1
|
McFadden E, Monticelli SR, Wang A, Ramamohan AR, Batchelor TG, Kuehne AI, Bakken RR, Tse AL, Chandran K, Herbert AS, McLellan JS. Engineering and structures of Crimean-Congo hemorrhagic fever virus glycoprotein complexes. Cell 2025; 188:303-315.e13. [PMID: 39701101 PMCID: PMC11761392 DOI: 10.1016/j.cell.2024.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 08/30/2024] [Accepted: 11/08/2024] [Indexed: 12/21/2024]
Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV) is a tickborne virus that can cause severe disease in humans with case fatality rates of 10%-40%. Although structures of CCHFV glycoproteins GP38 and Gc have provided insights into viral entry and defined epitopes of neutralizing and protective antibodies, the structure of glycoprotein Gn and its interactions with GP38 and Gc have remained elusive. Here, we use structure-guided protein engineering to produce a stabilized GP38-Gn-Gc heterotrimeric glycoprotein complex (GP38-GnH-DS-Gc). A cryo-electron microscopy (cryo-EM) structure of this complex provides the molecular basis for GP38's association on the viral surface, reveals the structure of Gn, and demonstrates that GP38-Gn restrains the Gc fusion loops in the prefusion conformation, facilitated by an N-linked glycan attached to Gn. Immunization with GP38-GnH-DS-Gc conferred 40% protection against lethal IbAr10200 challenge in mice. These data define the architecture of a GP38-Gn-Gc protomer and provide a template for structure-guided vaccine antigen development.
Collapse
Affiliation(s)
- Elizabeth McFadden
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Stephanie R Monticelli
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA; The Geneva Foundation, Tacoma, WA 98402, USA
| | - Albert Wang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ajit R Ramamohan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Thomas G Batchelor
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA; Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, USA
| | - Ana I Kuehne
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Russell R Bakken
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Alexandra L Tse
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Andrew S Herbert
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Jason S McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
2
|
Rissmann M, Noack D, Spliethof TM, Vaes VP, Stam R, van Run P, Clark JJ, Verjans GMGM, Haagmans BL, Krammer F, Koopmans MPG, van den Brand JMA, Rockx B. A pan-orthohantavirus human lung xenograft mouse model and its utility for preclinical studies. PLoS Pathog 2025; 21:e1012875. [PMID: 39841788 PMCID: PMC11774489 DOI: 10.1371/journal.ppat.1012875] [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: 08/06/2024] [Revised: 01/28/2025] [Accepted: 01/01/2025] [Indexed: 01/24/2025] Open
Abstract
Orthohantaviruses are emerging zoonotic viruses that can infect humans via the respiratory tract. There is an unmet need for an in vivo model to study infection of different orthohantaviruses in physiologically relevant tissue and to assess the efficacy of novel pan-orthohantavirus countermeasures. Here, we describe the use of a human lung xenograft mouse model to study the permissiveness for different orthohantavirus species and to assess its utility for preclinical testing of therapeutics. Following infection of xenografted human lung tissues, distinct orthohantavirus species differentially replicated in the human lung and subsequently spread systemically. The different orthohantaviruses primarily targeted the endothelium, respiratory epithelium and macrophages in the human lung. A proof-of-concept preclinical study showed treatment of these mice with a virus neutralizing antibody could block Andes orthohantavirus infection and dissemination. This pan-orthohantavirus model will facilitate progress in the fundamental understanding of pathogenesis and virus-host interactions for orthohantaviruses. Furthermore, it is an invaluable tool for preclinical evaluation of novel candidate pan-orthohantavirus intervention strategies.
Collapse
Affiliation(s)
- Melanie Rissmann
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Danny Noack
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Thomas M. Spliethof
- Division of Pathology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Vincent P. Vaes
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Rianne Stam
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Peter van Run
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jordan J. Clark
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | | | - Bart L. Haagmans
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Marion P. G. Koopmans
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Barry Rockx
- Department of Viroscience, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
3
|
Goens MM, Howard EL, Warner BM, Susta L, Wootton SK. Rapid Development of Small Rodent Animal Models for Infectious Disease Research Through Vectorized Receptor Molecule Expression. Viruses 2024; 16:1794. [PMID: 39599908 PMCID: PMC11599079 DOI: 10.3390/v16111794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/09/2024] [Accepted: 11/16/2024] [Indexed: 11/29/2024] Open
Abstract
The emergence and re-emergence of pathogens with pandemic potential has been a persistent issue throughout history. Recent decades have seen significant outbreaks of zoonotic viruses from members of the Coronaviridae, Filoviridae, Paramyxoviridae, Flaviviridae, and Togaviridae families, resulting in widespread infections. The continual emergence of zoonotic viral pathogens and associated infections highlights the need for prevention strategies and effective treatments. Central to this effort is the availability of suitable animal models, which are essential for understanding pathogenesis and assessing transmission dynamics. These animals are also critical for evaluating the safety and efficacy of novel vaccines or therapeutics and are essential in facilitating regulatory approval of new products. Rapid development of animal models is an integral aspect of pandemic response and preparedness; however, their establishment is fraught by several rate-limiting steps, including selection of a suitable species, the logistical challenges associated with sharing and disseminating transgenic animals (e.g., the time-intensive nature of breeding and maintaining colonies), the availability of technical expertise, as well as ethical and regulatory approvals. A method for the rapid development of relevant animal models that has recently gained traction, in large part due to the COVID-19 pandemic, is the use of gene therapy vectors to express human viral receptors in readily accessible laboratory animals to enable virus infection and development of clinical disease. These models can be developed rapidly on any genetic background, making mechanistic studies and accelerated evaluation of novel countermeasures possible. In this review, we will discuss important considerations for the effective development of animal models using viral vector approaches and review the current vector-based animal models for studying viral pathogenesis and evaluating prophylactic and therapeutic strategies, with an emphasis on models of SARS-CoV-2 infection based on the vectorized expression of human angiotensin-converting enzyme 2.
Collapse
Affiliation(s)
- Melanie M. Goens
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Erin L. Howard
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Bryce M. Warner
- Vaccine and Infectious Disease Organization, University of Saskatchewan, 120 Veterinary Rd, Saskatoon, SK S7N 5E3, Canada;
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, 107 Wiggins Rd, Saskatoon, SK S7N 5E5, Canada
| | - Leonardo Susta
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Sarah K. Wootton
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
4
|
Gonzalez V, Word C, Guerra-Pilaquinga N, Mazinani M, Fawcett S, Portfors C, Falzarano D, Kell AM, Jangra RK, Banerjee A, Seifert SN, Letko M. Viral susceptibility and innate immune competency of Carollia perspicillata bat cells produced for virological studies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.19.624190. [PMID: 39605657 PMCID: PMC11601607 DOI: 10.1101/2024.11.19.624190] [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: 11/29/2024]
Abstract
Multiple viruses that are highly pathogenic in humans are known to have evolved in bats. How bats tolerate infection with these viruses, however, is poorly understood. As viruses engage in a wide range of interactions with their hosts, it is essential to study bat viruses in a system that resembles their natural environment like bat-derived in vitro cellular models. However, stable and accessible bat cell lines are not widely available for the broader scientific community. Here, we generated in vitro reagents for the Seba's short-tailed bat (Carollia perspicillata), tested multiple methods of immortalization, and characterized their susceptibility to virus infection and response to immune stimulation. Using a pseudotyped virus library and authentic virus infections, we show that these C. perspicillata cell lines derived from a diverse array of tissues are susceptible to viruses bearing the glycoprotein of numerous orthohantaviruses, including Andes and Hantaan virus and are also susceptible to live hantavirus infection. Furthermore, stimulation with synthetic double-stranded RNA prior to infection with VSV and MERS-CoV induced a protective antiviral response, demonstrating the suitability of our cell lines to study the bat antiviral immune response. Taken together, the approaches outlined here will inform future efforts to develop in vitro tools for virology from non-model organisms and these C. perspicillata cell lines will enable studies on virus-host interactions in bats.
Collapse
Affiliation(s)
- Victoria Gonzalez
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, SK S7N 5E3 Canada
- Department of Veterinary Microbiology, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Cierra Word
- Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana. 71103, USA
| | | | - Mitra Mazinani
- Paul G. Allen School for Global Health, Washington State University, Pullman, Washington, 99163, USA
| | - Stephen Fawcett
- Paul G. Allen School for Global Health, Washington State University, Pullman, Washington, 99163, USA
| | | | - Darryl Falzarano
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, SK S7N 5E3 Canada
- Department of Veterinary Microbiology, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Alison M. Kell
- Department of Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Rohit K. Jangra
- Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana. 71103, USA
| | - Arinjay Banerjee
- Vaccine and Infectious Disease Organization (VIDO), University of Saskatchewan, Saskatoon, SK S7N 5E3 Canada
- Department of Veterinary Microbiology, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
- Department of Biology, University of Waterloo, Waterloo, ON N2L 3G1, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Stephanie N. Seifert
- Paul G. Allen School for Global Health, Washington State University, Pullman, Washington, 99163, USA
| | - Michael Letko
- Paul G. Allen School for Global Health, Washington State University, Pullman, Washington, 99163, USA
| |
Collapse
|
5
|
Tang M, Liang K, Duan W, Xia S, Shi D, Li E, Liu W, Wang Q. Reactive astrocytes promote tumor progression by up-regulating tumor protocadherin 1 expression in lung cancer brain metastasis. Biochem Biophys Res Commun 2024; 732:150431. [PMID: 39047401 DOI: 10.1016/j.bbrc.2024.150431] [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: 03/22/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024]
Abstract
Brain metastasis (BM) is one of the main causes of death in patients with non-small cell lung carcinoma. The specific pathological processes of BM, which are inextricably linked to the brain tumor microenvironment, such as the abundance of astrocytes, lead to limited treatment options and poor prognosis. Reactive astrocytes are acquired in the BM; however, the underlying mechanisms remain unclear. This study aimed to explore the mechanisms by which astrocytes promote BM development. We determined the crucial role of reactive astrocytes in promoting the proliferation and migration of brain metastatic lung tumor cells by upregulating protocadherin 1 (PCDH1) expression in an in vitro co-culture model. The overexpression of PCDH1 was confirmed in clinical BM samples using immunohistochemical staining. Survival analysis indicated that high-PCDH1 expression was associated with poor survival in patients with lung adenocarcinoma. In vivo assays further showed that silence of PCDH1 effectively inhibited the tumor progression of brain metastases and prolonged the survival of animals. RNA sequencing has revealed that PCDH1 plays an important role in cell proliferation and adhesion. In conclusion, the present study revealed the promoting role of astrocytes in enhancing the aggressive phenotype of brain metastatic tumor cells by regulating the expression of PCDH1, which might be a biomarker for BM diagnosis and prognosis, suggesting the potential efficacy of targeting important astrocyte-tumor interactions in the treatment of patients with non-small cell lung carcinoma with BM.
Collapse
Affiliation(s)
- Mengyi Tang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Kun Liang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Wenzhe Duan
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shengkai Xia
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dongmei Shi
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Encheng Li
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Wenwen Liu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, China; Cancer Translational Medicine Research Center, The Second Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Qi Wang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Dalian Medical University, Dalian, China; Cancer Translational Medicine Research Center, The Second Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
6
|
Tse AL, Acreman CM, Ricardo-Lax I, Berrigan J, Lasso G, Balogun T, Kearns FL, Casalino L, McClain GL, Chandran AM, Lemeunier C, Amaro RE, Rice CM, Jangra RK, McLellan JS, Chandran K, Miller EH. Distinct pathways for evolution of enhanced receptor binding and cell entry in SARS-like bat coronaviruses. PLoS Pathog 2024; 20:e1012704. [PMID: 39546542 PMCID: PMC11602109 DOI: 10.1371/journal.ppat.1012704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 11/27/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
Understanding the zoonotic risks posed by bat coronaviruses (CoVs) is critical for pandemic preparedness. Herein, we generated recombinant vesicular stomatitis viruses (rVSVs) bearing spikes from divergent bat CoVs to investigate their cell entry mechanisms. Unexpectedly, the successful recovery of rVSVs bearing the spike from SHC014-CoV, a SARS-like bat CoV, was associated with the acquisition of a novel substitution in the S2 fusion peptide-proximal region (FPPR). This substitution enhanced viral entry in both VSV and coronavirus contexts by increasing the availability of the spike receptor-binding domain to recognize its cellular receptor, ACE2. A second substitution in the S1 N-terminal domain, uncovered through the rescue and serial passage of a virus bearing the FPPR substitution, further enhanced spike:ACE2 interaction and viral entry. Our findings identify genetic pathways for adaptation by bat CoVs during spillover and host-to-host transmission, fitness trade-offs inherent to these pathways, and potential Achilles' heels that could be targeted with countermeasures.
Collapse
Affiliation(s)
- Alexandra L. Tse
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Cory M. Acreman
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Inna Ricardo-Lax
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York, United States of America
| | - Jacob Berrigan
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Gorka Lasso
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Toheeb Balogun
- Department of Molecular Biology, University of California San Diego, La Jolla, California, United States of America
| | - Fiona L. Kearns
- Department of Molecular Biology, University of California San Diego, La Jolla, California, United States of America
| | - Lorenzo Casalino
- Department of Molecular Biology, University of California San Diego, La Jolla, California, United States of America
| | - Georgia L. McClain
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York, United States of America
| | - Amartya Mudry Chandran
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Charlotte Lemeunier
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Rommie E. Amaro
- Department of Molecular Biology, University of California San Diego, La Jolla, California, United States of America
| | - Charles M. Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York, United States of America
| | - Rohit K. Jangra
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
- Present address: Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, United States of America
| | - Jason S. McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Kartik Chandran
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| | - Emily Happy Miller
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, New York, United States of America
| |
Collapse
|
7
|
Boegelein L, Schreiber P, Philipp A, Nusshag C, Essbauer S, Zeier M, Krautkrämer E. Replication kinetics of pathogenic Eurasian orthohantaviruses in human mesangial cells. Virol J 2024; 21:241. [PMID: 39354507 PMCID: PMC11446005 DOI: 10.1186/s12985-024-02517-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 09/23/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND Eurasian pathogenic orthohantaviruses cause hemorrhagic fever with renal syndrome (HFRS) characterized by acute kidney injury (AKI). The virulence of orthohantaviruses varies enormously and direct infection of different renal cell types contribute to pathogenesis. Glomerular mesangial cells play an essential role in the interplay between kidney cells and proper kidney function. Therefore, we analyzed the replication competence of different orthohantavirus species in primary mesangial cells and a mesangial cell line. METHODS We tested the suitability of the mesangial cell line CIHGM-1 (conditionally immortalized human glomerular mesangial cells) as cell culture model for orthohantavirus kidney infection by comparison with primary human renal mesangial cells (HRMCs). We analyzed infection with high pathogenic Hantaan virus (HTNV), moderate pathogenic Puumala virus (PUUV) and non-/low-pathogenic Tula virus (TULV). RESULTS Effective viral spread was observed for PUUV only, whereas infection with HTNV and TULV was abortive. However, in contrast to TULV, HTNV exhibits an initially high infection rate and declines afterwards. This replication pattern was observed in HRMCs and CIHGM-1 cells. Viability or adhesion was neither impaired for PUUV-infected CIHGM-1 nor HRMCs. A loss of migration capacity was observed in PUUV-infected CIHGM-1 cells, but not in HRMCs. CONCLUSIONS The identification of differences in the replication competence of pathogenic orthohantavirus strains in renal mesangial cells is of special interest and may provide useful insights in the virus-specific mechanisms of orthohantavirus induced AKI. The use of CIHGM-1 cells will facilitate the research in a relevant cell culture system.
Collapse
Affiliation(s)
- Lukas Boegelein
- Department of Nephrology, University of Heidelberg, Im Neuenheimer Feld 162, D-69120, Heidelberg, Germany
| | - Pamela Schreiber
- Department of Nephrology, University of Heidelberg, Im Neuenheimer Feld 162, D-69120, Heidelberg, Germany
| | - Alexandra Philipp
- Department of Nephrology, University of Heidelberg, Im Neuenheimer Feld 162, D-69120, Heidelberg, Germany
| | - Christian Nusshag
- Department of Nephrology, University of Heidelberg, Im Neuenheimer Feld 162, D-69120, Heidelberg, Germany
| | - Sandra Essbauer
- Department Virology and Intracellular Agents, Bundeswehr Institute of Microbiology, German Centre for Infection Research, Munich Partner Site, D-80937, Munich, Germany
| | - Martin Zeier
- Department of Nephrology, University of Heidelberg, Im Neuenheimer Feld 162, D-69120, Heidelberg, Germany
| | - Ellen Krautkrämer
- Department of Nephrology, University of Heidelberg, Im Neuenheimer Feld 162, D-69120, Heidelberg, Germany.
| |
Collapse
|
8
|
Li W, Plante JA, Lin C, Basu H, Plung JS, Fan X, Boeckers JM, Oros J, Buck TK, Anekal PV, Hanson WA, Varnum H, Wells A, Mann CJ, Tjang LV, Yang P, Reyna RA, Mitchell BM, Shinde DP, Walker JL, Choi SY, Brusic V, Llopis PM, Weaver SC, Umemori H, Chiu IM, Plante KS, Abraham J. Shifts in receptors during submergence of an encephalitic arbovirus. Nature 2024; 632:614-621. [PMID: 39048821 PMCID: PMC11324528 DOI: 10.1038/s41586-024-07740-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 06/19/2024] [Indexed: 07/27/2024]
Abstract
Western equine encephalitis virus (WEEV) is an arthropod-borne virus (arbovirus) that frequently caused major outbreaks of encephalitis in humans and horses in the early twentieth century, but the frequency of outbreaks has since decreased markedly, and strains of this alphavirus isolated in the past two decades are less virulent in mammals than strains isolated in the 1930s and 1940s1-3. The basis for this phenotypic change in WEEV strains and coincident decrease in epizootic activity (known as viral submergence3) is unclear, as is the possibility of re-emergence of highly virulent strains. Here we identify protocadherin 10 (PCDH10) as a cellular receptor for WEEV. We show that multiple highly virulent ancestral WEEV strains isolated in the 1930s and 1940s, in addition to binding human PCDH10, could also bind very low-density lipoprotein receptor (VLDLR) and apolipoprotein E receptor 2 (ApoER2), which are recognized by another encephalitic alphavirus as receptors4. However, whereas most of the WEEV strains that we examined bind to PCDH10, a contemporary strain has lost the ability to recognize mammalian PCDH10 while retaining the ability to bind avian receptors, suggesting WEEV adaptation to a main reservoir host during enzootic circulation. PCDH10 supports WEEV E2-E1 glycoprotein-mediated infection of primary mouse cortical neurons, and administration of a soluble form of PCDH10 protects mice from lethal WEEV challenge. Our results have implications for the development of medical countermeasures and for risk assessment for re-emerging WEEV strains.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- Birds/metabolism
- Birds/virology
- Communicable Diseases, Emerging/epidemiology
- Communicable Diseases, Emerging/virology
- Encephalitis Virus, Western Equine/classification
- Encephalitis Virus, Western Equine/metabolism
- Encephalitis Virus, Western Equine/pathogenicity
- Encephalomyelitis, Equine/epidemiology
- Encephalomyelitis, Equine/virology
- Host Specificity
- LDL-Receptor Related Proteins/metabolism
- Neurons/metabolism
- Neurons/virology
- Phenotype
- Protocadherins/metabolism
- Receptors, LDL/metabolism
- Receptors, LDL/genetics
- Receptors, Virus/metabolism
- Viral Envelope Proteins/metabolism
- Viral Zoonoses/epidemiology
- Viral Zoonoses/virology
Collapse
Affiliation(s)
- Wanyu Li
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Jessica A Plante
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - ChieYu Lin
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Himanish Basu
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Jesse S Plung
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Xiaoyi Fan
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Joshua M Boeckers
- Department of Neurology, F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jessica Oros
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Tierra K Buck
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Praju V Anekal
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- MicRoN Core, Harvard Medical School, Boston, MA, USA
| | - Wesley A Hanson
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Haley Varnum
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Adrienne Wells
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- MicRoN Core, Harvard Medical School, Boston, MA, USA
| | - Colin J Mann
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Laurentia V Tjang
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Pan Yang
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Rachel A Reyna
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Brooke M Mitchell
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Divya P Shinde
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Jordyn L Walker
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - So Yoen Choi
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Vesna Brusic
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Paula Montero Llopis
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- MicRoN Core, Harvard Medical School, Boston, MA, USA
| | - Scott C Weaver
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Hisashi Umemori
- Department of Neurology, F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Isaac M Chiu
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Kenneth S Plante
- World Reference Center for Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, TX, USA
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Jonathan Abraham
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA.
- Center for Integrated Solutions in Infectious Diseases, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
9
|
Tse AL, Acreman CM, Ricardo-Lax I, Berrigan J, Lasso G, Balogun T, Kearns FL, Casalino L, McClain GL, Chandran AM, Lemeunier C, Amaro RE, Rice CM, Jangra RK, McLellan JS, Chandran K, Miller EH. Distinct pathway for evolution of enhanced receptor binding and cell entry in SARS-like bat coronaviruses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600393. [PMID: 38979151 PMCID: PMC11230278 DOI: 10.1101/2024.06.24.600393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Understanding the zoonotic risks posed by bat coronaviruses (CoVs) is critical for pandemic preparedness. Herein, we generated recombinant vesicular stomatitis viruses (rVSVs) bearing spikes from divergent bat CoVs to investigate their cell entry mechanisms. Unexpectedly, the successful recovery of rVSVs bearing the spike from SHC014, a SARS-like bat CoV, was associated with the acquisition of a novel substitution in the S2 fusion peptide-proximal region (FPPR). This substitution enhanced viral entry in both VSV and coronavirus contexts by increasing the availability of the spike receptor-binding domain to recognize its cellular receptor, ACE2. A second substitution in the spike N-terminal domain, uncovered through forward-genetic selection, interacted epistatically with the FPPR substitution to synergistically enhance spike:ACE2 interaction and viral entry. Our findings identify genetic pathways for adaptation by bat CoVs during spillover and host-to-host transmission, fitness trade-offs inherent to these pathways, and potential Achilles' heels that could be targeted with countermeasures.
Collapse
|
10
|
Chen N, Wang X, Guo Y, Zhao M, Cao B, Zhan B, Li Y, Zhou T, Zhu F, Guo C, Shi Y, Wang Q, Zhang L, Li Y. IL-37d suppresses Rheb-mTORC1 axis independently of TCS2 to alleviate alcoholic liver disease. Commun Biol 2024; 7:756. [PMID: 38907105 PMCID: PMC11192940 DOI: 10.1038/s42003-024-06427-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 06/07/2024] [Indexed: 06/23/2024] Open
Abstract
Tuberous sclerosis complex 2 (TSC2) crucially suppresses Rheb activity to prevent mTORC1 activation. However, mutations in TSC genes lead to mTORC1 overactivation, thereby causing various developmental disorders and cancer. Therefore, the discovery of novel Rheb inhibitors is vital to prevent mTOR overactivation. Here, we reveals that the anti-inflammatory cytokine IL-37d can bind to lysosomal Rheb and suppress its activity independent of TSC2, thereby preventing mTORC1 activation. The binding of IL-37d to Rheb switch-II subregion destabilizes the Rheb-mTOR and mTOR-S6K interactions, further halting mTORC1 signaling. Unlike TSC2, IL-37d is reduced under ethanol stimulation, which results in mitigating the suppression of lysosomal Rheb-mTORC1 activity. Consequently, the recombinant human IL-37d protein (rh-IL-37d) with a TAT peptide greatly improves alcohol-induced liver disorders by hindering Rheb-mTORC1 axis overactivation in a TSC2- independent manner. Together, IL-37d emerges as a novel Rheb suppressor independent of TSC2 to terminate mTORC1 activation and improve abnormal lipid metabolism in the liver.
Collapse
Affiliation(s)
- Nuo Chen
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Xiaoyu Wang
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Yaxin Guo
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Ming Zhao
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Baihui Cao
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Bing Zhan
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Yubin Li
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Tian Zhou
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Faliang Zhu
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Chun Guo
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Yongyu Shi
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Qun Wang
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China
| | - Lining Zhang
- Department of Immunology, School of Basic Medical Science, Shandong University, Jinan, China.
| | - Yan Li
- Department of Pathogen Biology, School of Basic Medical Science, Shandong University, Jinan, China.
| |
Collapse
|
11
|
Zarate-Sanchez E, George SC, Moya ML, Robertson C. Vascular dysfunction in hemorrhagic viral fevers: opportunities for organotypic modeling. Biofabrication 2024; 16:032008. [PMID: 38749416 PMCID: PMC11151171 DOI: 10.1088/1758-5090/ad4c0b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/25/2024] [Accepted: 05/15/2024] [Indexed: 06/06/2024]
Abstract
The hemorrhagic fever viruses (HFVs) cause severe or fatal infections in humans. Named after their common symptom hemorrhage, these viruses induce significant vascular dysfunction by affecting endothelial cells, altering immunity, and disrupting the clotting system. Despite advances in treatments, such as cytokine blocking therapies, disease modifying treatment for this class of pathogen remains elusive. Improved understanding of the pathogenesis of these infections could provide new avenues to treatment. While animal models and traditional 2D cell cultures have contributed insight into the mechanisms by which these pathogens affect the vasculature, these models fall short in replicatingin vivohuman vascular dynamics. The emergence of microphysiological systems (MPSs) offers promising avenues for modeling these complex interactions. These MPS or 'organ-on-chip' models present opportunities to better mimic human vascular responses and thus aid in treatment development. In this review, we explore the impact of HFV on the vasculature by causing endothelial dysfunction, blood clotting irregularities, and immune dysregulation. We highlight how existing MPS have elucidated features of HFV pathogenesis as well as discuss existing knowledge gaps and the challenges in modeling these interactions using MPS. Understanding the intricate mechanisms of vascular dysfunction caused by HFV is crucial in developing therapies not only for these infections, but also for other vasculotropic conditions like sepsis.
Collapse
Affiliation(s)
- Evelyn Zarate-Sanchez
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States of America
| | - Steven C George
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States of America
| | - Monica L Moya
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
| | - Claire Robertson
- Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, CA, United States of America
- UC Davis Comprehensive Cancer Center, Davis, CA, United States of America
| |
Collapse
|
12
|
McFadden E, Monticelli SR, Wang A, Ramamohan AR, Batchelor TG, Kuehne AI, Bakken RR, Tse AL, Chandran K, Herbert AS, McLellan JS. Engineering, structure, and immunogenicity of a Crimean-Congo hemorrhagic fever virus pre-fusion heterotrimeric glycoprotein complex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.20.590419. [PMID: 38659837 PMCID: PMC11042304 DOI: 10.1101/2024.04.20.590419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV) is a tick-borne virus that can cause severe disease in humans with case fatality rates of 10-40%. Although structures of CCHFV glycoproteins GP38 and Gc have provided insights into viral entry and defined epitopes of neutralizing and protective antibodies, the structure of glycoprotein Gn and its interactions with GP38 and Gc have remained elusive. Here, we used structure-guided protein engineering to produce a stabilized GP38-Gn-Gc heterotrimeric glycoprotein complex (GP38-GnH-DS-Gc). A cryo-EM structure of this complex provides the molecular basis for GP38's association on the viral surface, reveals the structure of Gn, and demonstrates that GP38-Gn restrains the Gc fusion loops in the prefusion conformation, facilitated by an N-linked glycan attached to Gn. Immunization with GP38-GnH-DS-Gc conferred 40% protection against lethal IbAr10200 challenge in mice. These data define the architecture of a GP38-Gn-Gc protomer and provide a template for structure-guided vaccine antigen development.
Collapse
Affiliation(s)
- Elizabeth McFadden
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| | - Stephanie R. Monticelli
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
- The Geneva Foundation, Tacoma, WA, USA
| | - Albert Wang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ajit R. Ramamohan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| | - Thomas G. Batchelor
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
- Oak Ridge Institute of Science Education, Oak Ridge, TN, USA
| | - Ana I. Kuehne
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Russell R. Bakken
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Alexandra L. Tse
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Andrew S. Herbert
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Jason S. McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
13
|
Rani P, George B, V S, Biswas S, V M, Pal A, Rajmani RS, Das S. MicroRNA-22-3p displaces critical host factors from the 5' UTR and inhibits the translation of Coxsackievirus B3 RNA. J Virol 2024; 98:e0150423. [PMID: 38289119 PMCID: PMC10883805 DOI: 10.1128/jvi.01504-23] [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: 09/25/2023] [Accepted: 01/02/2024] [Indexed: 02/21/2024] Open
Abstract
Coxsackievirus B3 (CVB3) is known to cause acute myocarditis and pancreatitis in humans. We investigated the microRNAs (miRNAs) that can potentially govern the viral life cycle by binding to the untranslated regions (UTRs) of CVB3 RNA. MicroRNA-22-3p was short-listed, as its potential binding site overlapped with the region crucial for recruiting internal ribosome entry site trans-acting factors (ITAFs) and ribosomes. We demonstrate that miR-22-3p binds CVB3 5' UTR, hinders recruitment of key ITAFs on viral mRNA, disrupts the spatial structure required for ribosome recruitment, and ultimately blocks translation. Likewise, cells lacking miR-22-3p exhibited heightened CVB3 infection compared to wild type, confirming its role in controlling infection. Interestingly, miR-22-3p level was found to be increased at 4 hours post-infection, potentially due to the accumulation of viral 2A protease in the early phase of infection. 2Apro enhances the miR-22-3p level to dislodge the ITAFs from the SD-like sequence, rendering the viral RNA accessible for binding of replication factors to switch to replication. Furthermore, one of the cellular targets of miR-22-3p, protocadherin-1 (PCDH1), was significantly downregulated during CVB3 infection. Partial silencing of PCDH1 reduced viral replication, demonstrating its proviral role. Interestingly, upon CVB3 infection in mice, miR-22-3p level was found to be downregulated only in the small intestine, the primary target organ, indicating its possible role in influencing tissue tropism. It appears miR-22-3p plays a dual role during infection by binding viral RNA to aid its life cycle as a viral strategy and by targeting a proviral protein to restrict viral replication as a host response.IMPORTANCECVB3 infection is associated with the development of end-stage heart diseases. Lack of effective anti-viral treatments and vaccines for CVB3 necessitates comprehensive understanding of the molecular players during CVB3 infection. miRNAs have emerged as promising targets for anti-viral strategies. Here, we demonstrate that miR-22-3p binds to 5' UTR and inhibits viral RNA translation at the later stage of infection to promote viral RNA replication. Conversely, as host response, it targets PCDH1, a proviral factor, to discourage viral propagation. miR-22-3p also influences CVB3 tissue tropism. Deciphering the multifaced role of miR-22-3p during CVB3 infection unravels the necessary molecular insights, which can be exploited for novel intervening strategies to curb infection and restrict viral pathogenesis.
Collapse
Affiliation(s)
- Priya Rani
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Biju George
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Sabarishree V
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Somarghya Biswas
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Madhurya V
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Apala Pal
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
| | - Raju S. Rajmani
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - Saumitra Das
- Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, India
- National Institute of Biomedical Genomics, Kalyani, India
| |
Collapse
|
14
|
Meola A, Guardado-Calvo P. Production and Purification of Hantavirus Glycoproteins in Drosophila melanogaster S2 Cells. Methods Mol Biol 2024; 2762:3-16. [PMID: 38315356 DOI: 10.1007/978-1-0716-3666-4_1] [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] [Indexed: 02/07/2024]
Abstract
Hantaviruses, are rodent-borne viruses found worldwide that are transmitted to humans through inhalation of contaminated excreta. They can cause a renal or a pulmonary syndrome, depending on the virus, and no effective treatment is currently available for either of these diseases. Hantaviral particles are covered by a protein lattice composed of two glycoproteins (Gn and Gc) that mediate adsorption to target cells and fusion with endosomal membranes, making them prime targets for neutralizing antibodies. Here we present the methodology to produce soluble recombinant glycoproteins in different conformations, either alone or as a stabilized Gn/Gc complex, using stably transfected Drosophila S2 cells.
Collapse
Affiliation(s)
- Annalisa Meola
- G5 Structural Biology of Infectious Diseases, Institut Pasteur, Université Paris Cité, Paris, France
| | - Pablo Guardado-Calvo
- G5 Structural Biology of Infectious Diseases, Institut Pasteur, Université Paris Cité, Paris, France.
| |
Collapse
|
15
|
Du X, Yi X, Zou X, Chen Y, Tai Y, Ren X, He X. PCDH1, a poor prognostic biomarker and potential target for pancreatic adenocarcinoma metastatic therapy. BMC Cancer 2023; 23:1102. [PMID: 37957639 PMCID: PMC10642060 DOI: 10.1186/s12885-023-11474-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 10/03/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Pancreatic adenocarcinoma (PAAD) is an aggressive solid tumour characterised by few early symptoms, high mortality, and lack of effective treatment. Therefore, it is important to identify new potential therapeutic targets and prognostic biomarkers of PAAD. METHODS The Cancer Genome Atlas and Genotype-Tissue Expression databases were used to identify the expression and prognostic model of protocadherin 1 (PCDH1). The prognostic performance of risk factors and diagnosis of patients with PAAD were evaluated by regression analysis, nomogram, and receiver operating characteristic curve. Paraffin sections were collected from patients for immunohistochemistry (IHC) analysis. The expression of PCDH1 in cells obtained from primary tumours or metastatic biopsies was identified using single-cell RNA sequencing (scRNA-seq). Real-time quantitative polymerase chain reaction (qPCR) and western blotting were used to verify PCDH1 expression levels and the inhibitory effects of the compounds. RESULTS The RNA and protein levels of PCDH1 were significantly higher in PAAD cells than in normal pancreatic ductal cells, similar to those observed in tissue sections from patients with PAAD. Aberrant methylation of the CpG site cg19767205 and micro-RNA (miRNA) hsa-miR-124-1 may be important reasons for the high PCDH1 expression in PAAD. Up-regulated PCDH1 promotes pancreatic cancer cell metastasis. The RNA levels of PCDH1 were significantly down-regulated following flutamide treatment. Flutamide reduced the percentage of PCDH1 RNA level in PAAD cells Panc-0813 to < 50%. In addition, the PCDH1 protein was significantly down-regulated after Panc-0813 cells were incubated with 20 µM flutamide and proves to be a potential therapeutic intervention for PAAD. CONCLUSION PCDH1 is a key prognostic biomarker and promoter of PAAD metastasis. Additionally, flutamide may serve as a novel compound that down-regulates PCDH1 expression as a potential treatment for combating PAAD progression and metastasis.
Collapse
Affiliation(s)
- Xingyi Du
- Key Laboratory of Structure-Based Drug Design and Discovery, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, 100850, China
- Nanhu Laboratory, Jiaxing, 314002, China
| | - Xiaoyu Yi
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, 100850, China
- Nanhu Laboratory, Jiaxing, 314002, China
| | - Xiaocui Zou
- Key Laboratory of Structure-Based Drug Design and Discovery, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Yuan Chen
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, 100850, China
- Nanhu Laboratory, Jiaxing, 314002, China
| | - Yanhong Tai
- Department of Pathology, No.307 Hospital of PLA, Beijing, 100071, China
| | - Xuhong Ren
- Key Laboratory of Structure-Based Drug Design and Discovery, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| | - Xinhua He
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, 100850, China.
- Nanhu Laboratory, Jiaxing, 314002, China.
| |
Collapse
|
16
|
Hartman AL, Myler PJ. Bunyavirales: Scientific Gaps and Prototype Pathogens for a Large and Diverse Group of Zoonotic Viruses. J Infect Dis 2023; 228:S376-S389. [PMID: 37849397 PMCID: PMC10582323 DOI: 10.1093/infdis/jiac338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2023] Open
Abstract
Research directed at select prototype pathogens is part of the approach put forth by the National Institute of Allergy and Infectious Disease (NIAID) to prepare for future pandemics caused by emerging viruses. We were tasked with identifying suitable prototypes for four virus families of the Bunyavirales order (Phenuiviridae, Peribunyaviridae, Nairoviridae, and Hantaviridae). This is a challenge due to the breadth and diversity of these viral groups. While there are many differences among the Bunyavirales, they generally have complex ecological life cycles, segmented genomes, and cause a range of human clinical outcomes from mild to severe and even death. Here, we delineate potential prototype species that encompass the breadth of clinical outcomes of a given family, have existing reverse genetics tools or animal disease models, and can be amenable to a platform approach to vaccine testing. Suggested prototype pathogens outlined here can serve as a starting point for further discussions.
Collapse
Affiliation(s)
- Amy L Hartman
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Peter J Myler
- Department of Pediatrics and the Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, Washington, USA
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| |
Collapse
|
17
|
Chen RX, Gong HY, Wang X, Sun MH, Ji YF, Tan SM, Chen JM, Shao JW, Liao M. Zoonotic Hantaviridae with Global Public Health Significance. Viruses 2023; 15:1705. [PMID: 37632047 PMCID: PMC10459939 DOI: 10.3390/v15081705] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/04/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Hantaviridae currently encompasses seven genera and 53 species. Multiple hantaviruses such as Hantaan virus, Seoul virus, Dobrava-Belgrade virus, Puumala virus, Andes virus, and Sin Nombre virus are highly pathogenic to humans. They cause hemorrhagic fever with renal syndrome (HFRS) and hantavirus cardiopulmonary syndrome or hantavirus pulmonary syndrome (HCPS/HPS) in many countries. Some hantaviruses infect wild or domestic animals without causing severe symptoms. Rodents, shrews, and bats are reservoirs of various mammalian hantaviruses. Recent years have witnessed significant advancements in the study of hantaviruses including genomics, taxonomy, evolution, replication, transmission, pathogenicity, control, and patient treatment. Additionally, new hantaviruses infecting bats, rodents, shrews, amphibians, and fish have been identified. This review compiles these advancements to aid researchers and the public in better recognizing this zoonotic virus family with global public health significance.
Collapse
Affiliation(s)
- Rui-Xu Chen
- School of Life Science and Engineering, Foshan University, Foshan 528225, China; (R.-X.C.); (H.-Y.G.); (X.W.); (M.-H.S.); (Y.-F.J.); (S.-M.T.)
| | - Huan-Yu Gong
- School of Life Science and Engineering, Foshan University, Foshan 528225, China; (R.-X.C.); (H.-Y.G.); (X.W.); (M.-H.S.); (Y.-F.J.); (S.-M.T.)
| | - Xiu Wang
- School of Life Science and Engineering, Foshan University, Foshan 528225, China; (R.-X.C.); (H.-Y.G.); (X.W.); (M.-H.S.); (Y.-F.J.); (S.-M.T.)
| | - Ming-Hui Sun
- School of Life Science and Engineering, Foshan University, Foshan 528225, China; (R.-X.C.); (H.-Y.G.); (X.W.); (M.-H.S.); (Y.-F.J.); (S.-M.T.)
| | - Yu-Fei Ji
- School of Life Science and Engineering, Foshan University, Foshan 528225, China; (R.-X.C.); (H.-Y.G.); (X.W.); (M.-H.S.); (Y.-F.J.); (S.-M.T.)
| | - Su-Mei Tan
- School of Life Science and Engineering, Foshan University, Foshan 528225, China; (R.-X.C.); (H.-Y.G.); (X.W.); (M.-H.S.); (Y.-F.J.); (S.-M.T.)
| | - Ji-Ming Chen
- School of Life Science and Engineering, Foshan University, Foshan 528225, China; (R.-X.C.); (H.-Y.G.); (X.W.); (M.-H.S.); (Y.-F.J.); (S.-M.T.)
| | - Jian-Wei Shao
- School of Life Science and Engineering, Foshan University, Foshan 528225, China; (R.-X.C.); (H.-Y.G.); (X.W.); (M.-H.S.); (Y.-F.J.); (S.-M.T.)
| | - Ming Liao
- College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou 510230, China
| |
Collapse
|
18
|
Jeyachandran AV, Irudayam JI, Dubey S, Chakravarty N, Konda B, Shah A, Su B, Wang C, Cui Q, Williams KJ, Srikanth S, Shi Y, Deb A, Damoiseaux R, Stripp BR, Ramaiah A, Arumugaswami V. Comparative Analysis of Molecular Pathogenic Mechanisms and Antiviral Development Targeting Old and New World Hantaviruses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.04.552083. [PMID: 37577539 PMCID: PMC10418258 DOI: 10.1101/2023.08.04.552083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Background Hantaviruses - dichotomized into New World (i.e. Andes virus, ANDV; Sin Nombre virus, SNV) and Old-World viruses (i.e. Hantaan virus, HTNV) - are zoonotic viruses transmitted from rodents to humans. Currently, no FDA-approved vaccines against hantaviruses exist. Given the recent breakthrough to human-human transmission by the ANDV, an essential step is to establish an effective pandemic preparedness infrastructure to rapidly identify cell tropism, infective potential, and effective therapeutic agents through systematic investigation. Methods We established human cell model systems in lung (airway and distal lung epithelial cells), heart (pluripotent stem cell-derived (PSC-) cardiomyocytes), and brain (PSC-astrocytes) cell types and subsequently evaluated ANDV, HTNV and SNV tropisms. Transcriptomic, lipidomic and bioinformatic data analyses were performed to identify the molecular pathogenic mechanisms of viruses in different cell types. This cell-based infection system was utilized to establish a drug testing platform and pharmacogenomic comparisons. Results ANDV showed broad tropism for all cell types assessed. HTNV replication was predominantly observed in heart and brain cells. ANDV efficiently replicated in human and mouse 3D distal lung organoids. Transcriptomic analysis showed that ANDV infection resulted in pronounced inflammatory response and downregulation of cholesterol biosynthesis pathway in lung cells. Lipidomic profiling revealed that ANDV-infected cells showed reduced level of cholesterol esters and triglycerides. Further analysis of pathway-based molecular signatures showed that, compared to SNV and HTNV, ANDV infection caused drastic lung cell injury responses. A selective drug screening identified STING agonists, nucleoside analogues and plant-derived compounds that inhibited ANDV viral infection and rescued cellular metabolism. In line with experimental results, transcriptome data shows that the least number of total and unique differentially expressed genes were identified in urolithin B- and favipiravir-treated cells, confirming the higher efficiency of these two drugs in inhibiting ANDV, resulting in host cell ability to balance gene expression to establish proper cell functioning. Conclusions Overall, our study describes advanced human PSC-derived model systems and systems-level transcriptomics and lipidomic data to better understand Old and New World hantaviral tropism, as well as drug candidates that can be further assessed for potential rapid deployment in the event of a pandemic.
Collapse
Affiliation(s)
- Arjit Vijey Jeyachandran
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Joseph Ignatius Irudayam
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Swati Dubey
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Nikhil Chakravarty
- Department of Epidemiology, University of California, Los Angeles, CA, USA
| | - Bindu Konda
- Department of Medicine, Lung and Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Aayushi Shah
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Baolong Su
- Dept. of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, USA
- UCLA Lipidomics Lab, University of California, Los Angeles, Los Angeles, CA, USA
| | - Cheng Wang
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, CA, USA
| | - Qi Cui
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, CA, USA
| | - Kevin J. Williams
- Dept. of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, USA
- UCLA Lipidomics Lab, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sonal Srikanth
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Yanhong Shi
- Department of Neurodegenerative Diseases, Beckman Research Institute of City of Hope, CA, USA
| | - Arjun Deb
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA, USA
| | - Robert Damoiseaux
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA, USA
- California NanoSystems Institute, UCLA, Los Angeles, CA, USA
- Department of Bioengineering, Samueli School of Engineering, UCLA, Los Angeles, CA, USA
| | - Barry R. Stripp
- Department of Medicine, Lung and Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
- Eli & Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA, USA
- California NanoSystems Institute, UCLA, Los Angeles, CA, USA
| |
Collapse
|
19
|
Zhang L, Peng X, Wang Q, Li J, Lv S, Han S, Zhang L, Ding H, Wang CY, Xiao G, Du X, Peng K, Li H, Liu W. CCR2 is a host entry receptor for severe fever with thrombocytopenia syndrome virus. SCIENCE ADVANCES 2023; 9:eadg6856. [PMID: 37531422 PMCID: PMC10396298 DOI: 10.1126/sciadv.adg6856] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 06/30/2023] [Indexed: 08/04/2023]
Abstract
Severe fever with thrombocytopenia syndrome virus (SFTSV) is an emerging tick-borne bunyavirus causing a high fatality rate of up to 30%. To date, the receptor mediating SFTSV entry remained uncharacterized, hindering the understanding of disease pathogenesis. Here, C-C motif chemokine receptor 2 (CCR2) was identified as a host receptor for SFTSV based on a genome-wide CRISPR-Cas9 screen. Knockout of CCR2 substantially reduced viral binding and infection. CCR2 enhanced SFTSV binding through direct binding to SFTSV glycoprotein N (Gn), which is mediated by its N-terminal extracellular domain. Depletion of CCR2 in C57BL/6J mouse model attenuated SFTSV replication and pathogenesis. The peripheral blood primary monocytes from elderly individuals or subjects with underlying diabetes mellitus showed higher CCR2 surface expression and supported stronger binding and replication of SFTSV. Together, these data indicate that CCR2 is a host entry receptor for SFTSV infection and a novel target for developing anti-SFTSV therapeutics.
Collapse
Affiliation(s)
- Leike Zhang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
- Hubei Jiangxia Laboratory, Wuhan, Hubei 430200, China
| | - Xuefang Peng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Qingxing Wang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
| | - Jin Li
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Shouming Lv
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Shuo Han
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Lingyu Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Heng Ding
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Cong-Yi Wang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430040, China
| | - Gengfu Xiao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
| | - Xuguang Du
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Ke Peng
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430071, China
| | - Hao Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
- School of Public Health, Wuhan University, Wuhan, Hubei 430071, China
| | - Wei Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
- School of Public Health, Wuhan University, Wuhan, Hubei 430071, China
| |
Collapse
|
20
|
Schreiber P, Friedrich AK, Gruber G, Nusshag C, Boegelein L, Essbauer S, Uhrig J, Zeier M, Krautkrämer E. Differences in the Susceptibility of Human Tubular Epithelial Cells for Infection with Orthohantaviruses. Viruses 2023; 15:1670. [PMID: 37632012 PMCID: PMC10459294 DOI: 10.3390/v15081670] [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: 06/15/2023] [Revised: 07/21/2023] [Accepted: 07/30/2023] [Indexed: 08/27/2023] Open
Abstract
Diseases induced by infection with pathogenic orthohantaviruses are characterized by a pronounced organ-specific manifestation. Pathogenic Eurasian orthohantaviruses cause hemorrhagic fever with renal syndrome (HFRS) with often massive proteinuria. Therefore, the use of a relevant kidney cell culture would be favorable to analyze the underlying cellular mechanisms of orthohantavirus-induced acute kidney injury (AKI). We tested different human tubular epithelial cell lines for their suitability as an in vitro infection model. Permissiveness and replication kinetics of highly pathogenic Hantaan virus (HTNV) and non-/low-pathogenic Tula virus (TULV) were analyzed in tubular epithelial cell lines and compared to human primary tubular epithelial cells. Ana-lysis of the cell line HK-2 revealed the same results for viral replication, morphological and functional effects as observed for HTNV in primary cells. In contrast, the cell lines RPTEC/TERT1 and TH1 demonstrated only poor infection rates after inoculation with HTNV and are unusable as an infection model. While pathogenic HNTV infects primary tubular and HK-2 cells, non-/low-pathogenic TULV infects neither primary tubular cells nor the cell line HK-2. Our results show that permissiveness of renal cells varies between orthohantaviruses with differences in pathogenicity and that HK-2 cells demonstrate a suitable in vitro model to study viral tropism and pathogenesis of orthohantavirus-induced AKI.
Collapse
Affiliation(s)
- Pamela Schreiber
- Department of Nephrology, University of Heidelberg, D-69120 Heidelberg, Germany
| | | | - Gefion Gruber
- Department of Nephrology, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Christian Nusshag
- Department of Nephrology, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Lukas Boegelein
- Department of Nephrology, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Sandra Essbauer
- Bundeswehr Institute of Microbiology, Department Virology and Intracellular Agents, German Centre for Infection Research, Munich Partner Site, D-80937 Munich, Germany
| | - Josephine Uhrig
- Department of Nephrology, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Martin Zeier
- Department of Nephrology, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Ellen Krautkrämer
- Department of Nephrology, University of Heidelberg, D-69120 Heidelberg, Germany
| |
Collapse
|
21
|
Slough MM, Li R, Herbert AS, Lasso G, Kuehne AI, Monticelli SR, Bakken RR, Liu Y, Ghosh A, Moreau AM, Zeng X, Rey FA, Guardado-Calvo P, Almo SC, Dye JM, Jangra RK, Wang Z, Chandran K. Two point mutations in protocadherin-1 disrupt hantavirus recognition and afford protection against lethal infection. Nat Commun 2023; 14:4454. [PMID: 37488123 PMCID: PMC10366084 DOI: 10.1038/s41467-023-40126-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/06/2023] [Indexed: 07/26/2023] Open
Abstract
Andes virus (ANDV) and Sin Nombre virus (SNV) are the etiologic agents of severe hantavirus cardiopulmonary syndrome (HCPS) in the Americas for which no FDA-approved countermeasures are available. Protocadherin-1 (PCDH1), a cadherin-superfamily protein recently identified as a critical host factor for ANDV and SNV, represents a new antiviral target; however, its precise role remains to be elucidated. Here, we use computational and experimental approaches to delineate the binding surface of the hantavirus glycoprotein complex on PCDH1's first extracellular cadherin repeat domain. Strikingly, a single amino acid residue in this PCDH1 surface influences the host species-specificity of SNV glycoprotein-PCDH1 interaction and cell entry. Mutation of this and a neighboring residue substantially protects Syrian hamsters from pulmonary disease and death caused by ANDV. We conclude that PCDH1 is a bona fide entry receptor for ANDV and SNV whose direct interaction with hantavirus glycoproteins could be targeted to develop new interventions against HCPS.
Collapse
Affiliation(s)
- Megan M Slough
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Rong Li
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, USA
| | - Andrew S Herbert
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Gorka Lasso
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ana I Kuehne
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Stephanie R Monticelli
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
- The Geneva Foundation, Tacoma, WA, USA
| | - Russell R Bakken
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Yanan Liu
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, USA
| | - Agnidipta Ghosh
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Alicia M Moreau
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Xiankun Zeng
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Félix A Rey
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Structural Virology Unit, F-75015, Paris, France
| | - Pablo Guardado-Calvo
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Structural Virology Unit, F-75015, Paris, France
- Institut Pasteur, Université Paris Cité, Structural Biology of Infectious Diseases Unit, F-75015, Paris, France
| | - Steven C Almo
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, USA
| | - John M Dye
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Rohit K Jangra
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA.
- Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA.
| | - Zhongde Wang
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, UT, USA.
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
22
|
Stass R, Engdahl TB, Chapman NS, Wolters RM, Handal LS, Diaz SM, Crowe JE, Bowden TA. Mechanistic basis for potent neutralization of Sin Nombre hantavirus by a human monoclonal antibody. Nat Microbiol 2023:10.1038/s41564-023-01413-y. [PMID: 37322112 DOI: 10.1038/s41564-023-01413-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 05/17/2023] [Indexed: 06/17/2023]
Abstract
Rodent-borne hantaviruses are prevalent worldwide and upon spillover to human populations, cause severe disease for which no specific treatment is available. A potent antibody response is key for recovery from hantavirus infection. Here we study a highly neutralizing human monoclonal antibody, termed SNV-42, which was derived from a memory B cell isolated from an individual with previous Sin Nombre virus (SNV) infection. Crystallographic analysis demonstrates that SNV-42 targets the Gn subcomponent of the tetrameric (Gn-Gc)4 glycoprotein assembly that is relevant for viral entry. Integration of our 1.8 Å structure with the (Gn-Gc)4 ultrastructure arrangement indicates that SNV-42 targets the membrane-distal region of the virus envelope. Comparison of the SNV-42 paratope encoding variable genes with inferred germline gene segments reveals high sequence conservation, suggesting that germline-encoded antibodies inhibit SNV. Furthermore, mechanistic assays reveal that SNV-42 interferes with both receptor recognition and fusion during host-cell entry. This work provides a molecular-level blueprint for understanding the human neutralizing antibody response to hantavirus infection.
Collapse
Affiliation(s)
- Robert Stass
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Taylor B Engdahl
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nathaniel S Chapman
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rachael M Wolters
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Laura S Handal
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Summer M Diaz
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James E Crowe
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Thomas A Bowden
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
23
|
Mittler E, Serris A, Esterman ES, Florez C, Polanco LC, O’Brien CM, Slough MM, Tynell J, Gröning R, Sun Y, Abelson DM, Wec AZ, Haslwanter D, Keller M, Ye C, Bakken RR, Jangra RK, Dye JM, Ahlm C, Rappazzo CG, Ulrich RG, Zeitlin L, Geoghegan JC, Bradfute SB, Sidoli S, Forsell MN, Strandin T, Rey FA, Herbert AS, Walker LM, Chandran K, Guardado-Calvo P. Structural and mechanistic basis of neutralization by a pan-hantavirus protective antibody. Sci Transl Med 2023; 15:eadg1855. [PMID: 37315110 PMCID: PMC11721787 DOI: 10.1126/scitranslmed.adg1855] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 05/18/2023] [Indexed: 06/16/2023]
Abstract
Emerging rodent-borne hantaviruses cause severe diseases in humans with no approved vaccines or therapeutics. We recently isolated a monoclonal broadly neutralizing antibody (nAb) from a Puumala virus-experienced human donor. Here, we report its structure bound to its target, the Gn/Gc glycoprotein heterodimer comprising the viral fusion complex. The structure explains the broad activity of the nAb: It recognizes conserved Gc fusion loop sequences and the main chain of variable Gn sequences, thereby straddling the Gn/Gc heterodimer and locking it in its prefusion conformation. We show that the nAb's accelerated dissociation from the divergent Andes virus Gn/Gc at endosomal acidic pH limits its potency against this highly lethal virus and correct this liability by engineering an optimized variant that sets a benchmark as a candidate pan-hantavirus therapeutic.
Collapse
Affiliation(s)
- Eva Mittler
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Alexandra Serris
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Structural Virology Unit, F-75015 Paris, France
| | | | - Catalina Florez
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA
- The Geneva Foundation, Tacoma, WA 98402, USA
| | - Laura C. Polanco
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Cecilia M. O’Brien
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA
- The Geneva Foundation, Tacoma, WA 98402, USA
| | - Megan M. Slough
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Janne Tynell
- Department of Clinical Microbiology, Umeå University, 90187 Umeå, Sweden
- Zoonosis Unit, Department of Virology, Medical Faculty, University of Helsinki, 00290 Helsinki, Finland
| | - Remigius Gröning
- Department of Clinical Microbiology, Umeå University, 90187 Umeå, Sweden
| | - Yan Sun
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | - Denise Haslwanter
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Markus Keller
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, 17493 Greifswald-Insel Riems, Germany
| | - Chunyan Ye
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM 87131, USA
| | - Russel R. Bakken
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA
| | - Rohit K. Jangra
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - John M. Dye
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA
| | - Clas Ahlm
- Department of Clinical Microbiology, Umeå University, 90187 Umeå, Sweden
| | | | - Rainer G. Ulrich
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, 17493 Greifswald-Insel Riems, Germany
- Partner site: Hamburg-Lübeck-Borstel-Riems, German Centre for Infection Research (DZIF), 17493 Greifswald-Insel Riems, Germany
| | - Larry Zeitlin
- Mapp Biopharmaceutical Inc., San Diego, CA 92121, USA
| | | | - Steven B. Bradfute
- Center for Global Health, Department of Internal Medicine, University of New Mexico Health Science Center, Albuquerque, NM 87131, USA
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | - Tomas Strandin
- Zoonosis Unit, Department of Virology, Medical Faculty, University of Helsinki, 00290 Helsinki, Finland
| | - Felix A. Rey
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Structural Virology Unit, F-75015 Paris, France
| | - Andrew S. Herbert
- U.S. Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD 21702, USA
| | | | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Pablo Guardado-Calvo
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Structural Virology Unit, F-75015 Paris, France
| |
Collapse
|
24
|
LaPointe A, Gale M, Kell AM. Orthohantavirus Replication in the Context of Innate Immunity. Viruses 2023; 15:1130. [PMID: 37243216 PMCID: PMC10220641 DOI: 10.3390/v15051130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Orthohantaviruses are rodent-borne, negative-sense RNA viruses that are capable of causing severe vascular disease in humans. Over the course of viral evolution, these viruses have tailored their replication cycles in such a way as to avoid and/or antagonize host innate immune responses. In the rodent reservoir, this results in life long asymptomatic infections. However, in hosts other than its co-evolved reservoir, the mechanisms for subduing the innate immune response may be less efficient or absent, potentially leading to disease and/or viral clearance. In the case of human orthohantavirus infection, the interaction of the innate immune response with viral replication is thought to give rise to severe vascular disease. The orthohantavirus field has made significant advancements in understanding how these viruses replicate and interact with host innate immune responses since their identification by Dr. Ho Wang Lee and colleagues in 1976. Therefore, the purpose of this review, as part of this special issue dedicated to Dr. Lee, was to summarize the current knowledge of orthohantavirus replication, how viral replication activates innate immunity, and how the host antiviral response, in turn, impacts viral replication.
Collapse
Affiliation(s)
- Autumn LaPointe
- Department of Molecular Genetics and Microbiology, University of New Mexico, 915 Camino de Salud NE, Albuquerque, NM 87131, USA
| | - Michael Gale
- Department of Immunology, Center for Innate Immunity and Immune Disease, University of Washington, Seattle, WA 98109, USA
| | - Alison M. Kell
- Department of Molecular Genetics and Microbiology, University of New Mexico, 915 Camino de Salud NE, Albuquerque, NM 87131, USA
| |
Collapse
|
25
|
Engdahl TB, Binshtein E, Brocato RL, Kuzmina NA, Principe LM, Kwilas SA, Kim RK, Chapman NS, Porter MS, Guardado-Calvo P, Rey FA, Handal LS, Diaz SM, Zagol-Ikapitte IA, Tran MH, McDonald WH, Meiler J, Reidy JX, Trivette A, Bukreyev A, Hooper JW, Crowe JE. Antigenic mapping and functional characterization of human New World hantavirus neutralizing antibodies. eLife 2023; 12:e81743. [PMID: 36971354 PMCID: PMC10115451 DOI: 10.7554/elife.81743] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 03/27/2023] [Indexed: 03/29/2023] Open
Abstract
Hantaviruses are high-priority emerging pathogens carried by rodents and transmitted to humans by aerosolized excreta or, in rare cases, person-to-person contact. While infections in humans are relatively rare, mortality rates range from 1 to 40% depending on the hantavirus species. There are currently no FDA-approved vaccines or therapeutics for hantaviruses, and the only treatment for infection is supportive care for respiratory or kidney failure. Additionally, the human humoral immune response to hantavirus infection is incompletely understood, especially the location of major antigenic sites on the viral glycoproteins and conserved neutralizing epitopes. Here, we report antigenic mapping and functional characterization for four neutralizing hantavirus antibodies. The broadly neutralizing antibody SNV-53 targets an interface between Gn/Gc, neutralizes through fusion inhibition and cross-protects against the Old World hantavirus species Hantaan virus when administered pre- or post-exposure. Another broad antibody, SNV-24, also neutralizes through fusion inhibition but targets domain I of Gc and demonstrates weak neutralizing activity to authentic hantaviruses. ANDV-specific, neutralizing antibodies (ANDV-5 and ANDV-34) neutralize through attachment blocking and protect against hantavirus cardiopulmonary syndrome (HCPS) in animals but target two different antigenic faces on the head domain of Gn. Determining the antigenic sites for neutralizing antibodies will contribute to further therapeutic development for hantavirus-related diseases and inform the design of new broadly protective hantavirus vaccines.
Collapse
Affiliation(s)
- Taylor B Engdahl
- Department of Pathology, Microbiology and Immunology, Vanderbilt UniversityNashvilleUnited States
| | - Elad Binshtein
- Vanderbilt Vaccine Center, Vanderbilt University Medical CenterNashvilleUnited States
| | - Rebecca L Brocato
- Virology Division, United States Army Medical Research Institute of Infectious DiseasesFt DetrickUnited States
| | - Natalia A Kuzmina
- Department of Pathology, The University of Texas Medical Branch at GalvestonGalvestonUnited States
- Galveston National LaboratoryGalvestonUnited States
| | - Lucia M Principe
- Virology Division, United States Army Medical Research Institute of Infectious DiseasesFt DetrickUnited States
| | - Steven A Kwilas
- Virology Division, United States Army Medical Research Institute of Infectious DiseasesFt DetrickUnited States
| | - Robert K Kim
- Virology Division, United States Army Medical Research Institute of Infectious DiseasesFt DetrickUnited States
| | - Nathaniel S Chapman
- Department of Pathology, Microbiology and Immunology, Vanderbilt UniversityNashvilleUnited States
| | - Monique S Porter
- Department of Pathology, Microbiology and Immunology, Vanderbilt UniversityNashvilleUnited States
| | | | - Félix A Rey
- Institut Pasteur, Université Paris CitéParisFrance
| | - Laura S Handal
- Vanderbilt Vaccine Center, Vanderbilt University Medical CenterNashvilleUnited States
| | - Summer M Diaz
- Vanderbilt Vaccine Center, Vanderbilt University Medical CenterNashvilleUnited States
| | - Irene A Zagol-Ikapitte
- Department of Biochemistry and Mass Spectrometry Research Center, Vanderbilt UniversityNashvilleUnited States
| | - Minh H Tran
- Department of Biochemistry and Mass Spectrometry Research Center, Vanderbilt UniversityNashvilleUnited States
| | - W Hayes McDonald
- Department of Biochemistry and Mass Spectrometry Research Center, Vanderbilt UniversityNashvilleUnited States
| | - Jens Meiler
- Department of Chemistry, Vanderbilt UniversityNashvilleUnited States
| | - Joseph X Reidy
- Vanderbilt Vaccine Center, Vanderbilt University Medical CenterNashvilleUnited States
| | - Andrew Trivette
- Vanderbilt Vaccine Center, Vanderbilt University Medical CenterNashvilleUnited States
| | - Alexander Bukreyev
- Department of Pathology, The University of Texas Medical Branch at GalvestonGalvestonUnited States
- Galveston National LaboratoryGalvestonUnited States
- Department of Microbiology and Immunology, University of Texas Medical BranchGalvestonUnited States
| | - Jay W Hooper
- Virology Division, United States Army Medical Research Institute of Infectious DiseasesFt DetrickUnited States
| | - James E Crowe
- Department of Pathology, Microbiology and Immunology, Vanderbilt UniversityNashvilleUnited States
- Vanderbilt Vaccine Center, Vanderbilt University Medical CenterNashvilleUnited States
- Department of Pediatrics, Vanderbilt University Medical CenterNashvilleUnited States
| |
Collapse
|
26
|
Schlohsarczyk EK, Drewes S, Koteja P, Röhrs S, Ulrich RG, Teifke JP, Herden C. Tropism of Puumala orthohantavirus and Endoparasite Coinfection in the Bank Vole Reservoir. Viruses 2023; 15:v15030612. [PMID: 36992321 PMCID: PMC10058470 DOI: 10.3390/v15030612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 02/08/2023] [Accepted: 02/12/2023] [Indexed: 02/25/2023] Open
Abstract
In Europe, most cases of human hantavirus disease are caused by Puumala orthohantavirus (PUUV) transmitted by bank voles (Clethrionomys glareolus, syn. Myodes glareolus), in which PUUV causes inconspicuous infection. Little is known about tropism and endoparasite coinfections in PUUV-infected reservoir and spillover-infected rodents. Here, we characterized PUUV tropism, pathological changes and endoparasite coinfections. The voles and some non-reservoir rodents were examined histologically, immunohistochemically, by in situ hybridization, indirect IgG enzyme-linked immunosorbent assay and reverse transcription-polymerase chain reaction. PUUV RNA and anti-PUUV antibodies were detected simultaneously in a large proportion of the bank voles, indicating persistent infection. Although PUUV RNA was not detected in non-reservoir rodents, the detection of PUUV-reactive antibodies suggests virus contact. No specific gross and histological findings were detected in the infected bank voles. A broad organ tropism of PUUV was observed: kidney and stomach were most frequently infected. Remarkably, PUUV was detected in cells lacking the typical secretory capacity, which may contribute to the maintenance of virus persistence. PUUV-infected wild bank voles were found to be frequently coinfected with Hepatozoon spp. and Sarcocystis (Frenkelia) spp., possibly causing immune modulation that may influence susceptibility to PUUV infection or vice versa. The results are a prerequisite for a deeper understanding of virus–host interactions in natural hantavirus reservoirs.
Collapse
Affiliation(s)
- Elfi K. Schlohsarczyk
- Institute of Veterinary Pathology, FB10—Veterinary Medicine, Justus-Liebig-University Giessen, 35392 Giessen, Germany
- Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Stephan Drewes
- Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Paweł Koteja
- Institute of Environmental Sciences, Faculty of Biology, Jagiellonian University, 30-387 Kraków, Poland
| | - Susanne Röhrs
- Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Rainer G. Ulrich
- Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Jens P. Teifke
- Institute of Veterinary Pathology, FB10—Veterinary Medicine, Justus-Liebig-University Giessen, 35392 Giessen, Germany
- Friedrich-Loeffler-Institut, 17493 Greifswald-Insel Riems, Germany
| | - Christiane Herden
- Institute of Veterinary Pathology, FB10—Veterinary Medicine, Justus-Liebig-University Giessen, 35392 Giessen, Germany
- Correspondence: ; Tel.: +49-6419938201
| |
Collapse
|
27
|
Sehgal A, Mehta S, Sahay K, Martynova E, Rizvanov A, Baranwal M, Chandy S, Khaiboullina S, Kabwe E, Davidyuk Y. Hemorrhagic Fever with Renal Syndrome in Asia: History, Pathogenesis, Diagnosis, Treatment, and Prevention. Viruses 2023; 15:v15020561. [PMID: 36851775 PMCID: PMC9966805 DOI: 10.3390/v15020561] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/30/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Hemorrhagic Fever with Renal Syndrome (HFRS) is the most frequently diagnosed zoonosis in Asia. This zoonotic infection is the result of exposure to the virus-contaminated aerosols. Orthohantavirus infection may cause Hemorrhagic Fever with Renal Syndrome (HRFS), a disease that is characterized by acute kidney injury and increased vascular permeability. Several species of orthohantaviruses were identified as causing infection, where Hantaan, Puumala, and Seoul viruses are most common. Orthohantaviruses are endemic to several Asian countries, such as China, South Korea, and Japan. Along with those countries, HFRS tops the list of zoonotic infections in the Far Eastern Federal District of Russia. Recently, orthohantavirus circulation was demonstrated in small mammals in Thailand and India, where orthohantavirus was not believed to be endemic. In this review, we summarized the current data on orthohantaviruses in Asia. We gave the synopsis of the history and diversity of orthohantaviruses in Asia. We also described the clinical presentation and current understanding of the pathogenesis of orthohantavirus infection. Additionally, conventional and novel approaches for preventing and treating orthohantavirus infection are discussed.
Collapse
Affiliation(s)
- Ayushi Sehgal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | - Sanya Mehta
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | - Kritika Sahay
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | - Ekaterina Martynova
- OpenLab “Gene and Cell Technologies”, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
| | - Albert Rizvanov
- OpenLab “Gene and Cell Technologies”, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, India
| | - Sara Chandy
- Childs Trust Medical Research Foundation, Kanchi Kamakoti Childs Trust Hospital, Chennai 600034, India
| | - Svetlana Khaiboullina
- OpenLab “Gene and Cell Technologies”, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
| | - Emmanuel Kabwe
- OpenLab “Gene and Cell Technologies”, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
- Kazan Research Institute of Epidemiology and Microbiology, Kazan 420012, Russia
| | - Yuriy Davidyuk
- OpenLab “Gene and Cell Technologies”, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
- Correspondence:
| |
Collapse
|
28
|
Menke L, Sperber HS, Aji AK, Chiantia S, Schwarzer R, Sieben C. Advances in fluorescence microscopy for orthohantavirus research. Microscopy (Oxf) 2023:6987530. [PMID: 36639937 DOI: 10.1093/jmicro/dfac075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 11/30/2022] [Accepted: 01/12/2023] [Indexed: 01/15/2023] Open
Abstract
Orthohantaviruses are important zoonotic pathogens responsible for a considerable disease burden globally. Partly due to our incomplete understanding of orthohantavirus replication, there is currently no effective antiviral treatment available. Recently, novel microscopy techniques and cutting-edge, automated image analysis algorithms have emerged, enabling to study cellular, subcellular and even molecular processes in unprecedented detail and depth. To date, fluorescence light microscopy allows us to visualize viral and cellular components and macromolecular complexes in live cells which in turn enables the study of specific steps of the viral replication cycle such as particle entry or protein trafficking at high temporal and spatial resolution. In this review, we highlight how fluorescence microscopy has provided new insights and improved our understanding of orthohantavirus biology. We discuss technical challenges such as studying live infected cells, give alternatives with recombinant protein expression and highlight future opportunities for example the application of super-resolution microscopy techniques, which has shown great potential in studies of different cellular processes and viral pathogens.
Collapse
Affiliation(s)
- Laura Menke
- Nanoscale Infection Biology Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Hannah S Sperber
- Institute for Translational HIV Research, University Hospital Essen, Essen, Germany
| | - Amit Koikkarah Aji
- University of Potsdam, Institute of Biochemistry and Biology, Department of Physical Biochemistry, Potsdam, Germany
| | - Salvatore Chiantia
- University of Potsdam, Institute of Biochemistry and Biology, Department of Physical Biochemistry, Potsdam, Germany
| | - Roland Schwarzer
- Institute for Translational HIV Research, University Hospital Essen, Essen, Germany
| | - Christian Sieben
- Nanoscale Infection Biology Helmholtz Centre for Infection Research, Braunschweig, Germany.,Institute of Genetics, Technische Universität Braunschweig, Braunschweig, Germany
| |
Collapse
|
29
|
Yuan F, Zheng A. Replicating-Competent VSV-Vectored Pseudotyped Viruses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1407:329-348. [PMID: 36920706 DOI: 10.1007/978-981-99-0113-5_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Vesicular stomatitis virus (VSV) is prototype virus in the family of Rhabdoviridae. Reverse genetic platform has enabled the genetic manipulation of VSV as a powerful live viral vector. Replicating-competent VSV is constructed by replacing the original VSV glycoprotein gene with heterologous envelope genes. The resulting recombinant viruses are able to replicate in permissive cells and incorporate the foreign envelope proteins on the surface of the viral particle without changing the bullet-shape morphology. Correspondingly, the cell tropism of replicating-competent VSV is determined by the foreign envelope proteins. Replicating-competent VSVs have been successfully used for selecting critical viral receptors or host factors, screening mutants that escape therapeutic antibodies, and developing VSV-based live viral vaccines.
Collapse
Affiliation(s)
- Fei Yuan
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Aihua Zheng
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
30
|
Ning T, Huang W, Min L, Yang Y, Liu S, Xu J, Zhang N, Xie SA, Zhu S, Wang Y. Pseudotyped Viruses for Orthohantavirus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1407:229-252. [PMID: 36920700 DOI: 10.1007/978-981-99-0113-5_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Orthohantaviruses, members of the Orthohantavirus genus of Hantaviridae family of the Bunyavirales order, are enveloped, negative-sense, single-stranded, tripartite RNA viruses. They are emerging zoonotic pathogens carried by small mammals including rodents, moles, shrews, and bats and are the etiologic agents of hemorrhagic fever with renal syndrome (HFRS) and hantavirus cardiopulmonary syndrome (HCPS) among humans. With the characteristics of low biological risk but strong operability, a variety of pseudotyped viruses have been constructed as alternatives to authentic orthohantaviruses to help delineate the roles of host factors in viral entry and other virus-host interactions, to assist in deciphering mechanisms of immune response and correlates of protection, to enhance our understanding of viral antigenic property, to characterize viral entry inhibitors, and to be developed as vaccines. In this chapter, we will discuss the general property of orthohantavirus, construction of pseudotyped orthohantaviruses based on different packaging systems, and their current applications.
Collapse
Affiliation(s)
- Tingting Ning
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Weijin Huang
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| | - Li Min
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Yi Yang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Si Liu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Junxuan Xu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Nan Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Si-An Xie
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Shengtao Zhu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China.
| | - Youchun Wang
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China. .,Institute of Medical Biology, Chinese Academy of Medicine Sciences & Peking Union Medical College, Kunming, China.
| |
Collapse
|
31
|
Zhang R. Studying Virus-Host Interactions with CRISPR Technology. Methods Mol Biol 2023; 2585:105-117. [PMID: 36331769 DOI: 10.1007/978-1-0716-2760-0_11] [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] [Indexed: 06/16/2023]
Abstract
The mosquito-borne West Nile virus (WNV) poses a great threat to public health as no vaccine or specific antiviral treatment is available. Exploring virus-host interactions, specifically host factors that are required for virus infection, is important for better understanding the biology, pathogenesis, and transmission of WNV. Such essential host factors may also represent antiviral targets. The development of CRISPR technology has provided a powerful and convenient tool to perturbate host gene expression, allowing for unbiased, genome-wide screens of host factors for virus infection. Here we describe the necessary steps for performing a CRISPR knockout screen, which can also be applied to other viruses, to identify host factors critical for WNV infection.
Collapse
Affiliation(s)
- Rong Zhang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
32
|
Gallo G, Kotlik P, Roingeard P, Monot M, Chevreux G, Ulrich RG, Tordo N, Ermonval M. Diverse susceptibilities and responses of human and rodent cells to orthohantavirus infection reveal different levels of cellular restriction. PLoS Negl Trop Dis 2022; 16:e0010844. [PMID: 36223391 PMCID: PMC9591050 DOI: 10.1371/journal.pntd.0010844] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 10/24/2022] [Accepted: 09/23/2022] [Indexed: 11/19/2022] Open
Abstract
Orthohantaviruses are rodent-borne emerging viruses that may cause severe diseases in humans but no apparent pathology in their small mammal reservoirs. However, the mechanisms leading to tolerance or pathogenicity in humans and persistence in rodent reservoirs are poorly understood, as is the manner in which they spread within and between organisms. Here, we used a range of cellular and molecular approaches to investigate the interactions of three different orthohantaviruses-Puumala virus (PUUV), responsible for a mild to moderate form of hemorrhagic fever with renal syndrome in humans, Tula virus (TULV) with low pathogenicity, and non-pathogenic Prospect Hill virus (PHV)-with human and rodent host cell lines. Besides the fact that cell susceptibility to virus infection was shown to depend on the cell type and virus strain, the three orthohantaviruses were able to infect Vero E6 and HuH7 human cells, but only the former secreted infectious particles. In cells derived from PUUV reservoir, the bank vole (Myodes glareolus), PUUV achieved a complete viral cycle, while TULV did not enter the cells and PHV infected them but did not produce infectious particles, reflecting differences in host specificity. A search for mature virions by electron microscopy (EM) revealed that TULV assembly occurred in part at the plasma membrane, whereas PHV particles were trapped in autophagic vacuoles in cells of the heterologous rodent host. We described differential interactions of orthohantaviruses with cellular factors, as supported by the cellular distribution of viral nucleocapsid protein with cell compartments, and proteomics identification of cellular partners. Our results also showed that interferon (IFN) dependent gene expression was regulated in a cell and virus species dependent manner. Overall, our study highlighted the complexity of the host-virus relationship and demonstrated that orthohantaviruses are restricted at different levels of the viral cycle. In addition, the study opens new avenues to further investigate how these viruses differ in their interactions with cells to evade innate immunity and how it depends on tissue type and host species.
Collapse
Affiliation(s)
- Giulia Gallo
- Institut Pasteur, Université Paris Cité, Département de Virologie, Unité des Stratégies Antivirales, Paris, France
- Sorbonne Université, Ecole Doctorale Complexité du Vivant, Paris, France
- * E-mail: (ME); (GG)
| | - Petr Kotlik
- Laboratory of Molecular Ecology, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Liběchov, Czech Republic
| | - Philippe Roingeard
- INSERM U1259 et plateforme IBISA de Microscopie Electronique, Université et CHRU de Tours, Tours, France
| | - Marc Monot
- Institut Pasteur, Université Paris Cité, Biomics Platform, C2RT, Paris, France
| | | | - Rainer G. Ulrich
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Partner site Hamburg-Lübeck-Borstel-Riems, German Centre for Infection Research (DZIF), Greifswald-Insel Riems, Germany
| | - Noël Tordo
- Institut Pasteur, Université Paris Cité, Département de Virologie, Unité des Stratégies Antivirales, Paris, France
- Institut Pasteur de Guinée, Conakry, Guinée
| | - Myriam Ermonval
- Institut Pasteur, Université Paris Cité, Département de Virologie, Unité des Stratégies Antivirales, Paris, France
- * E-mail: (ME); (GG)
| |
Collapse
|
33
|
Boshra H. An Overview of the Infectious Cycle of Bunyaviruses. Viruses 2022; 14:2139. [PMID: 36298693 PMCID: PMC9610998 DOI: 10.3390/v14102139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/19/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
Bunyaviruses represent the largest group of RNA viruses and are the causative agent of a variety of febrile and hemorrhagic illnesses. Originally characterized as a single serotype in Africa, the number of described bunyaviruses now exceeds over 500, with its presence detected around the world. These predominantly tri-segmented, single-stranded RNA viruses are transmitted primarily through arthropod and rodent vectors and can infect a wide variety of animals and plants. Although encoding for a small number of proteins, these viruses can inflict potentially fatal disease outcomes and have even developed strategies to suppress the innate antiviral immune mechanisms of the infected host. This short review will attempt to provide an overall description of the order Bunyavirales, describing the mechanisms behind their infection, replication, and their evasion of the host immune response. Furthermore, the historical context of these viruses will be presented, starting from their original discovery almost 80 years ago to the most recent research pertaining to viral replication and host immune response.
Collapse
Affiliation(s)
- Hani Boshra
- Global Urgent and Advanced Research and Development (GUARD), 911 Rue Principale, Batiscan, QC G0X 1A0, Canada
| |
Collapse
|
34
|
The Bovine Seminal Plasma Protein PDC-109 Possesses Pan-Antiviral Activity. Viruses 2022; 14:v14092031. [PMID: 36146836 PMCID: PMC9504757 DOI: 10.3390/v14092031] [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: 08/22/2022] [Revised: 09/08/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
Mammalian seminal plasma contains a multitude of bioactive components, including lipids, glucose, mineral elements, metabolites, proteins, cytokines, and growth factors, with various functions during insemination and fertilization. The seminal plasma protein PDC-109 is one of the major soluble components of the bovine ejaculate and is crucially important for sperm motility, capacitation, and acrosome reaction. A hitherto underappreciated function of seminal plasma is its anti-microbial and antiviral activity, which may limit the sexual transmission of infectious diseases during intercourse. We have recently discovered that PDC-109 inhibits the membrane fusion activity of influenza virus particles and significantly impairs viral infections at micromolar concentrations. Here we investigated whether the antiviral activity of PDC-109 is restricted to Influenza or if other mammalian viruses are similarly affected. We focused on Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2), the etiological agent of the Coronavirus Disease 19 (COVID-19), thoroughly assessing PDC-109 inhibition with SARS-CoV-2 Spike (S)-pseudotyped reporter virus particles, but also live-virus infections. Consistent with our previous publications, we found significant virus inhibition, albeit accompanied by substantial cytotoxicity. However, using time-of-addition experiments we discovered a treatment regimen that enables virus suppression without affecting cell viability. We furthermore demonstrated that PDC-109 is also able to impair infections mediated by the VSV glycoprotein (VSVg), thus indicating a broad pan-antiviral activity against multiple virus species and families.
Collapse
|
35
|
Ye Z, Yang Y, Wei Y, Li L, Wang X, Zhang J. PCDH1 promotes progression of pancreatic ductal adenocarcinoma via activation of NF-κB signalling by interacting with KPNB1. Cell Death Dis 2022; 13:633. [PMID: 35864095 PMCID: PMC9304345 DOI: 10.1038/s41419-022-05087-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 07/07/2022] [Accepted: 07/07/2022] [Indexed: 01/21/2023]
Abstract
Uncontrolled growth, distant metastasis and chemoresistance are critical characteristics of pancreatic ductal adenocarcinoma (PDAC), and they result in high mortality; however, the mechanisms triggering these effects have not been fully investigated. In this study, we analysed a dataset in the Cancer Genome Atlas (TCGA) and identified PCDH1, a rarely studied transmembrane protein, as a novel prognostic marker in PDAC patients. We demonstrated that PCDH1 expression was upregulated in PDAC tissues, and its expression levels were associated with the depth of tumour invasion and lymph node metastasis. Patients with high PCDH1 levels showed poor overall survival (OS). We also investigated the biological significance of PCDH1 in PDAC cell growth, metastasis, and side population (SP) phenotype acquisition and explored the internal molecular mechanisms of PCDH1 action. Our results demonstrated that PCDH1 enhanced p65 nuclear localization by interacting with KPNB1, a well-characterized nuclear transporter, thereby activating the NF-κB signalling pathway and increasing its functional effects during PDAC progression. Hence, our results indicate that PCDH1 can be used as a negative prognostic marker and may be a potential therapeutic target for PDAC patients.
Collapse
Affiliation(s)
- Zhihua Ye
- Department of Medical Oncology Center, Zhongshan City People's Hospital, 528403, Zhongshan City, Guangdong Province, P. R. China
| | - Yingyu Yang
- Department of Medical Oncology Center, Zhongshan City People's Hospital, 528403, Zhongshan City, Guangdong Province, P. R. China
| | - Ying Wei
- Department of Medical Oncology Center, Zhongshan City People's Hospital, 528403, Zhongshan City, Guangdong Province, P. R. China
| | - Lamei Li
- Department of Medical Oncology Center, Zhongshan City People's Hospital, 528403, Zhongshan City, Guangdong Province, P. R. China
| | - Xinyi Wang
- Department of Medical Oncology Center, Zhongshan City People's Hospital, 528403, Zhongshan City, Guangdong Province, P. R. China
| | - Junkai Zhang
- Department of Medical Oncology Center, Zhongshan City People's Hospital, 528403, Zhongshan City, Guangdong Province, P. R. China.
| |
Collapse
|
36
|
Nusshag C, Boegelein L, Schreiber P, Essbauer S, Osberghaus A, Zeier M, Krautkrämer E. Expression Profile of Human Renal Mesangial Cells Is Altered by Infection with Pathogenic Puumala Orthohantavirus. Viruses 2022; 14:v14040823. [PMID: 35458553 PMCID: PMC9025590 DOI: 10.3390/v14040823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/10/2022] [Accepted: 04/14/2022] [Indexed: 11/24/2022] Open
Abstract
Acute kidney injury (AKI) with proteinuria is a hallmark of infections with Eurasian orthohantaviruses. Different kidney cells are identified as target cells of hantaviruses. Mesangial cells may play a central role in the pathogenesis of AKI by regulation of inflammatory mediators and signaling cascades. Therefore, we examined the characteristics of hantavirus infection on human renal mesangial cells (HRMCs). Receptor expression and infection with pathogenic Puumala virus (PUUV) and low-pathogenic Tula virus (TULV) were explored. To analyze changes in protein expression in infected mesangial cells, we performed a proteome profiler assay analyzing 38 markers of kidney damage. We compared the proteome profile of in vitro-infected HRMCs with the profile detected in urine samples of 11 patients with acute hantavirus infection. We observed effective productive infection of HRMCs with pathogenic PUUV, but only poor abortive infection for low-pathogenic TULV. PUUV infection resulted in the deregulation of proteases, adhesion proteins, and cytokines associated with renal damage. The urinary proteome profile of hantavirus patients demonstrated also massive changes, which in part correspond to the alterations observed in the in vitro infection of HRMCs. The direct infection of mesangial cells may induce a local environment of signal mediators that contributes to AKI in hantavirus infection.
Collapse
Affiliation(s)
- Christian Nusshag
- Department of Nephrology, University of Heidelberg, D-69120 Heidelberg, Germany; (C.N.); (L.B.); (P.S.); (A.O.); (M.Z.)
| | - Lukas Boegelein
- Department of Nephrology, University of Heidelberg, D-69120 Heidelberg, Germany; (C.N.); (L.B.); (P.S.); (A.O.); (M.Z.)
| | - Pamela Schreiber
- Department of Nephrology, University of Heidelberg, D-69120 Heidelberg, Germany; (C.N.); (L.B.); (P.S.); (A.O.); (M.Z.)
| | - Sandra Essbauer
- Bundeswehr Institute of Microbiology, Department Virology and Intracellular Agents, German Centre for Infection Research, Munich Partner Site, D-80937 Munich, Germany;
| | - Anja Osberghaus
- Department of Nephrology, University of Heidelberg, D-69120 Heidelberg, Germany; (C.N.); (L.B.); (P.S.); (A.O.); (M.Z.)
| | - Martin Zeier
- Department of Nephrology, University of Heidelberg, D-69120 Heidelberg, Germany; (C.N.); (L.B.); (P.S.); (A.O.); (M.Z.)
| | - Ellen Krautkrämer
- Department of Nephrology, University of Heidelberg, D-69120 Heidelberg, Germany; (C.N.); (L.B.); (P.S.); (A.O.); (M.Z.)
- Correspondence:
| |
Collapse
|
37
|
Mittler E, Wec AZ, Tynell J, Guardado-Calvo P, Wigren-Byström J, Polanco LC, O’Brien CM, Slough MM, Abelson DM, Serris A, Sakharkar M, Pehau-Arnaudet G, Bakken RR, Geoghegan JC, Jangra RK, Keller M, Zeitlin L, Vapalahti O, Ulrich RG, Bornholdt ZA, Ahlm C, Rey FA, Dye JM, Bradfute SB, Strandin T, Herbert AS, Forsell MN, Walker LM, Chandran K. Human antibody recognizing a quaternary epitope in the Puumala virus glycoprotein provides broad protection against orthohantaviruses. Sci Transl Med 2022; 14:eabl5399. [PMID: 35294259 PMCID: PMC9805701 DOI: 10.1126/scitranslmed.abl5399] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The rodent-borne hantavirus Puumala virus (PUUV) and related agents cause hemorrhagic fever with renal syndrome (HFRS) in humans. Other hantaviruses, including Andes virus (ANDV) and Sin Nombre virus, cause a distinct zoonotic disease, hantavirus cardiopulmonary syndrome (HCPS). Although these infections are severe and have substantial case fatality rates, no FDA-approved hantavirus countermeasures are available. Recent work suggests that monoclonal antibodies may have therapeutic utility. We describe here the isolation of human neutralizing antibodies (nAbs) against tetrameric Gn/Gc glycoprotein spikes from PUUV-experienced donors. We define a dominant class of nAbs recognizing the "capping loop" of Gn that masks the hydrophobic fusion loops in Gc. A subset of nAbs in this class, including ADI-42898, bound Gn/Gc complexes but not Gn alone, strongly suggesting that they recognize a quaternary epitope encompassing both Gn and Gc. ADI-42898 blocked the cell entry of seven HCPS- and HFRS-associated hantaviruses, and single doses of this nAb could protect Syrian hamsters and bank voles challenged with the highly virulent HCPS-causing ANDV and HFRS-causing PUUV, respectively. ADI-42898 is a promising candidate for clinical development as a countermeasure for both HCPS and HFRS, and its mode of Gn/Gc recognition informs the development of broadly protective hantavirus vaccines.
Collapse
Affiliation(s)
- Eva Mittler
- Department of Microbiology and Immunology, Albert Einstein College of Medicine; Bronx, NY 10461, USA
| | | | - Janne Tynell
- Department of Clinical Microbiology, Umeå University; Umeå, Sweden.,Zoonosis Unit, Department of Virology, University of Helsinki; Helsinki, Finland
| | - Pablo Guardado-Calvo
- Structural Virology Unit, Department of Virology, Institut Pasteur; Paris 75724, France
| | | | - Laura C. Polanco
- Department of Microbiology and Immunology, Albert Einstein College of Medicine; Bronx, NY 10461, USA
| | - Cecilia M. O’Brien
- U.S. Army Medical Research Institute of Infectious Diseases; Fort Detrick, MD 21702, USA.,The Geneva Foundation; Tacoma, WA 98402, USA
| | - Megan M. Slough
- Department of Microbiology and Immunology, Albert Einstein College of Medicine; Bronx, NY 10461, USA
| | | | - Alexandra Serris
- Structural Virology Unit, Department of Virology, Institut Pasteur; Paris 75724, France
| | | | - Gerard Pehau-Arnaudet
- Structural Virology Unit, Department of Virology, Institut Pasteur; Paris 75724, France
| | - Russell R. Bakken
- U.S. Army Medical Research Institute of Infectious Diseases; Fort Detrick, MD 21702, USA
| | | | - Rohit K. Jangra
- Department of Microbiology and Immunology, Albert Einstein College of Medicine; Bronx, NY 10461, USA
| | - Markus Keller
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health; 17493 Greifswald-Insel Riems, Germany
| | - Larry Zeitlin
- Mapp Biopharmaceutical, Inc.; San Diego, CA 92121, USA
| | - Olli Vapalahti
- Zoonosis Unit, Department of Virology, University of Helsinki; Helsinki, Finland.,Veterinary Biosciences, Veterinary Faculty, University of Helsinki; Helsinki, Finland
| | - Rainer G. Ulrich
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health; 17493 Greifswald-Insel Riems, Germany.,Deutsches Zentrum für Infektionsforschung, Partner site Hamburg-Lübeck-Borstel-Riems; Greifswald-Insel Riems, Germany
| | | | - Clas Ahlm
- Department of Clinical Microbiology, Umeå University; Umeå, Sweden
| | - Felix A. Rey
- Structural Virology Unit, Department of Virology, Institut Pasteur; Paris 75724, France
| | - John M. Dye
- U.S. Army Medical Research Institute of Infectious Diseases; Fort Detrick, MD 21702, USA
| | - Steven B. Bradfute
- University of New Mexico Health Science Center, Center for Global Health, Department of Internal Medicine; Albuquerque, NM 87131, USA
| | - Tomas Strandin
- Zoonosis Unit, Department of Virology, University of Helsinki; Helsinki, Finland.,Correspondence: (T.S.), (A.S.H.), (M.N.E.F.), (L.M.W.), (K.C.)
| | - Andrew S. Herbert
- U.S. Army Medical Research Institute of Infectious Diseases; Fort Detrick, MD 21702, USA.,The Geneva Foundation; Tacoma, WA 98402, USA.,Correspondence: (T.S.), (A.S.H.), (M.N.E.F.), (L.M.W.), (K.C.)
| | - Mattias N.E. Forsell
- Department of Clinical Microbiology, Umeå University; Umeå, Sweden.,Correspondence: (T.S.), (A.S.H.), (M.N.E.F.), (L.M.W.), (K.C.)
| | - Laura M. Walker
- Adimab, LLC; Lebanon, NH 03766, USA.,Adagio Therapeutics, Inc.; Waltham, MA 02451, USA.,Correspondence: (T.S.), (A.S.H.), (M.N.E.F.), (L.M.W.), (K.C.)
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine; Bronx, NY 10461, USA.,Correspondence: (T.S.), (A.S.H.), (M.N.E.F.), (L.M.W.), (K.C.)
| |
Collapse
|
38
|
Roman-Sosa G, Leske A, Ficht X, Dau TH, Holzerland J, Hoenen T, Beer M, Kammerer R, Schirmbeck R, Rey FA, Cordo SM, Groseth A. Immunization with GP1 but Not Core-like Particles Displaying Isolated Receptor-Binding Epitopes Elicits Virus-Neutralizing Antibodies against Junín Virus. Vaccines (Basel) 2022; 10:vaccines10020173. [PMID: 35214632 PMCID: PMC8874384 DOI: 10.3390/vaccines10020173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 02/01/2023] Open
Abstract
New World arenaviruses are rodent-transmitted viruses and include a number of pathogens that are responsible for causing severe human disease. This includes Junín virus (JUNV), which is the causative agent of Argentine hemorrhagic fever. The wild nature and mobility of the rodent reservoir host makes it difficult to control the disease, and currently passive immunization with high-titer neutralizing antibody-containing plasma from convalescent patients is the only specific therapy. However, dwindling supplies of naturally available convalescent plasma, and challenges in developing similar resources for other closely related viruses, have made the development of alternative antibody-based therapeutic approaches of critical importance. In this study, we sought to induce a neutralizing antibody response in rabbits against the receptor-binding subunit of the viral glycoprotein, GP1, and the specific peptide sequences in GP1 involved in cellular receptor contacts. While these specific receptor-interacting peptides did not efficiently induce the production of neutralizing antibodies when delivered as a particulate antigen (as part of hepatitis B virus core-like particles), we showed that recombinant JUNV GP1 purified from transfected mammalian cells induced virus-neutralizing antibodies at high titers in rabbits. Further, neutralization was observed across a range of unrelated JUNV strains, a feature that is critical for effectiveness in the field. These results underscore the potential of GP1 alone to induce a potent neutralizing antibody response and highlight the importance of epitope presentation. In addition, effective virus neutralization by rabbit antibodies supports the potential applicability of this species for the future development of immunotherapeutics (e.g., based on humanized monoclonal antibodies). Such information can be applied in the design of vaccines and immunogens for both prevention and specific therapies against this and likely also other closely related pathogenic New World arenaviruses.
Collapse
Affiliation(s)
- Gleyder Roman-Sosa
- Department of Internal Medicine I, Ulm University Hospital, 89081 Ulm, Germany; (X.F.); (R.S.)
- Correspondence: (G.R.-S.); (A.G.)
| | - Anne Leske
- Laboratory for Arenavirus Biology, Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, 17493 Greifswald, Germany; (A.L.); (J.H.)
| | - Xenia Ficht
- Department of Internal Medicine I, Ulm University Hospital, 89081 Ulm, Germany; (X.F.); (R.S.)
| | - Tung Huy Dau
- Laboratory for Immunogenetics, Institute of Immunology, Friedrich-Loeffler-Institut, 17493 Greifswald, Germany; (T.H.D.); (R.K.)
| | - Julia Holzerland
- Laboratory for Arenavirus Biology, Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, 17493 Greifswald, Germany; (A.L.); (J.H.)
| | - Thomas Hoenen
- Laboratory for Integrative Cell and Infection Biology, Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, 17493 Greifswald, Germany;
| | - Martin Beer
- National and OIE Reference Laboratory for BHV-1, Institute of Diagnostic Virology, Friedrich-Loeffler-Institut, 17493 Greifswald, Germany;
| | - Robert Kammerer
- Laboratory for Immunogenetics, Institute of Immunology, Friedrich-Loeffler-Institut, 17493 Greifswald, Germany; (T.H.D.); (R.K.)
| | - Reinhold Schirmbeck
- Department of Internal Medicine I, Ulm University Hospital, 89081 Ulm, Germany; (X.F.); (R.S.)
| | - Felix A. Rey
- Structural Virology Unit, CNRS UMR3569, Institut Pasteur, Université de Paris, 75015 Paris, France;
| | - Sandra M. Cordo
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), University of Buenos Aires, Ciudad Universitaria, Pabellón II, Piso 4, Buenos Aires 1428, Argentina;
| | - Allison Groseth
- Laboratory for Arenavirus Biology, Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, 17493 Greifswald, Germany; (A.L.); (J.H.)
- Correspondence: (G.R.-S.); (A.G.)
| |
Collapse
|
39
|
Koehler FC, Di Cristanziano V, Späth MR, Hoyer-Allo KJR, Wanken M, Müller RU, Burst V. OUP accepted manuscript. Clin Kidney J 2022; 15:1231-1252. [PMID: 35756741 PMCID: PMC9217627 DOI: 10.1093/ckj/sfac008] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Indexed: 01/18/2023] Open
Abstract
Hantavirus-induced diseases are emerging zoonoses with endemic appearances and frequent outbreaks in different parts of the world. In humans, hantaviral pathology is characterized by the disruption of the endothelial cell barrier followed by increased capillary permeability, thrombocytopenia due to platelet activation/depletion and an overactive immune response. Genetic vulnerability due to certain human leukocyte antigen haplotypes is associated with disease severity. Typically, two different hantavirus-caused clinical syndromes have been reported: hemorrhagic fever with renal syndrome (HFRS) and hantavirus cardiopulmonary syndrome (HCPS). The primarily affected vascular beds differ in these two entities: renal medullary capillaries in HFRS caused by Old World hantaviruses and pulmonary capillaries in HCPS caused by New World hantaviruses. Disease severity in HFRS ranges from mild, e.g. Puumala virus-associated nephropathia epidemica, to moderate, e.g. Hantaan or Dobrava virus infections. HCPS leads to a severe acute respiratory distress syndrome with high mortality rates. Due to novel insights into organ tropism, hantavirus-associated pathophysiology and overlapping clinical features, HFRS and HCPS are believed to be interconnected syndromes frequently involving the kidneys. As there are no specific antiviral treatments or vaccines approved in Europe or the USA, only preventive measures and public awareness may minimize the risk of hantavirus infection. Treatment remains primarily supportive and, depending on disease severity, more invasive measures (e.g., renal replacement therapy, mechanical ventilation and extracorporeal membrane oxygenation) are needed.
Collapse
Affiliation(s)
- Felix C Koehler
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- CECAD, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Veronica Di Cristanziano
- Institute of Virology, University of Cologne, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
| | - Martin R Späth
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- CECAD, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - K Johanna R Hoyer-Allo
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- CECAD, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Manuel Wanken
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Roman-Ulrich Müller
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- CECAD, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | | |
Collapse
|
40
|
Xian X, Wang Y, Liu G. Genetically Engineered Hamster Models of Dyslipidemia and Atherosclerosis. Methods Mol Biol 2022; 2419:433-459. [PMID: 35237980 DOI: 10.1007/978-1-0716-1924-7_26] [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] [Indexed: 06/14/2023]
Abstract
Animal models of human diseases play an extremely important role in biomedical research. Among them, mice are widely used animal models for translational research, especially because of ease of generation of genetically engineered mice. However, because of the great differences in biology between mice and humans, translation of findings to humans remains a major issue. Therefore, the exploration of models with biological and metabolic characteristics closer to those of humans has never stopped.Although pig and nonhuman primates are biologically similar to humans, their genetic engineering is technically difficult, the cost of breeding is high, and the experimental time is long. As a result, the application of these species as model animals, especially genetically engineered model animals, in biomedical research is greatly limited.In terms of lipid metabolism and cardiovascular diseases, hamsters have several characteristics different from rats and mice, but similar to those in humans. The hamster is therefore an ideal animal model for studying lipid metabolism and cardiovascular disease because of its small size and short reproduction period. However, the phenomenon of zygote division, which was unexpectedly blocked during the manipulation of hamster embryos for some unknown reasons, had plagued researchers for decades and no genetically engineered hamsters have therefore been generated as animal models of human diseases for a long time. After solving the problem of in vitro development of hamster zygotes, we successfully prepared enhanced green fluorescent protein (eGFP) transgenic hamsters by microinjection of lentiviral vectors into the zona pellucida space of zygotes. On this basis, we started the development of cardiovascular disease models using the hamster embryo culture system combined with the novel genome editing technique of clustered regularly interspaced short palindromic repeats (CRISPR )/CRISPR associated protein 9 (Cas9). In this chapter, we will introduce some of the genetically engineered hamster models with dyslipidemia and the corresponding characteristics of these models. We hope that the genetically engineered hamster models can be further recognized and complement other genetically engineered animal models such as mice, rats, and rabbits. This will lead to new avenues and pathways for the study of lipid metabolism and its related diseases.
Collapse
Affiliation(s)
- Xunde Xian
- Institute of Cardiovascular Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yuhui Wang
- Institute of Cardiovascular Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, School of Basic Medical Sciences, Peking University, Beijing, China
| | - George Liu
- Institute of Cardiovascular Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, School of Basic Medical Sciences, Peking University, Beijing, China.
| |
Collapse
|
41
|
Abstract
To identify novel host factors as putative targets to reverse HIV-1 latency, we performed an insertional mutagenesis genetic screen in a latent HIV-1 infected pseudohaploid KBM7 cell line (Hap-Lat). Following mutagenesis, insertions were mapped to the genome, and bioinformatic analysis resulted in the identification of 69 candidate host genes involved in maintaining HIV-1 latency. A select set of candidate genes was functionally validated using short hairpin RNA (shRNA)-mediated depletion in latent HIV-1 infected J-Lat A2 and 11.1 T cell lines. We confirmed ADK, CHD9, CMSS1, EVI2B, EXOSC8, FAM19A, GRIK5, IRF2BP2, NF1, and USP15 as novel host factors involved in the maintenance of HIV-1 latency. Chromatin immunoprecipitation assays indicated that CHD9, a chromodomain helicase DNA-binding protein, maintains HIV-1 latency via direct association with the HIV-1 5′ long terminal repeat (LTR), and its depletion results in increased histone acetylation at the HIV-1 promoter, concomitant with HIV-1 latency reversal. FDA-approved inhibitors 5-iodotubercidin, trametinib, and topiramate, targeting ADK, NF1, and GRIK5, respectively, were characterized for their latency reversal potential. While 5-iodotubercidin exhibited significant cytotoxicity in both J-Lat and primary CD4+ T cells, trametinib reversed latency in J-Lat cells but not in latent HIV-1 infected primary CD4+ T cells. Importantly, topiramate reversed latency in cell line models, in latently infected primary CD4+ T cells, and crucially in CD4+ T cells from three people living with HIV-1 (PLWH) under suppressive antiretroviral therapy, without inducing T cell activation or significant toxicity. Thus, using an adaptation of a haploid forward genetic screen, we identified novel and druggable host factors contributing to HIV-1 latency.
Collapse
|
42
|
Guardado-Calvo P, Rey FA. The Viral Class II Membrane Fusion Machinery: Divergent Evolution from an Ancestral Heterodimer. Viruses 2021; 13:v13122368. [PMID: 34960636 PMCID: PMC8706100 DOI: 10.3390/v13122368] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/24/2021] [Accepted: 11/25/2021] [Indexed: 02/06/2023] Open
Abstract
A key step during the entry of enveloped viruses into cells is the merger of viral and cell lipid bilayers. This process is driven by a dedicated membrane fusion protein (MFP) present at the virion surface, which undergoes a membrane–fusogenic conformational change triggered by interactions with the target cell. Viral MFPs have been extensively studied structurally, and are divided into three classes depending on their three-dimensional fold. Because MFPs of the same class are found in otherwise unrelated viruses, their intra-class structural homology indicates horizontal gene exchange. We focus this review on the class II fusion machinery, which is composed of two glycoproteins that associate as heterodimers. They fold together in the ER of infected cells such that the MFP adopts a conformation primed to react to specific clues only upon contact with a target cell, avoiding premature fusion in the producer cell. We show that, despite having diverged in their 3D fold during evolution much more than the actual MFP, the class II accompanying proteins (AP) also derive from a distant common ancestor, displaying an invariant core formed by a β-ribbon and a C-terminal immunoglobulin-like domain playing different functional roles—heterotypic interactions with the MFP, and homotypic AP/AP contacts to form spikes, respectively. Our analysis shows that class II APs are easily identifiable with modern structural prediction algorithms, providing useful information in devising immunogens for vaccine design.
Collapse
|
43
|
Kell AM. Innate Immunity to Orthohantaviruses: Could Divergent Immune Interactions Explain Host-specific Disease Outcomes? J Mol Biol 2021; 434:167230. [PMID: 34487792 PMCID: PMC8894506 DOI: 10.1016/j.jmb.2021.167230] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 10/20/2022]
Abstract
The genus Orthohantavirus (family Hantaviridae, order Bunyavirales) consists of numerous genetic and pathologically distinct viral species found within rodent and mammalian insectivore populations world-wide. Although reservoir hosts experience persistent asymptomatic infection, numerous rodent-borne orthohantaviruses cause severe disease when transmitted to humans, with case-fatality rates up to 40%. The first isolation of an orthohantavirus occurred in 1976 and, since then, the field has made significant progress in understanding the immune correlates of disease, viral interactions with the human innate immune response, and the immune kinetics of reservoir hosts. Much still remains elusive regarding the molecular mechanisms of orthohantavirus recognition by the innate immune response and viral antagonism within the reservoir host, however. This review provides a summary of the last 45 years of research into orthohantavirus interaction with the host innate immune response. This summary includes discussion of current knowledge involving human, non-reservoir rodent, and reservoir innate immune responses to viruses which cause hemorrhagic fever with renal syndrome and hantavirus cardio-pulmonary syndrome. Review of the literature concludes with a brief proposition for the development of novel tools needed to drive forward investigations into the molecular mechanisms of innate immune activation and consequences for disease outcomes in the various hosts for orthohantaviruses.
Collapse
Affiliation(s)
- Alison M Kell
- Department of Molecular Genetics and Microbiology, University of New Mexico, 915 Camino de Salud, Albuquerque, NM 87131, United States.
| |
Collapse
|
44
|
Guardado-Calvo P, Rey FA. The surface glycoproteins of hantaviruses. Curr Opin Virol 2021; 50:87-94. [PMID: 34418649 DOI: 10.1016/j.coviro.2021.07.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 11/30/2022]
Abstract
Hantaviruses are rodent-borne viruses distributed worldwide, transmitted through the air and with the ability to spread from person to person. They maintain a non-symptomatic persistent infection in their rodent hosts, but their spillover to humans produces a renal or pulmonary syndrome associated with high fatality rates. Hantavirus particles are lipid-enveloped and display a characteristic surface lattice built up of tetragonal spikes composed of two glycoproteins, Gn and Gc. The pleomorphism of these particles has hindered cryo-EM efforts to obtain detailed structural information and only by using a combination of X-ray crystallography and cryo-electron tomography it was possible to build an atomic model of the surface lattice. Here we review these structural efforts and the unanticipated evolutionary relations between hantaviruses and alphaviruses highlighted by these studies.
Collapse
Affiliation(s)
| | - Félix A Rey
- Institut Pasteur, Structural Virology Unit, and CNRS UMR 3569, Paris, France
| |
Collapse
|
45
|
Hägele S, Nusshag C, Müller A, Baumann A, Zeier M, Krautkrämer E. Cells of the human respiratory tract support the replication of pathogenic Old World orthohantavirus Puumala. Virol J 2021; 18:169. [PMID: 34404450 PMCID: PMC8369447 DOI: 10.1186/s12985-021-01636-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 08/09/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Transmission of all known pathogenic orthohantaviruses (family Hantaviridae) usually occurs via inhalation of aerosols contaminated with viral particles derived from infected rodents and organ manifestation of infections is characterized by lung and kidney involvement. Orthohantaviruses found in Eurasia cause hemorrhagic fever with renal syndrome (HFRS) and New World orthohantaviruses cause hantavirus cardiopulmonary syndrome (HCPS). However, cases of infection with Old World orthohantaviruses with severe pulmonary manifestations have also been observed. Therefore, human airway cells may represent initial targets for orthohantavirus infection and may also play a role in the pathogenesis of infections with Eurasian orthohantaviruses. METHODS We analyzed the permissiveness of primary endothelial cells of the human pulmonary microvasculature and of primary human epithelial cells derived from bronchi, bronchioles and alveoli for Old World orthohantavirus Puumala virus (PUUV) in vitro. In addition, we examined the expression of orthohantaviral receptors in these cell types. To minimize donor-specific effects, cells from two different donors were tested for each cell type. RESULTS Productive infection with PUUV was observed for endothelial cells of the microvasculature and for the three tested epithelial cell types derived from different sites of the respiratory tract. Interestingly, infection and particle release were also detected in bronchial and bronchiolar epithelial cells although expression of the orthohantaviral receptor integrin β3 was not detectable in these cell types. In addition, replication kinetics and viral release demonstrate enormous donor-specific variations. CONCLUSIONS The human respiratory epithelium is among the first targets of orthohantaviral infection and may contribute to virus replication, dissemination and pathogenesis of HFRS-causing orthohantaviruses. Differences in initial pulmonary infection due to donor-specific factors may play a role in the observed broad variance of severity and symptoms of orthohantavirus disease in patients. The absence of detectable levels of integrin αVβ3 surface expression on bronchial and small airway epithelial cells indicates an alternate mode of orthohantaviral entry in these cells that is independent from integrin β3.
Collapse
Affiliation(s)
- Stefan Hägele
- Department of Nephrology, University of Heidelberg, Im Neuenheimer Feld 162, 69120, Heidelberg, Germany
| | - Christian Nusshag
- Department of Nephrology, University of Heidelberg, Im Neuenheimer Feld 162, 69120, Heidelberg, Germany
| | - Alexander Müller
- Department of Nephrology, University of Heidelberg, Im Neuenheimer Feld 162, 69120, Heidelberg, Germany
| | - Alexandra Baumann
- Department of Nephrology, University of Heidelberg, Im Neuenheimer Feld 162, 69120, Heidelberg, Germany
| | - Martin Zeier
- Department of Nephrology, University of Heidelberg, Im Neuenheimer Feld 162, 69120, Heidelberg, Germany
| | - Ellen Krautkrämer
- Department of Nephrology, University of Heidelberg, Im Neuenheimer Feld 162, 69120, Heidelberg, Germany.
| |
Collapse
|
46
|
Meier K, Thorkelsson SR, Quemin ERJ, Rosenthal M. Hantavirus Replication Cycle-An Updated Structural Virology Perspective. Viruses 2021; 13:1561. [PMID: 34452426 PMCID: PMC8402763 DOI: 10.3390/v13081561] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/31/2021] [Accepted: 08/02/2021] [Indexed: 11/17/2022] Open
Abstract
Hantaviruses infect a wide range of hosts including insectivores and rodents and can also cause zoonotic infections in humans, which can lead to severe disease with possible fatal outcomes. Hantavirus outbreaks are usually linked to the population dynamics of the host animals and their habitats being in close proximity to humans, which is becoming increasingly important in a globalized world. Currently there is neither an approved vaccine nor a specific and effective antiviral treatment available for use in humans. Hantaviruses belong to the order Bunyavirales with a tri-segmented negative-sense RNA genome. They encode only five viral proteins and replicate and transcribe their genome in the cytoplasm of infected cells. However, many details of the viral amplification cycle are still unknown. In recent years, structural biology methods such as cryo-electron tomography, cryo-electron microscopy, and crystallography have contributed essentially to our understanding of virus entry by membrane fusion as well as genome encapsidation by the nucleoprotein. In this review, we provide an update on the hantavirus replication cycle with a special focus on structural virology aspects.
Collapse
Affiliation(s)
- Kristina Meier
- Department of Virology, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany;
| | - Sigurdur R. Thorkelsson
- Centre for Structural Systems Biology, Leibniz Institute for Experimental Virology, University of Hamburg, 22607 Hamburg, Germany;
| | - Emmanuelle R. J. Quemin
- Centre for Structural Systems Biology, Leibniz Institute for Experimental Virology, University of Hamburg, 22607 Hamburg, Germany;
| | - Maria Rosenthal
- Department of Virology, Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany;
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 22525 Hamburg, Germany
| |
Collapse
|
47
|
Dieterle ME, Solà-Riera C, Ye C, Goodfellow SM, Mittler E, Kasikci E, Bradfute SB, Klingström J, Jangra RK, Chandran K. Genetic depletion studies inform receptor usage by virulent hantaviruses in human endothelial cells. eLife 2021; 10:e69708. [PMID: 34232859 PMCID: PMC8263056 DOI: 10.7554/elife.69708] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/28/2021] [Indexed: 11/13/2022] Open
Abstract
Hantaviruses are RNA viruses with known epidemic threat and potential for emergence. Several rodent-borne hantaviruses cause zoonoses accompanied by severe illness and death. However, assessments of zoonotic risk and the development of countermeasures are challenged by our limited knowledge of the molecular mechanisms of hantavirus infection, including the identities of cell entry receptors and their roles in influencing viral host range and virulence. Despite the long-standing presumption that β3/β1-containing integrins are the major hantavirus entry receptors, rigorous genetic loss-of-function evidence supporting their requirement, and that of decay-accelerating factor (DAF), is lacking. Here, we used CRISPR/Cas9 engineering to knockout candidate hantavirus receptors, singly and in combination, in a human endothelial cell line that recapitulates the properties of primary microvascular endothelial cells, the major targets of viral infection in humans. The loss of β3 integrin, β1 integrin, and/or DAF had little or no effect on entry by a large panel of hantaviruses. By contrast, loss of protocadherin-1, a recently identified entry receptor for some hantaviruses, substantially reduced hantavirus entry and infection. We conclude that major host molecules necessary for endothelial cell entry by PCDH1-independent hantaviruses remain to be discovered.
Collapse
Affiliation(s)
- Maria Eugenia Dieterle
- Department of Microbiology and Immunology, Albert Einstein College of MedicineBronxUnited States
| | - Carles Solà-Riera
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska InstitutetStockholmSweden
| | - Chunyan Ye
- University of New Mexico Health Science Center, Center for Global Health, Department of Internal MedicineAlbuquerqueUnited States
| | - Samuel M Goodfellow
- University of New Mexico Health Science Center, Center for Global Health, Department of Internal MedicineAlbuquerqueUnited States
| | - Eva Mittler
- Department of Microbiology and Immunology, Albert Einstein College of MedicineBronxUnited States
| | - Ezgi Kasikci
- Department of Microbiology and Immunology, Albert Einstein College of MedicineBronxUnited States
| | - Steven B Bradfute
- University of New Mexico Health Science Center, Center for Global Health, Department of Internal MedicineAlbuquerqueUnited States
| | - Jonas Klingström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska InstitutetStockholmSweden
| | - Rohit K Jangra
- Department of Microbiology and Immunology, Albert Einstein College of MedicineBronxUnited States
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of MedicineBronxUnited States
| |
Collapse
|
48
|
Saavedra F, Díaz FE, Retamal‐Díaz A, Covián C, González PA, Kalergis AM. Immune response during hantavirus diseases: implications for immunotherapies and vaccine design. Immunology 2021; 163:262-277. [PMID: 33638192 PMCID: PMC8207335 DOI: 10.1111/imm.13322] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/05/2021] [Accepted: 02/15/2021] [Indexed: 12/18/2022] Open
Abstract
Orthohantaviruses, previously named hantaviruses, cause two emerging zoonotic diseases: haemorrhagic fever with renal syndrome (HFRS) in Eurasia and hantavirus cardiopulmonary syndrome (HCPS) in the Americas. Overall, over 200 000 cases are registered every year worldwide, with a fatality rate ranging between 0·1% and 15% for HFRS and between 20% and 40% for HCPS. No specific treatment or vaccines have been approved by the U.S. Food and Drug Administration (FDA) to treat or prevent hantavirus-caused syndromes. Currently, little is known about the mechanisms at the basis of hantavirus-induced disease. However, it has been hypothesized that an excessive inflammatory response plays an essential role in the course of the disease. Furthermore, the contributions of the cellular immune response to either viral clearance or pathology have not been fully elucidated. This article discusses recent findings relative to the immune responses elicited to hantaviruses in subjects suffering HFRS or HCPS, highlighting the similarities and differences between these two clinical diseases. Also, we summarize the most recent data about the cellular immune response that could be important for designing new vaccines to prevent this global public health problem.
Collapse
Affiliation(s)
- Farides Saavedra
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiagoChile
| | - Fabián E. Díaz
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiagoChile
| | - Angello Retamal‐Díaz
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiagoChile
| | - Camila Covián
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiagoChile
| | - Pablo A. González
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiagoChile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiagoChile
- Millennium Institute on Immunology and ImmunotherapyDepartamento de EndocrinologíaFacultad de MedicinaEscuela de MedicinaPontificia Universidad Católica de ChileSantiagoChile
| |
Collapse
|
49
|
Fukunaga K, Tanji M, Hanzawa N, Kuroda H, Inui M. Protocadherin-1 is expressed in the notochord of mouse embryo but is dispensable for its formation. Biochem Biophys Rep 2021; 27:101047. [PMID: 34189280 PMCID: PMC8219654 DOI: 10.1016/j.bbrep.2021.101047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 05/25/2021] [Accepted: 06/06/2021] [Indexed: 11/18/2022] Open
Abstract
Notochord is an embryonic midline structure that serves as mechanical support for axis elongation and the signaling center for the surrounding tissues. Precursors of notochord are initially induced in the dorsal most mesoderm region in gastrulating embryo and separate from the surrounding mesoderm/endoderm tissue to form an elongated rod-like structure, suggesting that cell adhesion molecules may play an important role in this step. In Xenopus embryo, axial protocadherin (AXPC), an orthologue of mammalian Protocadherin-1 (PCDH1), is indispensable for the assembly and separation from the surrounding tissue of the notochord cells. However, the role of PCDH1 in mammalian notochord remains unknown. We herein report that PCDH1 is expressed in the notochord of mouse embryo and that PCDH1-deficient mice form notochord normally. First, we examined the temporal expression pattern of pcdh1 and found that pcdh1 mRNA was expressed from embryonic day (E) 7.5, prior to the stage when notochord cells detach from the surrounding endoderm tissue. Second, we found that PCDH1 protein is expressed in the notochord of mouse embryos in addition to the previously reported expression in endothelial cells. To further investigate the role of PCDH1 in embryonic development, we generated PCDH1-deficient mice using the CRISPR-Cas9 system. In PCDH1-deficient embryos, notochord formation and separation from the surrounding tissue were normal. Structure and marker gene expression of notochord were also unaffected by loss of PCDH1. Major vascular patterns in PCDH1-deficient embryo were essentially normal. These results suggest that PCDH1 is dispensable for notochord formation, including the tissue separation process, in mammalian embryos. We successfully identified the evolutionary conserved expression of PCDH1 in notochord, but its function may differ among species.
Collapse
Affiliation(s)
- Kanako Fukunaga
- Systems Biology Program, Graduate School of Media and Governance, Keio University, Kanagawa, 252-0882, Japan
- Institute for Advanced Biosciences, Keio University, Kanagawa, 252-0882, Japan
- Laboratory of Animal Regeneration Systemology, Department of Life Sciences, School of Agriculture, Meiji University, Kanagawa, 214-8571, Japan
| | - Masafumi Tanji
- Laboratory of Animal Regeneration Systemology, Department of Life Sciences, School of Agriculture, Meiji University, Kanagawa, 214-8571, Japan
| | - Nana Hanzawa
- Laboratory of Animal Regeneration Systemology, Department of Life Sciences, School of Agriculture, Meiji University, Kanagawa, 214-8571, Japan
| | - Hiroki Kuroda
- Institute for Advanced Biosciences, Keio University, Kanagawa, 252-0882, Japan
- Faculty of Environment and Information Studies, Keio University, Kanagawa, 252-0882, Japan
| | - Masafumi Inui
- Laboratory of Animal Regeneration Systemology, Department of Life Sciences, School of Agriculture, Meiji University, Kanagawa, 214-8571, Japan
- Department of Systems BioMedicine, National Institute for Child Health and Development, Tokyo, 157-8535, Japan
- Corresponding author. Laboratory of Animal Regeneration Systemology, Department of Life Science, School of Agriculture, Meiji University, Kanagawa, 214-8571, Japan.
| |
Collapse
|
50
|
Engdahl TB, Kuzmina NA, Ronk AJ, Mire CE, Hyde MA, Kose N, Josleyn MD, Sutton RE, Mehta A, Wolters RM, Lloyd NM, Valdivieso FR, Ksiazek TG, Hooper JW, Bukreyev A, Crowe JE. Broad and potently neutralizing monoclonal antibodies isolated from human survivors of New World hantavirus infection. Cell Rep 2021; 35:109086. [PMID: 33951434 PMCID: PMC8142553 DOI: 10.1016/j.celrep.2021.109086] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 03/17/2021] [Accepted: 04/14/2021] [Indexed: 01/01/2023] Open
Abstract
New World hantaviruses (NWHs) are endemic in North and South America and cause hantavirus cardiopulmonary syndrome (HCPS), with a case fatality rate of up to 40%. Knowledge of the natural humoral immune response to NWH infection is limited. Here, we describe human monoclonal antibodies (mAbs) isolated from individuals previously infected with Sin Nombre virus (SNV) or Andes virus (ANDV). Most SNV-reactive antibodies show broad recognition and cross-neutralization of both New and Old World hantaviruses, while many ANDV-reactive antibodies show activity for ANDV only. mAbs ANDV-44 and SNV-53 compete for binding to a distinct site on the ANDV surface glycoprotein and show potently neutralizing activity to New and Old World hantaviruses. Four mAbs show therapeutic efficacy at clinically relevant doses in hamsters. These studies reveal a convergent and potently neutralizing human antibody response to NWHs and suggest therapeutic potential for human mAbs against HCPS. Engdahl et al. show that monoclonal antibodies isolated from human survivors of New World hantavirus infection display broad and potent neutralization across hantavirus species and recognize distinct sites on the glycoprotein spike. Multiple antibodies demonstrate potential therapeutic candidates for New World hantavirus infection. Some antibodies also neutralized Old World hantaviruses.
Collapse
Affiliation(s)
- Taylor B Engdahl
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Natalia A Kuzmina
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - Adam J Ronk
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - Chad E Mire
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA; Animal Resource Center, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Matthew A Hyde
- Animal Resource Center, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Nurgun Kose
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Matthew D Josleyn
- Virology Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Rachel E Sutton
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Apoorva Mehta
- Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Rachael M Wolters
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Nicole M Lloyd
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - Francisca R Valdivieso
- Programa Hantavirus, Instituto de Ciencias e Innovación en Medicina (ICIM), Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago 7590943, Chile
| | - Thomas G Ksiazek
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA
| | - Jay W Hooper
- Virology Division, US Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD 21702, USA
| | - Alexander Bukreyev
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, Galveston, TX 77550, USA.
| | - James E Crowe
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| |
Collapse
|