/therapy , Disease Models, Animal , Echovirus Infections/immunology , Echovirus Infections/physiopathology , Echovirus Infections/therapy , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/physiopathology , Epstein-Barr Virus Infections/therapy , Erythema Infectiosum/immunology , Erythema Infectiosum/physiopathology , Erythema Infectiosum/therapy , HIV Infections/physiopathology , Hepatitis C/immunology , Hepatitis C/physiopathology , Hepatitis C/therapy , Humans , Immunoglobulins, Intravenous/therapeutic use , Immunologic Factors/therapeutic use , Inflammation/diagnosis , Inflammation/immunology , Inflammation/therapy , Influenza, Human/immunology , Influenza, Human/physiopathology , Influenza, Human/therapy , Leukocytes/immunology , Myocarditis/diagnosis , Myocarditis/immunology , Myocarditis/therapy , Myocardium/pathology , Prognosis , Roseolovirus Infections/immunology , Roseolovirus Infections/physiopathology
18.
Biomed Res Int ; 2020: 9850351, 2020.
Article in English | MEDLINE | ID: mdl-33274234

ABSTRACT

BACKGROUND: Scarce information exists about immunity to hand, foot, and mouth disease (HFMD) among household contacts of index cases in Vietnam and what that means for reducing ongoing HFMD transmission in the community. METHODS: We analyzed neutralizing antibodies (NT) and the incidence of enterovirus (EVs) infection among household contacts of index cases in a province where HFMD remains endemic. Throat swab and 2 mL blood samples from household contacts were collected at enrollment, during and after 2 weeks follow-up. RESULTS: The incidence of EV-A71 infection among household contacts was 40/84 (47.6%, 95% Cl: 36.9-58.3%), compared with 106/336 (31.5%, 95% Cl: 26.6-36.5%) for CV-A6 and 36/107 (33.6%, 95% Cl: 24.7-42.6%) for CV-A16. The incidence of CV-A6 infection was fairly constant across ages; in contrast, CV-A71 and CV-A16 had some variation across ages. At baseline, higher geometric mean titer (GMT) of EV-A71, CV-A6, and CV-A16 antibody titers was found for 25-34-year groups (range 216.3 to 305.0) compared to the other age groups. There was a statistically significant difference in GMT values of CV-A6 and CV-A16 between those who had an infection or did not have infection among households with an index case of these serotypes. CONCLUSIONS: Our results indicated that adults were becoming infected with HFMD and could be contributing to the transmission. There is, therefore, a need for considering the household setting as an additional target for intervention programs for HFMD.


Subject(s)
Coxsackievirus Infections/epidemiology , Coxsackievirus Infections/virology , Enterovirus A, Human/physiology , Enterovirus Infections/epidemiology , Enterovirus Infections/virology , Enterovirus/physiology , Family Characteristics , Adolescent , Adult , Age Factors , Antibodies, Neutralizing , Child , Child, Preschool , Coxsackievirus Infections/immunology , Enterovirus/immunology , Enterovirus A, Human/immunology , Enterovirus Infections/immunology , Follow-Up Studies , Humans , Incidence , Middle Aged , Seroepidemiologic Studies , Serogroup , Vietnam/epidemiology , Viral Load , Young Adult
19.
Int Immunopharmacol ; 88: 106941, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33182061

ABSTRACT

Viral myocarditis (VMC) is characterized by cardiac inflammation and excessive inflammatory responses after viral infection. SENP2, a deSUMO-specific protease, has been reported to regulate antiviral innate immunity. This study aimed to investigate whether SENP2 affects CVB3-induced VMC. We generated a CVB3-induced VMC mouse model in 6-week-old cardiomyocyte-specific Senp2 knockout mice. The mice were sacrificed at days 0, 2, 4 and 6 after CVB3 infection. The survival rate, body weight, myocardial histopathological changes, viral load, cytokine levels and antiviral gene expression in cardiac tissues of both groups were investigated. Our study indicated that the expression of Senp2 in primary cardiomyocytes was upregulated by CVB3 infection. Moreover, deletion of Senp2 in the heart exacerbated CVB3 infection-induced myocarditis, facilitated CVB3 viral replication and downregulated the expression of antiviral proteins. In conclusion, our findings suggest a protective role for SENP2 in CVB3-induced VMC.


Subject(s)
Coxsackievirus Infections/immunology , Cysteine Endopeptidases/immunology , Enterovirus B, Human/physiology , Myocarditis/immunology , Myocytes, Cardiac/immunology , Animals , Cells, Cultured , Coxsackievirus Infections/complications , Coxsackievirus Infections/virology , Cysteine Endopeptidases/genetics , Male , Mice, Inbred C57BL , Mice, Transgenic , Myocarditis/etiology , Myocarditis/virology , Myocytes, Cardiac/pathology , Virus Replication
20.
Commun Biol ; 3(1): 580, 2020 10 16.
Article in English | MEDLINE | ID: mdl-33067530

ABSTRACT

Previous research suggests that hepatocytes catabolize chemical toxins but do not remove microbial agents, which are filtered out by other liver cells (Kupffer cells and endothelial cells). Here we show that, contrary to current understanding, hepatocytes trap and rapidly silence type B coxsackieviruses (CVBs). In genetically wildtype mice, this activity causes hepatocyte damage, which is alleviated in mice carrying a hepatocyte-specific deletion of the coxsackievirus-adenovirus receptor. However, in these mutant mice, there is a dramatic early rise in blood-borne virus, followed by accelerated systemic disease and increased mortality. Thus, wild type hepatocytes act similarly to a sponge for CVBs, protecting against systemic illness at the expense of their own survival. We speculate that hepatocytes may play a similar role in other viral infections as well, thereby explaining why hepatocytes have evolved their remarkable regenerative capacity. Our data also suggest that, in addition to their many other functions, hepatocytes might be considered an integral part of the innate immune system.


Subject(s)
Coxsackievirus Infections/virology , Disease Resistance , Enterovirus/physiology , Hepatocytes/metabolism , Hepatocytes/virology , Host-Pathogen Interactions , Animals , Coxsackie and Adenovirus Receptor-Like Membrane Protein/deficiency , Coxsackievirus Infections/genetics , Coxsackievirus Infections/immunology , Coxsackievirus Infections/pathology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Disease Resistance/genetics , Disease Resistance/immunology , Gene Expression , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Immunity, Innate , Interferon-alpha/metabolism , Liver/metabolism , Liver/pathology , Liver/virology , Mice , Mice, Knockout , Mortality , Viral Load , Viremia
SELECTION OF CITATIONS
SEARCH DETAIL
...