1
|
Rai P, Webb EM, Paulson SL, Kang L, Weger‐Lucarelli J. Obesity's Unexpected Influence: Reduced Alphavirus Transmission and Altered Immune Activation in the Vector. J Med Virol 2024; 96:e70032. [PMID: 39466902 PMCID: PMC11600488 DOI: 10.1002/jmv.70032] [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: 05/16/2024] [Revised: 09/26/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024]
Abstract
Chikungunya virus (CHIKV) and Mayaro virus (MAYV) are emerging/re-emerging alphaviruses transmitted by Aedes spp. mosquitoes and responsible for recent disease outbreaks in the Americas. The capacity of these viruses to cause epidemics is frequently associated with increased mosquito transmission, which in turn is governed by virus-host-vector interactions. Although many studies have explored virus-vector interactions, significant gaps remain in understanding how vertebrate host factors influence alphavirus transmission by mosquitoes. We previously showed that obesity, a ubiquitous vertebrate host biological factor, reduces alphavirus transmission potential in mosquitoes. We hypothesized that alphavirus-infected obese bloodmeals altered immune genes and/or pathways in mosquitoes, thereby inhibiting virus transmission. To test this, we conducted RNA sequencing (RNA-seq) and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) on midgut RNA from mosquitoes fed on alphavirus-infected lean and obese mice. This approach aimed to identify potential antiviral or proviral genes and pathways altered in mosquitoes after consuming infected obese bloodmeals. We found upregulation of the Toll pathway and downregulation of several metabolic and other genes in mosquitoes fed on alphavirus-infected obese bloodmeals. Through gene knockdown studies, we demonstrated the antiviral role of Toll pathway and proviral roles of AAEL009965 and fatty acid synthase (FASN) in the transmission of alphaviruses by mosquitoes. Therefore, this study utilized obesity to identify factors influencing alphavirus transmission by mosquitoes and this research approach may pave the way for designing broadly effective antiviral measures to combat mosquito-borne viruses, such as releasing transgenic mosquitoes deficient in the identified genes.
Collapse
Affiliation(s)
- Pallavi Rai
- Department of Biomedical Sciences and PathobiologyVirginia TechBlacksburgVirginiaUSA
- Center for Emerging, Zoonotic, and Arthropod‐Borne PathogensVirginia TechBlacksburgVirginiaUSA
| | - Emily M. Webb
- Center for Emerging, Zoonotic, and Arthropod‐Borne PathogensVirginia TechBlacksburgVirginiaUSA
- Department of EntomologyFralin Life Sciences Institute, Virginia TechBlacksburgVirginiaUSA
| | - Sally L. Paulson
- Department of EntomologyFralin Life Sciences Institute, Virginia TechBlacksburgVirginiaUSA
| | - Lin Kang
- Department of Biomedical Sciences and PathobiologyVirginia TechBlacksburgVirginiaUSA
- Biomedical ResearchEdward Via College of Osteopathic MedicineMonroeLos AngelesUSA
- College of PharmacyUniversity of Louisiana MonroeMonroeLos AngelesUSA
| | - James Weger‐Lucarelli
- Department of Biomedical Sciences and PathobiologyVirginia TechBlacksburgVirginiaUSA
- Center for Emerging, Zoonotic, and Arthropod‐Borne PathogensVirginia TechBlacksburgVirginiaUSA
| |
Collapse
|
2
|
Hameed M, Hossain MS, Daamen AR, Lipsky PE, Weger-Lucarelli J. Granulocyte colony-stimulating factor protects against arthritogenic alphavirus pathogenesis in a type I IFN-dependent manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.09.617470. [PMID: 39416071 PMCID: PMC11482922 DOI: 10.1101/2024.10.09.617470] [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
Arthritogenic alphaviruses cause disease characterized by fever, rash, and incapacitating joint pain. Alphavirus infection stimulates robust inflammatory responses in infected hosts, leading to the upregulation of several cytokines, including granulocyte colony-stimulating factor (G-CSF). G-CSF is secreted by endothelial cells, fibroblasts, macrophages, and monocytes and binds to colony stimulating factor 3 receptor (CSF3R, also known as G-CSFR) on the surface of myeloid cells. G-CSFR signaling initiates proliferation, differentiation, and maturation of myeloid cells, especially neutrophils. Importantly, G-CSF has been found at high levels in both the acute and chronic phases of chikungunya disease; however, the role of G-CSF in arthritogenic alphavirus disease remains unexplored. Here, we sought to test the effect of G-CSF on chikungunya virus (CHIKV) and Mayaro virus (MAYV) infection using G-CSFR-deficient mice (G-CSFR-/-). We observed sustained weight loss in G-CSFR-/- mice following viand MAYV infection compared to wild-type mice. Furthermore, G-CSFR-/- mice had a significantly higher percentage of inflammatory monocytes and reduction in neutrophils throughout infection. The difference in weight loss in G-CSFR-/- mice induced by alphavirus infection was corrected by blocking type I IFN signaling. In summary, these studies show that type I IFN signaling contributes to G-CSFR mediated control of arthritogenic alphavirus disease.
Collapse
Affiliation(s)
- Muddassar Hameed
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
- Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
- Department of Pathology & Immunology, Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Md Shakhawat Hossain
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
- Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Andrea R. Daamen
- AMPEL BioSolutions LLC and the RILITE Research Institute, Charlottesville, VA, United States
| | - Peter E. Lipsky
- AMPEL BioSolutions LLC and the RILITE Research Institute, Charlottesville, VA, United States
| | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
- Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| |
Collapse
|
3
|
Hameed M, Daamen AR, Hossain MS, Coutermarsh-Ott S, Lipsky PE, Weger-Lucarelli J. Obesity-Associated Changes in Immune Cell Dynamics During Alphavirus Infection Revealed by Single Cell Transcriptomic Analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.10.617696. [PMID: 39416014 PMCID: PMC11482886 DOI: 10.1101/2024.10.10.617696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Obesity induces diverse changes in host immunity, resulting in worse disease outcomes following infection with various pathogens, including arthritogenic alphaviruses. However, the impact of obesity on the functional landscape of immune cells during arthritogenic alphavirus infection remains unexplored. Here, we used single-cell RNA sequencing (scRNA-seq) to dissect the blood and tissue immune responses to Mayaro virus (MAYV) infection in lean and obese mice. Footpad injection of MAYV caused significant shifts in immune cell populations and induced robust expression of interferon response and proinflammatory cytokine genes and related pathways in both blood and tissue. In MAYV-infected lean mice, analysis of the local tissue response revealed a unique macrophage subset with high expression of IFN response genes that was not found in obese mice. This was associated with less severe inflammation in lean mice. These results provide evidence for a unique macrophage population that may contribute to the superior capacity of lean mice to control arthritogenic alphavirus infection.
Collapse
Affiliation(s)
- Muddassar Hameed
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
- Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
- Department of Pathology & Immunology, Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Andrea R. Daamen
- AMPEL BioSolutions LLC and the RILITE Research Institute, Charlottesville, VA, United States
| | - Md Shakhawat Hossain
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
- Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Sheryl Coutermarsh-Ott
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
| | - Peter E. Lipsky
- AMPEL BioSolutions LLC and the RILITE Research Institute, Charlottesville, VA, United States
| | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
- Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| |
Collapse
|
4
|
Hameed M, Solomon NA, Weger-Lucarelli J. Lack of pathogenic involvement of CCL4 and its receptor CCR5 in arthritogenic alphavirus disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.606106. [PMID: 39131287 PMCID: PMC11312581 DOI: 10.1101/2024.07.31.606106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Arthritogenic alphaviruses, including chikungunya virus (CHIKV), Mayaro virus (MAYV), Ross River virus (RRV), and O'nyong nyong virus (ONNV) are emerging and reemerging viruses that cause disease characterized by fever, rash, and incapacitating joint swelling. Alphavirus infection induces robust immune responses in infected hosts, leading to the upregulation of several cytokines and chemokines, including chemokine C ligand 4 (CCL4). CCL4 is a chemoattractant for immune cells such as T cells, natural killer cells, monocytes/macrophages, and dendritic cells, recruiting these cells to the site of infection, stimulating the release of proinflammatory mediators, and inducing T cell differentiation. CCL4 has been found at high levels in both the acute and chronic phases of chikungunya disease; however, the role of CCL4 in arthritogenic alphavirus disease development remains unexplored. Here, we tested the effect of CCL4 on MAYV infection in mice through antibody depletion and treatment with recombinant mouse CCL4. We observed no differences in mice depleted of CCL4 or treated with recombinant CCL4 in terms of disease progression such as weight loss and footpad swelling or the development of viremia. CCL4 uses the G protein-coupled receptor C-C chemokine receptor type 5 (CCR5). To determine whether CCR5 deficiency would alter disease outcomes or virus replication in mice, we inoculated CCR5 knockout (CCR5-/-) mice with MAYV and observed no effect on disease development and immune cell profile of blood and footpads between CCR5-/- and wild type mice. These studies failed to identify a clear role for CCL4 or its receptor CCR5 in MAYV infection.
Collapse
Affiliation(s)
- Muddassar Hameed
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24060, USA
- Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| | - Norman A. Solomon
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24060, USA
- Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24060, USA
- Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
- Lead contact
| |
Collapse
|
5
|
Honce R, Vazquez-Pagan A, Livingston B, Mandarano AH, Wilander BA, Cherry S, Hargest V, Sharp B, Brigleb PH, Kirkpatrick Roubidoux E, Van de Velde LA, Skinner RC, McGargill MA, Thomas PG, Schultz-Cherry S. Diet switch pre-vaccination improves immune response and metabolic status in formerly obese mice. Nat Microbiol 2024; 9:1593-1606. [PMID: 38637722 DOI: 10.1038/s41564-024-01677-y] [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: 04/11/2022] [Accepted: 03/20/2024] [Indexed: 04/20/2024]
Abstract
Metabolic disease is epidemiologically linked to severe complications upon influenza virus infection, thus vaccination is a priority in this high-risk population. Yet, vaccine responses are less effective in these same hosts. Here we examined how the timing of diet switching from a high-fat diet to a control diet affected influenza vaccine efficacy in diet-induced obese mice. Our results demonstrate that the systemic meta-inflammation generated by high-fat diet exposure limited T cell maturation to the memory compartment at the time of vaccination, impacting the recall of effector memory T cells upon viral challenge. This was not improved with a diet switch post-vaccination. However, the metabolic dysfunction of T cells was reversed if weight loss occurred 4 weeks before vaccination, restoring a functional recall response. This corresponded with changes in the systemic obesity-related biomarkers leptin and adiponectin, highlighting the systemic and specific effects of diet on influenza vaccine immunogenicity.
Collapse
Affiliation(s)
- Rebekah Honce
- Department of Host Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
- Vermont Lung Center, Division of Pulmonology and Critical Care, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Ana Vazquez-Pagan
- Department of Host Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
- Graduate School of Biomedical Sciences, St Jude Children's Research Hospital, Memphis, TN, USA
- Weill Cornell Medicine, New York City, NY, USA
- Noguchi Medical Research Institute (NMRI), Accra, Ghana
| | - Brandi Livingston
- Department of Host Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Benjamin A Wilander
- Graduate School of Biomedical Sciences, St Jude Children's Research Hospital, Memphis, TN, USA
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Sean Cherry
- Department of Host Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Virginia Hargest
- Department of Host Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Bridgett Sharp
- Department of Host Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Pamela H Brigleb
- Department of Host Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Lee-Ann Van de Velde
- Department of Host Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
| | - R Chris Skinner
- Division of Natural Sciences and Mathematics, University of the Ozarks, Clarksville, AR, USA
- Department of Nutrition and Food Sciences, College of Agriculture and Life Sciences, University of Vermont, Burlington, VT, USA
| | - Maureen A McGargill
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Paul G Thomas
- Department of Host Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Stacey Schultz-Cherry
- Department of Host Microbe Interactions, St Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
6
|
Chuong C, Cereghino C, Rai P, Bates TA, Oberer M, Weger-Lucarelli J. Enhanced attenuation of chikungunya vaccines expressing antiviral cytokines. NPJ Vaccines 2024; 9:59. [PMID: 38472211 PMCID: PMC10933427 DOI: 10.1038/s41541-024-00843-x] [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: 01/03/2023] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Alphaviruses are vector-borne, medically relevant, positive-stranded RNA viruses that cause disease in animals and humans worldwide. Of this group, chikungunya virus (CHIKV) is the most significant human pathogen, responsible for generating millions of infections leading to severe febrile illness and debilitating chronic joint pain. Currently, there are limited treatments to protect against alphavirus disease; thus, there is a tremendous need to generate safe and effective vaccines. Live-attenuated vaccines (LAVs) are cost-effective and potent immunization strategies capable of generating long-term protection in a single dose. However, LAVs often produce systemic viral replication, which can lead to unwanted post-vaccination side effects and pose a risk of reversion to a pathogenic phenotype and transmission to mosquitoes. Here, we utilized a chimeric infectious clone of CHIKV engineered with the domain C of the E2 gene of Semliki Forest virus (SFV) to express IFNγ and IL-21-two potent antiviral and immunomodulatory cytokines-in order to improve the LAV's attenuation while maintaining immunogenicity. The IFNγ- and IL-21-expressing vaccine candidates were stable during passage and significantly attenuated post-vaccination, as mice experienced reduced footpad swelling with minimal systemic replication and dissemination capacity compared to the parental vaccine. Additionally, these candidates provided complete protection to mice challenged with WT CHIKV. Our dual attenuation strategy represents an innovative way to generate safe and effective alphavirus vaccines that could be applied to other viruses.
Collapse
Affiliation(s)
- Christina Chuong
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA, USA
| | - Chelsea Cereghino
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA, USA
| | - Pallavi Rai
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA, USA
| | - Tyler A Bates
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Megan Oberer
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA.
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
7
|
Rai P, Webb EM, Kang L, Weger-Lucarelli J. Insulin reduces the transmission potential of chikungunya virus and activates the toll pathway in Aedes aegypti mosquitoes. INSECT MOLECULAR BIOLOGY 2023; 32:648-657. [PMID: 37334906 DOI: 10.1111/imb.12863] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/08/2023] [Indexed: 06/21/2023]
Abstract
Chikungunya virus (CHIKV) is an alphavirus that has re-emerged globally over the last two decades and has the potential to become endemic in the United States due to the presence of competent mosquito vectors, Aedes aegypti and Aedes albopictus. CHIK disease is characterised by fever, rash, and joint pain, and causes chronic debilitating joint pain and swelling in >50% of infected individuals. Given the disease severity caused by CHIKV and the global presence of vectors to facilitate its spread, strategies to reduce viral transmission are desperately needed; however, the human biological factors driving CHIKV transmission are poorly understood. Towards that end, we have previously shown that mosquitoes fed on alphavirus-infected obese mice have reduced infection and transmission rates compared to those fed on infected lean mice despite similar viremia in lean and obese mice. One of the many host factors that increase in obese hosts is insulin, which was previously shown to impact the infection of mosquitoes by several flaviviruses. However, insulin's impact on alphavirus infection of live mosquitoes is unknown and whether insulin influences mosquito-borne virus transmission has not been tested. To test this, we exposed A. aegypti mosquitoes to bloodmeals with CHIKV in the presence or absence of physiologically relevant levels of insulin and found that insulin significantly lowered both infection and transmission rates. RNA sequencing analysis on mosquito midguts isolated at 1-day-post-infectious-bloodmeal (dpbm) showed enrichment in genes in the Toll immune pathway in the presence of insulin, which was validated by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). We then sought to determine if the Toll pathway plays a role in CHIKV infection of Ae. aegypti mosquitoes; therefore, we knocked down Myd88, a critical immune adaptor molecule for the Toll pathway, in live mosquitoes, and found increased CHIKV infection compared to the mock knockdown control group. Overall, these data demonstrate that insulin reduces CHIKV transmission by Ae. aegypti and activates the Toll pathway in mosquitoes, suggesting that conditions resulting in higher serum insulin concentrations may reduce alphavirus transmission. Finally, these studies suggest that strategies to activate insulin or Toll signalling in mosquitoes may be an effective control strategy against medically relevant alphaviruses.
Collapse
Affiliation(s)
- Pallavi Rai
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD College of Veterinary Medicine, Blacksburg, Virginia, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia, USA
| | - Emily M Webb
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia, USA
- Department of Entomology, Fralin Life Sciences Institute, Virginia Tech, Blacksburg, Virginia, USA
| | - Lin Kang
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD College of Veterinary Medicine, Blacksburg, Virginia, USA
- Biomedical Research, Edward Via College of Osteopathic Medicine, Monroe, Louisiana, USA
| | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD College of Veterinary Medicine, Blacksburg, Virginia, USA
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia, USA
| |
Collapse
|
8
|
Hargrave AS, Sippy R, Cueva C, Polhemus M, Beltran E, Abbott MA, Stewart-Ibarra AM. Allergies, body mass, and hospitalization due to arbovirus infection: A prospective surveillance study in Machala, Ecuador. Epidemiol Infect 2023; 151:e181. [PMID: 37823310 PMCID: PMC10644055 DOI: 10.1017/s0950268823001656] [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: 10/03/2022] [Revised: 09/04/2023] [Accepted: 10/02/2023] [Indexed: 10/13/2023] Open
Abstract
Dengue, chikungunya, and Zika are arboviruses that cause 390 million infections annually. Risk factors for hospitalization are poorly understood. Communities affected by these diseases have an escalating prevalence of allergies and obesity, which are linked to immune dysfunction. We assessed the association of allergies or body mass with hospitalization for an arbovirus infection. From 2014 to 2017, we recruited participants with a clinical diagnosis of arbovirus infection. Arbovirus infections were laboratory-confirmed and allergies were self-reported. Mid-upper arm circumference (MUAC), weight, and height were measured. We used two logistic regression models to assess the relationships between hospitalization and allergies and between hospitalization and body mass (MUAC for participants <20 years old and body mass index (BMI) for adults ≥20 years old). Models were stratified by age group and adjusted for confounders. For allergies, 41 of 265 were hospitalized. There was no association between allergies and hospitalization. For body mass, 34 of 251 were hospitalized. There was a 43% decrease in hospitalization odds for each additional centimetre MUAC among children (aOR 0.566, 95% CI 0.252-1.019) and a 12% decrease in hospitalization odds for each additional BMI unit among adults (aOR 0.877, 95% CI 0.752-0.998). Our work encourages the exploration of the underlying mechanisms.
Collapse
Affiliation(s)
- Anita S. Hargrave
- Department of Internal Medicine, University of California San Francisco, San Francisco, California, USA
- Department of Internal Medicine, San Francisco VA Health Care System, San Francisco, California, United States
| | - Rachel Sippy
- Institute for Global Health and Translational Science, SUNY Upstate Medical University, Syracuse, New York, USA
- Department of Geography, University of Florida, Gainesville, Florida, United States
- Current affiliation: Department of Psychiatry, University of Cambridge, Cambridge, United Kingdom
| | - Cinthya Cueva
- Institute for Global Health and Translational Science, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Mark Polhemus
- Institute for Global Health and Translational Science, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Efrain Beltran
- Faculdad de Medicina, Universidad Técnica de Machala, El Oro, Ecuador
| | - Mark A. Abbott
- Institute for Global Health and Translational Science, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Anna M. Stewart-Ibarra
- Department of Medicine, SUNY Upstate Medical University, Syracuse, New York, United States
- Inter-American Institute for Global Change Research, Montevideo, Uruguay
| |
Collapse
|
9
|
Nguyen TV, Ngwe Tun MM, Cao MT, Dao HM, Luong CQ, Huynh TKL, Nguyen TTT, Hoang TND, Morita K, Le TQM, Pham QD, Takamatsu Y, Hasebe F. Serological and Molecular Epidemiology of Chikungunya Virus Infection in Vietnam, 2017-2019. Viruses 2023; 15:2065. [PMID: 37896842 PMCID: PMC10611313 DOI: 10.3390/v15102065] [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: 09/10/2023] [Revised: 09/30/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
Chikungunya fever is an acute febrile illness caused by the chikungunya virus (CHIKV), which is transmitted by Aedes mosquitoes. Since 1965, only a few studies with limited scope have been conducted on CHIKV in Vietnam. Thus, this study aimed to determine the seroprevalence and molecular epidemiology of CHIKV infection among febrile patients in Vietnam from 2017 to 2019. A total of 1063 serum samples from 31 provinces were collected and tested for anti-CHIKV IgM and IgG ELISA. The 50% focus reduction neutralization test (FRNT50) was used to confirm CHIKV-neutralizing antibodies. Quantitative real-time RT-PCR (RT-qPCR) was performed to confirm the presence of the CHIKV genome. The results showed that 15.9% (169/1063) of the patients had anti-CHIKV IgM antibodies, 20.1% (214/1063) had anti-CHIKV IgG antibodies, 10.4% (111/1063) had CHIKV-neutralizing antibodies, and 27.7% (130/469) of the samples were positive in RT-qPCR analysis. The E1 CHIKV genome sequences were detected among the positive RT-qPCR samples. Our identified sequences belonged to the East/Central/South/African (ECSA) genotype, which has been prevalent in Vietnam previously, suggesting CHIKV has been maintained and is endemic in Vietnam. This study demonstrates a high prevalence of CHIKV infection in Vietnam and calls for an annual surveillance program to understand its impact.
Collapse
Affiliation(s)
- Thanh Vu Nguyen
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, Nagasaki 852-8523, Japan; (T.V.N.); (K.M.)
- Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan
- Pasteur Institute in Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam; (M.T.C.); (H.M.D.); (C.Q.L.); (T.K.L.H.); (T.T.T.N.); (T.N.D.H.); (Q.D.P.)
| | - Mya Myat Ngwe Tun
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, Nagasaki 852-8523, Japan; (T.V.N.); (K.M.)
- DEJIMA Infectious Disease Research Alliance, Nagasaki University, Nagasaki 852-8523, Japan
- Center for Vaccines and Therapeutic Antibodies for Emerging Infectious Diseases, Shimane University, Izumo 690-8504, Japan
| | - Minh Thang Cao
- Pasteur Institute in Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam; (M.T.C.); (H.M.D.); (C.Q.L.); (T.K.L.H.); (T.T.T.N.); (T.N.D.H.); (Q.D.P.)
| | - Huy Manh Dao
- Pasteur Institute in Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam; (M.T.C.); (H.M.D.); (C.Q.L.); (T.K.L.H.); (T.T.T.N.); (T.N.D.H.); (Q.D.P.)
| | - Chan Quang Luong
- Pasteur Institute in Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam; (M.T.C.); (H.M.D.); (C.Q.L.); (T.K.L.H.); (T.T.T.N.); (T.N.D.H.); (Q.D.P.)
| | - Thi Kim Loan Huynh
- Pasteur Institute in Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam; (M.T.C.); (H.M.D.); (C.Q.L.); (T.K.L.H.); (T.T.T.N.); (T.N.D.H.); (Q.D.P.)
| | - Thi Thanh Thuong Nguyen
- Pasteur Institute in Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam; (M.T.C.); (H.M.D.); (C.Q.L.); (T.K.L.H.); (T.T.T.N.); (T.N.D.H.); (Q.D.P.)
| | - Thi Nhu Dao Hoang
- Pasteur Institute in Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam; (M.T.C.); (H.M.D.); (C.Q.L.); (T.K.L.H.); (T.T.T.N.); (T.N.D.H.); (Q.D.P.)
| | - Kouichi Morita
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, Nagasaki 852-8523, Japan; (T.V.N.); (K.M.)
- DEJIMA Infectious Disease Research Alliance, Nagasaki University, Nagasaki 852-8523, Japan
| | - Thi Quynh Mai Le
- National Institute of Hygiene and Epidemiology, Hanoi 100000, Vietnam;
| | - Quang Duy Pham
- Pasteur Institute in Ho Chi Minh City, Ho Chi Minh City 700000, Vietnam; (M.T.C.); (H.M.D.); (C.Q.L.); (T.K.L.H.); (T.T.T.N.); (T.N.D.H.); (Q.D.P.)
| | - Yuki Takamatsu
- Department of Virology, Institute of Tropical Medicine, Nagasaki University, Nagasaki 852-8523, Japan; (T.V.N.); (K.M.)
- DEJIMA Infectious Disease Research Alliance, Nagasaki University, Nagasaki 852-8523, Japan
| | - Futoshi Hasebe
- Vietnam Research Station, Institute of Tropical Medicine, Nagasaki University, Nagasaki 852-8523, Japan
| |
Collapse
|
10
|
Hameed M, Rai P, Makris M, Weger-Lucarelli J. Optimized protocol for mouse footpad immune cell isolation for single-cell RNA sequencing and flow cytometry. STAR Protoc 2023; 4:102409. [PMID: 37402171 PMCID: PMC10339044 DOI: 10.1016/j.xpro.2023.102409] [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: 05/04/2023] [Revised: 06/07/2023] [Accepted: 06/07/2023] [Indexed: 07/06/2023] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) requires the preparation of a highly viable single-cell suspension to get reliable sequencing results. Here, we present a protocol for isolating mouse footpad leukocytes while maintaining high viability. We describe steps for footpad collection, enzymatic tissue dissociation, leukocyte isolation and purification, and cell fixation and preservation. We then detail combinatorial barcoding, library preparation, scRNA-seq, and data analysis. Cells can be used to generate a complete molecular atlas at the single cell level.
Collapse
Affiliation(s)
- Muddassar Hameed
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24060, USA; Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| | - Pallavi Rai
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24060, USA; Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| | - Melissa Makris
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24060, USA
| | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24060, USA; Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA.
| |
Collapse
|
11
|
Cereghino C, Roesch F, Carrau L, Hardy A, Ribeiro-Filho HV, Henrion-Lacritick A, Koh C, Marano JM, Bates TA, Rai P, Chuong C, Akter S, Vallet T, Blanc H, Elliott TJ, Brown AM, Michalak P, LeRoith T, Bloom JD, Marques RE, Saleh MC, Vignuzzi M, Weger-Lucarelli J. The E2 glycoprotein holds key residues for Mayaro virus adaptation to the urban Aedes aegypti mosquito. PLoS Pathog 2023; 19:e1010491. [PMID: 37018377 PMCID: PMC10109513 DOI: 10.1371/journal.ppat.1010491] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/17/2023] [Accepted: 03/13/2023] [Indexed: 04/06/2023] Open
Abstract
Adaptation to mosquito vectors suited for transmission in urban settings is a major driver in the emergence of arboviruses. To better anticipate future emergence events, it is crucial to assess their potential to adapt to new vector hosts. In this work, we used two different experimental evolution approaches to study the adaptation process of an emerging alphavirus, Mayaro virus (MAYV), to Ae. aegypti, an urban mosquito vector of many other arboviruses. We identified E2-T179N as a key mutation increasing MAYV replication in insect cells and enhancing transmission after escaping the midgut of live Ae. aegypti. In contrast, this mutation decreased viral replication and binding in human fibroblasts, a primary cellular target of MAYV in humans. We also showed that MAYV E2-T179N generates reduced viremia and displays less severe tissue pathology in vivo in a mouse model. We found evidence in mouse fibroblasts that MAYV E2-T179N is less dependent on the Mxra8 receptor for replication than WT MAYV. Similarly, exogenous expression of human apolipoprotein receptor 2 and Mxra8 enhanced WT MAYV replication compared to MAYV E2-T179N. When this mutation was introduced in the closely related chikungunya virus, which has caused major outbreaks globally in the past two decades, we observed increased replication in both human and insect cells, suggesting E2 position 179 is an important determinant of alphavirus host-adaptation, although in a virus-specific manner. Collectively, these results indicate that adaptation at the T179 residue in MAYV E2 may result in increased vector competence-but coming at the cost of optimal replication in humans-and may represent a first step towards a future emergence event.
Collapse
Affiliation(s)
- Chelsea Cereghino
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, United States of America
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Ferdinand Roesch
- Institut Pasteur, Viral Populations and Pathogenesis Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
- UMR 1282 ISP, INRAE Centre Val de Loire, Nouzilly, France
| | - Lucía Carrau
- Institut Pasteur, Viral Populations and Pathogenesis Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
- Department of Microbiology, New York University Langone Medical Center, New York, New York, United States of America
| | - Alexandra Hardy
- Institut Pasteur, Viral Populations and Pathogenesis Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
| | - Helder V. Ribeiro-Filho
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Brazil
| | - Annabelle Henrion-Lacritick
- Institut Pasteur, Viruses and RNA Interference Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
| | - Cassandra Koh
- Institut Pasteur, Viruses and RNA Interference Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
| | - Jeffrey M. Marano
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, United States of America
- Translational Biology, Medicine, and Health Graduate Program, Virginia Tech, Roanoke, Virginia, United States of America
| | - Tyler A. Bates
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Pallavi Rai
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Christina Chuong
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Shamima Akter
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, United States of America
- Department of Bioinformatics and Computational Biology, School of Systems Biology, George Mason University, Fairfax, Virginia, United States of America
| | - Thomas Vallet
- Institut Pasteur, Viral Populations and Pathogenesis Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
| | - Hervé Blanc
- Institut Pasteur, Viruses and RNA Interference Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
| | - Truitt J. Elliott
- Program in Genetics, Bioinformatics, and Computational Biology (GBCB), Virginia Tech, Blacksburg, Virginia, United States of America
- Research and Informatics, University Libraries, Virginia Tech, Blacksburg, Virginia, United States of America
- Department of Biochemistry, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Anne M. Brown
- Program in Genetics, Bioinformatics, and Computational Biology (GBCB), Virginia Tech, Blacksburg, Virginia, United States of America
| | - Pawel Michalak
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, United States of America
- Edward Via College of Osteopathic Medicine, Monroe, Louisiana, United States of America
- Center for One Health Research, VA-MD Regional College of Veterinary Medicine, Blacksburg, Virginia, Untied States of Ameria
- Institute of Evolution, University of Haifa, Haifa, Israel
| | - Tanya LeRoith
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Jesse D. Bloom
- Basic Sciences Division and Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Howard Hughes Medical Institute, Chevy Chase, Maryland, United States of America
| | - Rafael Elias Marques
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, São Paulo, Brazil
| | - Maria-Carla Saleh
- Institut Pasteur, Viruses and RNA Interference Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
| | - Marco Vignuzzi
- Institut Pasteur, Viral Populations and Pathogenesis Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
| | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, United States of America
- Center for Emerging, Zoonotic, and Arthropod-borne Pathogens, Virginia Tech, Blacksburg, Virginia, United States of America
- Institut Pasteur, Viral Populations and Pathogenesis Unit, Centre National de la Recherche Scientifique UMR 3569, Paris, France
| |
Collapse
|
12
|
Krokovsky L, Lins CRB, Guedes DRD, Wallau GDL, Ayres CFJ, Paiva MHS. Dynamic of Mayaro Virus Transmission in Aedes aegypti, Culex quinquefasciatus Mosquitoes, and a Mice Model. Viruses 2023; 15:v15030799. [PMID: 36992508 PMCID: PMC10053307 DOI: 10.3390/v15030799] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/08/2023] [Accepted: 03/11/2023] [Indexed: 03/31/2023] Open
Abstract
Mayaro virus (MAYV) is transmitted by Haemagogus spp. mosquitoes and has been circulating in Amazon areas in the North and Central West regions of Brazil since the 1980s, with an increase in human case notifications in the last 10 years. MAYV introduction in urban areas is a public health concern as infections can cause severe symptoms similar to other alphaviruses. Studies with Aedes aegypti have demonstrated the potential vector competence of the species and the detection of MAYV in urban populations of mosquitoes. Considering the two most abundant urban mosquito species in Brazil, we investigated the dynamics of MAYV transmission by Ae. aegypti and Culex quinquefasciatus in a mice model. Mosquito colonies were artificially fed with blood containing MAYV and infection (IR) and dissemination rates (DR) were evaluated. On the 7th day post-infection (dpi), IFNAR BL/6 mice were made available as a blood source to both mosquito species. After the appearance of clinical signs of infection, a second blood feeding was performed with a new group of non-infected mosquitoes. RT-qPCR and plaque assays were carried out with animal and mosquito tissues to determine IR and DR. For Ae. aegypti, we found an IR of 97.5-100% and a DR reached 100% in both 7 and 14 dpi. While IR and DR for Cx. quinquefasciatus was 13.1-14.81% and 60% to 80%, respectively. A total of 18 mice were used (test = 12 and control = 6) for Ae. aegypti and 12 (test = 8 and control = 4) for Cx. quinquefasciatus to evaluate the mosquito-mice transmission rate. All mice that were bitten by infected Ae. aegypti showed clinical signs of infection while all mice exposed to infected Cx. quinquefasciatus mosquitoes remained healthy. Viremia in the mice from Ae. aegypti group ranged from 2.5 × 108 to 5 × 109 PFU/mL. Ae. aegypti from the second blood feeding showed a 50% IR. Our study showed the applicability of an efficient model to complete arbovirus transmission cycle studies and suggests that the Ae. aegypti population evaluated is a competent vector for MAYV, while highlighting the vectorial capacity of Ae. aegypti and the possible introduction into urban areas. The mice model employed here is an important tool for arthropod-vector transmission studies with laboratory and field mosquito populations, as well as with other arboviruses.
Collapse
Affiliation(s)
- Larissa Krokovsky
- Departamento de Entomologia, Instituto Aggeu Magalhães, Fundação Oswaldo Cruz, Av. Professor Moraes Rego, S/N, Campus da UFPE, Cidade Universitária, Recife 50740-465, PE, Brazil
| | - Carlos Ralph Batista Lins
- Biotério de Criação, Instituto Aggeu Magalhães, Fundação Oswaldo Cruz, Av. Professor Moraes Rego, S/N, Campus da UFPE, Cidade Universitária, Recife 50740-465, PE, Brazil
| | - Duschinka Ribeiro Duarte Guedes
- Departamento de Entomologia, Instituto Aggeu Magalhães, Fundação Oswaldo Cruz, Av. Professor Moraes Rego, S/N, Campus da UFPE, Cidade Universitária, Recife 50740-465, PE, Brazil
| | - Gabriel da Luz Wallau
- Departamento de Entomologia, Instituto Aggeu Magalhães, Fundação Oswaldo Cruz, Av. Professor Moraes Rego, S/N, Campus da UFPE, Cidade Universitária, Recife 50740-465, PE, Brazil
| | - Constância Flávia Junqueira Ayres
- Departamento de Entomologia, Instituto Aggeu Magalhães, Fundação Oswaldo Cruz, Av. Professor Moraes Rego, S/N, Campus da UFPE, Cidade Universitária, Recife 50740-465, PE, Brazil
| | - Marcelo Henrique Santos Paiva
- Departamento de Entomologia, Instituto Aggeu Magalhães, Fundação Oswaldo Cruz, Av. Professor Moraes Rego, S/N, Campus da UFPE, Cidade Universitária, Recife 50740-465, PE, Brazil
- Núcleo de Ciências da Vida, Centro Acadêmico do Agreste, Universidade Federal de Pernambuco (UFPE), Rodovia BR-104, km 59-Nova Caruaru, Caruaru 55002-970, PE, Brazil
| |
Collapse
|
13
|
Zimler RA, Alto BW. Vertical Transmission of Zika Virus by Florida Aedes aegypti and Ae. albopictus. INSECTS 2023; 14:289. [PMID: 36975974 PMCID: PMC10059064 DOI: 10.3390/insects14030289] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/08/2023] [Accepted: 03/14/2023] [Indexed: 06/18/2023]
Abstract
The Zika virus pandemic of 2015, with mosquitoes Aedes aegypti and Ae. albopictus as the putative vectors, prompted public health concerns and the need to improve our understanding of both the horizontal and vertical transmission of Zika virus. Local transmission is especially concerning for Florida, where these two mosquito species are abundant and widespread throughout much of the year. Here, we evaluate the relative vertical transmission and filial infection rate of progeny of Florida Ae. aegypti and Ae. albopictus following ingestion of infected blood by parental mosquitoes at either 6 or 7 log10 plaque forming units/mL of Zika virus. Florida Ae. aegypti exhibited higher rates of disseminated infection than Ae. albopictus, consistent with other studies indicating greater permissibility of Zika virus in Ae. aegypti. We observed low vertical transmission in both Ae. aegypti (1.1-3.2%) and Ae. albopictus (0-0.3%) mosquitoes, despite imbibing infected blood at titers that yielded high susceptibility to infection and modest horizontal transmission rates. Filial infection rates, testing individual mosquitoes for Ae. aegypti and Ae. albopictus, were 6-10% and 0-6.4%, respectively. Both these invasive Stegomyia mosquitoes were capable of vertically transmitting Zika virus under laboratory conditions, and approximately 5% of female progeny of Ae. aegypti were capable of transmitting Zika virus upon first bite.
Collapse
|
14
|
Geerling E, Hameed M, Weger-Lucarelli J, Pinto AK. Metabolic syndrome and aberrant immune responses to viral infection and vaccination: Insights from small animal models. Front Immunol 2022; 13:1015563. [PMID: 36532060 PMCID: PMC9747772 DOI: 10.3389/fimmu.2022.1015563] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/08/2022] [Indexed: 12/05/2022] Open
Abstract
This review outlines the propensity for metabolic syndrome (MetS) to induce elevated disease severity, higher mortality rates post-infection, and poor vaccination outcomes for viral pathogens. MetS is a cluster of conditions including high blood glucose, an increase in circulating low-density lipoproteins and triglycerides, abdominal obesity, and elevated blood pressure which often overlap in their occurrence. MetS diagnoses are on the rise, as reported cases have increased by greater than 35% since 1988, resulting in one-third of United States adults currently diagnosed as MetS patients. In the aftermath of the 2009 H1N1 pandemic, a link between MetS and disease severity was established. Since then, numerous studies have been conducted to illuminate the impact of MetS on enhancing virally induced morbidity and dysregulation of the host immune response. These correlative studies have emphasized the need for elucidating the mechanisms by which these alterations occur, and animal studies conducted as early as the 1940s have linked the conditions associated with MetS with enhanced viral disease severity and poor vaccine outcomes. In this review, we provide an overview of the importance of considering overall metabolic health in terms of cholesterolemia, glycemia, triglyceridemia, insulin and other metabolic molecules, along with blood pressure levels and obesity when studying the impact of metabolism-related malignancies on immune function. We highlight the novel insights that small animal models have provided for MetS-associated immune dysfunction following viral infection. Such animal models of aberrant metabolism have paved the way for our current understanding of MetS and its impact on viral disease severity, dysregulated immune responses to viral pathogens, poor vaccination outcomes, and contributions to the emergence of viral variants.
Collapse
Affiliation(s)
- Elizabeth Geerling
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Muddassar Hameed
- Department of Biomedical Science and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
- Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - James Weger-Lucarelli
- Department of Biomedical Science and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
- Center for Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Amelia K. Pinto
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
15
|
Hameed M, Geerling E, Pinto AK, Miraj I, Weger-Lucarelli J. Immune response to arbovirus infection in obesity. Front Immunol 2022; 13:968582. [PMID: 36466818 PMCID: PMC9716109 DOI: 10.3389/fimmu.2022.968582] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 11/04/2022] [Indexed: 12/26/2023] Open
Abstract
Obesity is a global health problem that affects 650 million people worldwide and leads to diverse changes in host immunity. Individuals with obesity experience an increase in the size and the number of adipocytes, which function as an endocrine organ and release various adipocytokines such as leptin and adiponectin that exert wide ranging effects on other cells. In individuals with obesity, macrophages account for up to 40% of adipose tissue (AT) cells, three times more than in adipose tissue (10%) of healthy weight individuals and secrete several cytokines and chemokines such as interleukin (IL)-1β, chemokine C-C ligand (CCL)-2, IL-6, CCL5, and tumor necrosis factor (TNF)-α, leading to the development of inflammation. Overall, obesity-derived cytokines strongly affect immune responses and make patients with obesity more prone to severe symptoms than patients with a healthy weight. Several epidemiological studies reported a strong association between obesity and severe arthropod-borne virus (arbovirus) infections such as dengue virus (DENV), chikungunya virus (CHIKV), West Nile virus (WNV), and Sindbis virus (SINV). Recently, experimental investigations found that DENV, WNV, CHIKV and Mayaro virus (MAYV) infections cause worsened disease outcomes in infected diet induced obese (DIO) mice groups compared to infected healthy-weight animals. The mechanisms leading to higher susceptibility to severe infections in individuals with obesity remain unknown, though a better understanding of the causes will help scientists and clinicians develop host directed therapies to treat severe disease. In this review article, we summarize the effects of obesity on the host immune response in the context of arboviral infections. We have outlined that obesity makes the host more susceptible to infectious agents, likely by disrupting the functions of innate and adaptive immune cells. We have also discussed the immune response of DIO mouse models against some important arboviruses such as CHIKV, MAYV, DENV, and WNV. We can speculate that obesity-induced disruption of innate and adaptive immune cell function in arboviral infections ultimately affects the course of arboviral disease. Therefore, further studies are needed to explore the cellular and molecular aspects of immunity that are compromised in obesity during arboviral infections or vaccination, which will be helpful in developing specific therapeutic/prophylactic interventions to prevent immunopathology and disease progression in individuals with obesity.
Collapse
Affiliation(s)
- Muddassar Hameed
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Elizabeth Geerling
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO, United States
| | - Amelia K. Pinto
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO, United States
| | - Iqra Miraj
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
| | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
16
|
Liu Z, Ding X, Haider MS, Ali F, Yu H, Chen X, Tan S, Zu Y, Liu W, Ding B, Zheng A, Zheng J, Qian Z, Ashfaq H, Yu D, Li K. A metagenomic insight into the Yangtze finless porpoise virome. Front Vet Sci 2022; 9:922623. [PMID: 36118360 PMCID: PMC9478467 DOI: 10.3389/fvets.2022.922623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
The Yangtze finless porpoise (Neophocaena phocaenoides asiaeorientalis) inhabiting the Yantze River, China is critically endangered because of the influences of infectious disease, human activity, and water contamination. Viral diseases are one of the crucial factors that threatening the health of Yangtze finless porpoise. However, there are few studies which elaborate the viral diversity of Yangtze finless. Therefore, this study was performed to investigate the viral diversity of Yangtze finless by metagenomics. Results indicated that a total of 12,686,252 high-quality valid sequences were acquired and 2,172 virus reads were recognized. Additionally, we also obtained a total of 10,600 contigs. Phages was the most abundant virus in the samples and the ratio of DNA and RNA viruses were 69.75 and 30.25%, respectively. Arenaviridae, Ackermannviridae and Siphoviridae were the three most predominant families in all the samples. Moreover, the majority of viral genus were Mammarenavirus, Limestonevirus and Lambdavirus. The results of gene prediction indicated that these viruses play vital roles in biological process, cellular component, molecular function, and disease. To the best of our knowledge, this is the first report on the viral diversity of Yangtze finless porpoise, which filled the gaps in its viral information. Meanwhile, this study can also provide a theoretical basis for the establishment of the prevention and protection system for virus disease of Yangtze finless porpoise.
Collapse
Affiliation(s)
- Zhigang Liu
- College of Life Science, Anqing Normal University, Anqing, China
- Research Center of Aquatic Organism Conservation and Water Ecosystem Restoration in Anhui Province, Anqing Normal University, Anqing, China
- Zhigang Liu
| | - Xin Ding
- College of Life Science, Anqing Normal University, Anqing, China
| | | | - Farah Ali
- Department of Theriogenology, Faculty of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Han Yu
- College of Life Science, Anqing Normal University, Anqing, China
| | - Xin Chen
- College of Life Science, Anqing Normal University, Anqing, China
| | - Shuaishuai Tan
- College of Life Science, Anqing Normal University, Anqing, China
| | - Yuan Zu
- College of Life Science, Anqing Normal University, Anqing, China
| | - Wenlong Liu
- College of Life Science, Anqing Normal University, Anqing, China
| | - Bangzhi Ding
- College of Life Science, Anqing Normal University, Anqing, China
| | - Aifang Zheng
- College of Life Science, Anqing Normal University, Anqing, China
| | - Jinsong Zheng
- Institute of Hydrobiology, Chinese Academy of Sciences, Beijing, China
| | - Zhengyi Qian
- Hubei Yangtze River Ecological Protection Foundation, Wuhan, China
| | - Hassan Ashfaq
- Institute of Continuing Education and Extension, University of Veterinary Animal Sciences, Lahore, Pakistan
| | - Daoping Yu
- College of Life Science, Anqing Normal University, Anqing, China
- Research Center of Aquatic Organism Conservation and Water Ecosystem Restoration in Anhui Province, Anqing Normal University, Anqing, China
| | - Kun Li
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing, China
- *Correspondence: Kun Li
| |
Collapse
|
17
|
Aedes aegypti Shows Increased Susceptibility to Zika Virus via Both In Vitro and In Vivo Models of Type II Diabetes. Viruses 2022; 14:v14040665. [PMID: 35458395 PMCID: PMC9024453 DOI: 10.3390/v14040665] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/11/2022] [Accepted: 03/18/2022] [Indexed: 11/16/2022] Open
Abstract
Chronic conditions like type II diabetes (T2DM) have long been known to exacerbate many infectious diseases. For many arboviruses, including Zika virus (ZIKV), severe outcomes, morbidity and mortality usually only occur in patients with such pre-existing conditions. However, the effects of T2DM and other pre-existing conditions on human blood (e.g., hypo/hyperinsulinemia, hyperglycemia and hyperlipidemia) that may impact infectivity of arboviruses for vectors is largely unexplored. We investigated whether the susceptibility of Aedes aegypti mosquitoes was affected when the mosquitoes fed on “diabetic” bloodmeals, such as bloodmeals composed of artificially glycosylated erythrocytes or those from viremic, diabetic mice (LEPRDB/DB). Increasing glycosylation of erythrocytes from hemoglobin A1c (HgbA1c) values of 5.5–5.9 to 6.2 increased the infection rate of a Galveston, Texas strain of Ae. aegypti to ZIKV strain PRVABC59 at a bloodmeal titer of 4.14 log10 FFU/mL from 0.0 to 40.9 and 42.9%, respectively. ZIKV was present in the blood of viremic LEPRDB/DB mice at similar levels as isogenic control C57BL/6J mice (3.3 log10 FFU/mL and 3.6 log10 FFU/mL, respectively. When mice sustained a higher ZIKV viremia of 4.6 log10 FFU/mL, LEPRDB/DB mice infected 36.3% of mosquitoes while control C57BL/6J mice with a viremia of 4.2 log10 FFU/mL infected only 4.1%. Additionally, when highly susceptible Ae. aegypti Rockefeller mosquitoes fed on homozygous LEPRDB/DB, heterozygous LEPRWT/DB, and control C57BL/6J mice with viremias of ≈ 4 log10 FFU/mL, 54%, 15%, and 33% were infected, respectively. In total, these data suggest that the prevalence of T2DM in a population may have a significant impact on ZIKV transmission and indicates the need for further investigation of the impacts of pre-existing metabolic conditions on arbovirus transmission.
Collapse
|
18
|
Lucas CJ, Morrison TE. Animal models of alphavirus infection and human disease. Adv Virus Res 2022; 113:25-88. [DOI: 10.1016/bs.aivir.2022.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
|
19
|
Geerling E, Stone ET, Steffen TL, Hassert M, Brien JD, Pinto AK. Obesity Enhances Disease Severity in Female Mice Following West Nile Virus Infection. Front Immunol 2021; 12:739025. [PMID: 34531877 PMCID: PMC8439568 DOI: 10.3389/fimmu.2021.739025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/09/2021] [Indexed: 11/21/2022] Open
Abstract
A rise in adiposity in the United States has resulted in more than 70% of adults being overweight or obese, and global obesity rates have tripled since 1975. Following the 2009 H1N1 pandemic, obesity was characterized as a risk factor that could predict severe infection outcomes to viral infection. Amidst the SARS-CoV-2 pandemic, obesity has remained a significant risk factor for severe viral disease as obese patients have a higher likelihood for developing severe symptoms and requiring hospitalization. However, the mechanism by which obesity enhances viral disease is unknown. In this study, we utilized a diet-induced obesity mouse model of West Nile virus (WNV) infection, a flavivirus that cycles between birds and mosquitoes and incidentally infects both humans and mice. Likelihood for severe WNV disease is associated with risk factors such as diabetes that are comorbidities also linked to obesity. Utilizing this model, we showed that obesity-associated chronic inflammation increased viral disease severity as obese female mice displayed higher mortality rates and elevated viral titers in the central nervous system. In addition, our studies highlighted that obesity also dysregulates host acute adaptive immune responses, as obese female mice displayed significant dysfunction in neutralizing antibody function. These studies highlight that obesity-induced immunological dysfunction begins at early time points post infection and is sustained through memory phase, thus illuminating a potential for obesity to alter the differentiation landscape of adaptive immune cells.
Collapse
Affiliation(s)
- Elizabeth Geerling
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO, United States
| | - E Taylor Stone
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO, United States
| | - Tara L Steffen
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO, United States
| | - Mariah Hassert
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO, United States
| | - James D Brien
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO, United States
| | - Amelia K Pinto
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO, United States
| |
Collapse
|
20
|
Rai P, Chuong C, LeRoith T, Smyth JW, Panov J, Levi M, Kehn-Hall K, Duggal NK, Lucarelli JW. Adenovirus transduction to express human ACE2 causes obesity-specific morbidity in mice, impeding studies on the effect of host nutritional status on SARS-CoV-2 pathogenesis. Virology 2021; 563:98-106. [PMID: 34509029 PMCID: PMC8414371 DOI: 10.1016/j.virol.2021.08.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 01/08/2023]
Abstract
The COVID-19 pandemic has paralyzed the global economy and resulted in millions of deaths globally. People with co-morbidities like obesity, diabetes and hypertension are at an increased risk for severe COVID-19 illness. This is of overwhelming concern because 42% of Americans are obese, 30% are pre-diabetic and 9.4% have clinical diabetes. Here, we investigated the effect of obesity on disease severity following SARS-CoV-2 infection using a well-established mouse model of diet-induced obesity. Diet-induced obese and lean control C57BL/6 N mice, transduced for ACE2 expression using replication-defective adenovirus, were infected with SARS-CoV-2, and monitored for lung pathology, viral titers, and cytokine expression. No significant differences in tissue pathology or viral replication was observed between AdV transduced lean and obese groups, infected with SARS-CoV-2, but certain cytokines were expressed more significantly in infected obese mice compared to the lean ones. Notably, significant weight loss was observed in obese mice treated with the adenovirus vector, independent of SARS-CoV-2 infection, suggesting an obesity-dependent morbidity induced by the vector. These data indicate that the adenovirus-transduced mouse model of SARS-CoV-2 infection, as described here and elsewhere, may be inappropriate for nutrition studies.
Collapse
Affiliation(s)
- Pallavi Rai
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Christina Chuong
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Tanya LeRoith
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - James W Smyth
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, Roanoke, VA, USA; Department of Biological Sciences, College of Science, Virginia Polytechnic State Institute and State University, Blacksburg, VA, USA; Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| | - Julia Panov
- Tauber Bioinformatics Research Center, Haifa 3498838, Israel; Sagol Department of Neurobiology, University of Haifa, Haifa 3498838, Israel
| | - Moshe Levi
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, USA
| | - Kylene Kehn-Hall
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Nisha K Duggal
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - James-Weger Lucarelli
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA.
| |
Collapse
|
21
|
Sugar feeding protects against arboviral infection by enhancing gut immunity in the mosquito vector Aedes aegypti. PLoS Pathog 2021; 17:e1009870. [PMID: 34473801 PMCID: PMC8412342 DOI: 10.1371/journal.ppat.1009870] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 08/06/2021] [Indexed: 12/27/2022] Open
Abstract
As mosquito females require a blood meal to reproduce, they can act as vectors of numerous pathogens, such as arboviruses (e.g. Zika, dengue and chikungunya viruses), which constitute a substantial worldwide public health burden. In addition to blood meals, mosquito females can also take sugar meals to get carbohydrates for their energy reserves. It is now recognised that diet is a key regulator of health and disease outcome through interactions with the immune system. However, this has been mostly studied in humans and model organisms. So far, the impact of sugar feeding on mosquito immunity and in turn, how this could affect vector competence for arboviruses has not been explored. Here, we show that sugar feeding increases and maintains antiviral immunity in the digestive tract of the main arbovirus vector Aedes aegypti. Our data demonstrate that the gut microbiota does not mediate the sugar-induced immunity but partly inhibits it. Importantly, sugar intake prior to an arbovirus-infected blood meal further protects females against infection with arboviruses from different families. Sugar feeding blocks arbovirus initial infection and dissemination from the gut and lowers infection prevalence and intensity, thereby decreasing the transmission potential of female mosquitoes. Finally, we show that the antiviral role of sugar is mediated by sugar-induced immunity. Overall, our findings uncover a crucial role of sugar feeding in mosquito antiviral immunity which in turn decreases vector competence for arboviruses. Since Ae. aegypti almost exclusively feed on blood in some natural settings, our findings suggest that this lack of sugar intake could increase the spread of mosquito-borne arboviral diseases.
Collapse
|
22
|
Silverio R, Gonçalves DC, Andrade MF, Seelaender M. Coronavirus Disease 2019 (COVID-19) and Nutritional Status: The Missing Link? Adv Nutr 2021; 12:682-692. [PMID: 32975565 PMCID: PMC7543263 DOI: 10.1093/advances/nmaa125] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 08/19/2020] [Accepted: 09/09/2020] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is an emerging disease that has reached pandemic status by rapidly spreading worldwide. Elderly individuals and patients with comorbidities such as obesity, diabetes, and hypertension show a higher risk of hospitalization, severe disease, and mortality by acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. These patients frequently show exacerbated secretion of proinflammatory cytokines associated with an overreaction of the immune system, the so-called cytokine storm. Host nutritional status plays a pivotal role in the outcome of a variety of different infectious diseases. It is known that the immune system is highly affected by malnutrition, leading to decreased immune responses with consequent augmented risk of infection and disease severity. Body composition, especially low lean mass and high adiposity, has consistently been linked to worsened prognosis in many different diseases. In this review, evidence concerning the impact of nutritional status on viral infection outcomes is discussed.
Collapse
Affiliation(s)
- Renata Silverio
- Cancer Metabolism Research Group, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Multicenter Graduate Program in Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Daniela Caetano Gonçalves
- Cancer Metabolism Research Group, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Biosciences Department, Universidade Federal de São Paulo (UNIFESP), Campus Baixada Santista, Santos, Brazil
| | - Márcia Fábia Andrade
- Cancer Metabolism Research Group, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Marilia Seelaender
- Cancer Metabolism Research Group, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Department of Clinical Surgery, LIM 26-HC, Faculdade de Medicina, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
23
|
Louca P, Murray B, Klaser K, Graham MS, Mazidi M, Leeming ER, Thompson E, Bowyer R, Drew DA, Nguyen LH, Merino J, Gomez M, Mompeo O, Costeira R, Sudre CH, Gibson R, Steves CJ, Wolf J, Franks PW, Ourselin S, Chan AT, Berry SE, Valdes AM, Calder PC, Spector TD, Menni C. Modest effects of dietary supplements during the COVID-19 pandemic: insights from 445 850 users of the COVID-19 Symptom Study app. BMJ Nutr Prev Health 2021; 4:149-157. [PMID: 34308122 PMCID: PMC8061565 DOI: 10.1136/bmjnph-2021-000250] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES Dietary supplements may ameliorate SARS-CoV-2 infection, although scientific evidence to support such a role is lacking. We investigated whether users of the COVID-19 Symptom Study app who regularly took dietary supplements were less likely to test positive for SARS-CoV-2 infection. DESIGN App-based community survey. SETTING 445 850 subscribers of an app that was launched to enable self-reported information related to SARS-CoV-2 infection for use in the general population in the UK (n=372 720), the USA (n=45 757) and Sweden (n=27 373). MAIN EXPOSURE Self-reported regular dietary supplement usage (constant use during previous 3 months) in the first waves of the pandemic up to 31 July 2020. MAIN OUTCOME MEASURES SARS-CoV-2 infection confirmed by viral RNA reverse transcriptase PCR test or serology test before 31 July 2020. RESULTS In 372 720 UK participants (175 652 supplement users and 197 068 non-users), those taking probiotics, omega-3 fatty acids, multivitamins or vitamin D had a lower risk of SARS-CoV-2 infection by 14% (95% CI (8% to 19%)), 12% (95% CI (8% to 16%)), 13% (95% CI (10% to 16%)) and 9% (95% CI (6% to 12%)), respectively, after adjusting for potential confounders. No effect was observed for those taking vitamin C, zinc or garlic supplements. On stratification by sex, age and body mass index (BMI), the protective associations in individuals taking probiotics, omega-3 fatty acids, multivitamins and vitamin D were observed in females across all ages and BMI groups, but were not seen in men. The same overall pattern of association was observed in both the US and Swedish cohorts. CONCLUSION In women, we observed a modest but significant association between use of probiotics, omega-3 fatty acid, multivitamin or vitamin D supplements and lower risk of testing positive for SARS-CoV-2. We found no clear benefits for men nor any effect of vitamin C, garlic or zinc. Randomised controlled trials are required to confirm these observational findings before any therapeutic recommendations can be made.
Collapse
Affiliation(s)
- Panayiotis Louca
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Benjamin Murray
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK
| | - Kerstin Klaser
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK
| | - Mark S Graham
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK
| | - Mohsen Mazidi
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Emily R Leeming
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Ellen Thompson
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Ruth Bowyer
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - David A Drew
- Clinical & Translational Epidemiology Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Long H Nguyen
- Clinical & Translational Epidemiology Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jordi Merino
- Clinical & Translational Epidemiology Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Maria Gomez
- Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Olatz Mompeo
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Ricardo Costeira
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Carole H Sudre
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London, UK
| | - Rachel Gibson
- Department of Nutritional Sciences, King's College London, London, UK
| | - Claire J Steves
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | | | - Paul W Franks
- Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Sebastien Ourselin
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK
| | - Andrew T Chan
- Clinical & Translational Epidemiology Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sarah E Berry
- Department of Nutritional Sciences, King's College London, London, UK
| | - Ana M Valdes
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- Division of Rheumatology, Orthopaedics and Dermatology, School of Medicine, University of Nottingham, Nottingham, Nottinghamshire, UK
| | - Philip C Calder
- Human Development & Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| |
Collapse
|
24
|
Umeoguaju FU, Ephraim-Emmanuel BC, Patrick-Iwuanyanwu KC, Zelikoff JT, Orisakwe OE. Plant-Derived Food Grade Substances (PDFGS) Active Against Respiratory Viruses: A Systematic Review of Non-clinical Studies. Front Nutr 2021; 8:606782. [PMID: 33634160 PMCID: PMC7900554 DOI: 10.3389/fnut.2021.606782] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 01/15/2021] [Indexed: 12/11/2022] Open
Abstract
Human diet comprises several classes of phytochemicals some of which are potentially active against human pathogenic viruses. This study examined available evidence that identifies existing food plants or constituents of edible foods that have been reported to inhibit viral pathogenesis of the human respiratory tract. SCOPUS and PUBMED databases were searched with keywords designed to retrieve articles that investigated the effect of plant-derived food grade substances (PDFGS) on the activities of human pathogenic viruses. Eligible studies for this review were those done on viruses that infect the human respiratory tract. Forty six (46) studies met the specified inclusion criteria from the initial 5,734 hits. The selected studies investigated the effects of different PDFGS on the infectivity, proliferation and cytotoxicity of different respiratory viruses including influenza A virus (IAV), influenza B virus (IBV), Respiratory syncytial virus (RSV), human parainfluenza virus (hPIV), Human coronavirus NL63 (HCoV-NL63), and rhinovirus (RV) in cell lines and mouse models. This review reveals that PDFGS inhibits different stages of the pathological pathways of respiratory viruses including cell entry, replication, viral release and viral-induced dysregulation of cellular homeostasis and functions. These alterations eventually lead to the reduction of virus titer, viral-induced cellular damages and improved survival of host cells. Major food constituents active against respiratory viruses include flavonoids, phenolic acids, tannins, lectins, vitamin D, curcumin, and plant glycosides such as glycyrrhizin, acteoside, geniposide, and iridoid glycosides. Herbal teas such as guava tea, green and black tea, adlay tea, cistanche tea, kuding tea, licorice extracts, and edible bird nest extracts were also effective against respiratory viruses in vitro. The authors of this review recommend an increased consumption of foods rich in these PDFGS including legumes, fruits (e.g berries, citrus), tea, fatty fish and curcumin amongst human populations with high prevalence of respiratory viral infections in order to prevent, manage and/or reduce the severity of respiratory virus infections.
Collapse
Affiliation(s)
- Francis U. Umeoguaju
- World Bank Africa Centre of Excellence in Public Health and Toxicological Research (ACE-PUTOR), University of Port Harcourt, Port Harcourt, Nigeria
| | - Benson C. Ephraim-Emmanuel
- World Bank Africa Centre of Excellence in Public Health and Toxicological Research (ACE-PUTOR), University of Port Harcourt, Port Harcourt, Nigeria
- Department of Dental Health Sciences, Ogbia, Bayelsa State College of Health Technology, Otakeme, Nigeria
| | - Kingsley C. Patrick-Iwuanyanwu
- World Bank Africa Centre of Excellence in Public Health and Toxicological Research (ACE-PUTOR), University of Port Harcourt, Port Harcourt, Nigeria
| | - Judith T. Zelikoff
- Department of Environmental Medicine, New York University Grossman School of Medicine, New York, NY, United States
| | - Orish Ebere Orisakwe
- World Bank Africa Centre of Excellence in Public Health and Toxicological Research (ACE-PUTOR), University of Port Harcourt, Port Harcourt, Nigeria
- Department of Experimental Pharmacology and Toxicology, Faculty of Pharmacy, University of Port Harcourt, Port Harcourt, Nigeria
| |
Collapse
|
25
|
Wang ZH, Lin YW, Wei XB, Li F, Liao XL, Yuan HQ, Huang DZ, Qin TH, Geng H, Wang SH. Predictive Value of Prognostic Nutritional Index on COVID-19 Severity. Front Nutr 2021; 7:582736. [PMID: 33521032 PMCID: PMC7840577 DOI: 10.3389/fnut.2020.582736] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 12/17/2020] [Indexed: 02/05/2023] Open
Abstract
Background: The prognostic nutritional index (PNI) has been described as a simple risk-stratified tool for several diseases. We explored the predictive role of the PNI on coronavirus disease 2019 (COVID-19) severity. Methods: A total of 101 patients with COVID-19 were included in this retrospective study from January 2020 to March 2020. They were divided into two groups according to COVID-19 severity: non-critical (n = 56) and critical (n = 45). The PNI was calculated upon hospital admission: 10 × serum albumin (g/dL) + 0.005 × total lymphocyte count (/mm3). Critical COVID-19 was defined as having one of the following features: respiratory failure necessitating mechanical ventilation; shock; organ dysfunction necessitating admission to the intensive care unit (ICU). The correlation between the PNI with COVID-19 severity was analyzed. Results: The PNI was significantly lower in critically ill than that in non-critically ill patients (P < 0.001). The receiver operating characteristic curve indicated that the PNI was a good discrimination factor for identifying COVID-19 severity (P < 0.001). Multivariate logistic regression analysis showed the PNI to be an independent risk factor for critical illness due to COVID-19 (P = 0.002). Conclusions: The PNI is a valuable biomarker that could be used to discriminate COVID-19 severity.
Collapse
Affiliation(s)
- Zhong-hua Wang
- Department of Critical Care Medicine, Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ying-Wen Lin
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Xue-biao Wei
- Department of Critical Care Medicine, Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Fei Li
- Emergency Department, The First People's Hospital of Jingzhou, Jingzhou, China
| | - Xiao-Long Liao
- Department of Critical Care Medicine, Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hui-qing Yuan
- Department of Respiratory and Critical Care Medicine, The First People's Hospital of Shaoguan, Shaoguan, China
| | - Dao-zheng Huang
- Department of Critical Care Medicine, Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Tie-he Qin
- Department of Critical Care Medicine, Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Heng Geng
- Department of Critical Care Medicine, The First People's Hospital of Jingzhou, Jingzhou, China
| | - Shou-hong Wang
- Department of Critical Care Medicine, Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
26
|
Fedele D, De Francesco A, Riso S, Collo A. Obesity, malnutrition, and trace element deficiency in the coronavirus disease (COVID-19) pandemic: An overview. Nutrition 2021; 81:111016. [PMID: 33059127 PMCID: PMC7832575 DOI: 10.1016/j.nut.2020.111016] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/24/2020] [Accepted: 08/29/2020] [Indexed: 02/06/2023]
Abstract
The world is currently facing the coronavirus disease (COVID-19) pandemic which places great pressure on health care systems and workers, often presents with severe clinical features, and sometimes requires admission into intensive care units. Derangements in nutritional status, both for obesity and malnutrition, are relevant for the clinical outcome in acute illness. Systemic inflammation, immune system impairment, sarcopenia, and preexisting associated conditions, such as respiratory, cardiovascular, and metabolic diseases related to obesity, could act as crucial factors linking nutritional status and the course and outcome of COVID-19. Nevertheless, vitamins and trace elements play an essential role in modulating immune response and inflammatory status. Overall, evaluation of the patient's nutritional status is not negligible for its implications on susceptibility, course, severity, and responsiveness to therapies, in order to perform a tailored nutritional intervention as an integral part of the treatment of patients with COVID-19. The aim of this study was to review the current data on the relevance of nutritional status, including trace elements and vitamin status, in influencing the course and outcome of the disease 3 mo after the World Health Organization's declaration of COVID-19 as a pandemic.
Collapse
Affiliation(s)
- Debora Fedele
- Dietetic and Clinical Nutrition Unit, San Giovanni Battista Hospital, Città della Salute e della Scienza, Turin, Italy.
| | - Antonella De Francesco
- Dietetic and Clinical Nutrition Unit, San Giovanni Battista Hospital, Città della Salute e della Scienza, Turin, Italy
| | - Sergio Riso
- Dietetic and Clinical Nutrition Unit, Maggiore della Carità Hospital, Novara, Italy
| | - Alessandro Collo
- Dietetic and Clinical Nutrition Unit, Maggiore della Carità Hospital, Novara, Italy
| |
Collapse
|
27
|
Bates TA, Chuong C, Hawks SA, Rai P, Duggal NK, Weger-Lucarelli J. Development and characterization of infectious clones of two strains of Usutu virus. Virology 2020; 554:28-36. [PMID: 33352463 DOI: 10.1016/j.virol.2020.12.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/02/2020] [Accepted: 12/07/2020] [Indexed: 12/23/2022]
Abstract
Usutu virus (USUV; genus Flavivirus; family Flaviviridae) is a mosquito-borne, positive-sense RNA virus that is currently causing significant die-offs in numerous bird species throughout Europe and has caused infections in humans. Currently, there are no molecular clones for USUV, hence, hindering studies on the pathogenesis and transmission of USUV. Here, we demonstrate the development and characterization of infectious clones for two modern strains of USUV isolated from Europe and Africa. We show that the infectious clone-derived viruses replicated similarly to the parental strains in mammalian and insect cells. Additionally, we observed similar levels of replication and disease in two mouse models. These clones will aid the study of USUV infection, transmission, diagnostics, and vaccines.
Collapse
Affiliation(s)
- Tyler A Bates
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Christina Chuong
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Seth A Hawks
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Pallavi Rai
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - Nisha K Duggal
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA
| | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, VA-MD Regional College of Veterinary Medicine, Blacksburg, VA, USA.
| |
Collapse
|
28
|
Vodnar DC, Mitrea L, Teleky BE, Szabo K, Călinoiu LF, Nemeş SA, Martău GA. Coronavirus Disease (COVID-19) Caused by (SARS-CoV-2) Infections: A Real Challenge for Human Gut Microbiota. Front Cell Infect Microbiol 2020; 10:575559. [PMID: 33363049 PMCID: PMC7756003 DOI: 10.3389/fcimb.2020.575559] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 11/16/2020] [Indexed: 01/08/2023] Open
Abstract
The current COVID-19 pandemic is a great challenge for worldwide researchers in the human microbiota area because the mechanisms and long-term effects of the infection at the GI level are not yet deeply understood. In the current review, scientific literature including original research articles, clinical studies, epidemiological reports, and review-type articles concerning human intestinal infection with SARS-CoV-2 and the possible consequences on the microbiota were reviewed. Moreover, the following aspects pertaining to COVID-19 have also been discussed: transmission, resistance in the human body, the impact of nutritional status in relation to the intestinal microbiota, and the impact of comorbid metabolic disorders such as inflammatory bowel disease (IBS), obesity, and type two diabetes (T2D). The articles investigated show that health, age, and nutritional status are associated with specific communities of bacterial species in the gut, which could influence the clinical course of COVID-19 infection. Fecal microbiota alterations were associated with fecal concentrations of SARS-CoV-2 and COVID-19 severity. Patients suffering from metabolic and gastrointestinal (GI) disorders are thought to be at a moderate-to-high risk of infection with SARS-CoV-2, indicating the direct implication of gut dysbiosis in COVID-19 severity. However, additional efforts are required to identify the initial GI symptoms of COVID-19 for possible early intervention.
Collapse
Affiliation(s)
- Dan-Cristian Vodnar
- Institute of Life Sciences, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania
- Faculty of Food Science and Technology, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania
| | - Laura Mitrea
- Institute of Life Sciences, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania
| | - Bernadette-Emoke Teleky
- Institute of Life Sciences, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania
| | - Katalin Szabo
- Institute of Life Sciences, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania
| | - Lavinia-Florina Călinoiu
- Institute of Life Sciences, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania
| | - Silvia-Amalia Nemeş
- Institute of Life Sciences, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania
- Faculty of Food Science and Technology, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania
| | - Gheorghe-Adrian Martău
- Institute of Life Sciences, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania
- Faculty of Food Science and Technology, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania
| |
Collapse
|
29
|
Chuong C, Bates TA, Akter S, Werre SR, LeRoith T, Weger-Lucarelli J. Nutritional status impacts dengue virus infection in mice. BMC Biol 2020; 18:106. [PMID: 32854687 PMCID: PMC7453574 DOI: 10.1186/s12915-020-00828-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 07/11/2020] [Indexed: 01/09/2023] Open
Abstract
Background Dengue virus (DENV) is estimated to infect 390 million people annually. However, few host factors that alter disease severity are known. Malnutrition, defined as both over- and undernutrition, is a growing problem worldwide and has long been linked to dengue disease severity by epidemiological and anecdotal observations. Accordingly, we sought to establish a mouse model to assess the impact of nutritional status on DENV disease severity. Results Using transiently immunocompromised mice, we established a model of mild dengue disease with measurable viremia. We then applied it to study the effects of healthy weight, obese, and low-protein diets representing normal, over-, and undernutrition, respectively. Upon infection with DENV serotype 2, obese mice experienced more severe morbidity in the form of weight loss and thrombocytopenia compared to healthy weight groups. Additionally, obesity altered cytokine expression following DENV infection. Although low protein-fed mice did not lose significant weight after DENV2 infection, they also experienced a reduction in platelets as well as increased spleen pathology and viral titers. Conclusions Our results indicate that obese or undernourished mice incur greater disease severity after DENV infection. These studies establish a role for nutritional status in DENV disease severity.
Collapse
Affiliation(s)
- Christina Chuong
- Department of Biomedical Sciences and Pathobiology, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA
| | - Tyler A Bates
- Department of Biomedical Sciences and Pathobiology, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA
| | - Shamima Akter
- Department of Biomedical Sciences and Pathobiology, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA.,Present Address: Department of Bioinformatics and Computational Biology, School of Systems Biology, George Mason University, Fairfax, VA, USA
| | - Stephen R Werre
- Department of Population Health Sciences, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA
| | - Tanya LeRoith
- Department of Biomedical Sciences and Pathobiology, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA
| | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, VA-MD College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
30
|
They are what you eat: Shaping of viral populations through nutrition and consequences for virulence. PLoS Pathog 2020; 16:e1008711. [PMID: 32790755 PMCID: PMC7425860 DOI: 10.1371/journal.ppat.1008711] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
31
|
Vidal OM, Acosta-Reyes J, Padilla J, Navarro-Lechuga E, Bravo E, Viasus D, Arcos-Burgos M, Vélez JI. Chikungunya outbreak (2015) in the Colombian Caribbean: Latent classes and gender differences in virus infection. PLoS Negl Trop Dis 2020; 14:e0008281. [PMID: 32492017 PMCID: PMC7304630 DOI: 10.1371/journal.pntd.0008281] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 06/19/2020] [Accepted: 04/08/2020] [Indexed: 12/26/2022] Open
Abstract
Chikungunya virus (CHIKV), a mosquito-borne alphavirus of the Togaviridae family, is part of a group of emergent diseases, including arbovirus, constituting an increasing public health problem in tropical areas worldwide. CHIKV causes a severe and debilitating disease with high morbidity. The first Colombian autochthonous case was reported in the Colombian Caribbean region in September 2014. Within the next two to three months, the CHIKV outbreak reached its peak. Although the CHIKV pattern of clinical symptomatology has been documented in different epidemiological studies, understanding of the relationship between clinical symptomatology and variation in phenotypic response to CHIKV infection in humans remains limited. We performed a cross sectional study following 1160 individuals clinically diagnosed with CHIKV at the peak of the Chikungunya outbreak in the Colombian Caribbean region. We examined the relationship between symptomatology and diverse phenotypic responses. Latent Class Cluster Analysis (LCCA) models were used to characterize patients’ symptomatology and further identify subgroups of individuals with differential phenotypic response. We found that most individuals presented fever (94.4%), headache (73.28%) and general discomfort (59.4%), which are distinct clinical symptoms of a viral infection. Furthermore, 11/26 (43.2%) of the categorized symptoms were more frequent in women than in men. LCCA disclosed seven distinctive phenotypic response profiles in this population of CHIKV infected individuals. Interestingly, 282 (24.3%) individuals exhibited a lower symptomatic “extreme” phenotype and 74 (6.4%) patients were within the severe complex “extreme” phenotype. Although clinical symptomatology may be diverse, there are distinct symptoms or group of symptoms that can be correlated with differential phenotypic response and perhaps susceptibility to CHIKV infection, especially in the female population. This suggests that, comparatively to men, women are a CHIKV at-risk population. Further study is needed to validate these results and determine whether the distinct LCCA profiles are a result of the immune response or a mixture of genetic, lifestyle and environmental factors. Our findings could contribute to the development of machine learning approaches to characterizing CHIKV infection in other populations. Preliminary results have shown prediction models achieving up to 92% accuracy overall, with substantial sensitivity, specificity and accuracy values per LCCA-derived cluster. The Chikungunya virus (CHIKV) infection is a mosquito-borne virus of the Togaviridae family, part of the arbovirus group of mosquito-transmitted pathogens. CHIKV causes a severe and debilitating disease with high morbidity. In this study, we comprehensively analysed clinical data from 1160 individuals from the Colombian Caribbean, who were diagnosed with CHIKV infection during the 2014 epidemic peak and before the Zika epidemic (registered back in 2015). Further, the presence of latent classes and predictors of CHIKV susceptibility and severity of the CHIKV infection were analysed. Although it is well known that people respond differently to infection, our results showed that these differences are not arbitrary and may come from the specific orchestration of our immune response and specific genetic makeup. For example, we identified that females infected with CHIKV exhibited significant and heterogeneous phenotypic response patterns compared to men. Overall, these results inform about potential predictors and outlining strategies to study the natural history of CHIKV infection. Future studies assessing the contribution of demographic, immunological and genetic factors to symptom co-occurrence could shed some light on the severity of the clinical symptomatology and, ultimately, lead to more accurate, more efficient and differential diagnosis. These results could contribute to the development of machine learning approaches to characterizing CHIKV infection in other populations and provide more accurate and differential diagnosis.
Collapse
Affiliation(s)
- Oscar M. Vidal
- Universidad del Norte, Barranquilla, Colombia
- * E-mail: (OMV); (JIV)
| | | | | | | | - Elsa Bravo
- Epidemiological Surveillance Team, Health Secretary Program, Barranquilla, Colombia
| | | | - Mauricio Arcos-Burgos
- Grupo de Investigación en Psiquiatría (GIPSI), Departamento de Psiquiatría, Instituto de Investigaciones Médicas, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Jorge I. Vélez
- Universidad del Norte, Barranquilla, Colombia
- * E-mail: (OMV); (JIV)
| |
Collapse
|
32
|
Caccialanza R, Laviano A, Lobascio F, Montagna E, Bruno R, Ludovisi S, Corsico AG, Di Sabatino A, Belliato M, Calvi M, Iacona I, Grugnetti G, Bonadeo E, Muzzi A, Cereda E. Early nutritional supplementation in non-critically ill patients hospitalized for the 2019 novel coronavirus disease (COVID-19): Rationale and feasibility of a shared pragmatic protocol. Nutrition 2020; 74:110835. [PMID: 32280058 PMCID: PMC7194616 DOI: 10.1016/j.nut.2020.110835] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Beginning in December 2019, the 2019 novel coronavirus disease (COVID-19) has caused a pneumonia epidemic that began in Wuhan, China, and is rapidly spreading throughout the whole world. Italy is the hardest hit country after China. Considering the deleterious consequences of malnutrition, which certainly can affect patients with COVID-19, the aim of this article is to present a pragmatic protocol for early nutritional supplementation of non-critically ill patients hospitalized for COVID-19 disease. It is based on the observation that most patients present at admission with severe inflammation and anorexia leading to a drastic reduction of food intake, and that a substantial percentage develops respiratory failure requiring non-invasive ventilation or even continuous positive airway pressure. METHODS High-calorie dense diets in a variety of different consistencies with highly digestible foods and snacks are available for all patients. Oral supplementation of whey proteins as well as intravenous infusion of multivitamin, multimineral trace elements solutions are implemented at admission. In the presence of 25-hydroxyvitamin D deficit, cholecalciferol is promptly supplied. If nutritional risk is detected, two to three bottles of protein-calorie oral nutritional supplements (ONS) are provided. If <2 bottles/d of ONS are consumed for 2 consecutive days and/or respiratory conditions are worsening, supplemental/total parenteral nutrition is prescribed. CONCLUSION We are aware that our straight approach may be debatable. However, to cope with the current emergency crisis, its aim is to promptly and pragmatically implement nutritional care in patients with COVID-19, which might be overlooked despite being potentially beneficial to clinical outcomes and effective in preventing the consequences of malnutrition in this patient population.
Collapse
Affiliation(s)
- Riccardo Caccialanza
- Clinical Nutrition and Dietetics Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy.
| | - Alessandro Laviano
- Department of Translational and Precision Medicine, La Sapienza University, Rome, Italy
| | - Federica Lobascio
- Clinical Nutrition and Dietetics Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Elisabetta Montagna
- Clinical Nutrition and Dietetics Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Raffaele Bruno
- Division of Infectious Diseases Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Department of Medical, Surgical, Diagnostic and Paediatric Science, University of Pavia, Pavia, Italy
| | - Serena Ludovisi
- Division of Infectious Diseases Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Department of Medical, Surgical, Diagnostic and Paediatric Science, University of Pavia, Pavia, Italy
| | - Angelo Guido Corsico
- Pneumology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - Antonio Di Sabatino
- First Department of Internal Medicine, Fondazione IRCCS Policlinico San Matteo and University of Pavia, Pavia, Italy
| | - Mirko Belliato
- UOS Advanced Respiratory Intensive Care Unit, UOC Anestesia e Rianimazione 1, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Monica Calvi
- Pharmacy Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Isabella Iacona
- Pharmacy Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Giuseppina Grugnetti
- Nursing Technical and Rehabilitation Service, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Elisa Bonadeo
- UOC Direzione Medica di Presidio, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Alba Muzzi
- UOC Direzione Medica di Presidio, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Emanuele Cereda
- Clinical Nutrition and Dietetics Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
33
|
Briguglio M, Pregliasco FE, Lombardi G, Perazzo P, Banfi G. The Malnutritional Status of the Host as a Virulence Factor for New Coronavirus SARS-CoV-2. Front Med (Lausanne) 2020; 7:146. [PMID: 32391367 PMCID: PMC7191079 DOI: 10.3389/fmed.2020.00146] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 04/03/2020] [Indexed: 01/08/2023] Open
Affiliation(s)
- Matteo Briguglio
- IRCCS Orthopedic Institute Galeazzi, Scientific Direction, Milan, Italy
| | - Fabrizio Ernesto Pregliasco
- Health Management, IRCCS Orthopedic Institute Galeazzi, Milan, Italy
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Orthopedic Institute Galeazzi, Milan, Italy
- Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Poland
| | - Paolo Perazzo
- Post-operative Intensive Care Unit & Anesthesia, IRCCS Orthopedic Institute Galeazzi, Milan, Italy
| | - Giuseppe Banfi
- IRCCS Orthopedic Institute Galeazzi, Scientific Direction, Milan, Italy
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|