1
|
Ding X, Lai X, Klaestrup IH, Jensen SRN, Nielsen MM, Thorsen K, Romero-Ramos M, Luo Y, Lin L, Reinert LS, Paludan SR. Temporally resolved single-cell RNA sequencing reveals protective and pathological responses during herpes simplex virus CNS infection. J Neuroinflammation 2025; 22:146. [PMID: 40450318 DOI: 10.1186/s12974-025-03471-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/19/2025] [Indexed: 06/03/2025] Open
Abstract
BACKGROUND Herpes Simplex Virus 1 (HSV-1) is a neurotropic virus causing encephalitis and post-infectious complications. Infections can induce a range of acute, subacute, and progressing brain disease, and in recent years it has emerged that immune responses are involved in the pathogenesis of these diseases. METHODS Mice were infected with HSV-1 through corneal infection, and the brain stem was analyzed using single-cell and GeoMx spatial transcriptomics. Through these technologies we profiled temporal transcriptomic changes in cell populations, pathways, and cell-cell communication associated with antiviral activity and inflammation-induced disturbance of physiological brain structures and activities. RESULTS We found that microglia proportions increased early after HSV-1 infection, followed by monocyte influx and later by T cells. The blood-brain barrier was disrupted, and transcriptomic profiles associated with homeostatic brain transcriptional activities were altered. Early transcriptional responses were dominated by antiviral and inflammatory activities. A microglia subpopulation with high type I interferon and chemokine expression localized to infection sites, likely mediating antiviral defense and immune recruitment. Monocyte subpopulations displayed a broader activation profile than microglia and was a central mediator of crosstalk between immune cells. Cytokines from microglia, monocytes, and T cells reprogrammed brain cells, notably endothelial cells and oligodendrocytes, disrupting brain functions. Comparing datasets from various brain diseases revealed the identified microglia subpopulation as specific to viral infections. CONCLUSIONS This study identifies a unique population of virus-activated microglia with antiviral and proinflammatory properties and reveals monocytes to be a key driver of interactions driving pathology in the virus-infected brain.
Collapse
Affiliation(s)
- Xiangning Ding
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark
| | - Xin Lai
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark
| | - Ida H Klaestrup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- DANDRITE, Aarhus University, Aarhus, Denmark
| | - Sara R N Jensen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Morten M Nielsen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Kasper Thorsen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Marina Romero-Ramos
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- DANDRITE, Aarhus University, Aarhus, Denmark
| | - Yonglun Luo
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Lin Lin
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Line S Reinert
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark
| | - Søren R Paludan
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- Center for Immunology of Viral Infections, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
2
|
Zhang R, Qiao H, Zhou K, Ju X, Cao X, Dong J, Wu M, Yu L, Zhang S. An immune-based predictive model for HBV clearance: validation in multicenter cohorts and mechanistic insights from in vivo studies. Virol J 2025; 22:153. [PMID: 40399947 PMCID: PMC12096729 DOI: 10.1186/s12985-025-02792-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 05/13/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND Chronic HBV infection is a major risk factor for hepatocellular carcinoma, posing a significant global health burden. However, predictive models for HBV clearance based on immune biomarkers remain limited. METHODS We systematically developed a predictive tool by quantifying mRNA expression levels of CD4⁺ T-cell subset transcription factors, cytokines, and immune checkpoints in PBMCs from chronic HBV patients and resolved HBV individuals using RT-qPCR. A binary logistic regression model was constructed in the training cohort, with performance evaluated by ROC and calibration curves, followed by internal and external validation in independent cohorts. For in vivo validation, an HBV-transfected mouse model was established via rapid tail vein injection of pGL3-CP-Fluc-HBV1.2C2 plasmid. Outcomes included body weight, HBsAg/HBV DNA levels, and luciferase activity. Kaplan-Meier analysis assessed cumulative clearance rates, while RT-qPCR tracked model-related mRNA dynamics in PBMCs. RESULTS The model identified GATA3, FOXP3, IFNG, TNF, and HAVCR2 as key genes, demonstrating robust predictive accuracy for HBV clearance. Dose-specific temporal patterns of immune gene regulation were observed, revealing distinct immunomodulatory mechanisms between groups. CONCLUSION This study establishes a reliable immune-based predictive model for HBV clearance and highlights divergent immune responses in chronic versus resolved infection.
Collapse
Affiliation(s)
- Rongzheng Zhang
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
| | - Han Qiao
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
| | - Kun Zhou
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
- Department of Clinical Laboratory, Beidahuang Industry Group General Hospital, Harbin, 150000, China
| | - Xiaomei Ju
- Department of Clinical Laboratory, The First Clinical Hospital of Jilin Academy of Traditional Chinese Medicine, Changchun, 130000, China
| | - Xinyang Cao
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
| | - Jianming Dong
- Department of Immunology, Harbin Medical University, Harbin, 150000, China
| | - Meng Wu
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
| | - Le Yu
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, China
| | - Shuyun Zhang
- Scientific Research Center, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150000, China.
| |
Collapse
|
3
|
Raha JR, Kim KH, Le CTT, Bhatnagar N, Pal SS, Liu R, Grovenstein P, Yeasmin M, Racheal F, Shin CH, Wang BZ, Kang SM. Intranasal vaccination with multi-neuraminidase and M2e virus-like particle vaccine results in greater mucosal immunity and protection against influenza than intramuscular injection. Vaccine 2025; 57:127206. [PMID: 40339180 DOI: 10.1016/j.vaccine.2025.127206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/10/2025]
Abstract
Intramuscular injection of seasonal influenza vaccines provides strain-specific neutralizing antibodies, but not against variants, and no effective mucosal immunity. Here, we report that multi-subtype neuraminidase (NA) and M2 ectodomain repeat (5xM2e) virus-like particle vaccine (NA-M2e) conferred higher efficacy of broad cross-protection after two doses of intranasal delivery than intramuscular injection. The intranasally vaccinated mice displayed high levels of IgA antibodies, IFN-γ+ CD4 and CD8 T cells, germinal center B cells, plasma cells, and early innate immune cells locally in the lungs. In contrast, intramuscular vaccination systemically induced innate and adaptive immune responses in the spleen. Our findings demonstrate that the intranasal delivery of NA-M2e vaccine induces enhanced mucosal immunity and comparable serum IgG antibodies, offering improved efficacy of cross-protection against diverse influenza virus strains compared to the intramuscular injection in a mouse model.
Collapse
Affiliation(s)
- Jannatul Ruhan Raha
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Chau Thuy Tien Le
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Noopur Bhatnagar
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Surya Sekhar Pal
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Rong Liu
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Phillip Grovenstein
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Mahmuda Yeasmin
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Farayola Racheal
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Chong Hyun Shin
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Bao-Zhong Wang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30302, USA.
| |
Collapse
|
4
|
Abduljaleel Z. Decoding SARS-CoV-2 variants: Mutations, viral stability, and breakthroughs in vaccines and therapies. Biophys Chem 2025; 320-321:107413. [PMID: 39987705 DOI: 10.1016/j.bpc.2025.107413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/06/2025] [Accepted: 02/13/2025] [Indexed: 02/25/2025]
Abstract
This study investigates the infectivity of SARS-CoV-2 and its immune evasion mechanisms, particularly through mutations in the spike protein that enable the virus to escape host immune responses. As global vaccination efforts continue, understanding viral evolution and immune evasion strategies remains critical. This analysis focuses on fourteen key mutations (Arg346Lys, Lys417Asp, Leu452Glu, Leu452Arg, Phe456Leu, Ser477Asp, Thr478Lys, Glu484Ala, Glu484Lys, Glu484Gln, Gln493Arg, Gly496Ser, Glu498Arg, and His655Y) within the receptor-binding domain (RBD) of the spike protein. The results reveal consistent patterns of immune escape across various SARS-CoV-2 variants, with specific mutations influencing protein stability, binding affinity to the hACE2 receptor, and antibody recognition. These findings demonstrate how single-point mutations can destabilize the spike protein and reduce the efficacy of the immune response. By correlating expression levels and thermodynamic stability with immune evasion, this study provides valuable insights into the functional characteristics of the spike protein. The findings contribute to the understanding of immune escape variants and identify potential targets for enhancing vaccine efficacy and developing therapeutic approaches in response to the evolving SARS-CoV-2 landscape. SHORT SUMMARY: The study investigates the infectivity of SARS-CoV-2 and its implications for immune evasion. It focuses on fourteen key mutations within the spike protein's Receptor-Binding Domain (S-RBD) and reveals consistent patterns associated with immune escape in various SARS-CoV-2 variants. The research highlights the influence of factors such as protein fold stability, hACE2 binding, and antibody evasion on spike protein evolution. Single-point immune escape variants alter virus stability, impacting antibody response success. The study provides valuable insights into immune escape variants and suggests avenues for enhancing vaccine efficacy. It also opens the way for novel therapeutic approaches in the context of SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Zainularifeen Abduljaleel
- Faculty of Medicine, Department of Medical Genetics, Umm Al-Qura University, P.O. Box 715, Makkah 21955, Saudi Arabia.
| |
Collapse
|
5
|
Myburgh L, Karsjens H, Blanas A, de Ligt A, van Loon K, Huijbers EJM, van Beijnum JR, Engbersen DJM, Rekiki A, Mignon C, Vratskikh O, Griffioen AW. Targeting the early life stages of SARS-CoV-2 using a multi-peptide conjugate vaccine. Vaccine 2025; 54:126989. [PMID: 40088511 DOI: 10.1016/j.vaccine.2025.126989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 02/26/2025] [Accepted: 03/01/2025] [Indexed: 03/17/2025]
Abstract
The spike glycoprotein is a key factor in the infection cycle of SARS-CoV-2, as it mediates both receptor recognition and membrane fusion by the virus. Therefore, in this study, we aimed to design a multi-peptide conjugate vaccine against SARS-CoV-2, targeting the early stages of the virus's life cycle. We used iBoost technology, which is designed to induce immune responses against low- or non-immunogenic epitopes. We selected six peptide sequences, each representing a key domain of the spike protein (i.e., receptor binding domain (RBM), subdomain 1 (SD1), subdomain 2 (SD2), S1/S2, fusion peptide and the S2' sequences (FP + S2'), heptad repeat 1 (HR1)). Immunization studies in mice displayed targeted humoral and cellular immune responses against specific peptides of the spike protein simultaneously, while inducing cross-protection against the Delta and Omicron coronavirus variants. Moreover, vaccinated hamsters challenged with SARS-CoV-2 elicited high antibody levels against key peptides, induced early neutralizing antibody responses and resulted in less weight loss compared to controls. This highlights the potential for improving viral control and disease outcomes when utilizing this strategy. Therefore, by using iBoost technology in conjunction with our peptide design strategy, we were able to successfully target non-immunodominant regions in the spike protein while activating both arms of the adaptive immune system.
Collapse
MESH Headings
- Animals
- SARS-CoV-2/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/chemistry
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Mice
- COVID-19/prevention & control
- COVID-19/immunology
- Vaccines, Subunit/immunology
- Vaccines, Subunit/administration & dosage
- Cricetinae
- Vaccines, Conjugate/immunology
- Vaccines, Conjugate/administration & dosage
- Female
- Mice, Inbred BALB C
- Immunity, Cellular
- Humans
- Cross Protection
- Immunity, Humoral
- Epitopes/immunology
Collapse
Affiliation(s)
- Lauren Myburgh
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Haiko Karsjens
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Athanasios Blanas
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Aafke de Ligt
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Karlijn van Loon
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands
| | - Elisabeth J M Huijbers
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands; CimCure BV, Amsterdam, the Netherlands
| | - Judy R van Beijnum
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands; CimCure BV, Amsterdam, the Netherlands
| | | | | | | | | | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Amsterdam, the Netherlands; CimCure BV, Amsterdam, the Netherlands.
| |
Collapse
|
6
|
Liu M, Brodeur KE, Bledsoe JR, Harris CN, Joerger J, Weng R, Hsu EE, Lam MT, Rimland CA, LeSon CE, Yue J, Henderson LA, Dedeoglu F, Newburger JW, Nigrovic PA, Son MBF, Lee PY. Features of hyperinflammation link the biology of Epstein-Barr virus infection and cytokine storm syndromes. J Allergy Clin Immunol 2025; 155:1346-1356.e9. [PMID: 39622297 DOI: 10.1016/j.jaci.2024.11.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 01/24/2025]
Abstract
BACKGROUND Overt immune activation by viral infections can lead to cytokine storm syndromes, such as hemophagocytic lymphohistiocytosis (HLH) and macrophage activation syndrome (MAS). OBJECTIVE We aimed to compare the immune response to different viral pathogens to understand the connection between infections and cytokine storm syndromes. METHODS We recruited children who sought care at the emergency department with fever for ≥3 days. We performed immune profiling using Olink proximity extension assay and flow cytometry. We compared the findings with cases of HLH, MAS, Kawasaki disease (KD), and multisystem inflammatory syndrome in children (MIS-C). RESULTS We enrolled 352 febrile patients and studied 110 cases of confirmed common viral infections. We found that Epstein-Barr virus (EBV) uniquely triggered high levels of multiple cytokines (IL-18, IL-27, TNF, FLT3 ligand, and lymphotoxin alpha) and IFN-γ-induced chemokines (CXCL9/10/11 and CCL19). These patterns are similar to the hyperinflammatory response associated with HLH/MAS but are less consistent with the findings in KD and MIS-C. Flow cytometry analysis revealed that CD38+HLA-DR+ T lymphocytes, which are pathogenic cells responsible for IFN-γ production in HLH/MAS, are vastly expanded in patients with acute EBV infection. Cell sorting identified CD38+HLA-DR+ T cells as atypical lymphocytes that are classically associated with acute EBV infection. CONCLUSION This work broadens our understanding of common viral infections in children and provides an immunologic basis for the link between EBV infection and HLH/MAS.
Collapse
Affiliation(s)
- Meng Liu
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass; Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Kailey E Brodeur
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Jacob R Bledsoe
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Claudia N Harris
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Jill Joerger
- Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Rachel Weng
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Evan E Hsu
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Michael T Lam
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Casey A Rimland
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass; Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - Courtney E LeSon
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Jian Yue
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass; Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Lauren A Henderson
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Fatma Dedeoglu
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Jane W Newburger
- Division of Cardiology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Peter A Nigrovic
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass; Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass
| | - Mary Beth F Son
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Pui Y Lee
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass.
| |
Collapse
|
7
|
Prakruthi J, Kori D, Paramshetti S, Ahamed SFN, Panchagnula R, Chandrashekara S. Old Age, B Cell Function and Count Are the Critical Factors for Predicting Infection Risk in Patients With Autoimmune Rheumatic Diseases Undergoing Immunosuppression: A Cohort Study. Int J Rheum Dis 2025; 28:e70187. [PMID: 40123284 DOI: 10.1111/1756-185x.70187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/11/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025]
Abstract
OBJECTIVE The study aims to assess baseline immune parameters that predict infection risk in autoimmune rheumatic disease (ARD) patients, with the goal of identifying high-risk individuals requiring immunosuppressive therapy escalation, based on infection rates during a one-year follow-up. METHODS The independent cohort study was conducted at a tertiary rheumatology center in India from December 2019 to March 2022. It included adult participants with ARDs undergoing immunosuppression. Ethics approval and informed consent were obtained. Patients underwent detailed history, clinical examination, and baseline investigations, which included complete hemogram, inflammatory parameters, immunoglobulin levels, cellular levels of the immune system, complement levels, and viral markers. Descriptive statistics, ANOVA, chi-squared tests, t-tests, and Fisher's exact tests were used. OLS regression analyses identified significant predictors of infection risk. They were followed up for a period of 1 year for any infection episodes. RESULTS Of the 106 participants recruited, 4 were excluded due to disease-related complications during the 3-month period of follow-up. The mean age of the participants was 38.21 ± 12.73 years, with an average follow-up duration of 13.1 ± 8.35 months. Among the remaining 102 participants, younger age was associated with a lower infection risk (OR 1.047). Protective factors against infection included lower levels of immunoglobulin E (IgE) (OR 0.379), methotrexate (MTX) use (OR 0.247), and biologics (OR 0.543). Conversely, lower Immunoglobulin G (IgG), elevated neutrophil counts (OR 3.588), higher neutrophil-to-lymphocyte ratios (NLR) (OR 2.577), low platelet counts (OR 0.546), and steroid use, which increased the risk fivefold (OR 5.686), were identified as risk factors. Ordinary Least Squares (OLS) regression analysis highlighted age, IgG levels, CD19 lymphocyte counts, WBC counts, and ESR as significant predictors of infection risk between the groups. CONCLUSION Older age, low IgG, low B cell count (CD19) predict susceptibility to infections; high neutrophil counts, low platelets, and elevated NLR are key predictors of developing infection in ARDs patients. Careful monitoring and tailored treatment strategies are essential to reduce infection risks. Further research is needed in this direction to develop predictive algorithms.
Collapse
Affiliation(s)
- Jaladhar Prakruthi
- ChanRe Rheumatology & Immunology Center & Research, Bengaluru, Karnataka, India
| | - Devaraj Kori
- ChanRe Rheumatology & Immunology Center & Research, Bengaluru, Karnataka, India
| | - Shruti Paramshetti
- ChanRe Rheumatology & Immunology Center & Research, Bengaluru, Karnataka, India
| | | | | | - S Chandrashekara
- ChanRe Rheumatology & Immunology Center & Research, Bengaluru, Karnataka, India
| |
Collapse
|
8
|
Reikvam H, Tsykunova G, Sandnes M, Wendelbo Ø. Infectious complications and the utility of serum and cellular markers of infections in the setting of allogeneic hematopoietic stem cell transplantation. Expert Rev Clin Immunol 2025; 21:291-303. [PMID: 39760208 DOI: 10.1080/1744666x.2025.2450014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 12/24/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
INTRODUCTION Allogeneic hematopoietic stem cell transplantation (allo-HSCT) recipients are severely immunocompromised and susceptible to bacterial, viral, and fungal infections. Despite improved anti-microbial prophylaxis and preemptive strategies, bacterial bloodstream infections (BSIs) occur frequently in allo-HSCT recipients and are associated with increased morbidity and mortality. Cytomegalovirus (CMV) and Epstein Barr virus (EBV) are the most relevant viruses following allo-HSCT and remain major concerns. Fungal infections, including those caused by Candida and Aspergillus species, are persistent and feared complications. AREAS COVERED We aim to provide clinicians caring for allo-HSCT recipients with a comprehensive overview of the risk factors that predispose patients to common bacterial, fungal, and viral infections during the first years post-transplant. The focus is on the value of noninvasive diagnostic biomarkers and serological assays in enhancing the early detection and management of these infections. EXPERT OPINION Effective management of infectious complications following allo-HSCT relies on continuous immune recovery monitoring and the implementation of advanced diagnostic methods. Utilizing noninvasive diagnostic methods is crucial for early detection and different intervention strategies. The development and integration of reliable microbiological markers into clinical practice is essential for enhancing patient outcomes and mitigating infection-related risks. Emphasizing diagnostic innovation will be pivotal in advancing patient care post-allo-HSCT.
Collapse
Affiliation(s)
- Håkon Reikvam
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
- K.G. Jebsen Center for Myeloid Blood Cancer, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Galina Tsykunova
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Hemato- oncology, Østfold Hospital, Grålum, Norway
| | - Miriam Sandnes
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Øystein Wendelbo
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
- Faculty of Health, VID Specialized University, Bergen, Norway
| |
Collapse
|
9
|
Cyr Y, Gourvest M, Ciabattoni GO, Zhang T, Newman AA, Zahr T, Delbare S, Schlamp F, Dittmann M, Moore KJ, van Solingen C. lncRNA CARINH regulates expression and function of innate immune transcription factor IRF1 in macrophages. Life Sci Alliance 2025; 8:e202403021. [PMID: 39773901 PMCID: PMC11707381 DOI: 10.26508/lsa.202403021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025] Open
Abstract
The discovery of long non-coding RNAs (lncRNAs) has provided a new perspective on the centrality of RNA in gene regulation and genome organization. Here, we screened for lncRNAs with putative functions in the host response to single-stranded RNA respiratory viruses. We identify CARINH as a conserved cis-acting lncRNA up-regulated in three respiratory diseases to control the expression of its antisense gene IRF1, a key transcriptional regulator of the antiviral response. CARINH and IRF1 are coordinately increased in the circulation of patients infected with human metapneumovirus, influenza A virus, or SARS-CoV-2, and in macrophages in response to viral infection or TLR3 agonist treatment. Targeted depletion of CARINH or its mouse ortholog Carinh in macrophages reduces the expression of IRF1/Irf1 and their associated target gene networks, increasing susceptibility to viral infection. Accordingly, CRISPR-mediated deletion of Carinh in mice reduces antiviral immunity, increasing viral burden upon sublethal challenge with influenza A virus. Together, these findings identify a conserved role of lncRNA CARINH in coordinating interferon-stimulated genes and antiviral immune responses.
Collapse
Affiliation(s)
- Yannick Cyr
- Department of Medicine, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Morgane Gourvest
- Department of Medicine, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Grace O Ciabattoni
- Department of Microbiology, New York University Langone Health, New York, NY, USA
| | - Tracy Zhang
- Department of Medicine, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Alexandra Ac Newman
- Department of Medicine, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Tarik Zahr
- Department of Medicine, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Sofie Delbare
- Department of Medicine, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Florencia Schlamp
- Department of Medicine, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Meike Dittmann
- Department of Microbiology, New York University Langone Health, New York, NY, USA
| | - Kathryn J Moore
- Department of Medicine, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA
- Department of Cell Biology, New York University Langone Health, New York, NY, USA
| | - Coen van Solingen
- Department of Medicine, Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
10
|
Cavalcante MSB, Santos DS, Araújo LM, Freitas PL, Silva CAM, Carvalho KGB, Araújo MTF, da Silva EVP, Rodrigues de Farias APD, Guerreiro Diniz D, Picanço Diniz CW, Diniz JAP. Inflammatory and neuropathological responses to Vesiculovirus carajas encephalitis in adult mice: variability, tolerance and resistance. Front Cell Infect Microbiol 2025; 15:1499658. [PMID: 40078875 PMCID: PMC11897020 DOI: 10.3389/fcimb.2025.1499658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 02/05/2025] [Indexed: 03/14/2025] Open
Abstract
Vesiculovirus carajas (CARV) is a pathogen with neuroinvasive potential, yet its impact on neuroinflammation and sickness behavior remains poorly understood. In this study, we investigated the neuropathological and immunological responses to CARV encephalitis in adult BALB/c mice. Mice were intranasally inoculated with either infected or uninfected brain homogenates, and clinical, histopathological, and cytokine profiles were analyzed. CARV antigens were primarily detected in necrotic neurons, with prominent microglial activation near the ventricles and blood vessels. By day 10 post-infection, infected mice exhibited significantly elevated levels of MCP-1, IFN-γ, IL-12 p70, TNF-α, IL-6, and IL-10 in the brain, indicating a strong inflammatory response. These findings highlight the inflammatory modulation associated with CARV infection and suggest a hematogenous route of neuroinvasion, distinguishing CARV from other vesiculovirus species. This study provides new insights into the pathogenesis of CARV encephalitis and its potential impact on neuroimmune dynamics.
Collapse
Affiliation(s)
- Maria Sueli Barbosa Cavalcante
- Programa de Pós-Graduação em Neurociências e Biologia Celular, Universidade Federal do Pará, Belém, Pará, Brazil
- Laboratório de Análises Clínicas do Hospital Universitário João de Barros Barreto, Empresa Brasileira de Serviços Hospitalares, Belém, Pará, Brazil
| | - Diego Siqueira Santos
- Laboratório de Análises Clínicas do Hospital Universitário João de Barros Barreto, Empresa Brasileira de Serviços Hospitalares, Belém, Pará, Brazil
| | - Lidineuza Machado Araújo
- Laboratório de Análises Clínicas do Hospital Universitário João de Barros Barreto, Empresa Brasileira de Serviços Hospitalares, Belém, Pará, Brazil
| | - Priscilla Lieuthier Freitas
- Laboratório de Análises Clínicas do Hospital Universitário João de Barros Barreto, Empresa Brasileira de Serviços Hospitalares, Belém, Pará, Brazil
| | | | | | | | | | | | - Daniel Guerreiro Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil
| | - Cristovam Wanderley Picanço Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Hospital Universitário João de Barros Barreto, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Pará, Brazil
| | | |
Collapse
|
11
|
Ni J, Kong D, Chen Z, Zeng W, Zhan B, Gong Z. Epidemiological Characteristics of Hemorrhagic Fever with Renal Syndrome in Longyou County, China. Viruses 2025; 17:313. [PMID: 40143244 PMCID: PMC11946407 DOI: 10.3390/v17030313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/07/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
(1) Background: We aimed to analyze the epidemiological characteristics of hemorrhagic fever with renal syndrome (HFRS) in Longyou County and to provide a basis for the future response to this disease. (2) Methods: Data on hemorrhagic fever and host animals were collected from 2011 to 2023. Descriptive methods were used to analyze the epidemic. The R4.4.1 software was used to show how the host density relates to the virus levels, temperature, and rainfall and to predict the host density. (3) Results: We observed 58 cases of hemorrhagic fever, the majority of which occurred in farmers. There were two incidence peaks each year during the spring and winter seasons, accounting for 22.41% and 43.10% of the total cases, respectively. The outdoor rodent population density was significantly and positively correlated with the outdoor rodent virus prevalence (R2 = 0.9411), serving as a robust predictor of the outdoor rodent virus prevalence. Additionally, the density of outdoor rodents exhibited a strong nonlinear relationship with the temperature and precipitation. (4) Conclusions: After hemorrhagic fever vaccination, rodent population density control, and rodent carrier rodent control from 1995 to 2000, the hemorrhagic fever epidemic was generally stable, and the epidemiological characteristics remained stable. In the future, we should continue to take active and effective comprehensive measures to intervene, further realize the effective control of HFRS, and prevent the recurrence of hemorrhagic fever epidemics.
Collapse
Affiliation(s)
- Jing Ni
- School of Public Health, Hangzhou Medical College, Hangzhou 310013, China;
- Department of Communicable Disease Control and Prevention, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, China
| | - Dejun Kong
- Longyou Centre for Disease Control and Prevention, Quzhou 324400, China; (D.K.); (Z.C.); (W.Z.)
| | - Zhongbing Chen
- Longyou Centre for Disease Control and Prevention, Quzhou 324400, China; (D.K.); (Z.C.); (W.Z.)
| | - Weiming Zeng
- Longyou Centre for Disease Control and Prevention, Quzhou 324400, China; (D.K.); (Z.C.); (W.Z.)
| | - Bingdong Zhan
- Quzhou Centre for Disease Control and Prevention, Quzhou 324000, China
| | - Zhenyu Gong
- Department of Communicable Disease Control and Prevention, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou 310051, China
- Zhejiang Key Lab of Vaccine, Infectious Disease Prevention and Control, Hangzhou 310051, China
| |
Collapse
|
12
|
Jang YS, Yoon SY, Krishnan R, Oh MJ. Temperature-dependent viral hemorrhagic septicemia virus (VHSV) infection kinetics and immune response in primary olive flounder spleen cell culture and the host. Virology 2025; 603:110390. [PMID: 39765021 DOI: 10.1016/j.virol.2025.110390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/24/2024] [Accepted: 01/02/2025] [Indexed: 02/04/2025]
Abstract
The replication and mortality caused by the viral hemorrhagic septicemia virus (VHSV) in fish vary depending on temperature. VHSV causes mortality at the temperatures below 15 °C, while infection is not established in olive flounder at temperatures above 25 °C. However, how VHSV infection manifests at the cellular level under different temperature conditions is not understood. In this study, we aimed to elucidate the mechanism by which VHSV infection is controlled by comparing viral replication and immune responses in vitro and in vivo. In the in vitro experiment, viral mRNA levels on day 5 post-challenge differed more than 10-fold between temperatures, highest at 15 °C, followed by 20 °C, and lowest at 25 °C, with replication gradually increasing over 5 days. In vivo, replication peaked by day 2 and then declined at all temperatures. VHSV infection showed a controlled tendency in the fish, whereas the virus exhibited a continuously increasing infection pattern for up to 5 days in olive flounder spleen cell cultures at a temperature of 25 °C. The level of viral infection within the cells (25 °C) was lower compared to conditions below 20 °C; however, the expression levels of interferon-related genes and pro-inflammatory cytokine genes were relatively low. The expression of ISG15 and Mx genes in spleen (25 °C) was significantly high at 24 h post challenge in the in vivo experiment.
Collapse
Affiliation(s)
- Yo-Seb Jang
- Department of Aqualife Medicine, Chonnam National University, Yeosu, Republic of Korea
| | - Su-Young Yoon
- Department of Aqualife Medicine, Chonnam National University, Yeosu, Republic of Korea
| | - Rahul Krishnan
- Department of Aquatic Animal Health Management, Faculty of Fisheries, Kerala University of Fisheries and Ocean Studies, Kerala, India
| | - Myung-Joo Oh
- Department of Aqualife Medicine, Chonnam National University, Yeosu, Republic of Korea.
| |
Collapse
|
13
|
Kalaichelvan A, Nadarajapillai K, Sellaththurai SR, Arachchi UPE, Kim MJ, Jung S, Lee J. CRISPR/Cas9-induced knockout of tumor necrosis factor-alpha-type I augments viral infection in zebrafish. FISH & SHELLFISH IMMUNOLOGY 2025; 157:110092. [PMID: 39716581 DOI: 10.1016/j.fsi.2024.110092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/02/2024] [Accepted: 12/20/2024] [Indexed: 12/25/2024]
Abstract
Tumor necrosis factor-alpha (TNF-α) is a pleiotropic cytokine with critical roles in inflammation, cell survival, and defense. As a member of the TNF superfamily, it exerts its effects by binding to transmembrane receptors and triggering various downstream signaling pathways. Although TNF-α's involvement in antiviral responses in mammals is well-established, its role in teleost remains poorly understood. This study investigated the contribution of TNF-α1 to antiviral immunity in zebrafish using a tnf-α1(-/-) knockout (KO) line. We challenged both wild-type and tnf-α1(-/-) zebrafish with viral hemorrhagic septicemia virus (VHSV) at both embryonic and adult stages. Mortality was observed at 4 days post-infection (dpi) in tnf-α1-deficient adult fish challenged with 5 × 106 TCID50 (VHSV) and at 5 dpi in adult wild fish challenged with the same concentration. In addition, tnf-α1(-/-) KO adult fish reached the maximum mortality of 100 % at 20 dpi, whereas wild adult fish reached 54 % mortality at the same time point. This increased susceptibility to early mortality was associated with a higher viral burden and altered expression of key immune genes, including the pro-inflammatory cytokines il-6 and il-1β, the anti-inflammatory cytokine il-10, and interferon-related genes such as irf1 and ifn-γ. Our findings demonstrate the crucial role of TNF-α1 in antiviral defense mechanisms in zebrafish and provide valuable insights into the functional conservation of TNF-α signaling across vertebrate species. This knowledge may contribute to the development of strategies to combat viral diseases in fish.
Collapse
Affiliation(s)
- Arthika Kalaichelvan
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - Kishanthini Nadarajapillai
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - Sarithaa Raguvaran Sellaththurai
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - U P E Arachchi
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea
| | - Myoung-Jin Kim
- Nakdonggang National Institute of Biological Resources, Sangju-si, Gyeongsangbuk-do, 37242, Republic of Korea
| | - Sumi Jung
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Life Research Institute, Gidang Marine Research Institute, Jeju National University, Jeju, 63333, Republic of Korea.
| | - Jehee Lee
- Department of Marine Life Sciences & Center for Genomic Selection in Korean Aquaculture, Jeju National University, Jeju, 63243, Republic of Korea; Marine Life Research Institute, Gidang Marine Research Institute, Jeju National University, Jeju, 63333, Republic of Korea.
| |
Collapse
|
14
|
Mielcarska MB, Rouse BT. Viruses and the Brain-A Relationship Prone to Trouble. Viruses 2025; 17:203. [PMID: 40006958 PMCID: PMC11860391 DOI: 10.3390/v17020203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Neurological disorders, some of which are associated with viral infections, are growing due to the aging and expanding population. Despite strong defenses of the central nervous system, some viruses have evolved ways to breach them, which often result in dire consequences. In this review, we recount the various ways by which different viruses can enter the CNS, and we describe the consequences of such invasions. Consequences may manifest as acute disease, such as encephalitis, meningitis, or result in long-term effects, such as neuromuscular dysfunction, as occurs in poliomyelitis. We discuss evidence for viral involvement in the causation of well-known chronic neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis, as well as vascular dementia in the elderly. We also describe the approaches currently available to control a few of the neural viral infections. These include antivirals that are effective against human immunodeficiency virus and herpes simplex virus, as well as vaccines valuable for controlling rabies virus, poliomyelitis virus, and some flavivirus infections. There is an urgent need to better understand, at a molecular level, how viruses contribute to acute and, especially, chronic neurological diseases and to develop more precise and effective vaccines and therapies.
Collapse
Affiliation(s)
- Matylda Barbara Mielcarska
- Department of Preclinical Sciences, Institute of Veterinary Sciences, Warsaw University of Life Sciences–SGGW, Jana Ciszewskiego 8, 02-786 Warsaw, Poland
| | - Barry T. Rouse
- College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
15
|
Suwanpakdee S, Wiratsudakul A, Chaisilp N, Prasittichai L, Skulpong A, Maneeorn P, Bhusri B, Mongkolpan C, Buddhirongawatr R, Taowan J, Wongluechai P, Arya N, Suwannaprapha P, Ngamwongsatit N, Wiriyarat W, Sangkachai N. Canine distemper outbreak and laryngeal paralysis in captive tigers (Panthera tigris). BMC Vet Res 2025; 21:33. [PMID: 39856666 PMCID: PMC11760695 DOI: 10.1186/s12917-025-04490-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
The canine distemper virus (CDV) could infect various wildlife species worldwide. The viral infection in large felids directly impacts wildlife conservation. This study aimed to understand better the burden of CDV outbreaks in captive tiger populations in Thailand and a novel discovery of their clinical signs with a history of CDV exposure. We followed up on their infection from May 2016 to October 2020 with laboratory testing and veterinary medical records. The cumulative morbidity and mortality rates were relatively high. Moreover, 50% of the tigers survived at 2 years after infection. All suspected and confirmed cases of CDV infections were significantly associated with laryngeal inflammation, which developed into paralysis in almost 50% of cases. Altogether, 50% of all tiger cases with chronic infection developed stridor at 314 days after virus infection [95% CI: 302-320]. Therefore, laryngeal paralysis may result from CDV infection and degeneration, potentially affecting the peripheral and central nervous systems. This condition could pose a life-threatening risk to tigers. The virus could spread quickly by contact with bodily excretion among tigers and fomite contamination once it affects a specific population. Implementation of biosecurity measures and vaccination is essential to mitigate the risk of disease spread and infection rates in tiger populations.
Collapse
Affiliation(s)
- Sarin Suwanpakdee
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Salaya, Nakhon Pathom, Thailand
- The Monitoring and Surveillance Center for Zoonotic Diseases in Wildlife and Exotic Animals, Faculty of Veterinary Science, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | - Anuwat Wiratsudakul
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Salaya, Nakhon Pathom, Thailand
- The Monitoring and Surveillance Center for Zoonotic Diseases in Wildlife and Exotic Animals, Faculty of Veterinary Science, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | - Nattarun Chaisilp
- The Monitoring and Surveillance Center for Zoonotic Diseases in Wildlife and Exotic Animals, Faculty of Veterinary Science, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | - Luxsana Prasittichai
- Department of National Parks, Wildlife and Plant Conservation, Ministry of Natural Resources and Environment, Chatuchak, Bangkok, Thailand
| | - Anurux Skulpong
- Department of National Parks, Wildlife and Plant Conservation, Ministry of Natural Resources and Environment, Chatuchak, Bangkok, Thailand
| | - Patarapol Maneeorn
- Department of National Parks, Wildlife and Plant Conservation, Ministry of Natural Resources and Environment, Chatuchak, Bangkok, Thailand
| | - Benjaporn Bhusri
- The Monitoring and Surveillance Center for Zoonotic Diseases in Wildlife and Exotic Animals, Faculty of Veterinary Science, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | - Chalisa Mongkolpan
- The Monitoring and Surveillance Center for Zoonotic Diseases in Wildlife and Exotic Animals, Faculty of Veterinary Science, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | - Ruangrat Buddhirongawatr
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Salaya, Nakhon Pathom, Thailand
- The Monitoring and Surveillance Center for Zoonotic Diseases in Wildlife and Exotic Animals, Faculty of Veterinary Science, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | - Jarupa Taowan
- The Monitoring and Surveillance Center for Zoonotic Diseases in Wildlife and Exotic Animals, Faculty of Veterinary Science, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | - Peerawat Wongluechai
- The Monitoring and Surveillance Center for Zoonotic Diseases in Wildlife and Exotic Animals, Faculty of Veterinary Science, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | - Nlin Arya
- Department of Pre-clinic and Applied Animal Science, Faculty of Veterinary Science, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | - Parin Suwannaprapha
- Department of Pre-clinic and Applied Animal Science, Faculty of Veterinary Science, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | - Natharin Ngamwongsatit
- Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Salaya, Nakhon Pathom, Thailand
- Laboratory of Bacteria, Veterinary Diagnostic Center, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Witthawat Wiriyarat
- The Monitoring and Surveillance Center for Zoonotic Diseases in Wildlife and Exotic Animals, Faculty of Veterinary Science, Mahidol University, Salaya, Nakhon Pathom, Thailand
- Department of Pre-clinic and Applied Animal Science, Faculty of Veterinary Science, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | - Nareerat Sangkachai
- The Monitoring and Surveillance Center for Zoonotic Diseases in Wildlife and Exotic Animals, Faculty of Veterinary Science, Mahidol University, Salaya, Nakhon Pathom, Thailand.
| |
Collapse
|
16
|
Major-Styles CT, Munns J, Zeng A, Vanden Oever M, O'Neill JS, Edgar RS. Chronic CRYPTOCHROME deficiency enhances cell-intrinsic antiviral defences. Philos Trans R Soc Lond B Biol Sci 2025; 380:20230344. [PMID: 39842480 PMCID: PMC11753882 DOI: 10.1098/rstb.2023.0344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/19/2024] [Accepted: 11/27/2024] [Indexed: 01/24/2025] Open
Abstract
The within-host environment changes over circadian time and influences the replication and severity of viruses. Genetic knockout of the circadian transcription factors CRYPTOCHROME 1 and CRYPTOCHROME 2 (CRY1-/-/CRY2-/-; CKO) leads to altered protein homeostasis and chronic activation of the integrated stress response (ISR). The adaptive ISR signalling pathways help restore cellular homeostasis by downregulating protein synthesis in response to endoplasmic reticulum overloading or viral infections. By quantitative mass spectrometry analysis, we reveal that many viral recognition proteins and type I interferon (IFN) effectors are significantly upregulated in lung fibroblast cells from CKO mice compared with wild-type (WT) mice. This basal 'antiviral state' restricts the growth of influenza A virus and is governed by the interaction between proteotoxic stress response pathways and constitutive type I IFN signalling. CKO proteome composition and type I IFN signature were partially phenocopied upon sustained depletion of CRYPTOCHROME (CRY) proteins using a small-molecule CRY degrader, with modest differential gene expression consistent with differences seen between CKO and WT cells. Our results highlight the crosstalk between circadian rhythms, cell-intrinsic antiviral defences and protein homeostasis, providing a tractable molecular model to investigate the interface of these key contributors to human health and disease.This article is part of the Theo Murphy meeting issue 'Circadian rhythms in infection and immunity'.
Collapse
Affiliation(s)
- Christine T. Major-Styles
- Department of Infectious Disease, Imperial College London, LondonSW7 2AZ, UK
- Francis Crick Institute, LondonNW1 1AT, UK
| | - Jack Munns
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, CambridgeCB2 0QH, UK
| | - Aiwei Zeng
- Department of Infectious Disease, Imperial College London, LondonSW7 2AZ, UK
- Francis Crick Institute, LondonNW1 1AT, UK
| | | | - John S. O'Neill
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, CambridgeCB2 0QH, UK
| | - Rachel S. Edgar
- Department of Infectious Disease, Imperial College London, LondonSW7 2AZ, UK
- Francis Crick Institute, LondonNW1 1AT, UK
| |
Collapse
|
17
|
Lydon E, Osborne CM, Wagner BD, Ambroggio L, Harris JK, Reeder R, Carpenter TC, Maddux AB, Leroue MK, Yehya N, DeRisi JL, Hall MW, Zuppa AF, Carcillo J, Meert K, Sapru A, Pollack MM, McQuillen P, Notterman DA, Langelier CR, Mourani PM, for the Eunice Kennedy Shriver National Institute of Child Health and Human Development Collaborative Pediatric Critical Care Research Network (CPCCRN). Proteomic profiling of the local and systemic immune response to pediatric respiratory viral infections. mSystems 2025; 10:e0133524. [PMID: 39611811 PMCID: PMC11748518 DOI: 10.1128/msystems.01335-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/11/2024] [Indexed: 11/30/2024] Open
Abstract
Viral lower respiratory tract infection (vLRTI) is a leading cause of hospitalization and death in children worldwide. Despite this, no studies have employed proteomics to characterize host immune responses to severe pediatric vLRTI in both the lower airway and systemic circulation. To address this gap, gain insights into vLRTI pathophysiology, and test a novel diagnostic approach, we assayed 1,305 proteins in tracheal aspirate (TA) and plasma from 62 critically ill children using SomaScan. We performed differential expression (DE) and pathway analyses comparing vLRTI (n = 40) to controls with non-infectious acute respiratory failure (n = 22), developed a diagnostic classifier using LASSO regression, and analyzed matched TA and plasma samples. We further investigated the impact of viral load and bacterial coinfection on the proteome. The TA signature of vLRTI was characterized by 200 DE proteins (Padj <0.05) with upregulation of interferons and T cell responses and downregulation of inflammation-modulating proteins including FABP and MIP-5. A nine-protein TA classifier achieved an area under the receiver operator curve (AUC) of 0.96 (95% CI: 0.90-1.00) for identifying vLRTI. In plasma, the host response to vLRTI was more muted with 56 DE proteins. Correlation between TA and plasma was limited, although ISG15 was elevated in both compartments. In bacterial coinfection, we observed increases in the TNF-stimulated protein TSG-6, as well as CRP, and interferon-related proteins. Viral load correlated positively with interferon signaling and negatively with neutrophil-activation pathways. Taken together, our study provides fresh insights into the lower airway and systemic proteome of severe pediatric vLRTI and identifies novel protein biomarkers with diagnostic potential.IMPORTANCEWe describe the first proteomic profiling of the lower airway and blood in critically ill children with severe viral lower respiratory tract infection (vLRTI). From tracheal aspirate (TA), we defined a proteomic signature of vLRTI characterized by increased expression of interferon signaling proteins and decreased expression of proteins involved in immune modulation including FABP and MIP-5. Using machine learning, we developed a parsimonious diagnostic classifier that distinguished vLRTI from non-infectious respiratory failure with high accuracy. Comparative analysis of paired TA and plasma specimens demonstrated limited concordance, although the interferon-stimulated protein ISG15 was significantly upregulated with vLRTI in both compartments. We further identified TSG-6 and CRP as airway biomarkers of bacterial-viral coinfection, and viral load analyses demonstrated a positive correlation with interferon-related protein expression and a negative correlation with the expression of neutrophil activation proteins. Taken together, our study provides new insights into the lower airway and systemic proteome of severe pediatric vLRTI.
Collapse
Affiliation(s)
- Emily Lydon
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Christina M. Osborne
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Brandie D. Wagner
- Department of Biostatistics and Informatics, University of Colorado, Colorado School of Public Health, Aurora, Colorado, USA
| | - Lilliam Ambroggio
- Sections of Emergency Medicine and Hospital Medicine, Children’s Hospital Colorado, Aurora, Colorado, USA
- Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, Colorado, USA
| | - J. Kirk Harris
- Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, Colorado, USA
| | - Ron Reeder
- Department of Pediatrics, University of Utah, Salt Lake City, Utah, USA
| | - Todd C. Carpenter
- Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, Colorado, USA
| | - Aline B. Maddux
- Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, Colorado, USA
| | - Matthew K. Leroue
- Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, Colorado, USA
| | - Nadir Yehya
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Joseph L. DeRisi
- Chan Zuckerberg Biohub, San Francisco, California, USA
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California, USA
| | - Mark W. Hall
- Department of Pediatrics, Nationwide Children’s Hospital, Columbus, Ohio, USA
| | - Athena F. Zuppa
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Joseph Carcillo
- Departments of Pediatrics and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kathleen Meert
- Department of Pediatrics, Children’s Hospital of Michigan, Central Michigan University, Detroit, Michigan, USA
| | - Anil Sapru
- Department of Pediatrics, University of California Los Angeles, Los Angeles, California, USA
| | - Murray M. Pollack
- Department of Pediatrics,, Children’s National Medical Center and George Washington School of Medicine and Health Sciences, Washington, DC, USA
| | - Patrick McQuillen
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Daniel A. Notterman
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| | - Charles R. Langelier
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Chan Zuckerberg Biohub, San Francisco, California, USA
| | - Peter M. Mourani
- Department of Pediatrics, Critical Care, University of Arkansas for Medical Sciences and Arkansas Children’s Hospital, Little Rock, Arkansas, USA
| | - for the Eunice Kennedy Shriver National Institute of Child Health and Human Development Collaborative Pediatric Critical Care Research Network (CPCCRN)
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Biostatistics and Informatics, University of Colorado, Colorado School of Public Health, Aurora, Colorado, USA
- Sections of Emergency Medicine and Hospital Medicine, Children’s Hospital Colorado, Aurora, Colorado, USA
- Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, Colorado, USA
- Department of Pediatrics, University of Utah, Salt Lake City, Utah, USA
- Chan Zuckerberg Biohub, San Francisco, California, USA
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California, USA
- Department of Pediatrics, Nationwide Children’s Hospital, Columbus, Ohio, USA
- Departments of Pediatrics and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pediatrics, Children’s Hospital of Michigan, Central Michigan University, Detroit, Michigan, USA
- Department of Pediatrics, University of California Los Angeles, Los Angeles, California, USA
- Department of Pediatrics,, Children’s National Medical Center and George Washington School of Medicine and Health Sciences, Washington, DC, USA
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
- Department of Pediatrics, Critical Care, University of Arkansas for Medical Sciences and Arkansas Children’s Hospital, Little Rock, Arkansas, USA
| |
Collapse
|
18
|
Pradhan SS, Balena V, Bera BC, Anand T, Khetmalis R, Madhwal A, Kandasamy S, Pavulraj S, Bernela M, Mor P, Tripathi BN, Virmani N. Multiple Gene Deletion Mutants of Equine Herpesvirus 1 Exhibit Strong Protective Efficacy Against Wild Virus Challenge in a Murine Model. Vaccines (Basel) 2025; 13:45. [PMID: 39852824 PMCID: PMC11768829 DOI: 10.3390/vaccines13010045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Equine herpesvirus type 1 (EHV1) is a ubiquitous viral pathogen infecting the equine population worldwide. EHV1 infection causes respiratory illness, abortion, neonatal foal mortality, and myeloencephalopathy. The currently available modified live EHV1 vaccines have safety and efficacy limitations. The two mutant EHV1 viruses (vToH-DMV (∆IR6/gE) and vToH-QMV (∆IR6/UL43/gE/UL56)), generated by the deletion of genes responsible for virulence (gE and IR6) and immunosuppression (uL43 and uL56), have been previously characterized by our group and found to generate good immune responses. The present study aimed to determine the safety and protective efficacy of the above mutants against a virulent EHV1 challenge in a murine model. METHODS BALB/c mice were intranasally immunized with a live vToH-QMV or vToH-DMV vaccine. Intranasal booster immunization was given at 14 days post-vaccination (dpv). Both mutants induced an optimal level of EHV1-specific humoral and cell-mediated immune responses, as determined by virus neutralization assay, ELISA, and immunophenotyping. At 35 dpv, the mice were intranasally challenged with wild-type EHV1 (vRaj strain). RESULTS Amongst the two mutants, vToH-QMV induced a better immune response than the vToH-DMV vaccine. Furthermore, vToH-QMV provided good protection in mice against the virulent challenge. It specifically exhibited less severe clinical disease in terms of clinical signs, body weight reduction, and gross and histopathological lung lesions accompanied by early virus clearance. CONCLUSIONS These studies are suggestive of vToH-QMV EHV1 being a potential vaccine candidate against EHV1 infection, which needs to be finally tested in the main host, i.e., horses.
Collapse
Affiliation(s)
- Stephanie S. Pradhan
- ICAR-National Research Centre on Equines, Sirsa Road, Hisar 125001, Haryana, India; (S.S.P.); (A.M.); (S.K.); (P.M.)
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly 243122, Uttar Pradesh, India
| | - Vekataramireddy Balena
- ICAR-National Research Centre on Equines, Sirsa Road, Hisar 125001, Haryana, India; (S.S.P.); (A.M.); (S.K.); (P.M.)
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly 243122, Uttar Pradesh, India
| | - Bidhan Chandra Bera
- ICAR-National Research Centre on Equines, Sirsa Road, Hisar 125001, Haryana, India; (S.S.P.); (A.M.); (S.K.); (P.M.)
| | - Taruna Anand
- ICAR-National Research Centre on Equines, Sirsa Road, Hisar 125001, Haryana, India; (S.S.P.); (A.M.); (S.K.); (P.M.)
| | - Rhushikesh Khetmalis
- ICAR-National Research Centre on Equines, Sirsa Road, Hisar 125001, Haryana, India; (S.S.P.); (A.M.); (S.K.); (P.M.)
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly 243122, Uttar Pradesh, India
| | - Aashwina Madhwal
- ICAR-National Research Centre on Equines, Sirsa Road, Hisar 125001, Haryana, India; (S.S.P.); (A.M.); (S.K.); (P.M.)
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly 243122, Uttar Pradesh, India
| | - Supriya Kandasamy
- ICAR-National Research Centre on Equines, Sirsa Road, Hisar 125001, Haryana, India; (S.S.P.); (A.M.); (S.K.); (P.M.)
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly 243122, Uttar Pradesh, India
| | - Selvaraj Pavulraj
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Manju Bernela
- ICAR-National Research Centre on Equines, Sirsa Road, Hisar 125001, Haryana, India; (S.S.P.); (A.M.); (S.K.); (P.M.)
| | - Priya Mor
- ICAR-National Research Centre on Equines, Sirsa Road, Hisar 125001, Haryana, India; (S.S.P.); (A.M.); (S.K.); (P.M.)
| | | | - Nitin Virmani
- ICAR-National Research Centre on Equines, Sirsa Road, Hisar 125001, Haryana, India; (S.S.P.); (A.M.); (S.K.); (P.M.)
| |
Collapse
|
19
|
Nardo D, Maddox EG, Riley JL. Cell therapies for viral diseases: a new frontier. Semin Immunopathol 2025; 47:5. [PMID: 39747475 PMCID: PMC11695571 DOI: 10.1007/s00281-024-01031-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025]
Abstract
Despite advances in medicine and antimicrobial research, viral infections continue to pose a major threat to human health. While major strides have been made in generating vaccines and small molecules to combat emerging pathogens, new modalities of treatment are warranted in diseases where there is a lack of treatment options, or where treatment cannot fully eradicate pathogens, as in HIV infection. Cellular therapies, some of which are FDA approved for treating cancer, take advantage of our developing understanding of the immune system, and harness this knowledge to enhance, or direct, immune responses toward infectious agents. As with cancer, viruses that evade immunity, do so by avoiding immune recognition or by redirecting the cellular responses that would eradicate them. As such, infusing virus specific immune cells has the potential to improve patient outcomes and should be investigated as a potential tool in the arsenal to fight infection. The present manuscript summarizes key findings made using cellular therapies for the treatment of viral infections, focusing on the potential that these strategies might have in controlling disease.
Collapse
Affiliation(s)
- David Nardo
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Emileigh G Maddox
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - James L Riley
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
20
|
La Frazia S, Pauciullo S, Zulian V, Garbuglia AR. Viral Oncogenesis: Synergistic Role of Genome Integration and Persistence. Viruses 2024; 16:1965. [PMID: 39772271 PMCID: PMC11728759 DOI: 10.3390/v16121965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
Persistence is a strategy used by many viruses to evade eradication by the immune system, ensuring their permanence and transmission within the host and optimizing viral fitness. During persistence, viruses can trigger various phenomena, including target organ damage, mainly due to an inflammatory state induced by infection, as well as cell proliferation and/or immortalization. In addition to immune evasion and chronic inflammation, factors contributing to viral persistence include low-level viral replication, the accumulation of viral mutants, and, most importantly, maintenance of the viral genome and reliance on viral oncoprotein production. This review focuses on the process of genome integration, which may occur at different stages of infection (e.g., HBV), during the chronic phase of infection (e.g., HPV, EBV), or as an essential part of the viral life cycle, as seen in retroviruses (HIV, HTLV-1). It also explores the close relationship between integration, persistence, and oncogenesis. Several models have been proposed to describe the genome integration process, including non-homologous recombination, looping, and microhomology models. Integration can occur either randomly or at specific genomic sites, often leading to genome destabilization. In some cases, integration results in the loss of genomic regions or impairs the regulation of oncogene and/or oncosuppressor expression, contributing to tumor development.
Collapse
Affiliation(s)
- Simone La Frazia
- Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Silvia Pauciullo
- Laboratory of Virology, National Institute for Infectious Diseases “Lazzaro Spallanzani” (IRCCS), 00149 Rome, Italy; (S.P.); (V.Z.); (A.R.G.)
| | - Verdiana Zulian
- Laboratory of Virology, National Institute for Infectious Diseases “Lazzaro Spallanzani” (IRCCS), 00149 Rome, Italy; (S.P.); (V.Z.); (A.R.G.)
| | - Anna Rosa Garbuglia
- Laboratory of Virology, National Institute for Infectious Diseases “Lazzaro Spallanzani” (IRCCS), 00149 Rome, Italy; (S.P.); (V.Z.); (A.R.G.)
| |
Collapse
|
21
|
Pedicino D, Volpe M. Weekly Journal Scan: increased incidence of acute myocardial infarction in patients with laboratory-confirmed influenza infection. Eur Heart J 2024; 45:5102-5103. [PMID: 39319692 DOI: 10.1093/eurheartj/ehae569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/26/2024] Open
Affiliation(s)
- Daniela Pedicino
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Largo A. Gemelli 8, Rome 00168, Italy
| | - Massimo Volpe
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Via di Grottarossa 1035, Rome, Italy
- IRCCS San Raffaele Roma, Via di Valcannuta 250, Rome, Italy
| |
Collapse
|
22
|
Tang A, Zhu M, Zhu J, Zhang D, Zhu S, Meng C, Li C, Liu G. The recombinant feline herpesvirus 1 expressing feline Calicivirus VP1 protein is safe and effective in cats. Vaccine 2024; 42:126468. [PMID: 39467408 DOI: 10.1016/j.vaccine.2024.126468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 07/07/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024]
Abstract
Feline herpesvirus type 1 (FHV) and feline calicivirus (FCV) are significant pathogens causing upper respiratory tract disease in cats. Existing inactivated or modified live vaccines against FCV and FHV face limitations in safety and efficacy. To overcome these challenges, a recombinant strain FHV ΔgI/gE-FCV VP1 was developed by deleting the gI/gE gene and concurrently expressing FCV VP1, using the FHV WX19 strain as the parental virus. Results indicated the presence of FCV VP1 in FHV ΔgI/gE-FCV VP1-infected CRFK cells, confirmed through protein blotting and immunofluorescence assays and virus-like particles (VLPs) of FCV were observed using transmission electron microscopy. For efficacy in cats, each animal received intranasal vaccination with 1 mL of FHV ΔgI/gE-FCV VP1 at 106 TCID50. Following completion of vaccination on day 28, animals were exposed to a potent FCV strain. Assessments included clinical signs, nasal shedding, virus neutralizing antibodies, cytokine expression and postmortem histological testing. All vaccinations with FHV ΔgI/gE-FCV VP1 were deemed safe, with significantly reduced clinical disease scores, pathological changes and viral nasal shedding following infection and robust immune responses were induced. These findings collectively suggest the effectiveness of FHV-based recombinant vaccines in preventing FCV infections.
Collapse
MESH Headings
- Animals
- Cats
- Caliciviridae Infections/prevention & control
- Caliciviridae Infections/veterinary
- Calicivirus, Feline/immunology
- Calicivirus, Feline/genetics
- Cat Diseases/prevention & control
- Cat Diseases/virology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Viral Vaccines/immunology
- Viral Vaccines/genetics
- Viral Vaccines/administration & dosage
- Virus Shedding
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Administration, Intranasal
- Varicellovirus/immunology
- Varicellovirus/genetics
- Capsid Proteins/immunology
- Capsid Proteins/genetics
- Cytokines/metabolism
- Female
- Herpesviridae Infections/prevention & control
- Herpesviridae Infections/veterinary
- Herpesviridae Infections/immunology
Collapse
Affiliation(s)
- Aoxing Tang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Meng Zhu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Jie Zhu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Da Zhang
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Shiqiang Zhu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Chunchun Meng
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Chuanfeng Li
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Guangqing Liu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China.
| |
Collapse
|
23
|
Hsieh SJ, Tsai TH, Lin JH, Lin TY, Chang FL, Chiang CW, Li PJ, Zheng JH, Tsai KC, Hung CS, Lee YC. Characterization of anti-EBNA-1 antibodies and exploration of their molecular mimicry potential in an EBV-infected Sjögren's syndrome patient. Biochem Biophys Res Commun 2024; 735:150839. [PMID: 39427375 DOI: 10.1016/j.bbrc.2024.150839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
There is a potential link between autoimmune diseases and Epstein-Barr virus (EBV) infections, with EBV playing a substantial role in the onset of Sjögren's syndrome (SjS). Some EBV proteins could mimic host self-antigens post-infection, leading to molecular mimicry. This similarity may cause the immune system to attack its tissues mistakenly. Among the various proteins associated with EBV, nuclear antigen 1 (EBNA-1) is essential for the latent replication of infected cells and is prevalent in all EBV-related diseases. In the study, single-chain variable fragment (scFv) antibodies targeting EBNA-1 were isolated using phage display technology from a primary SjS patient who also had a chronic active EBV infection. The specific clones were enriched after panning, and the binding activity of selected scFvs targeting EBNA-1 was confirmed. Sequence analysis indicated that the scFvs exhibiting positive signals could be grouped into five clones, all of which used homologous heavy chain V regions derived from germline Vh4-39, and two types of light chain V regions stemming from germline Vλ1-44 and Vλ3-15. These scFvs were found to exhibit a high degree of somatic mutations, likely indicative of antigen selection. Of the scFvs, P1-3 demonstrated the strongest binding affinity to EBNA-1, exhibiting a determined value of 7.3 x 10-8 M, and showed cross-reactivity to the SjS associated La/SSB self-antigen. The experimental results combined with AlphaFold 3 predictions revealed a potential epitope for scFv P1-3 binding to EBNA-1. Additionally, scFv P1-3 could also cross-bind to the modeled structure of La/SSB. We inferred a possible structural correlation between EBNA-1 and La/SSB involving an X2AX6PG epitope motif. This research contributes to our understanding of the structural basis of the interactions between antibodies and EBNA-1, shedding light on the VH and VL gene usage of anti-EBNA-1 antibodies in EBV-infected SjS patients and the potential origins of autoantibodies.
Collapse
Affiliation(s)
- Shang-Ju Hsieh
- Division of Urology, Department of Surgery, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Tsung-Hsun Tsai
- Department of Psychiatry, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Jiun-Han Lin
- Department of Industrial Technology, Ministry of Economic Affairs, Taipei, Taiwan; Food Industry Research and Development Institute, Hsinchu City, Taiwan
| | - Tsai-Yu Lin
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Fu-Ling Chang
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chen-Wei Chiang
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Pei Jhen Li
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jia Huei Zheng
- Taiwan Autoantibody Biobank Initiative, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Keng-Chang Tsai
- National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan; Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Ching-Sheng Hung
- Department of Laboratory Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| | - Yu-Ching Lee
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, Taiwan; Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
24
|
Beltrán JF, Belén LH, Yáñez AJ, Jimenez L. Predicting viral proteins that evade the innate immune system: a machine learning-based immunoinformatics tool. BMC Bioinformatics 2024; 25:351. [PMID: 39522017 PMCID: PMC11550529 DOI: 10.1186/s12859-024-05972-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Viral proteins that evade the host's innate immune response play a crucial role in pathogenesis, significantly impacting viral infections and potential therapeutic strategies. Identifying these proteins through traditional methods is challenging and time-consuming due to the complexity of virus-host interactions. Leveraging advancements in computational biology, we present VirusHound-II, a novel tool that utilizes machine learning techniques to predict viral proteins evading the innate immune response with high accuracy. We evaluated a comprehensive range of machine learning models, including ensemble methods, neural networks, and support vector machines. Using a dataset of 1337 viral proteins known to evade the innate immune response (VPEINRs) and an equal number of non-VPEINRs, we employed pseudo amino acid composition as the molecular descriptor. Our methodology involved a tenfold cross-validation strategy on 80% of the data for training, followed by testing on an independent dataset comprising the remaining 20%. The random forest model demonstrated superior performance metrics, achieving 0.9290 accuracy, 0.9283 F1 score, 0.9354 precision, and 0.9213 sensitivity in the independent testing phase. These results establish VirusHound-II as an advancement in computational virology, accessible via a user-friendly web application. We anticipate that VirusHound-II will be a crucial resource for researchers, enabling the rapid and reliable prediction of viral proteins evading the innate immune response. This tool has the potential to accelerate the identification of therapeutic targets and enhance our understanding of viral evasion mechanisms, contributing to the development of more effective antiviral strategies and advancing our knowledge of virus-host interactions.
Collapse
Affiliation(s)
- Jorge F Beltrán
- Department of Chemical Engineering, Faculty of Engineering and Science, Universidad de La Frontera, Ave. Francisco Salazar 01145, Temuco, Chile.
| | - Lisandra Herrera Belén
- Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad Santo Tomas, Temuco, Chile
| | - Alejandro J Yáñez
- Departamento de Investigación y Desarrollo, Greenvolution SpA., Puerto Varas, Chile
- Interdisciplinary Center for Aquaculture Research (INCAR), Concepcion, Chile
| | - Luis Jimenez
- Department of Chemical Engineering, Faculty of Engineering and Science, Universidad de La Frontera, Ave. Francisco Salazar 01145, Temuco, Chile
| |
Collapse
|
25
|
James SA, Joshua IA. Charting Peptide Shared Sequences Between 'Diabetes-Viruses' and Human Pancreatic Proteins, Their Structural and Autoimmune Implications. Bioinform Biol Insights 2024; 18:11779322241289936. [PMID: 39502449 PMCID: PMC11536397 DOI: 10.1177/11779322241289936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 08/21/2024] [Indexed: 11/08/2024] Open
Abstract
Diabetes mellitus (DM) is a metabolic syndrome characterized by hyperglycaemia, polydipsia, polyuria, and weight loss, among others. The pathophysiology for the disorders is complex and results in pancreatic abnormal function. Viruses have also been implicated in the metabolic syndrome. This study charted peptides to investigate and predict the autoimmune potential of shared sequences between 8 viral species proteins (which we termed 'diabetes-viruses') and the human pancreatic proteins. The structure and immunological relevance of shared sequences between viruses reported in DM onset and human pancreatic proteins were analysed. At nonapeptide mapping between human pancreatic protein and 'diabetic-viruses', reveal 1064 shared sequences distributed among 454 humans and 4288 viral protein sequences. The viral results showed herpesviruses, enterovirus (EV), human endogenous retrovirus, influenza A viruses, rotavirus, and rubivirus sequences are hosted by the human pancreatic protein. The most common shared nonapeptide was AAAAAAAAA, present in 30 human nonredundant sequences. Among the viral species, the shared sequence NSLEVLFQG occurred in 18 nonredundant EVs protein, while occurring merely in 1 human protein, whereas LGLDIEIAT occurred in 8 influenza A viruses overlapping to 1 human protein and KDELSEARE occurred in 2 rotaviruses. The prediction of the location of the shared sequences in the protein structures, showed most of the shared sequences are exposed and located either on the surface or cleft relative to the entire protein structure. Besides, the peptides in the viral protein shareome were predicted computationally for binding to MHC molecules. Here analyses showed that the entire 1064 shared sequences predicted 203 to be either HLA-A or B supertype-restricted epitopes. Fifty-one of the putative epitopes matched reported HLA ligands/T-cell epitopes majorly coming from EV B supertype representative allele restrictions. These data, shared sequences, and epitope charts provide important insight into the role of viruses on the onset of DM and its implications.
Collapse
Affiliation(s)
- Stephen A James
- Department of Biochemistry, Kaduna State University, Kaduna, Nigeria
- School of Data Sciences, Centre of Bioinformatics, Perdana University, Kuala Lumpur, Malaysia
| | - Istifanus A Joshua
- Department of Community Medicine, College of Medicine, Kaduna State University, Kaduna, Nigeria
- Department of Community Medicine, College of Health Sciences, Federal University Wukari, Wukari, Nigeria
| |
Collapse
|
26
|
Ayuti SR, Khairullah AR, Lamid M, Al-Arif MA, Warsito SH, Silaen OSM, Moses IB, Hermawan IP, Yanestria SM, Delima M, Ferasyi TR, Aryaloka S. Avian influenza in birds: Insights from a comprehensive review. Vet World 2024; 17:2544-2555. [PMID: 39829652 PMCID: PMC11736375 DOI: 10.14202/vetworld.2024.2544-2555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 10/09/2024] [Indexed: 01/22/2025] Open
Abstract
One of the worst zoonotic illnesses, avian influenza (AI), or commonly referred to as bird flu, is caused by viruses belonging to the genus Influenza viruses, which are members of the Orthomyxoviridae family. The harmful effects of AI illness can affect both human and animal health and cause financial losses. Globally, the AI virus lacks political purpose and is not limited by geographical limits. It has been isolated from poultry, wild birds, and captive birds in Asia, North America, Europe, Australia, and South America. Their virulence is divided into highly pathogenic AI (HPAI) and low pathogenic AI (LPAI). The AI virus can also be diagnosed in a laboratory setting using molecular tests like real-time polymerase chain reaction or serological tests like the hemagglutinin inhibition test, agar gel immunodiffusion, antigen detection enzyme-linked immunosorbent assay, and other immunoassays. The type of AI virus and host species determines the clinical manifestations, severity, and fatality rates of AI. Human infection with AI viruses typically results from direct transmission from infected birds to humans. AI outbreaks in domestic and wild birds are uncommon; however, an infection can pose a significant threat to public, veterinary, and medical health. Successful vaccination reduces the probability of AI H5N1 virus infection in meat and other poultry products and prevents systemic infection in chickens. This review will provide information that can be used as a reference for recognizing the dangers of AI and for preventing and controlling the disease, considering its potential to become a serious pandemic outbreak.
Collapse
Affiliation(s)
- Siti Rani Ayuti
- Doctoral Program of Veterinary Science, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, East Java, Indonesia
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Universitas Syiah Kuala, Banda Aceh, Aceh, Indonesia
| | - Aswin Rafif Khairullah
- Research Center for Veterinary Science, National Research and Innovation Agency, Bogor, West Java, Indonesia
| | - Mirni Lamid
- Division of Animal Husbandry, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Mohammad Anam Al-Arif
- Division of Animal Husbandry, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Sunaryo Hadi Warsito
- Division of Animal Husbandry, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, East Java, Indonesia
| | - Otto Sahat Martua Silaen
- Doctoral Program in Biomedical Science, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Ikechukwu Benjamin Moses
- Department of Applied Microbiology, Faculty of Science, Ebonyi State University, Abakaliki, Nigeria
| | - Intan Permatasari Hermawan
- Laboratory of Internal Medicine, Faculty of Veterinary Medicine, Universitas Wijaya Kusuma Surabaya, Surabaya, East Java, Indonesia
| | - Sheila Marty Yanestria
- Laboratory of Veterinary Public Health, Faculty of Veterinary Medicine, Universitas Wijaya Kusuma Surabaya, Surabaya, East Java, Indonesia
| | - Mira Delima
- Department of Animal Husbandry, Faculty of Agriculture, Universitas Syiah Kuala, Banda Aceh, Indonesia
| | - Teuku Reza Ferasyi
- Laboratory of Veterinary Public Health, Faculty of Veterinary Medicine, Universitas Syiah Kuala, Banda Aceh, Indonesia
- Center for Tropical Veterinary Studies, One Health Collaboration Center, Universitas Syiah Kuala, Banda Aceh, Aceh, Indonesia
| | - Suhita Aryaloka
- Master Program of Veterinary Agribusiness, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, East Java, Indonesia
| |
Collapse
|
27
|
Liu W, Wilke CO, Arnold JJ, Sotoudegan MS, Cameron CE. Single-Cell Virology: On-Chip, Quantitative Characterization of the Dynamics of Virus Spread from One Single Cell to Another. Viruses 2024; 16:1659. [PMID: 39599774 PMCID: PMC11598947 DOI: 10.3390/v16111659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024] Open
Abstract
Virus spread at the single-cell level is largely uncharacterized. We have designed and constructed a microfluidic device in which each nanowell contains a single, infected cell (donor) and a single, uninfected cell (recipient). Using a GFP-expressing poliovirus as our model, we observed both lytic and non-lytic spread. Donor cells supporting lytic spread established infection earlier than those supporting non-lytic spread. However, non-lytic spread established infections in recipient cells substantially faster than lytic spread and yielded higher rates of genome replication. While lytic spread was sensitive to the presence of capsid entry/uncoating inhibitors, non-lytic spread was not. Consistent with emerging models for non-lytic spread of enteroviruses using autophagy, reduction in LC3 levels in cells impaired non-lytic spread and elevated the fraction of virus in donor cells spreading lytically. The ability to distinguish lytic and non-lytic spread unambiguously will enable discovery of viral and host factors and host pathways used for non-lytic spread of enteroviruses and other viruses as well.
Collapse
Affiliation(s)
- Wu Liu
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
- School of Pharmaceutical Sciences, Shandong University, Jinan 250100, China
| | - Claus O. Wilke
- Center for Computational Biology and Bioinformatics, Institute for Cellular and Molecular Biology, and Department of Integrative Biology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jamie J. Arnold
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mohamad S. Sotoudegan
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Craig E. Cameron
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
28
|
Anfossi S, Darbaniyan F, Quinlan J, Calin S, Shimizu M, Chen M, Rausseo P, Winters M, Bogatenkova E, Do KA, Martinez I, Li Z, Antal L, Olariu TR, Wistuba I, Calin GA. MicroRNAs are enriched at COVID-19 genomic risk regions, and their blood levels correlate with the COVID-19 prognosis of cancer patients infected by SARS-CoV-2. Mol Cancer 2024; 23:235. [PMID: 39434078 PMCID: PMC11492698 DOI: 10.1186/s12943-024-02094-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 08/18/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Cancer patients are more susceptible to an aggressive course of COVID-19. Developing biomarkers identifying cancer patients at high risk of COVID-19-related death could help determine who needs early clinical intervention. The miRNAs hosted in the genomic regions associated with the risk of aggressive COVID-19 could represent potential biomarkers for clinical outcomes. PATIENTS AND METHODS Plasma samples were collected at The University of Texas MD Anderson Cancer Center from cancer patients (N = 128) affected by COVID-19. Serum samples were collected from vaccinated healthy individuals (n = 23) at the Municipal Clinical Emergency Teaching Hospital in Timisoara, Romania. An in silico positional cloning approach was used to identify the presence of miRNAs at COVID-19 risk-associated genomic regions: CORSAIRs (COvid-19 RiSk AssocIated genomic Regions). The miRNA levels were measured by RT-qPCR. RESULTS We found that miRNAs were enriched in CORSAIR. Low plasma levels of hsa-miR-150-5p and hsa-miR-93-5p were associated with higher COVID-19-related death. The levels of hsa-miR-92b-3p were associated with SARS-CoV-2 test positivity. Peripheral blood mononuclear cells (PBMC) increased secretion of hsa-miR-150-5p, hsa-miR-93-5p, and hsa-miR-92b-3p after in vitro TLR7/8- and T cell receptor (TCR)-mediated activation. Increased levels of these three miRNAs were measured in the serum samples of healthy individuals between one and nine months after the second dose of the Pfizer-BioNTech COVID-19 vaccine. SARS-CoV-2 infection of human airway epithelial cells influenced the miRNA levels inside their secreted extracellular vesicles. CONCLUSIONS MiRNAs are enriched at CORSAIR. Plasma miRNA levels can represent a potential blood biomarker for predicting COVID-19-related death in cancer patients.
Collapse
Affiliation(s)
- Simone Anfossi
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA.
| | - Faezeh Darbaniyan
- Department of Hematopoietic Biology & Malignancy, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Joseph Quinlan
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Steliana Calin
- Department of Hemopathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Masayoshi Shimizu
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Meng Chen
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Paola Rausseo
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Michael Winters
- Department of Microbiology, Immunology and Cell Biology, West Virginia University Cancer Institute, Morgantown, USA
| | - Elena Bogatenkova
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Kim-Anh Do
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Ivan Martinez
- Department of Microbiology, Immunology and Cell Biology, West Virginia University Cancer Institute, Morgantown, USA
| | - Ziyi Li
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Loredana Antal
- Clinical Laboratory, Municipal Clinical Emergency Hospital, Timisoara, Romania
| | - Tudor Rares Olariu
- Clinical Laboratory, Municipal Clinical Emergency Hospital, Timisoara, Romania
- Department of Infectious Diseases, Center for Diagnosis and Study of Parasitic Diseases, Victor Babes University of Medicine and Pharmacy, Timisoara, Romania
| | - Ignacio Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - George A Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA.
- The Non-coding RNA Center, The University of Texas MD Anderson Cancer Center, Houston, USA.
| |
Collapse
|
29
|
Luo Q, Lan P, Lin Y, Zhang P, Ma X. Effect of physical activity on anxiety and depression in COVID-19 adults: A systematic review and meta-analysis. iScience 2024; 27:110844. [PMID: 39429776 PMCID: PMC11490742 DOI: 10.1016/j.isci.2024.110844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 02/22/2024] [Accepted: 08/27/2024] [Indexed: 10/22/2024] Open
Abstract
While the benefits of physical activity on mental health are well-known, systematic reviews and meta-analyses on its impact on mental illness in adults with COVID-19 are scarce. This study of 25 randomized controlled trials shows that physical activity significantly reduces anxiety (standardized mean difference [SMD] = -0.915; 95% confidence interval [CI] = -1.182 to -0.648; I2 = 82.0%; p < 0.001) and depression (SMD = -0.752; 95% CI = -1.034 to -0.470; I2 = 81.4%; p < 0.001). Traditional Chinese ethnic sports are notably effective. Interventions under 3 weeks best reduce depression, while 3 ≤ 7 weeks optimally reduce anxiety. Sessions ≤5 times weekly, with 30 ≤ 60 min for anxiety and >60 min for depression, yield the best outcomes. These results highlight the specific effectiveness of physical activity in alleviating anxiety and depression in COVID-19 patients.
Collapse
Affiliation(s)
- Qingyuan Luo
- School of Wushu, Chengdu Sport University, Chengdu 610041, China
| | - Peng Lan
- School of Wushu, Chengdu Sport University, Chengdu 610041, China
| | | | - Peng Zhang
- College of Physical Education and Sports, Beijing Normal University, Beijing 100875, China
| | - Xiujie Ma
- School of Wushu, Chengdu Sport University, Chengdu 610041, China
- Chinese GuoShu Academy, Chengdu Sports University, Chengdu 610041, China
| |
Collapse
|
30
|
Hederman AP, Remmel CA, Sharma S, Natarajan H, Weiner JA, Wrapp D, Donner C, Delforge ML, d’Angelo P, Furione M, Fornara C, McLellan JS, Lilleri D, Marchant A, Ackerman ME. Discrimination of primary and chronic cytomegalovirus infection based on humoral immune profiles in pregnancy. J Clin Invest 2024; 134:e180560. [PMID: 39207860 PMCID: PMC11473158 DOI: 10.1172/jci180560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUNDMost humans have been infected with cytomegalovirus (CMV) by midlife without clinical signs of disease. However, in settings in which the immune system is undeveloped or compromised, the virus is not adequately controlled and consequently presents a major infectious cause of both congenital disease during pregnancy as well as opportunistic infection in children and adults. With clear evidence that risk to the fetus varies with gestational age at the time of primary maternal infection, further research on humoral responses to primary CMV infection during pregnancy is needed.METHODSHere, systems serology tools were applied to characterize antibody responses to CMV infection in pregnant and nonpregnant women experiencing either primary or chronic infection.RESULTSWhereas strikingly different antibody profiles were observed depending on infection status, limited differences were associated with pregnancy status. Beyond known differences in IgM responses used clinically for identification of primary infection, distinctions observed in IgA and FcγR-binding antibodies and among antigen specificities accurately predicted infection status. Machine learning was used to define the transition from primary to chronic states and predict time since infection with high accuracy. Humoral responses diverged over time in an antigen-specific manner, with IgG3 responses toward tegument decreasing over time as typical of viral infections, while those directed to pentamer and glycoprotein B were lower during acute and greatest during chronic infection.CONCLUSIONIn sum, this work provides insights into the antibody response associated with CMV infection status in the context of pregnancy, revealing aspects of humoral immunity that have the potential to improve CMV diagnostics.FUNDINGCYMAF consortium and NIH NIAID.
Collapse
Affiliation(s)
- Andrew P. Hederman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | | | - Shilpee Sharma
- European Plotkin Institute for Vaccinology, Université libre de Bruxelles, Brussels, Belgium
| | - Harini Natarajan
- Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, New Hampshire, USA
| | - Joshua A. Weiner
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | - Daniel Wrapp
- Department of Molecular Biosciences, The University of Texas, Austin, Texas, USA
| | - Catherine Donner
- Université Libre de Bruxelles (ULB), Hôpital Universitaire de Bruxelles (H.U.B.), CUB Hôpital Erasme, Department of Obstetrics and Gynecology, Brussels, Belgium
| | - Marie-Luce Delforge
- ULB, H.U.B., CUB Hôpital Erasme, National Reference Center for Congenital Infections, Brussels, Belgium
| | - Piera d’Angelo
- Microbiology and Virology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Milena Furione
- Microbiology and Virology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Chiara Fornara
- Microbiology and Virology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Jason S. McLellan
- Department of Molecular Biosciences, The University of Texas, Austin, Texas, USA
| | - Daniele Lilleri
- Microbiology and Virology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Arnaud Marchant
- European Plotkin Institute for Vaccinology, Université libre de Bruxelles, Brussels, Belgium
| | - Margaret E. Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
- Department of Microbiology and Immunology, Geisel School of Medicine, Hanover, New Hampshire, USA
| |
Collapse
|
31
|
Tehseen A, Kumar D, Dubey A, Sarkar R, Singh S, Sehrawat S. Glucocorticoid-mediated Suppression of Effector Programming Assists the Memory Transition of Virus-specific CD8+ T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1170-1186. [PMID: 39212406 DOI: 10.4049/jimmunol.2300513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 08/02/2024] [Indexed: 09/04/2024]
Abstract
We demonstrate the role of signaling via the glucocorticoid receptor, NR3C1, in differentiation of CD8+ T cell memory. Pharmacological inhibition as well as the short hairpin RNA-mediated knockdown of the receptor hindered memory transition and limited the homeostatic turnover of the activated CD8+ T cells. Dexamethasone exposure of CD8+ T cells expanded during a resolving infection with influenza A virus or a γ-herpesvirus promoted conversion of effector cells into memory cells by modulating cellular metabolism and lowering the accumulation of reactive oxygen species. Reduced reactive oxygen species levels in the responding effector cells upregulated Bcl2 and enhanced survival. The generated virus-specific memory CD8+ T cells were efficiently recalled following challenge of animals with a secondary infection to control it better. The memory-enhancing effect was predominantly evident at low doses of dexamethasone. Therefore, controlled glucocorticoid signaling within the effector CD8+ T cells is crucial for optimal memory differentiation.
Collapse
Affiliation(s)
- Azeez Tehseen
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab
| | - Dhaneshwar Kumar
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab
| | - Abhishek Dubey
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab
| | - Roman Sarkar
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab
| | - Sudhakar Singh
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab
| | - Sharvan Sehrawat
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Punjab
| |
Collapse
|
32
|
Chang T, Alvarez J, Chappidi S, Crockett S, Sorouri M, Orchard RC, Hancks DC. Metabolic reprogramming tips vaccinia virus infection outcomes by stabilizing interferon-γ induced IRF1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.10.617691. [PMID: 39416205 PMCID: PMC11482883 DOI: 10.1101/2024.10.10.617691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Interferon (IFN) induced activities are critical, early determinants of immune responses and infection outcomes. A key facet of IFN responses is the upregulation of hundreds of mRNAs termed interferon-stimulated genes (ISGs) that activate intrinsic and cell-mediated defenses. While primary interferon signaling is well-delineated, other layers of regulation are less explored but implied by aberrant ISG expression signatures in many diseases in the absence of infection. Consistently, our examination of tonic ISG levels across uninfected human tissues and individuals revealed three ISG subclasses. As tissue identity and many comorbidities with increased virus susceptibility are characterized by differences in metabolism, we characterized ISG responses in cells grown in media known to favor either aerobic glycolysis (glucose) or oxidative phosphorylation (galactose supplementation). While these conditions over time had a varying impact on the expression of ISG RNAs, the differences were typically greater between treatments than between glucose/galactose. Interestingly, extended interferon-priming led to divergent expression of two ISG proteins: upregulation of IRF1 in IFN-γ/glucose and increased IFITM3 in galactose by IFN-α and IFN-γ. In agreement with a hardwired response, glucose/galactose regulation of interferon-γ induced IRF1 is conserved in unrelated mouse and cat cell types. In galactose conditions, proteasome inhibition restored interferon-γ induced IRF1 levels to that of glucose/interferon-γ. Glucose/interferon-γ decreased replication of the model poxvirus vaccinia at low MOI and high MOIs. Vaccinia replication was restored by IRF1 KO. In contrast, but consistent with differential regulation of IRF1 protein by glucose/galactose, WT and IRF1 KO cells in galactose media supported similar levels of vaccinia replication regardless of IFN-γ priming. Also associated with glucose/galactose is a seemingly second block at a very late stage in viral replication which results in reductions in herpes- and poxvirus titers but not viral protein expression. Collectively, these data illustrate a novel layer of regulation for the key ISG protein, IRF1, mediated by glucose/galactose and imply unappreciated subprograms embedded in the interferon response. In principle, such cellular circuitry could rapidly adapt immune responses by sensing changing metabolite levels consumed during viral replication and cell proliferation.
Collapse
|
33
|
Lydon E, Osborne CM, Wagner BD, Ambroggio L, Kirk Harris J, Reeder R, Carpenter TC, Maddux AB, Leroue MK, Yehya N, DeRisi JL, Hall MW, Zuppa AF, Carcillo J, Meert K, Sapru A, Pollack MM, McQuillen P, Notterman DA, Langelier CR, Mourani PM. Proteomic profiling of the local and systemic immune response to pediatric respiratory viral infections. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.08.617294. [PMID: 39416167 PMCID: PMC11482837 DOI: 10.1101/2024.10.08.617294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Viral lower respiratory tract infection (vLRTI) is a leading cause of hospitalization and death in children worldwide. Despite this, no studies have employed proteomics to characterize host immune responses to severe pediatric vLRTI in both the lower airway and systemic circulation. To address this gap, gain insights into vLRTI pathophysiology, and test a novel diagnostic approach, we assayed 1,305 proteins in tracheal aspirate (TA) and plasma from 62 critically ill children using SomaScan. We performed differential expression (DE) and pathway analyses comparing vLRTI (n=40) to controls with non-infectious acute respiratory failure (n=22), developed a diagnostic classifier using LASSO regression, and analyzed matched TA and plasma samples. We further investigated the impact of viral load and bacterial coinfection on the proteome. The TA signature of vLRTI was characterized by 200 DE proteins (Padj<0.05) with upregulation of interferons and T cell responses and downregulation of inflammation-modulating proteins including FABP and MIP-5. A nine-protein TA classifier achieved an AUC of 0.96 (95% CI 0.90-1.00) for identifying vLRTI. In plasma, the host response to vLRTI was more muted with 56 DE proteins. Correlation between TA and plasma was limited, although ISG15 was elevated in both compartments. In bacterial coinfection, we observed increases in the TNF-stimulated protein TSG-6, as well as CRP, and interferon-related proteins. Viral load correlated positively with interferon signaling and negatively with neutrophil-activation pathways. Taken together, our study provides fresh insight into the lower airway and systemic proteome of severe pediatric vLRTI, and identifies novel protein biomarkers with diagnostic potential.
Collapse
Affiliation(s)
- Emily Lydon
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Christina M Osborne
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Brandie D Wagner
- Department of Biostatistics and Informatics, University of Colorado, Colorado School of Public Health, Aurora, CO
| | - Lilliam Ambroggio
- Sections of Emergency Medicine and Hospital Medicine, Children's Hospital Colorado, Aurora, CO
- Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO
| | - J Kirk Harris
- Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO
| | - Ron Reeder
- Department of Pediatrics, University of Utah, Salt Lake City, UT
| | - Todd C Carpenter
- Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO
| | - Aline B Maddux
- Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO
| | - Matthew K Leroue
- Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO
| | - Nadir Yehya
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Joseph L DeRisi
- Chan Zuckerberg Biohub, San Francisco, CA
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA
| | - Mark W Hall
- Department of Pediatrics, Nationwide Children's Hospital, Columbus, OH
| | - Athena F Zuppa
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Joseph Carcillo
- Departments of Pediatrics and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Kathleen Meert
- Department of Pediatrics, Children's Hospital of Michigan, Central Michigan University, Detroit, MI
| | - Anil Sapru
- Department of Pediatrics, University of California Los Angeles, Los Angeles, CA
| | - Murray M Pollack
- Department of Pediatrics, Children's National Medical Center and George Washington School of Medicine and Health Sciences, Washington, DC
| | - Patrick McQuillen
- Department of Pediatrics, University of California San Francisco, San Francisco, CA
| | | | - Charles R Langelier
- Department of Medicine, University of California San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub, San Francisco, CA
| | - Peter M Mourani
- Department of Pediatrics, Critical Care, University of Arkansas for Medical Sciences and Arkansas Children's Hospital, Little Rock, AR
| |
Collapse
|
34
|
Usha T, Hemavathi KN, Goyal AK, Abhinand C, Dhivya S, Cholarajan A, Joshi N, Babu D, Middha SK. Investigating emodin derivatives against SARS-CoV-2 found in medicinal herbs. KUWAIT JOURNAL OF SCIENCE 2024; 51:100265. [DOI: 10.1016/j.kjs.2024.100265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
|
35
|
Chang T, Alvarez J, Chappidi S, Crockett S, Sorouri M, Orchard RC, Hancks DC. Metabolic reprogramming tips vaccinia virus infection outcomes by stabilizing interferon-γ induced IRF1. PLoS Pathog 2024; 20:e1012673. [PMID: 39475961 PMCID: PMC11554218 DOI: 10.1371/journal.ppat.1012673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/11/2024] [Accepted: 10/16/2024] [Indexed: 11/06/2024] Open
Abstract
Interferon (IFN) induced activities are critical, early determinants of immune responses and infection outcomes. A key facet of IFN responses is the upregulation of hundreds of mRNAs termed interferon-stimulated genes (ISGs) that activate intrinsic and cell-mediated defenses. While primary interferon signaling is well-delineated, other layers of regulation are less explored but implied by aberrant ISG expression signatures in many diseases in the absence of infection. Consistently, our examination of tonic ISG levels across uninfected human tissues and individuals revealed three ISG subclasses. As tissue identity and many comorbidities with increased virus susceptibility are characterized by differences in metabolism, we characterized ISG responses in cells grown in media known to favor either aerobic glycolysis (glucose) or oxidative phosphorylation (galactose supplementation). While these conditions over time had a varying impact on the expression of ISG RNAs, the differences were typically greater between treatments than between glucose/galactose. Interestingly, extended interferon-priming led to divergent expression of two ISG proteins: upregulation of IRF1 in IFN-γ/glucose and increased IFITM3 in galactose by IFN-α and IFN-γ. In agreement with a hardwired response, glucose/galactose regulation of interferon-γ induced IRF1 is conserved in unrelated mouse and cat cell types. In galactose conditions, proteasome inhibition restored interferon-γ induced IRF1 levels to that of glucose/interferon-γ. Glucose/interferon-γ decreased replication of the model poxvirus vaccinia at low MOI and high MOIs. Vaccinia replication was restored by IRF1 KO. In contrast, but consistent with differential regulation of IRF1 protein by glucose/galactose, WT and IRF1 KO cells in galactose media supported similar levels of vaccinia replication regardless of IFN-γ priming. Also associated with glucose/galactose is a seemingly second block at a very late stage in viral replication which results in reductions in herpes- and poxvirus titers but not viral protein expression. Collectively, these data illustrate a novel layer of regulation for the key ISG protein, IRF1, mediated by glucose/galactose and imply unappreciated subprograms embedded in the interferon response. In principle, such cellular circuitry could rapidly adapt immune responses by sensing changing metabolite levels consumed during viral replication and cell proliferation.
Collapse
Affiliation(s)
- Tyron Chang
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Genetics, Development, and Disease Ph.D. program, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Jessica Alvarez
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Molecular Microbiology Ph.D. program, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Sruthi Chappidi
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Stacey Crockett
- Molecular Microbiology Ph.D. program, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Mahsa Sorouri
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Robert C. Orchard
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Dustin C. Hancks
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| |
Collapse
|
36
|
Lee SH, Bonifacio F, Prudente AS, Choi YI, Roh J, Adjafre BL, Park CK, Jung SJ, Cunha TM, Berta T. STING recognition of viral dsDNA by nociceptors mediates pain in mice. Brain Behav Immun 2024; 121:29-42. [PMID: 39025416 DOI: 10.1016/j.bbi.2024.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/05/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024] Open
Abstract
Pain is often one of the initial indicators of a viral infection, yet our understanding of how viruses induce pain is limited. Immune cells typically recognize viral nucleic acids, which activate viral receptors and signaling, leading to immunity. Interestingly, these viral receptors and signals are also present in nociceptors and are associated with pain. Here, we investigate the response of nociceptors to nucleic acids during viral infections, specifically focusing on the role of the viral signal, Stimulator of Interferon Genes (STING). Our research shows that cytosolic double-stranded DNA (dsDNA) from viruses, like herpes simplex virus 1 (HSV-1), triggers pain responses through STING expression in nociceptors. In addition, STING agonists alone can elicit pain responses. Notably, these responses involve the direct activation of STING in nociceptors through TRPV1. We also provided a proof-of-concept showing that STING and TRPV1 significantly contribute to the mechanical hypersensitivity induced by HSV-1 infection. These findings suggest that STING could be a potential therapeutic target for relieving pain during viral infections.
Collapse
Affiliation(s)
- Sang Hoon Lee
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, United States
| | - Fabio Bonifacio
- Center for Research in Inflammatory Diseases, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Arthur Silveira Prudente
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, United States
| | - Y I Choi
- Department of Physiology, Medical School, Hanyang University, Seoul, South Korea
| | - Jueun Roh
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, United States; Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon, South Korea
| | - Beatriz Lima Adjafre
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, United States; Center for Research in Inflammatory Diseases, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Chul-Kyu Park
- Gachon Pain Center and Department of Physiology, College of Medicine, Gachon University, Incheon, South Korea
| | - Sung Jun Jung
- Department of Physiology, Medical School, Hanyang University, Seoul, South Korea
| | - Thiago M Cunha
- Center for Research in Inflammatory Diseases, Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.
| | - Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, United States.
| |
Collapse
|
37
|
Melkonian AK, Hakobyan GV. Evaluation of the therapeutic action of original antiviral drug in SARS-CoV-2. Biotechnol Appl Biochem 2024; 71:1057-1069. [PMID: 38710664 DOI: 10.1002/bab.2597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/23/2024] [Indexed: 05/08/2024]
Abstract
Purpose of this article is to study the possible direct antiviral effect of "Armenikum" on SARS-CoV-2, conduct an in vitro study on the SARS-CoV-2 encephalomocarditis virus, and an in vivo study on the Syrian hamster model. Human coronavirus SARS-CoV-2 (delta strain) was used as the virus. Two groups of four-specimen hamsters were used to study the therapeutic activity of the drug during 48 h after infecting. One group of hamsters served as positive control and was infected with the virus at a similar dose as experimental one and was used as a control of pathology induced by the viral infection till the end of the experiment. Another group of hamsters (four of them) was injected physiological solution and was used as a control. The Syrian hamsters underwent a clinical blood test and computed tomography. "Armenikum" in the form of an injection has a significant antiviral effect on the human coronavirus SARS-CoV-2, credibly reducing the titers of the virus and the time of its elimination from the Syrian hamsters, significantly mitigating the viral infection. "Armenikum" in the form of an injection drug almost completely removes the pathological effect of the virus in the lungs of the hamsters.
Collapse
Affiliation(s)
| | - Gagik V Hakobyan
- Department of Oral and Maxillofacial Surgery, University of Yerevan State Medical University, Yerevan, Armenia
| |
Collapse
|
38
|
Liu W, Wilke CO, Arnold JJ, Sotoudegan MS, Cameron CE. Single-cell virology: On-chip, quantitative characterization of the dynamics of virus spread from one single cell to another. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.615011. [PMID: 39386720 PMCID: PMC11463428 DOI: 10.1101/2024.09.25.615011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Virus spread at the single-cell level is largely uncharacterized. We have designed and constructed a microfluidic device in which each nanowell contained a single, infected cell (donor) and a single, uninfected cell (recipient). Using a GFP-expressing poliovirus as our model, we observed both lytic and non-lytic spread. Donor cells supporting lytic spread established infection earlier than those supporting non-lytic spread. However, non-lytic spread established infections in recipient cells substantially faster than lytic spread and yielded higher rates of genome replication. While lytic spread was sensitive to the presence of capsid entry/uncoating inhibitors, non-lytic spread was not. Consistent with emerging models for non-lytic spread of enteroviruses using autophagy, reduction of LC3 levels in cells impaired non-lytic spread and elevated the fraction of virus in donor cells spreading lytically. The ability to distinguish lytic and non-lytic spread unambiguously will enable discovery of viral and host factors and host pathways used for non-lytic spread of enteroviruses and other viruses as well.
Collapse
Affiliation(s)
- Wu Liu
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
- Present address: School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong 250100, China
| | - Claus O Wilke
- Center for Computational Biology and Bioinformatics, Institute for Cellular and Molecular Biology, and Department of Integrative Biology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jamie J Arnold
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mohamad S Sotoudegan
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Craig E Cameron
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
39
|
Antony F, Kinha D, Nowińska A, Rouse BT, Suryawanshi A. The immunobiology of corneal HSV-1 infection and herpetic stromal keratitis. Clin Microbiol Rev 2024; 37:e0000624. [PMID: 39078136 PMCID: PMC11391706 DOI: 10.1128/cmr.00006-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2024] Open
Abstract
SUMMARYHuman alphaherpesvirus 1 (HSV-1) is a highly successful neurotropic pathogen that primarily infects the epithelial cells lining the orofacial mucosa. After primary lytic replication in the oral, ocular, and nasal mucosal epithelial cells, HSV-1 establishes life-long latency in neurons within the trigeminal ganglion. Patients with compromised immune systems experience frequent reactivation of HSV-1 from latency, leading to virus entry in the sensory neurons, followed by anterograde transport and lytic replication at the innervated mucosal epithelial surface. Although recurrent infection of the corneal mucosal surface is rare, it can result in a chronic immuno-inflammatory condition called herpetic stromal keratitis (HSK). HSK leads to gradual vision loss and can cause permanent blindness in severe untreated cases. Currently, there is no cure or successful vaccine to prevent latent or recurrent HSV-1 infections, posing a significant clinical challenge to managing HSK and preventing vision loss. The conventional clinical management of HSK primarily relies on anti-virals to suppress HSV-1 replication, anti-inflammatory drugs (such as corticosteroids) to provide symptomatic relief from pain and inflammation, and surgical interventions in more severe cases to replace damaged cornea. However, each clinical treatment strategy has limitations, such as local and systemic drug toxicities and the emergence of anti-viral-resistant HSV-1 strains. In this review, we summarize the factors and immune cells involved in HSK pathogenesis and highlight alternate therapeutic strategies for successful clinical management of HSK. We also discuss the therapeutic potential of immunoregulatory cytokines and immunometabolism modulators as promising HSK therapies against emerging anti-viral-resistant HSV-1 strains.
Collapse
Affiliation(s)
- Ferrin Antony
- Department of Molecular and Cell Biology, University of California, Berkeley, California, USA
| | - Divya Kinha
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Anna Nowińska
- Clinical Department of Ophthalmology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, Katowice, Poland
- Ophthalmology Department, Railway Hospital in Katowice, Katowice, Poland
| | - Barry T. Rouse
- College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| | - Amol Suryawanshi
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
40
|
Chikopela T, Mwesigwa N, Masenga SK, Kirabo A, Shibao CA. The Interplay of HIV and Long COVID in Sub-Saharan Africa: Mechanisms of Endothelial Dysfunction. Curr Cardiol Rep 2024; 26:859-871. [PMID: 38958890 DOI: 10.1007/s11886-024-02087-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/23/2024] [Indexed: 07/04/2024]
Abstract
PURPOSE OF REVIEW Long COVID affects approximately 5 million people in Africa. This disease is characterized by persistent symptoms or new onset of symptoms after an acute SARS-CoV-2 infection. Specifically, the most common symptoms include a range of cardiovascular problems such as chest pain, orthostatic intolerance, tachycardia, syncope, and uncontrolled hypertension. Importantly, these conditions appear to have endothelial dysfunction as the common denominator, which is often due to impaired nitric oxide (NO) mechanisms. This review discusses the role of mechanisms contributing to endothelial dysfunction in Long COVID, particularly in people living with HIV. RECENT FINDINGS Recent studies have reported that increased inflammation and oxidative stress, frequently observed in Long COVID, may contribute to NO dysfunction, ultimately leading to decreased vascular reactivity. These mechanisms have also been reported in people living with HIV. In regions like Africa, where HIV infection is still a major public health challenge with a prevalence of approximately 26 million people in 2022. Specifically, endothelial dysfunction has been reported as a major mechanism that appears to contribute to cardiovascular diseases and the intersection with Long COVID mechanisms is of particular concern. Further, it is well established that this population is more likely to develop Long COVID following infection with SARS-CoV-2. Therefore, concomitant infection with SARS-CoV-2 may lead to accelerated cardiovascular disease. We outline the details of the worsening health problems caused by Long COVID, which exacerbate pre-existing conditions such as endothelial dysfunction. The overlapping mechanisms of HIV and SARS-CoV-2, particularly the prolonged inflammatory response and chronic hypoxia, may increase susceptibility to Long COVID. Addressing these overlapping health issues is critical as it provides clinical entry points for interventions that could improve and enhance outcomes and quality of life for those affected by both HIV and Long COVID in the region.
Collapse
Affiliation(s)
- Theresa Chikopela
- Department of Human Physiology, Faculty of Medicine, Lusaka Apex Medical University, Lusaka, Zambia
| | - Naome Mwesigwa
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37332-0615, USA
| | - Sepiso K Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Livingstone Campus, Livingstone, Zambia
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37332-0615, USA
| | - Cyndya A Shibao
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37332-0615, USA.
| |
Collapse
|
41
|
Kim SH, Choi HN, Jo MG, Lee B, Kim YJ, Seong H, Song C, Yoo HS, Lee JH, Seong D, Park HJ, Roh IS, Yang J, Lee MY, Kim HJ, Park SW, Kim M, Kim SJ, Kim M, Kim HJ, Hong KW, Yun SP. Activation of neurotoxic A1-reactive astrocytes by SFTS virus infection accelerates fatal brain damage in IFNAR1 -/- mice. J Med Virol 2024; 96:e29854. [PMID: 39135475 DOI: 10.1002/jmv.29854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 07/08/2024] [Accepted: 08/01/2024] [Indexed: 09/26/2024]
Abstract
Severe fever with thrombocytopenia syndrome (SFTS) has a high mortality rate compared to other infectious diseases. SFTS is particularly associated with a high risk of mortality in immunocompromised individuals, while most patients who die of SFTS exhibit symptoms of severe encephalitis before death. However, the region of brain damage and mechanisms by which the SFTS virus (SFTSV) causes encephalitis remains unknown. Here, we revealed that SFTSV infects the brainstem and spinal cord, which are regions of the brain associated with respiratory function, and motor nerves in IFNAR1-/- mice. Further, we show that A1-reactive astrocytes are activated, causing nerve cell death, in infected mice. Primary astrocytes of SFTSV-infected IFNAR1-/- mice also induced neuronal cell death through the activation of A1-reactive astrocytes. Herein, we showed that SFTSV induces fatal neuroinflammation in the brain regions important for respiratory function and motor nerve, which may underlie mortality in SFTS patients. This study provides new insights for the treatment of SFTS, for which there is currently no therapeutic approach.
Collapse
Affiliation(s)
- Seon-Hee Kim
- Department of Pharmacology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Ha Nyeoung Choi
- Department of Pharmacology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
- Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Min Gi Jo
- Department of Pharmacology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
- Department of Pathology, College of Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Bina Lee
- Department of Pharmacology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Young Jin Kim
- Department of Pharmacology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
- Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Hyemin Seong
- Department of Ophthalmology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Chieun Song
- Department of Ophthalmology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Han Sol Yoo
- Department of Pharmacology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Jeong Hyun Lee
- Department of Pharmacology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Daseul Seong
- Division of foreign Animal Disease, Animal and Plant Quarantine Agency, Gimcheon, Republic of Korea
| | - Hyun-Jin Park
- Division of foreign Animal Disease, Animal and Plant Quarantine Agency, Gimcheon, Republic of Korea
| | - In-Soon Roh
- Division of foreign Animal Disease, Animal and Plant Quarantine Agency, Gimcheon, Republic of Korea
| | - Jinsung Yang
- Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
- Department of Biochemistry, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Min Young Lee
- College of Pharmacy, Kyungpook National University, Daegu, South Korea
| | - Hye Jung Kim
- Department of Pharmacology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
- Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Sang Won Park
- Department of Pharmacology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
- Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Mingyo Kim
- Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
- Department of Rheumatology Internal Medicine, Gyeongsang National University Hospital, Jinju, Republic of Korea
| | - Seong Jae Kim
- Department of Ophthalmology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Minkyeong Kim
- Department of Neurology, Gyeongsang National University Hospital, Jinju, Republic of Korea
| | - Hyun-Jeong Kim
- Division of foreign Animal Disease, Animal and Plant Quarantine Agency, Gimcheon, Republic of Korea
- Laboratory Animal Research Center, Central Scientific Instrumentation Facility, Gyeongsang National University, Jinju, Republic of Korea
| | - Kyung-Wook Hong
- Division of Infectious Diseases, Department of Internal Medicine, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju, Republic of Korea
| | - Seung Pil Yun
- Department of Pharmacology, Institute of Medical Sciences, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
- Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
| |
Collapse
|
42
|
Rani A, Stadler JT, Marsche G. HDL-based therapeutics: A promising frontier in combating viral and bacterial infections. Pharmacol Ther 2024; 260:108684. [PMID: 38964560 DOI: 10.1016/j.pharmthera.2024.108684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/03/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
Low levels of high-density lipoprotein (HDL) and impaired HDL functionality have been consistently associated with increased susceptibility to infection and its serious consequences. This has been attributed to the critical role of HDL in maintaining cellular lipid homeostasis, which is essential for the proper functioning of immune and structural cells. HDL, a multifunctional particle, exerts pleiotropic effects in host defense against pathogens. It functions as a natural nanoparticle, capable of sequestering and neutralizing potentially harmful substances like bacterial lipopolysaccharides. HDL possesses antiviral activity, preventing viruses from entering or fusing with host cells, thereby halting their replication cycle. Understanding the complex relationship between HDL and the immune system may reveal innovative targets for developing new treatments to combat infectious diseases and improve patient outcomes. This review aims to emphasize the role of HDL in influencing the course of bacterial and viral infections and its and its therapeutic potential.
Collapse
Affiliation(s)
- Alankrita Rani
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Styria, Austria
| | - Julia T Stadler
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Styria, Austria
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Styria, Austria; BioTechMed-Graz, Mozartgasse 12/II, 8010 Graz, Styria, Austria.
| |
Collapse
|
43
|
Macamo ED, Mkhize-Kwitshana ZL, Mthombeni J, Naidoo P. The Impact of HIV and Parasite Single Infection and Coinfection on Telomere Length: A Systematic Review. Curr Issues Mol Biol 2024; 46:7258-7290. [PMID: 39057072 PMCID: PMC11275449 DOI: 10.3390/cimb46070431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
HIV and parasite infections accelerate biological aging, resulting in immune senescence, apoptosis and cellular damage. Telomere length is considered to be one of the most effective biomarkers of biological aging. HIV and parasite infection have been reported to shorten telomere length in the host. This systematic review aimed to highlight work that explored the influence of HIV and parasite single infections and coinfection on telomere length. Using specific keywords related to the topic of interest, an electronic search of several online databases (Google Scholar, Web of Science, Scopus, Science Direct and PubMed) was conducted to extract eligible articles. The association between HIV infection or parasite infection and telomere length and the association between HIV and parasite coinfection and telomere length were assessed independently. The studies reported were mostly conducted in the European countries. Of the 42 eligible research articles reviewed, HIV and parasite single infections were independently associated with telomere length shortening. Some studies found no association between antiretroviral therapy (ART) and telomere length shortening, while others found an association between ART and telomere length shortening. No studies reported on the association between HIV and parasite coinfection and telomere length. HIV and parasite infections independently accelerate telomere length shortening and biological aging. It is possible that coinfection with HIV and parasites may further accelerate telomere length shortening; however, this is a neglected field of research with no reported studies to date.
Collapse
Affiliation(s)
- Engelinah D. Macamo
- Department of Medical Microbiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, Nelson R. Mandela Medical School Campus, University of KwaZulu-Natal, Durban 4001, South Africa
- Division of Research Capacity Development (RCD), South African Medical Research Council (SAMRC), Tygerberg, Cape Town 7505, South Africa
| | - Zilungile L. Mkhize-Kwitshana
- Department of Medical Microbiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, Nelson R. Mandela Medical School Campus, University of KwaZulu-Natal, Durban 4001, South Africa
- Division of Research Capacity Development (RCD), South African Medical Research Council (SAMRC), Tygerberg, Cape Town 7505, South Africa
- Department of Biomedical Sciences, Doorfontein Campus, University of Johannesburg, Johannesburg 1710, South Africa
- Biomedical Sciences Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Florida Campus, Johannesburg 1710, South Africa
| | - Julian Mthombeni
- Department of Biomedical Sciences, Doorfontein Campus, University of Johannesburg, Johannesburg 1710, South Africa
| | - Pragalathan Naidoo
- Department of Medical Microbiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, Nelson R. Mandela Medical School Campus, University of KwaZulu-Natal, Durban 4001, South Africa
- Division of Research Capacity Development (RCD), South African Medical Research Council (SAMRC), Tygerberg, Cape Town 7505, South Africa
| |
Collapse
|
44
|
Kinyenje E, Hokororo J, Ngowi R, Kiremeji M, Mnunga E, Samwel A, Sylvanus E, Mnken E, Yango M, Mtalika M, Mmbaga V, Saitoti N, Malecha A, Kundy F, Rwabilimbo M, Kaniki I, Mwisomba G, Charles E, Mughanga P, Kitambi M, Paul R, Richard E, Musyani A, Rabiel I, Haule G, Marandu L, Mwakapasa E, Manasseh G, Sindato C, Beyanga M, Kapyolo E, Jacob F, Mcharo J, Mayige M, Msemwa F, Saguti G, Kauki G, Masuma J, Mrema G, Kohi M, Yoti Z, Habtu M, Mwengee W, Mukurasi K, Gatei W, Ruggajo P, Kwesi E, Eliakimu E, Horumpende P, Magembe G, Nagu T. Infection prevention and control of highly infectious pathogens in resource-limited countries: an experience from Marburg viral disease outbreak in Kagera Region - Tanzania. BMC Infect Dis 2024; 24:628. [PMID: 38914946 PMCID: PMC11197355 DOI: 10.1186/s12879-024-09508-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 06/13/2024] [Indexed: 06/26/2024] Open
Abstract
Marburg viral disease (MVD) is a highly infectious disease with a case fatality rate of up to 90%, particularly impacting resource-limited countries where implementing Infection Prevention and Control (IPC) measures is challenging. This paper shares the experience of how Tanzania has improved its capacity to prevent and control highly infectious diseases, and how this capacity was utilized during the outbreak of the MVD disease that occurred for the first time in the country in 2023.In 2016 and the subsequent years, Tanzania conducted self and external assessments that revealed limited IPC capacity in responding to highly infectious diseases. To address these gaps, initiatives were undertaken, including the enhancement of IPC readiness through the development and dissemination of guidelines, assessments of healthcare facilities, supportive supervision and mentorship, procurement of supplies, and the renovation or construction of environments to bolster IPC implementation.The official confirmation and declaration of MVD on March 21, 2023, came after five patients had already died of the disease. MVD primarily spreads through contact and presents with severe symptoms, which make patient care and prevention challenging, especially in resource-limited settings. However, with the use of a trained workforce; IPC rapid needs assessment was conducted, identifying specific gaps. Based on the results; mentorship programs were carried out, specific policies and guidelines were developed, security measures were enhanced, all burial activities in the area were supervised, and both patients and staff were monitored across all facilities. By the end of the outbreak response on June 1, 2023, a total of 212 contacts had been identified, with the addition of only three deaths. Invasive procedures like dialysis and Manual Vacuum Aspiration prevented some deaths in infected patients, procedures previously discouraged.In summary, this experience underscores the critical importance of strict adherence to IPC practices in controlling highly infectious diseases. Recommendations for low-income countries include motivating healthcare providers and improving working conditions to enhance commitment in challenging environments. This report offers valuable insights and practical interventions for preparing for and addressing highly infectious disease outbreaks through implementation of IPC measures.
Collapse
Affiliation(s)
- Erick Kinyenje
- Health Quality Assurance Unit, Ministry of Health, P. O. Box 743, Dodoma, Tanzania.
| | - Joseph Hokororo
- Health Quality Assurance Unit, Ministry of Health, P. O. Box 743, Dodoma, Tanzania
| | - Ruth Ngowi
- Health Quality Assurance Unit, Ministry of Health, P. O. Box 743, Dodoma, Tanzania
| | - Michael Kiremeji
- Emergency Preparedness and Response Unit, Ministry of Health, P. O. Box 743, Dodoma, Tanzania
| | - Elice Mnunga
- Department of Pediatrics and Child Health, Bugando Medical Center, P. O. Box 1370, Mwanza, Tanzania
| | - Angela Samwel
- Emergency Preparedness and Response Unit, Ministry of Health, P. O. Box 743, Dodoma, Tanzania
| | - Erasto Sylvanus
- Emergency Preparedness and Response Unit, Ministry of Health, P. O. Box 743, Dodoma, Tanzania
| | - Emmanuel Mnken
- Health Promotion Section, Department of Preventive Services, Ministry of Health, P. O. Box 743, Dodoma, Tanzania
| | - Missana Yango
- Department of Internal Medicine, Dodoma Regional Referral Hospital, P. O. Box 904, Dodoma, Tanzania
| | - Mikidadi Mtalika
- Wildlife Research Institute, Kingupira Wildlife Research Centre, P. O. Box 16, Utete- Rufiji, Tanzania
| | - Vida Mmbaga
- Epidemiology Section, Department of Preventive Services, Ministry of Health, P. O. Box 743, Dodoma, Tanzania
| | - Noel Saitoti
- Bukoba District Council Hospital, Kagera Region, P. O. Box 491, Bukoba, Tanzania
| | - Alex Malecha
- Chato Zonal Hospital, P. O. Box 43, Chato- Geita, Tanzania
| | - Faith Kundy
- Department of Internal Medicine, Iringa Region Referral Hospital, P. O. Box 1260, Iringa, Tanzania
| | - Martin Rwabilimbo
- Bukoba Regional Referral Hospital, P. O. Box 299, Bukoba, Kagera, Tanzania
| | | | - Godwin Mwisomba
- Mirembe National Mental Health Hospital, P. O. Box 910, Dodoma, Tanzania
| | - Erica Charles
- Singida Regional Referral Hospital, P. O. Box 104, Singida, Tanzania
| | - Patrick Mughanga
- Department Emergency Medicine, Bugando Medical Center, P. O. Box 1370, Mwanza, Tanzania
| | - Mary Kitambi
- Emergency Preparedness and Response Unit, Ministry of Health, P. O. Box 743, Dodoma, Tanzania
| | - Radenta Paul
- Health Quality Assurance Unit, Ministry of Health, P. O. Box 743, Dodoma, Tanzania
| | - Erick Richard
- Emergency Preparedness and Response Unit, Ministry of Health, P. O. Box 743, Dodoma, Tanzania
| | - Atuganile Musyani
- Amref Health Africa in Tanzania, P. O. Box 2773, Dar es Salaam, Tanzania
| | - Irene Rabiel
- Amref Health Africa in Tanzania, P. O. Box 2773, Dar es Salaam, Tanzania
| | - Gift Haule
- Quality Assurance Department, Bugando Medical Center, P. O. Box 1370, Mwanza, Tanzania
| | - Laura Marandu
- Health Quality Assurance Unit, Ministry of Health, P. O. Box 743, Dodoma, Tanzania
| | - Emmanuel Mwakapasa
- Epidemiology Section, Department of Preventive Services, Ministry of Health, P. O. Box 743, Dodoma, Tanzania
| | - Gerald Manasseh
- President's Office - Regional Administration and Local Government, Dodoma, Tanzania
| | - Calvin Sindato
- Department of Clinical Research, National Institute for Medical Research, Tabora Medical Research Centre, P. O. Box 482, Tabora, Tanzania
| | - Medard Beyanga
- National Public Health Laboratory, P. O. Box 60000, Dar es Salaam, Tanzania
| | - Eliakimu Kapyolo
- Department of Clinical Research, Dodoma Medical Research Centre, National Institute for Medical Research, P. O. Box 805, Dodoma, Tanzania
| | - Frank Jacob
- Epidemiology Section, Department of Preventive Services, Ministry of Health, P. O. Box 743, Dodoma, Tanzania
| | - Jonathan Mcharo
- National Institute of Medical Research, Head Quarters, P. O. Box 2769, Dar es Salaam, Tanzania
| | - Mary Mayige
- National Institute of Medical Research, Head Quarters, P. O. Box 2769, Dar es Salaam, Tanzania
| | | | - Grace Saguti
- World Health Organization, Dar es Salaam, Tanzania
| | - George Kauki
- World Health Organization, Dar es Salaam, Tanzania
| | | | - George Mrema
- Epidemiology Section, Department of Preventive Services, Ministry of Health, P. O. Box 743, Dodoma, Tanzania
| | - Mugendi Kohi
- University of Iringa, P. O. Box 200, Iringa, Tanzania
| | - Zabulon Yoti
- World Health Organization, Dar es Salaam, Tanzania
| | - Michael Habtu
- World Health Organization Regional Office for Africa, Brazzaville, Congo
| | | | | | - Wangeci Gatei
- Centers for Disease Control and Prevention, Dar es Salaam, Tanzania
| | - Paschal Ruggajo
- Directorate of Curative Services, Ministry of Health, P. O. Box 743, Dodoma, Tanzania
| | - Elias Kwesi
- Emergency Preparedness and Response Unit, Ministry of Health, P. O. Box 743, Dodoma, Tanzania
| | - Eliudi Eliakimu
- Health Quality Assurance Unit, Ministry of Health, P. O. Box 743, Dodoma, Tanzania
| | - Pius Horumpende
- Unit of Research Coordination, Directorate of Curative Services, Ministry of Health, P. O Box 743, Dodoma, Tanzania
- Department of Research and Innovation, Peoples' Defence Forces (TPDF) es Salaam, Lugalo Military College of Medical Sciences (MCMS) and General Military Hospital (GMH), P. O. Box Dar, Dar es Salaam, Tanzania
| | - Grace Magembe
- Office of Permanent Secretary, Ministry of Health, P. O. Box 743, Dodoma, Tanzania
| | - Tumaini Nagu
- Office of Chief Medical Officer, Ministry of Health, P. O. Box 743, Dodoma, Tanzania
| |
Collapse
|
45
|
Gopal V, Koh MCY, Ngiam JN, Hang-Cheng O, Somani J, Tambyah PA, Tey J. Does Prior Respiratory Viral Infection Provide Cross-Protection Against Subsequent Respiratory Viral Infections? A Systematic Review and Meta-Analysis. Viruses 2024; 16:982. [PMID: 38932273 PMCID: PMC11209343 DOI: 10.3390/v16060982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/10/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
The epidemiology of different respiratory viral infections is believed to be affected by prior viral infections in addition to seasonal effects. This PROSPERO-registered systematic review identified 7388 studies, of which six met our criteria to answer the question specifically. The purpose of this review was to compare the prevalence of sequential viral infections in those with previously documented positive versus negative swabs. The pooled prevalence of sequential viral infections over varying periods from 30-1000 days of follow-up was higher following a negative respiratory viral swab at 0.15 than following a positive swab at 0.08, indicating the potential protective effects of prior respiratory viral infections. However, significant heterogeneity and publication biases were noted. There is some evidence, albeit of low quality, of a possible protective effect of an initial viral infection against subsequent infections by a different virus, which is possibly due to broad, nonspecific innate immunity. Future prospective studies are needed to validate our findings.
Collapse
Affiliation(s)
- Vennila Gopal
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore (P.A.T.)
- Faculty of Medicine and Health Sciences, University Malaysia Sabah, Kota Kinabalu 88400, Malaysia
| | - Matthew Chung Yi Koh
- Division of Infectious Diseases, Department of Medicine, National University Health System, Singapore 119228, Singapore; (M.C.Y.K.); (J.N.N.)
| | - Jinghao Nicholas Ngiam
- Division of Infectious Diseases, Department of Medicine, National University Health System, Singapore 119228, Singapore; (M.C.Y.K.); (J.N.N.)
| | - Ong Hang-Cheng
- Division of Infectious Diseases, Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia;
| | - Jyoti Somani
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore (P.A.T.)
- Division of Infectious Diseases, Department of Medicine, National University Health System, Singapore 119228, Singapore; (M.C.Y.K.); (J.N.N.)
| | - Paul Anatharajah Tambyah
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore (P.A.T.)
- Division of Infectious Diseases, Department of Medicine, National University Health System, Singapore 119228, Singapore; (M.C.Y.K.); (J.N.N.)
| | - Jeremy Tey
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore (P.A.T.)
- Department of Radiation Oncology, National University Cancer Institute, Singapore 119074, Singapore
| |
Collapse
|
46
|
Kalemera MD, Maher AK, Dominguez-Villar M, Maertens GN. Cell Culture Evaluation Hints Widely Available HIV Drugs Are Primed for Success if Repurposed for HTLV-1 Prevention. Pharmaceuticals (Basel) 2024; 17:730. [PMID: 38931397 PMCID: PMC11206710 DOI: 10.3390/ph17060730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 05/15/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
With an estimated 10 million people infected, the deltaretrovirus human T-cell lymphotropic virus type 1 (HTLV-1) is the second most prevalent pathogenic retrovirus in humans after HIV-1. Like HIV-1, HTLV-1 overwhelmingly persists in a host via a reservoir of latently infected CD4+ T cells. Although most patients are asymptomatic, HTLV-1-associated pathologies are often debilitating and include adult T-cell leukaemia/lymphoma (ATLL), which presents in mature adulthood and is associated with poor prognosis with short overall survival despite treatment. Curiously, the strongest indicator for the development of ATLL is the acquisition of HTLV-1 through breastfeeding. There are no therapeutic or preventative regimens for HTLV-1. However, antiretrovirals (ARVs), which target the essential retrovirus enzymes, have been developed for and transformed HIV therapy. As the architectures of retroviral enzyme active sites are highly conserved, some HIV-specific compounds are active against HTLV-1. Here, we expand on our work, which showed that integrase strand transfer inhibitors (INSTIs) and some nucleoside reverse transcriptase inhibitors (NRTIs) block HTLV-1 transmission in cell culture. Specifically, we find that dolutegravir, the INSTI currently recommended as the basis of all new combination antiretroviral therapy prescriptions, and the latest prodrug formula of the NRTI tenofovir, tenofovir alafenamide, also potently inhibit HTLV-1 infection. Our results, if replicated in a clinical setting, could see transmission rates of HTLV-1 and future caseloads of HTLV-1-associated pathologies like ATLL dramatically cut via the simple repurposing of already widely available HIV pills in HTLV-1 endemic areas. Considering our findings with the old medical saying "it is better to prevent than cure", we highly recommend the inclusion of INSTIs and tenofovir prodrugs in upcoming HTLV-1 clinical trials as potential prophylactics.
Collapse
Affiliation(s)
| | | | | | - Goedele N. Maertens
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK; (M.D.K.); (A.K.M.); (M.D.-V.)
| |
Collapse
|
47
|
Noel S, LaFrancois R, Scott ME. Gastrointestinal nematode infection during pregnancy and lactation enhances spatial reference memory and reduces indicators of anxiety-like behaviour in uninfected adult female mouse offspring. Parasitology 2024; 151:722-731. [PMID: 38808523 PMCID: PMC11474017 DOI: 10.1017/s0031182024000696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 05/30/2024]
Abstract
Maternal bacterial and viral infections that induce neuroinflammation in the developing brain are associated with impaired cognitive function and increased anxiety in the offspring. In contrast, maternal infection with the immunoregulatory murine gastrointestinal (GI) nematode, Heligmosomoides bakeri, appears to benefit neurodevelopment as juvenile 2- and 3-week-old male and female offspring had enhanced spatial memory, which may be due to a Th2/Treg biased neuroimmune environment. Here, the impact of maternal H. bakeri infection during pregnancy and lactation on the spatial and anxiety-like behaviours of adult, 3-month-old uninfected male and female offspring was explored for the first time. It was observed that adult female offspring of H. bakeri-infected dams had enhanced spatial reference memory and reduced anxiety-like behaviour compared to females of uninfected dams. These effects were not observed in adult male offspring. Thus, the positive influence of a maternal GI nematode infection on spatial memory of juvenile offspring persists in adult female offspring.
Collapse
Affiliation(s)
- Sophia Noel
- Institute of Parasitology, McGill University (Macdonald Campus), Quebec H9X 3V9, Canada
| | - Ryan LaFrancois
- Institute of Parasitology, McGill University (Macdonald Campus), Quebec H9X 3V9, Canada
| | - Marilyn E. Scott
- Institute of Parasitology, McGill University (Macdonald Campus), Quebec H9X 3V9, Canada
| |
Collapse
|
48
|
Noureddine R, Baba H, Aqillouch S, Abounouh K, Laazaazia O, Elmessaoudi-Idrissi M, Bahmani FZ, Tanouti IA, Ouladlahsen A, Sarih M, Dehbi H, Ezzikouri S. The Interleukin-6 gene variants may protect against SARS-CoV-2 infection and the severity of COVID-19: a case-control study in a Moroccan population. BMC Med Genomics 2024; 17:139. [PMID: 38783290 PMCID: PMC11112821 DOI: 10.1186/s12920-024-01911-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024] Open
Abstract
The symptoms of SARS-CoV-2 infection vary widely, ranging from asymptomatic cases to severe forms marked by acute respiratory distress syndrome, multi-organ damage, and fatalities. Studies indicate a correlation between specific genes and susceptibility to SARS-CoV-2 infection and disease severity, particularly involving variants in genes linked to inflammation and immune responses. The objective of this study is to investigate the association between rs1800795 (- 174 G > C) and rs1800797 (- 597 A > G) variants in the interleukin-6 (IL-6) promoter region and susceptibility to SARS-CoV-2 infection. Additionally, we aim to explore their correlation with COVID-19 severity in a Moroccan population. In this case-control study, we enrolled 270 unvaccinated COVID-19 patients, consisting of 132 with severe COVID-19 and 138 with asymptomatic-moderate COVID-19. Additionally, we included 339 SARS-CoV-2-negative group. Genotyping of rs1800795 and rs1800797 polymorphisms of the IL-6 gene was performed using predesigned TaqMan SNP genotyping. The median age of SARS-CoV-2-negative controls was 50 years, while severe COVID-19 cases exhibited a median age of 61 years. Additionally, individuals with asymptomatic to moderate COVID-19 had a median age of 36 years. We observed a significant age difference between severe and mild COVID-19 patients (p < 0.0001), and an association was noted between gender and the severity of COVID-19 (p = 0.011). The allele and genotype frequencies of the IL-6 - 597G > A and - 174G > C variants did not show significant associations with susceptibility to SARS-CoV-2 infection (p > 0.05). However, further analysis revealed that the linkage disequilibrium between rs1800797 and rs1800795 indicated that individuals with the GC* haplotype (OR = 0.04, 95% CI 0.01-0.30, p = 0.001) and AG* haplotype (OR = 0.11, 95% CI 0.03-0.46, p = 0.002) were significantly associated with protection against SARS-CoV-2 infection. Moreover, in the overdominant model, the IL-6 - 174 G/C genotype was found to be protective against the development of severe disease compared to those with the G/G-C/C genotypes (p = 0.03; OR = 0.41, 95% CI 0.18-0.96). However, correlations between complete blood count markers, hematological markers, D-dimer, C-reactive protein, and ferritin levels according to - 597 A > G and - 174G > C genotypes showed no significant differences (all p > 0.05). Our findings provide valuable insights into the pathogenesis of COVID-19, suggesting that genetic variations at the IL-6 gene may contribute to the susceptibility to severe SARS-CoV-2 infection within the Moroccan population.
Collapse
Affiliation(s)
- Rachid Noureddine
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, 1 Place Louis Pasteur, Casablanca, 20360, Maroc
- Laboratory of Genetics and Molecular Pathology, Medical School, University Hassan II, Casablanca, Maroc
- Laboratoire Morizgo d'analyses médicales, Casablanca, Maroc
| | - Hanâ Baba
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, 1 Place Louis Pasteur, Casablanca, 20360, Maroc
| | - Safaa Aqillouch
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, 1 Place Louis Pasteur, Casablanca, 20360, Maroc
| | - Karima Abounouh
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, 1 Place Louis Pasteur, Casablanca, 20360, Maroc
| | - Oumaima Laazaazia
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, 1 Place Louis Pasteur, Casablanca, 20360, Maroc
| | - Mohcine Elmessaoudi-Idrissi
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, 1 Place Louis Pasteur, Casablanca, 20360, Maroc
| | | | - Ikram Allah Tanouti
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, 1 Place Louis Pasteur, Casablanca, 20360, Maroc
| | - Ahd Ouladlahsen
- Service des maladies Infectieuses, CHU Ibn Rochd, Casablanca, Maroc
| | - M'hammed Sarih
- Service de Parasitologie et des Maladies Vectorielles, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Hind Dehbi
- Laboratory of Genetics and Molecular Pathology, Medical School, University Hassan II, Casablanca, Maroc
| | - Sayeh Ezzikouri
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, 1 Place Louis Pasteur, Casablanca, 20360, Maroc.
| |
Collapse
|
49
|
Casella V, Cebollada Rica P, Argilaguet J, Vidal E, González-Cao M, Güerri-Fernandez R, Bocharov G, Meyerhans A. Anti-PD-L1 Immunotherapy of Chronic Virus Infection Improves Virus Control without Augmenting Tissue Damage by Fibrosis. Viruses 2024; 16:799. [PMID: 38793680 PMCID: PMC11125757 DOI: 10.3390/v16050799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/25/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Immunotherapy with checkpoint inhibitors, albeit commonly used against tumors, is still at its infancy against chronic virus infections. It relies on the reinvigoration of exhausted T lymphocytes to eliminate virus-infected cells. Since T cell exhaustion is a physiological process to reduce immunopathology, the reinvigoration of these cells might be associated with an augmentation of pathological changes. To test this possibility, we here analyzed in the model system of chronic lymphocytic choriomeningitis virus (LCMV)-infected mice whether treatment with the checkpoint inhibitor anti-PD-L1 antibody would increase CD8 T cell-dependent fibrosis. We show that pre-existing spleen fibrosis did not worsen under conditions that increase CD8 T cell functionality and reduce virus loads suggesting that the CD8 T cell functionality increase remained below its pathogenicity threshold. These promising findings should further encourage immunotherapeutic trials against chronic virus infections.
Collapse
Affiliation(s)
- Valentina Casella
- Infection Biology Laboratory, Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra, 08003 Barcelona, Spain;
| | - Paula Cebollada Rica
- Infection Biology Laboratory, Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra, 08003 Barcelona, Spain;
| | - Jordi Argilaguet
- Institute of Agrifood Research and Technology (IRTA), Centre de Recerca en Sanitat Animal (CReSA), 08193 Barcelona, Spain; (J.A.); (E.V.)
- Unitat Mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), 08193 Barcelona, Spain
- WOAH Collaborating Centre for Emerging and Re-Emerging Pig Diseases in Europe, IRTA-CReSA, 08193 Barcelona, Spain
| | - Enric Vidal
- Institute of Agrifood Research and Technology (IRTA), Centre de Recerca en Sanitat Animal (CReSA), 08193 Barcelona, Spain; (J.A.); (E.V.)
- Unitat Mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), 08193 Barcelona, Spain
- WOAH Collaborating Centre for Emerging and Re-Emerging Pig Diseases in Europe, IRTA-CReSA, 08193 Barcelona, Spain
| | - María González-Cao
- Instituto Oncologico Dr. Rosell, Hospital Quiron-Dexeus Barcelona, 08028 Barcelona, Spain;
| | - Roberto Güerri-Fernandez
- Infectious Diseases Unit, Hospital del Mar, Institute of Medical Research (IMIM), 08003 Barcelona, Spain;
| | - Gennady Bocharov
- Marchuk Institute of Numerical Mathematics, Russian Academy of Sciences, 119991 Moscow, Russia;
- Institute of Computer Science and Mathematical Modeling, Sechenov First Moscow State Medical University, 119635 Moscow, Russia
| | - Andreas Meyerhans
- Infection Biology Laboratory, Department of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra, 08003 Barcelona, Spain;
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| |
Collapse
|
50
|
Peng X, Zhu X, Liu X, Huang Y, Zhu B. Increase in HIV reservoir and T cell immune response after CoronaVac vaccination in people living with HIV. Heliyon 2024; 10:e30394. [PMID: 38720759 PMCID: PMC11076980 DOI: 10.1016/j.heliyon.2024.e30394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/12/2024] Open
Abstract
Introduction CoronaVac, an inactivated vaccine developed by Sinovac Life Sciences, has been widely used for protection against Coronavirus Disease 2019 (COVID-19). This study investigates its effect on the HIV reservoir and T cell repertoires in people living with HIV (PLWHs). Methods Blood samples were collected from fifteen PLWHs who were administered at least two doses of CoronaVac between April 2021 and February 2022. The levels of cell-associated HIV RNA (CA HIV RNA) and HIV DNA, as well as the T cell receptor (TCR) repertoire profiles, TCR clustering and TCRβ annotation, were studied. Results A significant increase was observed in CA HIV RNA at 2 weeks (431.5 ± 164.2 copies/106 cells, P = 0.039) and 12 weeks (330.2 ± 105.9 copies/106 cells, P = 0.019) after the second dose, when compared to the baseline (0 weeks) (73.6 ± 23.7 copies/106 cells). Various diversity indices of the TCRβ repertoire, including Shannon index, Pielou's evenness index, and Hvj Index, revealed a slight increase (P < 0.05) following CoronaVac vaccination. The proportion of overlapping TCRβ clonotypes increased from baseline (31.9 %) to 2 weeks (32.5 %) and 12 weeks (40.4 %) after the second dose. We also found that the breadth and depth of COVID-19-specific T cells increased from baseline (0.003 and 0.0035) to 12 weeks (0.0066 and 0.0058) post the second dose. Conclusions Our study demonstrated an initial increase in HIV reservoir and TCR repertoire diversity, as well as an expansion in the depth and breadth of COVID-19-specific T-cell clones among CoronaVac-vaccinated PLWHs. These findings provide important insights into the effects of COVID-19 vaccination in PLWHs.
Collapse
Affiliation(s)
- Xiaorong Peng
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Xueling Zhu
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Xiang Liu
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Ying Huang
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Biao Zhu
- The Department of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| |
Collapse
|