1
|
Souza LRQ, Pedrosa CGDS, Puig-Pijuan T, da Silva Dos Santos C, Vitória G, Delou JMA, Setti-Perdigão P, Higa LM, Tanuri A, Rehen SK, Guimarães MZP. Saxitoxin potentiates human neuronal cell death induced by Zika virus while sparing neural progenitors and astrocytes. Sci Rep 2024; 14:22809. [PMID: 39354036 PMCID: PMC11445263 DOI: 10.1038/s41598-024-73873-z] [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: 10/04/2024] Open
Abstract
The Zika virus (ZIKV) epidemic declared in Brazil between 2015 and 2016 was associated with an increased prevalence of severe congenital malformations, including microcephaly. The distribution of microcephaly cases was not uniform across the country, with a disproportionately higher incidence in the Northeast region (NE). Our previous work demonstrated that saxitoxin (STX), a toxin present in the drinking water reservoirs of the NE, exacerbated the damaging effects of ZIKV on the developing brain. We hypothesized that the impact of STX might vary among different neural cell types. While ZIKV infection caused severe damages on astrocytes and neural stem cells (NSCs), the addition of STX did not exacerbate these effects. We observed that neurons subjected to STX exposure were more prone to apoptosis and displayed higher ZIKV infection rate. These findings suggest that STX exacerbates the harmful effects of ZIKV on neurons, thereby providing a plausible explanation for the heightened severity of ZIKV-induced congenital malformations observed in Brazil's NE. This study highlights the importance of understanding the interactive effects of environmental toxins and infectious pathogens on neural development, with potential implications for public health policies.
Collapse
Affiliation(s)
- Leticia R Q Souza
- Instituto D'Or de Pesquisa e Ensino, Rua Diniz Cordeiro, 30, Rio de Janeiro, CEP 22281-100, Brazil
| | - Carolina G da S Pedrosa
- Instituto D'Or de Pesquisa e Ensino, Rua Diniz Cordeiro, 30, Rio de Janeiro, CEP 22281-100, Brazil
| | - Teresa Puig-Pijuan
- Instituto D'Or de Pesquisa e Ensino, Rua Diniz Cordeiro, 30, Rio de Janeiro, CEP 22281-100, Brazil
| | | | - Gabriela Vitória
- Instituto D'Or de Pesquisa e Ensino, Rua Diniz Cordeiro, 30, Rio de Janeiro, CEP 22281-100, Brazil
| | - João M A Delou
- Instituto D'Or de Pesquisa e Ensino, Rua Diniz Cordeiro, 30, Rio de Janeiro, CEP 22281-100, Brazil
| | - Pedro Setti-Perdigão
- Instituto D'Or de Pesquisa e Ensino, Rua Diniz Cordeiro, 30, Rio de Janeiro, CEP 22281-100, Brazil
| | - Luiza M Higa
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Amilcar Tanuri
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Stevens Kastrup Rehen
- Instituto D'Or de Pesquisa e Ensino, Rua Diniz Cordeiro, 30, Rio de Janeiro, CEP 22281-100, Brazil
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Marília Zaluar P Guimarães
- Instituto D'Or de Pesquisa e Ensino, Rua Diniz Cordeiro, 30, Rio de Janeiro, CEP 22281-100, Brazil.
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.
| |
Collapse
|
2
|
Adonias GL, Siljak H, Balasubramaniam S, Barros MT. In silico modelling of neuron signal impact of cytokine storm-induced demyelination. Open Biol 2024; 14:240138. [PMID: 39226928 PMCID: PMC11371429 DOI: 10.1098/rsob.240138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 09/05/2024] Open
Abstract
In this study, we develop an in silico model of a neuron's behaviour under demyelination caused by a cytokine storm to investigate the effects of viral infections in the brain. We use a comprehensive model to measure how cytokine-induced demyelination affects the propagation of action potential (AP) signals within a neuron. We analysed the effects of neuron-neuron communications by applying information and communication theory at different levels of demyelination. Our simulations demonstrate that virus-induced degeneration can play a role in the signal power and spiking rate, which compromise the propagation and processing of information between neurons. We propose a transfer function to model the weakening effects on the AP. Our results show that demyelination induced by a cytokine storm not only degrades the signal but also impairs its propagation within the axon. Our proposed in silico model can analyse virus-induced neurodegeneration and enhance our understanding of virus-induced demyelination.
Collapse
Affiliation(s)
- Geoflly L. Adonias
- Walton Institute for Information and Communication Systems Science, South East Technology University, Waterford, Ireland
| | - Harun Siljak
- Department of Electronic and Electrical Engineering, Trinity College Dublin, Dublin, Ireland
| | | | - Michael Taynnan Barros
- School of Computer Science and Electronic Engineering, University of Essex, Colchester, UK
| |
Collapse
|
3
|
Lebov JF, LaForett DR, Gajewski A, Browne EN, Zambrana JV, Balmaseda A, Harris E, Hooper SR. Cohort study: Neurological and cognitive-behavioral sequelae of acquired Zika virus infection among Nicaraguan children. Pediatr Res 2024:10.1038/s41390-024-03160-0. [PMID: 38956218 DOI: 10.1038/s41390-024-03160-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/29/2023] [Accepted: 01/11/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND ZIKV has neuroinvasive properties, and in utero exposure can cause birth defects, but little is known about the neurological and neurocognitive impacts of acquired ZIKV infection, particularly in children. METHODS We assessed neurological symptoms frequency among ZIKV-infected children within one year after ZIKV infection. Three to 5 years post-infection, these children and a matched group of uninfected children were assessed via questionnaires, neurological exams, and neuropsychological testing to evaluate the association between prior ZIKV infection and subsequent neurological symptoms, and cognitive-behavioral function. RESULTS Among 194 ZIKV-infected children, 3 reported asthenia, 4 reported neck pain, and 10 reported back pain within one year post-infection. At follow-up, clinician-observed cranial nerve abnormalities were significantly more common among ZIKV-infected vs. uninfected children (16 vs. 3; p < 0.01), with vestibulocochlear nerve abnormalities observed most frequently. While ZIKV-infected children scored better than uninfected on cognitive measures, this difference was not clinically meaningful. CONCLUSIONS Neurological signs, including paresthesia and cranial nerve abnormalities, were observed among ZIKV-infected participants in our study. However, we did not observe a meaningful link between acquired ZIKV infection and subsequent neurological, cognitive, or behavioral outcomes in a representative sample. An exception may be hearing impairment and loss, which should be explored further in future studies. IMPACT Neurological symptoms, though rare, were observed and reported more frequently among ZIKV-infected vs. uninfected children. These included: asthenia, neck pain, back pain, paresthesia, and cranial nerve abnormalities. Neurocognitive and behavioral test scores were similar among ZIKV-infected and uninfected children. Our study suggests that ZIKV-infected children should be monitored for neurological symptoms and cranial neuropathy to better understand the full burden of acquired ZIKV infection among children.
Collapse
Affiliation(s)
- Jill F Lebov
- Social, Statistical, and Environmental Sciences, RTI International, Durham, NC, USA.
| | - Doré R LaForett
- FPG Child Development Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Erica N Browne
- Social, Statistical, and Environmental Sciences, RTI International, Durham, NC, USA
| | - José Victor Zambrana
- Sustainable Sciences Institute, Managua, Nicaragua
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Angel Balmaseda
- Sustainable Sciences Institute, Managua, Nicaragua
- Laboratorio Nacional de Virología, Centro Nacional de Diagnósis y Referencia, Ministerio de Salud, Managua, Nicaragua
| | - Eva Harris
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Stephen R Hooper
- Department of Health Sciences, School of Medicine, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
4
|
Metzler AD, Tang H. Zika Virus Neuropathogenesis-Research and Understanding. Pathogens 2024; 13:555. [PMID: 39057782 PMCID: PMC11279898 DOI: 10.3390/pathogens13070555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/19/2024] [Accepted: 06/22/2024] [Indexed: 07/28/2024] Open
Abstract
Zika virus (ZIKV), a mosquito-borne flavivirus, is prominently associated with microcephaly in babies born to infected mothers as well as Guillain-Barré Syndrome in adults. Each cell type infected by ZIKV-neuronal cells (radial glial cells, neuronal progenitor cells, astrocytes, microglia cells, and glioblastoma stem cells) and non-neuronal cells (primary fibroblasts, epidermal keratinocytes, dendritic cells, monocytes, macrophages, and Sertoli cells)-displays its own characteristic changes to their cell physiology and has various impacts on disease. Here, we provide an in-depth review of the ZIKV life cycle and its cellular targets, and discuss the current knowledge of how infections cause neuropathologies, as well as what approaches researchers are currently taking to further advance such knowledge. A key aspect of ZIKV neuropathogenesis is virus-induced neuronal apoptosis via numerous mechanisms including cell cycle dysregulation, mitochondrial fragmentation, ER stress, and the unfolded protein response. These, in turn, result in the activation of p53-mediated intrinsic cell death pathways. A full spectrum of infection models including stem cells and co-cultures, transwells to simulate blood-tissue barriers, brain-region-specific organoids, and animal models have been developed for ZIKV research.
Collapse
Affiliation(s)
| | - Hengli Tang
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
5
|
Sung CYW, Li M, Jonjic S, Sanchez V, Britt WJ. Cytomegalovirus infection lengthens the cell cycle of granule cell precursors during postnatal cerebellar development. JCI Insight 2024; 9:e175525. [PMID: 38855871 PMCID: PMC11382886 DOI: 10.1172/jci.insight.175525] [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: 09/07/2023] [Accepted: 04/23/2024] [Indexed: 06/11/2024] Open
Abstract
Human cytomegalovirus (HCMV) infection in infants infected in utero can lead to a variety of neurodevelopmental disorders. However, mechanisms underlying altered neurodevelopment in infected infants remain poorly understood. We have previously described a murine model of congenital HCMV infection in which murine CMV (MCMV) spreads hematogenously and establishes a focal infection in all regions of the brain of newborn mice, including the cerebellum. Infection resulted in disruption of cerebellar cortical development characterized by reduced cerebellar size and foliation. This disruption was associated with altered cell cycle progression of the granule cell precursors (GCPs), which are the progenitors that give rise to granule cells (GCs), the most abundant neurons in the cerebellum. In the current study, we have demonstrated that MCMV infection leads to prolonged GCP cell cycle, premature exit from the cell cycle, and reduced numbers of GCs resulting in cerebellar hypoplasia. Treatment with TNF-α neutralizing antibody partially normalized the cell cycle alterations of GCPs and altered cerebellar morphogenesis induced by MCMV infection. Collectively, our results argue that virus-induced inflammation altered the cell cycle of GCPs resulting in a reduced numbers of GCs and cerebellar cortical hypoplasia, thus providing a potential mechanism for altered neurodevelopment in fetuses infected with HCMV.
Collapse
Affiliation(s)
- Cathy Yea Won Sung
- Department of Microbiology, University of Alabama at Birmingham, School of Medicine, Birmingham, Alabama, USA
- Laboratory of Hearing Biology and Therapeutics, National Institute on Deafness and Other Communication Disorders (NIDCD), NIH, Bethesda, Maryland, USA
| | - Mao Li
- Department of Pediatrics, University of Alabama at Birmingham, School of Medicine, Birmingham, Alabama, USA
| | - Stipan Jonjic
- Department of Histology and Embryology and
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Veronica Sanchez
- Department of Pediatrics, University of Alabama at Birmingham, School of Medicine, Birmingham, Alabama, USA
| | - William J Britt
- Department of Microbiology, University of Alabama at Birmingham, School of Medicine, Birmingham, Alabama, USA
- Department of Pediatrics, University of Alabama at Birmingham, School of Medicine, Birmingham, Alabama, USA
- Department of Neurobiology, University of Alabama at Birmingham, School of Medicine, Birmingham, Alabama, USA
| |
Collapse
|
6
|
Ye M, Liu Y, Wang F, Yang X, Yang X, Gao X, Liu W, Yu J. Polysaccharide extracted from Sarcandra glabra residue attenuate cognitive impairment by regulating gut microbiota in diabetic mice. Int J Biol Macromol 2024; 270:132121. [PMID: 38719002 DOI: 10.1016/j.ijbiomac.2024.132121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 05/16/2024]
Abstract
Diabetic encephalopathy (DE), characterized by cognitive impairment, currently lacks targeted treatment. Previous studies have shown that Sarcandra glabra extracted residue polysaccharide (SERP) exhibited hypoglycemic effects either in vitro or in streptozotocin-induced diabetes mice. However, the therapeutic effect of SERP on DE was not elucidated. This study investigated the therapeutic effect of SERP on DE and its underlying mechanism. Our results revealed that SERP regulates glucose and lipid metabolism, improves cognitive function, and exhibits diminished activity post-antibiotic intervention. Importantly, we discovered a novel mechanism by which SERP modulates the gut microbiota, specifically enriching Bacteroidales S24-7, resulting in elevated levels of butyric acid in the intestine. This regulation modulates the intestinal endocrine cell lipid metabolism level, restores damaged intestinal barriers and neural epithelial circuits, thus exhibiting cure effects. Our findings suggest that SERP could become a candidate for treating DE, potentially involving the regulation mechanism of the "microbiota-gut-brain axis". This study underscores the unique therapeutic efficacy of SERP in managing DE, offering fresh drug candidates and innovative treatment strategies for this challenging condition.
Collapse
Affiliation(s)
- Meng Ye
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Yameng Liu
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Feng Wang
- Simcere Pharmaceutical Group Limited, Nanjing 210042, PR China
| | - Xiyuchen Yang
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xiaobing Yang
- Biology and Medicine Department, Jiangsu Industrial Technology Research Institute, Nanjing 210031, PR China
| | - Xiangdong Gao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Wei Liu
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Juping Yu
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
7
|
Ferrão Maciel-Fiuza M, Rengel BD, Wachholz GE, do Amaral Gomes J, de Oliveira MR, Kowalski TW, Roehe PM, Luiz Vianna FS, Schüler-Faccini L, Mayer FQ, Varela APM, Fraga LR. New candidate genes potentially involved in Zika virus teratogenesis. Comput Biol Med 2024; 173:108259. [PMID: 38522248 DOI: 10.1016/j.compbiomed.2024.108259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/15/2024] [Accepted: 03/06/2024] [Indexed: 03/26/2024]
Abstract
Despite efforts to elucidate Zika virus (ZIKV) teratogenesis, still several issues remain unresolved, particularly on the molecular mechanisms behind the pathogenesis of Congenital Zika Syndrome (CZS). To answer this question, we used bioinformatics tools, animal experiments and human gene expression analysis to investigate genes related to brain development potentially involved in CZS. Searches in databases for genes related to brain development and CZS were performed, and a protein interaction network was created. The expression of these genes was analyzed in a CZS animal model and secondary gene expression analysis (DGE) was performed in human cells exposed to ZIKV. A total of 2610 genes were identified in the databases, of which 1013 were connected. By applying centrality statistics of the global network, 36 candidate genes were identified, which, after selection resulted in nine genes. Gene expression analysis revealed distinctive expression patterns for PRKDC, PCNA, ATM, SMC3 as well as for FGF8 and SHH in the CZS model. Furthermore, DGE analysis altered expression of ATM, PRKDC, PCNA. In conclusion, systems biology are helpful tools to identify candidate genes to be validated in vitro and in vivo. PRKDC, PCNA, ATM, SMC3, FGF8 and SHH have altered expression in ZIKV-induced brain malformations.
Collapse
Affiliation(s)
- Miriãn Ferrão Maciel-Fiuza
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Instituto Nacional de Genética Médica Populacional, Porto Alegre, Brazil; Genomics Medicine Laboratory, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Bruna Duarte Rengel
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Genomics Medicine Laboratory, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Gabriela Elis Wachholz
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Genomics Medicine Laboratory, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Julia do Amaral Gomes
- Instituto Nacional de Genética Médica Populacional, Porto Alegre, Brazil; Genomics Medicine Laboratory, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Maikel Rosa de Oliveira
- Department of Morphological Sciences, Institute of Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Thayne Woycinck Kowalski
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Genomics Medicine Laboratory, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Teratogen Information System, Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Bioinformatics Core, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Centro Universitário CESUCA, Cachoeirinha, Brazil
| | - Paulo Michel Roehe
- Department of Microbiology, Immunology and Parasitology, Institute of Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernanda Sales Luiz Vianna
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Instituto Nacional de Genética Médica Populacional, Porto Alegre, Brazil; Genomics Medicine Laboratory, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Teratogen Information System, Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Lavínia Schüler-Faccini
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Instituto Nacional de Genética Médica Populacional, Porto Alegre, Brazil; Teratogen Information System, Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Fabiana Quoos Mayer
- Graduate Program in Molecular and Cellular Biology, Biotechnology Center, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Ana Paula Muterle Varela
- Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil.
| | - Lucas Rosa Fraga
- Genomics Medicine Laboratory, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Department of Morphological Sciences, Institute of Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Teratogen Information System, Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.
| |
Collapse
|
8
|
Calado AM, Seixas F, Dos Anjos Pires M. Virus as Teratogenic Agents. Methods Mol Biol 2024; 2753:105-142. [PMID: 38285335 DOI: 10.1007/978-1-0716-3625-1_4] [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: 01/30/2024]
Abstract
Viral infectious diseases are important causes of reproductive disorders, as abortion, fetal mummification, embryonic mortality, stillbirth, and congenital abnormalities in animals and in humans. In this chapter, we provide an overview of some virus, as important agents in teratology.We begin by describing the Zika virus, whose infection in humans had a very significant impact in recent years and has been associated with major health problems worldwide. This virus is a teratogenic agent in humans and has been classified as a public health emergency of international concern (PHEIC).Then, some viruses associated with reproductive abnormalities on animals, which have a significant economic impact on livestock, are described, as bovine herpesvirus, bovine viral diarrhea virus, Schmallenberg virus, Akabane virus, and Aino virus.For all viruses mentioned in this chapter, the teratogenic effects and the congenital malformations associated with fetus and newborn are described, according to the most recent scientific publications.
Collapse
Affiliation(s)
- Ana Margarida Calado
- Animal and Veterinary Research Centre (CECAV), UTAD, and Associate Laboratory for Animal and Veterinary Science (AL4Animals), Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences (ECAV), University of Trás-os-Montes e Alto Douro (UTAD), Vila Real, Portugal
| | - Fernanda Seixas
- Animal and Veterinary Research Centre (CECAV), UTAD, and Associate Laboratory for Animal and Veterinary Science (AL4Animals), Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences (ECAV), University of Trás-os-Montes e Alto Douro (UTAD), Vila Real, Portugal
| | - Maria Dos Anjos Pires
- Animal and Veterinary Research Centre (CECAV), UTAD, and Associate Laboratory for Animal and Veterinary Science (AL4Animals), Department of Veterinary Sciences, School of Agrarian and Veterinary Sciences (ECAV), University of Trás-os-Montes e Alto Douro (UTAD), Vila Real, Portugal.
| |
Collapse
|
9
|
Ren F, Narita R, Rashidi AS, Fruhwürth S, Gao Z, Bak RO, Thomsen MK, Verjans GMGM, Reinert LS, Paludan SR. ER stress induces caspase-2-tBID-GSDME-dependent cell death in neurons lytically infected with herpes simplex virus type 2. EMBO J 2023; 42:e113118. [PMID: 37646198 PMCID: PMC10548179 DOI: 10.15252/embj.2022113118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 08/14/2023] [Accepted: 08/14/2023] [Indexed: 09/01/2023] Open
Abstract
Neurotropic viruses, including herpes simplex virus (HSV) types 1 and 2, have the capacity to infect neurons and can cause severe diseases. This is associated with neuronal cell death, which may contribute to morbidity or even mortality if the infection is not controlled. However, the mechanistic details of HSV-induced neuronal cell death remain enigmatic. Here, we report that lytic HSV-2 infection of human neuron-like SH-SY5Y cells and primary human and murine brain cells leads to cell death mediated by gasdermin E (GSDME). HSV-2-induced GSDME-mediated cell death occurs downstream of replication-induced endoplasmic reticulum stress driven by inositol-requiring kinase 1α (IRE1α), leading to activation of caspase-2, cleavage of the pro-apoptotic protein BH3-interacting domain death agonist (BID), and mitochondria-dependent activation of caspase-3. Finally, necrotic neurons released alarmins, which activated inflammatory responses in human iPSC-derived microglia. In conclusion, lytic HSV infection in neurons activates an ER stress-driven pathway to execute GSDME-mediated cell death and promote inflammation.
Collapse
Affiliation(s)
- Fanghui Ren
- Department of BiomedicineAarhus UniversityAarhus CDenmark
| | - Ryo Narita
- Department of BiomedicineAarhus UniversityAarhus CDenmark
| | - Ahmad S Rashidi
- Department of ViroscienceErasmus Medical CentreRotterdamThe Netherlands
| | - Stefanie Fruhwürth
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and PhysiologySahlgrenska Academy at the University of GothenburgGothenburgSweden
| | - Zongliang Gao
- Department of BiomedicineAarhus UniversityAarhus CDenmark
| | - Rasmus O Bak
- Department of BiomedicineAarhus UniversityAarhus CDenmark
| | | | | | - Line S Reinert
- Department of BiomedicineAarhus UniversityAarhus CDenmark
| | - Søren R Paludan
- Department of BiomedicineAarhus UniversityAarhus CDenmark
- Department of Rheumatology and Inflammation Research, Institute of MedicineSahlgrenska Academy, University of GothenburgGothenburgSweden
| |
Collapse
|
10
|
Pagani I, Ottoboni L, Panina-Bordignon P, Martino G, Poli G, Taylor S, Turnbull JE, Yates E, Vicenzi E. Heparin Precursors with Reduced Anticoagulant Properties Retain Antiviral and Protective Effects That Potentiate the Efficacy of Sofosbuvir against Zika Virus Infection in Human Neural Progenitor Cells. Pharmaceuticals (Basel) 2023; 16:1385. [PMID: 37895856 PMCID: PMC10609960 DOI: 10.3390/ph16101385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 10/29/2023] Open
Abstract
Zika virus (ZIKV) infection during pregnancy can result in severe birth defects, such as microcephaly, as well as a range of other related health complications. Heparin, a clinical-grade anticoagulant, is shown to protect neural progenitor cells from death following ZIKV infection. Although heparin can be safely used during pregnancy, it retains off-target anticoagulant effects if directly employed against ZIKV infection. In this study, we investigated the effects of chemically modified heparin derivatives with reduced anticoagulant activities. These derivatives were used as experimental probes to explore the structure-activity relationships. Precursor fractions of porcine heparin, obtained during the manufacture of conventional pharmaceutical heparin with decreased anticoagulant activities, were also explored. Interestingly, these modified heparin derivatives and precursor fractions not only prevented cell death but also inhibited the ZIKV replication of infected neural progenitor cells grown as neurospheres. These effects were observed regardless of the specific sulfation position or overall charge. Furthermore, the combination of heparin with Sofosbuvir, an antiviral licensed for the treatment of hepatitis C (HCV) that also belongs to the same Flaviviridae family as ZIKV, showed a synergistic effect. This suggested that a combination therapy approach involving heparin precursors and Sofosbuvir could be a potential strategy for the prevention or treatment of ZIKV infections.
Collapse
Affiliation(s)
- Isabel Pagani
- Viral Pathogenesis and Biosafety Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Linda Ottoboni
- Neuroimmunology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Paola Panina-Bordignon
- Neuroimmunology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- School of Medicine, Vita-Salute San Raffaele University, Via Olgettina 58, 20132 Milan, Italy
| | - Gianvito Martino
- Neuroimmunology Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- School of Medicine, Vita-Salute San Raffaele University, Via Olgettina 58, 20132 Milan, Italy
| | - Guido Poli
- School of Medicine, Vita-Salute San Raffaele University, Via Olgettina 58, 20132 Milan, Italy
- Human Immuno-Virology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Sarah Taylor
- Department of Biochemistry & Systems Biology, ISMIB, University of Liverpool, Liverpool L69 7ZB, UK
| | - Jeremy E Turnbull
- Department of Biochemistry & Systems Biology, ISMIB, University of Liverpool, Liverpool L69 7ZB, UK
- Department of Life Sciences, Keele University, Keele, Staffs ST5 5BG, UK
| | - Edwin Yates
- Department of Biochemistry & Systems Biology, ISMIB, University of Liverpool, Liverpool L69 7ZB, UK
- Department of Life Sciences, Keele University, Keele, Staffs ST5 5BG, UK
| | - Elisa Vicenzi
- Viral Pathogenesis and Biosafety Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| |
Collapse
|
11
|
Bocciarelli C, Cordel N, Leschiera R, Talagas M, Le Gall-Ianotto C, Hu W, Marcorelles P, Bellemere G, Bredif S, Fluhr J, Misery L, Lebonvallet N. New human in vitro co-culture model of keratinocytes and sensory neurons like cells releasing substance P with an evaluation of the expression of ZIKV entry receptors: A potent opportunity to test Zika virus entry and to study Zika virus' infection in neurons? Exp Dermatol 2023; 32:1563-1568. [PMID: 37395585 DOI: 10.1111/exd.14870] [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: 07/29/2022] [Revised: 05/11/2023] [Accepted: 06/19/2023] [Indexed: 07/04/2023]
Abstract
During the course of acute ZIKV infection, pruritus is a cardinal symptom widely documented in the literature. Its frequent association with dysesthesia and several dysautonomic manifestations, suggests a pathophysiological mechanism involving the peripheral nervous system. The aim of this study was to develop a functional human model to potentially able to be infected by ZIKV: by demonstrating the functionality on a new human model of co-culture of keratinocyte and sensory neuron derived from induced pluripotent stem cells using a classical method of capsaicin induction and SP release, and verify the presence of ZIKV entry receptor in these cells. Depending of cellular type, receptors of the TAMs family, TIMs (TIM1, TIM3 and TIM4) and DC-SIGN and RIG1 were present/detected. The cells incubations with capsaicin resulted in an increase of the substance P. Hence, this study demonstrated the possibility to obtain co-cultures of human keratinocytes and human sensory neurons that release substance P in the same way than previously published in animal models which can be used as a model of neurogenic skin inflammation. The demonstration of the expression of ZIKV entry receptors in these cells allows to considerate the potent possibility that ZIKV is able to infect cells.
Collapse
Affiliation(s)
| | - Nadège Cordel
- Service de Dermatologie-Immunologie clinique, CHU de Guadeloupe Pointe à Pitre, Guadeloupe et Université de Normandie UNIROUEN, IRIB, Inserm, U1234, Rouen, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Chen Q, Li N, Zeng S, Wu S, Luo X, Zhang S, Zhu L, Wu J, Xie T, Bai S, Zhang H, Jiang Z, Lin S, Wu N, Jiang Y, Fang S, Wang X, Shu Y, Luo H. ZIKV infection differentially affects the transcriptional profiles in HTR8 and U251 cells. Virus Res 2023; 334:199166. [PMID: 37390859 PMCID: PMC10410584 DOI: 10.1016/j.virusres.2023.199166] [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: 12/29/2022] [Revised: 06/27/2023] [Accepted: 06/28/2023] [Indexed: 07/02/2023]
Abstract
The mechanism by which Zika virus (ZIKV) causes severe birth defects in pregnant women remains unclear. Cell tropisms in placenta and brain play a crucial role in ZIKV pathogenesis, leading to congenital Zika syndrome (CZS). To identify the host factors involved in ZIKV infection, we compared the transcriptional profiles of ZIKV-infected human first-trimester placental trophoblast cells HTR8/SVneo and a human glioblastoma astrocytoma cell line U251. Our results demonstrated that ZIKV exhibited lower rates of mRNA replication and protein expression in HTR8 than in U251 cells, while showing a higher release of infectious viral particles. However, a greater number of differentially expressed genes (DEGs) were found in ZIKV-infected U251 cells than in ZIKV-infected HTR8 cells. Several of these DEGs were enriched in distinct biological processes related to the characteristics of each cell type that may contribute to foetal damage. Both cell types exhibited activation of common interferons, inflammatory cytokines, and chemokine production upon ZIKV infection. Moreover, the neutralization of tumour necrosis factor-alpha (TNF-α) promoted ZIKV infection in both trophoblasts and glioblastoma astrocytoma cells. Overall, we identified multiple DEGs associated with ZIKV pathogenesis.
Collapse
Affiliation(s)
- Qiqi Chen
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Nina Li
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Shike Zeng
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; Huizhou Municipal Central Hospital, Huizhou 516001, PR China
| | - Shu Wu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; Cancer Hospital Chinese Academy of Medical Sciences, Shenzhen Center, Shenzhen 518172, PR China
| | - Xin Luo
- The Emergency Department, Eighth People's Hospital of Nanyang City, Nanyang 473000, PR China
| | - Shengze Zhang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Lin Zhu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Jiani Wu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; Shaoxing Center for Disease Control and Prevention, Shaoxing 312075, PR China
| | - Ting Xie
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Shaohui Bai
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Hao Zhang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Zhiyuan Jiang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Shaoli Lin
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China
| | - Nan Wu
- Shenzhen Nanshan Center for Disease Control and Prevention, Shenzhen 518054, PR China
| | - Ying Jiang
- Shenzhen Nanshan Center for Disease Control and Prevention, Shenzhen 518054, PR China
| | - Shisong Fang
- Shenzhen Center for Disease Control and Prevention, Shenzhen 518073, PR China
| | - Xin Wang
- Shenzhen Center for Disease Control and Prevention, Shenzhen 518073, PR China
| | - Yuelong Shu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100176, PR China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, PR China
| | - Huanle Luo
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China; School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou 510275, PR China; Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, PR China.
| |
Collapse
|
13
|
Niu D, Zhang X, Zhang S, Fan T, Zhou X, Wang H, Zhang X, Nan F, Jiang S, Liu F, Wang Y, Wang B. Human Cytomegalovirus IE2 Disrupts Neural Progenitor Development and Induces Microcephaly in Transgenic Mouse. Mol Neurobiol 2023; 60:3883-3897. [PMID: 36991278 DOI: 10.1007/s12035-023-03310-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/25/2023] [Indexed: 03/31/2023]
Abstract
Human cytomegalovirus (HCMV) is a significant contributor to congenital birth defects. Limited by the lack of animal models, the pathogenesis of neurological damage in vivo caused by HCMV infection and the role of individual viral genes remain to be elucidated. Immediate early (IE2) protein may play a function in neurodevelopmental problems caused by HCMV infection. Here, this study intended to investigate IE2's long-term effects on development of the brain in IE2-expressing transgenic mice (Rosa26-LSL-IE2+/-, Camk2α-Cre) aimed to observe the phenotype of postnatal mice. The expression of IE2 in transgenic mice was confirmed by PCR and Western blot technology. We collected mouse brain tissue at 2, 4, 6, 8, and 10 days postpartum to analyze the developmental process of neural stem cells by immunofluorescence. We discovered that transgenic mice (Rosa26-LSL-IE2+/-, Camk2α-Cre) can reliably produce IE2 in the brain at various postpartum phases. Furthermore, we also observed the symptoms of microcephaly in postnatal transgenic mice, and IE2 can damage the amount of neural stem cells, prevent them from proliferating and differentiating, and activate microglia and astrocytes, creating an unbalanced environment in the brain's neurons. In conclusion, we demonstrate that long-term expression of HCMV-IE2 can cause microcephaly through molecular mechanisms affecting the differentiation and development of neural stem cells in vivo. This work establishes a theoretical and experimental foundation for elucidating the molecular mechanism of fetal microcephaly brought by HCMV infection in throughout the period of neural development of pregnancy.
Collapse
Affiliation(s)
- Delei Niu
- Department of Pathogenic Biology, College of Basic Medicine, Qingdao University, Qingdao, 266000, Shandong, China
| | - Xianjuan Zhang
- Department of Pathogenic Biology, College of Basic Medicine, Qingdao University, Qingdao, 266000, Shandong, China
| | - Shuyun Zhang
- Department of Pathogenic Biology, College of Basic Medicine, Qingdao University, Qingdao, 266000, Shandong, China
| | - Tianyu Fan
- Department of Immunology, College of Basic Medicine, Qingdao University, Qingdao, 266000, Shandong, China
| | - Xiaoqiong Zhou
- Department of Pathogenic Biology, College of Basic Medicine, Qingdao University, Qingdao, 266000, Shandong, China
| | - Hui Wang
- Department of Special Medicine, College of Basic Medicine, Qingdao University, Qingdao, 266000, Shandong, China
| | - Xueming Zhang
- Department of Pathogenic Biology, College of Basic Medicine, Qingdao University, Qingdao, 266000, Shandong, China
| | - Fulong Nan
- Department of Special Medicine, College of Basic Medicine, Qingdao University, Qingdao, 266000, Shandong, China
| | - Shasha Jiang
- Department of Pathogenic Biology, College of Basic Medicine, Qingdao University, Qingdao, 266000, Shandong, China
| | - Fengjun Liu
- Department of Special Medicine, College of Basic Medicine, Qingdao University, Qingdao, 266000, Shandong, China
| | - Yunyang Wang
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China.
| | - Bin Wang
- Department of Pathogenic Biology, College of Basic Medicine, Qingdao University, Qingdao, 266000, Shandong, China.
- Department of Special Medicine, College of Basic Medicine, Qingdao University, Qingdao, 266000, Shandong, China.
| |
Collapse
|
14
|
Quincozes-Santos A, Bobermin LD, Costa NLF, Thomaz NK, Almeida RRDS, Beys-da-Silva WO, Santi L, Rosa RL, Capra D, Coelho-Aguiar JM, DosSantos MF, Heringer M, Cirne-Lima EO, Guimarães JA, Schuler-Faccini L, Gonçalves CA, Moura-Neto V, Souza DO. The role of glial cells in Zika virus-induced neurodegeneration. Glia 2023. [PMID: 36866453 DOI: 10.1002/glia.24353] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
Zika virus (ZIKV) is a strongly neurotropic flavivirus whose infection has been associated with microcephaly in neonates. However, clinical and experimental evidence indicate that ZIKV also affects the adult nervous system. In this regard, in vitro and in vivo studies have shown the ability of ZIKV to infect glial cells. In the central nervous system (CNS), glial cells are represented by astrocytes, microglia, and oligodendrocytes. In contrast, the peripheral nervous system (PNS) constitutes a highly heterogeneous group of cells (Schwann cells, satellite glial cells, and enteric glial cells) spread through the body. These cells are critical in both physiological and pathological conditions; as such, ZIKV-induced glial dysfunctions can be associated with the development and progression of neurological complications, including those related to the adult and aging brain. This review will address the effects of ZIKV infection on CNS and PNS glial cells, focusing on cellular and molecular mechanisms, including changes in the inflammatory response, oxidative stress, mitochondrial dysfunction, Ca2+ and glutamate homeostasis, neural metabolism, and neuron-glia communication. Of note, preventive and therapeutic strategies that focus on glial cells may emerge to delay and/or prevent the development of ZIKV-induced neurodegeneration and its consequences.
Collapse
Affiliation(s)
- André Quincozes-Santos
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Larissa Daniele Bobermin
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Naithan Ludian Fernandes Costa
- Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Natalie K Thomaz
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Rômulo Rodrigo de Souza Almeida
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | - Lucélia Santi
- Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Rafael L Rosa
- Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Daniela Capra
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Juliana M Coelho-Aguiar
- Laboratório de Morfogênese Celular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Marcos Fabio DosSantos
- Laboratório de Propriedades Mecânicas e Biologia Celular, Faculdade de Odontologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Manoela Heringer
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | | | | | - Carlos-Alberto Gonçalves
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Vivaldo Moura-Neto
- Instituto Estadual do Cérebro Paulo Niemeyer, Secretaria Estadual de Saúde do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.,Laboratório de Morfogênese Celular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Diogo Onofre Souza
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
15
|
Teulière J, Bernard C, Bonnefous H, Martens J, Lopez P, Bapteste E. Interactomics: Dozens of Viruses, Co-evolving With Humans, Including the Influenza A Virus, may Actively Distort Human Aging. Mol Biol Evol 2023; 40:msad012. [PMID: 36649176 PMCID: PMC9897028 DOI: 10.1093/molbev/msad012] [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: 06/10/2022] [Revised: 12/07/2022] [Accepted: 01/09/2023] [Indexed: 01/18/2023] Open
Abstract
Some viruses (e.g., human immunodeficiency virus 1 and severe acute respiratory syndrome coronavirus 2) have been experimentally proposed to accelerate features of human aging and of cellular senescence. These observations, along with evolutionary considerations on viral fitness, raised the more general puzzling hypothesis that, beyond documented sources in human genetics, aging in our species may also depend on virally encoded interactions distorting our aging to the benefits of diverse viruses. Accordingly, we designed systematic network-based analyses of the human and viral protein interactomes, which unraveled dozens of viruses encoding proteins experimentally demonstrated to interact with proteins from pathways associated with human aging, including cellular senescence. We further corroborated our predictions that specific viruses interfere with human aging using published experimental evidence and transcriptomic data; identifying influenza A virus (subtype H1N1) as a major candidate age distorter, notably through manipulation of cellular senescence. By providing original evidence that viruses may convergently contribute to the evolution of numerous age-associated pathways through co-evolution, our network-based and bipartite network-based methodologies support an ecosystemic study of aging, also searching for genetic causes of aging outside a focal aging species. Our findings, predicting age distorters and targets for anti-aging therapies among human viruses, could have fundamental and practical implications for evolutionary biology, aging study, virology, medicine, and demography.
Collapse
Affiliation(s)
- Jérôme Teulière
- Institut de Systématique, Evolution, Biodiversité (ISYEB), Sorbonne Université, CNRS, Museum National d’Histoire Naturelle, EPHE, Université des Antilles, Paris, France
| | - Charles Bernard
- Institut de Systématique, Evolution, Biodiversité (ISYEB), Sorbonne Université, CNRS, Museum National d’Histoire Naturelle, EPHE, Université des Antilles, Paris, France
| | - Hugo Bonnefous
- Institut de Systématique, Evolution, Biodiversité (ISYEB), Sorbonne Université, CNRS, Museum National d’Histoire Naturelle, EPHE, Université des Antilles, Paris, France
| | - Johannes Martens
- Sciences, Normes, Démocratie (SND), Sorbonne Université, CNRS, Paris, France
| | - Philippe Lopez
- Institut de Systématique, Evolution, Biodiversité (ISYEB), Sorbonne Université, CNRS, Museum National d’Histoire Naturelle, EPHE, Université des Antilles, Paris, France
| | - Eric Bapteste
- Institut de Systématique, Evolution, Biodiversité (ISYEB), Sorbonne Université, CNRS, Museum National d’Histoire Naturelle, EPHE, Université des Antilles, Paris, France
| |
Collapse
|
16
|
The RNA polymerase of cytoplasmically replicating Zika virus binds with chromatin DNA in nuclei and regulates host gene transcription. Proc Natl Acad Sci U S A 2022; 119:e2205013119. [PMID: 36442102 PMCID: PMC9894162 DOI: 10.1073/pnas.2205013119] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Zika virus (ZIKV) targets the neural progenitor cells (NPCs) in brain during intrauterine infections and consequently causes severe neurological disorders, such as microcephaly in neonates. Although replicating in the cytoplasm, ZIKV dysregulates the expression of thousands of host genes, yet the detailed mechanism remains elusive. Herein, we report that ZIKV encodes a unique DNA-binding protein to regulate host gene transcription in the nucleus. We found that ZIKV NS5, the viral RNA polymerase, associates tightly with host chromatin DNA through its methyltransferase domain and this interaction could be specifically blocked by GTP. Further study showed that expression of ZIKV NS5 in human NPCs markedly suppressed the transcription of its target genes, especially the genes involved in neurogenesis. Mechanistically, ZIKV NS5 binds onto the gene body of its target genes and then blocks their transcriptional elongation. The utero electroporation in pregnant mice showed that NS5 expression significantly disrupts the neurogenesis by reducing the number of Sox2- and Tbr2-positive cells in the fetal cortex. Together, our findings demonstrate a molecular clue linking to the abnormal neurodevelopment caused by ZIKV infection and also provide intriguing insights into the interaction between the host cell and the pathogenic RNA virus, where the cytoplasmic RNA virus encodes a DNA-binding protein to control the transcription of host cell in the nuclei.
Collapse
|
17
|
Lyoo KS, Kim HM, Lee B, Che YH, Kim SJ, Song D, Hwang W, Lee S, Park JH, Na W, Yun SP, Kim YJ. Direct neuronal infection of SARS-CoV-2 reveals cellular and molecular pathology of chemosensory impairment of COVID-19 patients. Emerg Microbes Infect 2022; 11:406-411. [PMID: 34962444 PMCID: PMC8803065 DOI: 10.1080/22221751.2021.2024095] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/25/2021] [Indexed: 12/17/2022]
Abstract
Patients with recent pandemic coronavirus disease 19 (COVID-19) complain of neurological abnormalities in sensory functions such as smell and taste in the early stages of infection. Determining the cellular and molecular mechanism of sensory impairment is critical to understand the pathogenesis of clinical manifestations, as well as in setting therapeutic targets for sequelae and recurrence. The absence of studies utilizing proper models of human peripheral nerve hampers an understanding of COVID-19 pathogenesis. Here, we report that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) directly infects human peripheral sensory neurons, leading to molecular pathogenesis for chemosensory impairments. An in vitro system utilizing human embryonic stem cell (hESC)-derived peripheral neurons was used to model the cellular and molecular pathologies responsible for symptoms that most COVID-19 patients experience early in infection or may develop as sequelae. Peripheral neurons differentiated from hESCs expressed viral entry factor ACE2, and were directly infected with SARS-CoV-2 via ACE2. Human peripheral neurons infected with SARS-CoV-2 exhibited impaired molecular features of chemosensory function associated with abnormalities in sensory neurons of the olfactory or gustatory organs. Our results provide new insights into the pathogenesis of chemosensory dysfunction in patients with COVID-19.
Collapse
Affiliation(s)
- Kwang-Soo Lyoo
- Korea Zoonosis Research Institute, Jeonbuk National University, Iksan, Korea
| | - Hyeon Myeong Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea
| | - Bina Lee
- Department of Pharmacology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Korea
| | - Young Hyun Che
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea
| | - Seong-Jae Kim
- Department of Ophthalmology, Institute of Health Sciences, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, Korea
| | - Daesub Song
- Department of Pharmacy, Korea University, Sejong, Korea
| | - Woochang Hwang
- Biostatistical Consulting and Research Lab, Medical Research Collaborating Center, Hanyang University, Seoul, Korea
| | - Sun Lee
- Department of Pathology, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Jae-Hoon Park
- Department of Pathology, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Woonsung Na
- Department of Veterinary Virology, College of Veterinary Medicine, Chonnam National University, Gwangju, Korea
| | - Seung Pil Yun
- Department of Pharmacology, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, Korea
| | - Yong Jun Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul, Korea
- Department of Pathology, School of Medicine, Kyung Hee University, Seoul, Korea
| |
Collapse
|
18
|
Davies AJ, Lleixà C, Siles AM, Gourlay DS, Berridge G, Dejnirattisai W, Ramírez-Santana C, Anaya JM, Falconar AK, Romero-Vivas CM, Osorio L, Parra B, Screaton GR, Mongkolsapaya J, Fischer R, Pardo CA, Halstead SK, Willison HJ, Querol L, Rinaldi S. Guillain-Barré Syndrome Following Zika Virus Infection Is Associated With a Diverse Spectrum of Peripheral Nerve Reactive Antibodies. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2022; 10:10/1/e200047. [PMID: 36411078 PMCID: PMC9679884 DOI: 10.1212/nxi.0000000000200047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 09/01/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND OBJECTIVES Recent outbreaks of Zika virus (ZIKV) in South and Central America have highlighted significant neurologic side effects. Concurrence with the inflammatory neuropathy Guillain-Barré syndrome (GBS) is observed in 1:4,000 ZIKV cases. Whether the neurologic symptoms of ZIKV infection are immune mediated is unclear. We used rodent and human live cellular models to screen for anti-peripheral nerve reactive IgG and IgM autoantibodies in the sera of patients with ZIKV with and without GBS. METHODS In this study, 52 patients with ZIKV-GBS were compared with 134 ZIKV-infected patients without GBS and 91 non-ZIKV controls. Positive sera were taken forward for target identification by immunoprecipitation and mass spectrometry, and candidate antigens were validated by ELISA and cell-based assays. Autoantibody reactions against glycolipid antigens were also screened on an array. RESULTS Overall, IgG antibody reactivities to rat Schwann cells (SCs) (6.5%) and myelinated cocultures (9.6%) were significantly higher, albeit infrequent, in the ZIKV-GBS group compared with all controls. IgM antibody immunoreactivity to dorsal root ganglia neurones (32.3%) and SCs (19.4%) was more frequently observed in the ZIKV-GBS group compared with other controls, whereas IgM reactivity to cocultures was as common in ZIKV and non-ZIKV sera. Strong axonal-binding ZIKV-GBS serum IgG antibodies from 1 patient were confirmed to react with neurofascin 155 and 186. Serum from a ZIKV-infected patient without GBS displayed strong myelin-binding and putative antilipid antigen reaction characteristics. There was, however, no significant association of ZIKV-GBS with any known antiglycolipid antibodies. DISCUSSION Autoantibody responses in ZIKV-GBS target heterogeneous peripheral nerve antigens suggesting heterogeneity of the humoral immune response despite a common prodromal infection.
Collapse
Affiliation(s)
- Alexander J Davies
- From the Nuffield Department of Clinical Neurosciences (A.J.D., S.R.), University of Oxford, John Radcliffe Hospital, UK; Neuromuscular Diseases Unit (C.L., A.M.S., L.Q.), Neurology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Spain; Centro para la Investigación Biomédica en red en Enfermedades Raras-(CIBERER) Madrid (C.L., A.M.S., L.Q.), Spain; Institute of Infection (D.S.G., S.K.H., H.J.W.), Immunity & Inflammation, University of Glasgow, University Place, UK; Target Discovery Institute (G.B., R.F.), NDM Research Building, University of Oxford, Old Road Campus, UK; Wellcome Centre for Human Genetics (W.D., G.R.S., J.M.), Nuffield Department of Medicine, University of Oxford, UK; Center for Autoimmune Diseases Research (CREA) (C.R.-S., J.-M.A.), Universidad del Rosario, Bogotá, Colombia; Departamento de Medicina (A.K.F., C.M.R.-V.), Universidad del Norte, Barranquilla, Colombia; Grupo de Epidemiología y Salud Poblacional (GESP) (L.O.,), School of Public Health, Universidad del Valle, Cali, Colombia; Department of Microbiology (B.P.), School of Basic Sciences, Universidad del Valle, Cali, Colombia; Dengue Hemorrhagic Fever Research Unit (J.M.), Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol Univeristy, Bangkok, Thailand; Department of Neurology (C.A.P.), Johns Hopkins University School of Medicine, Baltimore, MD; and LifeFactors (J.-M.A.), Rionegro, Colombia; Division of Emerging Infectious Disease (W.D.), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Cinta Lleixà
- From the Nuffield Department of Clinical Neurosciences (A.J.D., S.R.), University of Oxford, John Radcliffe Hospital, UK; Neuromuscular Diseases Unit (C.L., A.M.S., L.Q.), Neurology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Spain; Centro para la Investigación Biomédica en red en Enfermedades Raras-(CIBERER) Madrid (C.L., A.M.S., L.Q.), Spain; Institute of Infection (D.S.G., S.K.H., H.J.W.), Immunity & Inflammation, University of Glasgow, University Place, UK; Target Discovery Institute (G.B., R.F.), NDM Research Building, University of Oxford, Old Road Campus, UK; Wellcome Centre for Human Genetics (W.D., G.R.S., J.M.), Nuffield Department of Medicine, University of Oxford, UK; Center for Autoimmune Diseases Research (CREA) (C.R.-S., J.-M.A.), Universidad del Rosario, Bogotá, Colombia; Departamento de Medicina (A.K.F., C.M.R.-V.), Universidad del Norte, Barranquilla, Colombia; Grupo de Epidemiología y Salud Poblacional (GESP) (L.O.,), School of Public Health, Universidad del Valle, Cali, Colombia; Department of Microbiology (B.P.), School of Basic Sciences, Universidad del Valle, Cali, Colombia; Dengue Hemorrhagic Fever Research Unit (J.M.), Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol Univeristy, Bangkok, Thailand; Department of Neurology (C.A.P.), Johns Hopkins University School of Medicine, Baltimore, MD; and LifeFactors (J.-M.A.), Rionegro, Colombia; Division of Emerging Infectious Disease (W.D.), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Ana M Siles
- From the Nuffield Department of Clinical Neurosciences (A.J.D., S.R.), University of Oxford, John Radcliffe Hospital, UK; Neuromuscular Diseases Unit (C.L., A.M.S., L.Q.), Neurology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Spain; Centro para la Investigación Biomédica en red en Enfermedades Raras-(CIBERER) Madrid (C.L., A.M.S., L.Q.), Spain; Institute of Infection (D.S.G., S.K.H., H.J.W.), Immunity & Inflammation, University of Glasgow, University Place, UK; Target Discovery Institute (G.B., R.F.), NDM Research Building, University of Oxford, Old Road Campus, UK; Wellcome Centre for Human Genetics (W.D., G.R.S., J.M.), Nuffield Department of Medicine, University of Oxford, UK; Center for Autoimmune Diseases Research (CREA) (C.R.-S., J.-M.A.), Universidad del Rosario, Bogotá, Colombia; Departamento de Medicina (A.K.F., C.M.R.-V.), Universidad del Norte, Barranquilla, Colombia; Grupo de Epidemiología y Salud Poblacional (GESP) (L.O.,), School of Public Health, Universidad del Valle, Cali, Colombia; Department of Microbiology (B.P.), School of Basic Sciences, Universidad del Valle, Cali, Colombia; Dengue Hemorrhagic Fever Research Unit (J.M.), Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol Univeristy, Bangkok, Thailand; Department of Neurology (C.A.P.), Johns Hopkins University School of Medicine, Baltimore, MD; and LifeFactors (J.-M.A.), Rionegro, Colombia; Division of Emerging Infectious Disease (W.D.), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Dawn S Gourlay
- From the Nuffield Department of Clinical Neurosciences (A.J.D., S.R.), University of Oxford, John Radcliffe Hospital, UK; Neuromuscular Diseases Unit (C.L., A.M.S., L.Q.), Neurology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Spain; Centro para la Investigación Biomédica en red en Enfermedades Raras-(CIBERER) Madrid (C.L., A.M.S., L.Q.), Spain; Institute of Infection (D.S.G., S.K.H., H.J.W.), Immunity & Inflammation, University of Glasgow, University Place, UK; Target Discovery Institute (G.B., R.F.), NDM Research Building, University of Oxford, Old Road Campus, UK; Wellcome Centre for Human Genetics (W.D., G.R.S., J.M.), Nuffield Department of Medicine, University of Oxford, UK; Center for Autoimmune Diseases Research (CREA) (C.R.-S., J.-M.A.), Universidad del Rosario, Bogotá, Colombia; Departamento de Medicina (A.K.F., C.M.R.-V.), Universidad del Norte, Barranquilla, Colombia; Grupo de Epidemiología y Salud Poblacional (GESP) (L.O.,), School of Public Health, Universidad del Valle, Cali, Colombia; Department of Microbiology (B.P.), School of Basic Sciences, Universidad del Valle, Cali, Colombia; Dengue Hemorrhagic Fever Research Unit (J.M.), Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol Univeristy, Bangkok, Thailand; Department of Neurology (C.A.P.), Johns Hopkins University School of Medicine, Baltimore, MD; and LifeFactors (J.-M.A.), Rionegro, Colombia; Division of Emerging Infectious Disease (W.D.), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Georgina Berridge
- From the Nuffield Department of Clinical Neurosciences (A.J.D., S.R.), University of Oxford, John Radcliffe Hospital, UK; Neuromuscular Diseases Unit (C.L., A.M.S., L.Q.), Neurology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Spain; Centro para la Investigación Biomédica en red en Enfermedades Raras-(CIBERER) Madrid (C.L., A.M.S., L.Q.), Spain; Institute of Infection (D.S.G., S.K.H., H.J.W.), Immunity & Inflammation, University of Glasgow, University Place, UK; Target Discovery Institute (G.B., R.F.), NDM Research Building, University of Oxford, Old Road Campus, UK; Wellcome Centre for Human Genetics (W.D., G.R.S., J.M.), Nuffield Department of Medicine, University of Oxford, UK; Center for Autoimmune Diseases Research (CREA) (C.R.-S., J.-M.A.), Universidad del Rosario, Bogotá, Colombia; Departamento de Medicina (A.K.F., C.M.R.-V.), Universidad del Norte, Barranquilla, Colombia; Grupo de Epidemiología y Salud Poblacional (GESP) (L.O.,), School of Public Health, Universidad del Valle, Cali, Colombia; Department of Microbiology (B.P.), School of Basic Sciences, Universidad del Valle, Cali, Colombia; Dengue Hemorrhagic Fever Research Unit (J.M.), Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol Univeristy, Bangkok, Thailand; Department of Neurology (C.A.P.), Johns Hopkins University School of Medicine, Baltimore, MD; and LifeFactors (J.-M.A.), Rionegro, Colombia; Division of Emerging Infectious Disease (W.D.), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Wanwisa Dejnirattisai
- From the Nuffield Department of Clinical Neurosciences (A.J.D., S.R.), University of Oxford, John Radcliffe Hospital, UK; Neuromuscular Diseases Unit (C.L., A.M.S., L.Q.), Neurology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Spain; Centro para la Investigación Biomédica en red en Enfermedades Raras-(CIBERER) Madrid (C.L., A.M.S., L.Q.), Spain; Institute of Infection (D.S.G., S.K.H., H.J.W.), Immunity & Inflammation, University of Glasgow, University Place, UK; Target Discovery Institute (G.B., R.F.), NDM Research Building, University of Oxford, Old Road Campus, UK; Wellcome Centre for Human Genetics (W.D., G.R.S., J.M.), Nuffield Department of Medicine, University of Oxford, UK; Center for Autoimmune Diseases Research (CREA) (C.R.-S., J.-M.A.), Universidad del Rosario, Bogotá, Colombia; Departamento de Medicina (A.K.F., C.M.R.-V.), Universidad del Norte, Barranquilla, Colombia; Grupo de Epidemiología y Salud Poblacional (GESP) (L.O.,), School of Public Health, Universidad del Valle, Cali, Colombia; Department of Microbiology (B.P.), School of Basic Sciences, Universidad del Valle, Cali, Colombia; Dengue Hemorrhagic Fever Research Unit (J.M.), Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol Univeristy, Bangkok, Thailand; Department of Neurology (C.A.P.), Johns Hopkins University School of Medicine, Baltimore, MD; and LifeFactors (J.-M.A.), Rionegro, Colombia; Division of Emerging Infectious Disease (W.D.), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Carolina Ramírez-Santana
- From the Nuffield Department of Clinical Neurosciences (A.J.D., S.R.), University of Oxford, John Radcliffe Hospital, UK; Neuromuscular Diseases Unit (C.L., A.M.S., L.Q.), Neurology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Spain; Centro para la Investigación Biomédica en red en Enfermedades Raras-(CIBERER) Madrid (C.L., A.M.S., L.Q.), Spain; Institute of Infection (D.S.G., S.K.H., H.J.W.), Immunity & Inflammation, University of Glasgow, University Place, UK; Target Discovery Institute (G.B., R.F.), NDM Research Building, University of Oxford, Old Road Campus, UK; Wellcome Centre for Human Genetics (W.D., G.R.S., J.M.), Nuffield Department of Medicine, University of Oxford, UK; Center for Autoimmune Diseases Research (CREA) (C.R.-S., J.-M.A.), Universidad del Rosario, Bogotá, Colombia; Departamento de Medicina (A.K.F., C.M.R.-V.), Universidad del Norte, Barranquilla, Colombia; Grupo de Epidemiología y Salud Poblacional (GESP) (L.O.,), School of Public Health, Universidad del Valle, Cali, Colombia; Department of Microbiology (B.P.), School of Basic Sciences, Universidad del Valle, Cali, Colombia; Dengue Hemorrhagic Fever Research Unit (J.M.), Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol Univeristy, Bangkok, Thailand; Department of Neurology (C.A.P.), Johns Hopkins University School of Medicine, Baltimore, MD; and LifeFactors (J.-M.A.), Rionegro, Colombia; Division of Emerging Infectious Disease (W.D.), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Juan-Manuel Anaya
- From the Nuffield Department of Clinical Neurosciences (A.J.D., S.R.), University of Oxford, John Radcliffe Hospital, UK; Neuromuscular Diseases Unit (C.L., A.M.S., L.Q.), Neurology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Spain; Centro para la Investigación Biomédica en red en Enfermedades Raras-(CIBERER) Madrid (C.L., A.M.S., L.Q.), Spain; Institute of Infection (D.S.G., S.K.H., H.J.W.), Immunity & Inflammation, University of Glasgow, University Place, UK; Target Discovery Institute (G.B., R.F.), NDM Research Building, University of Oxford, Old Road Campus, UK; Wellcome Centre for Human Genetics (W.D., G.R.S., J.M.), Nuffield Department of Medicine, University of Oxford, UK; Center for Autoimmune Diseases Research (CREA) (C.R.-S., J.-M.A.), Universidad del Rosario, Bogotá, Colombia; Departamento de Medicina (A.K.F., C.M.R.-V.), Universidad del Norte, Barranquilla, Colombia; Grupo de Epidemiología y Salud Poblacional (GESP) (L.O.,), School of Public Health, Universidad del Valle, Cali, Colombia; Department of Microbiology (B.P.), School of Basic Sciences, Universidad del Valle, Cali, Colombia; Dengue Hemorrhagic Fever Research Unit (J.M.), Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol Univeristy, Bangkok, Thailand; Department of Neurology (C.A.P.), Johns Hopkins University School of Medicine, Baltimore, MD; and LifeFactors (J.-M.A.), Rionegro, Colombia; Division of Emerging Infectious Disease (W.D.), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Andrew K Falconar
- From the Nuffield Department of Clinical Neurosciences (A.J.D., S.R.), University of Oxford, John Radcliffe Hospital, UK; Neuromuscular Diseases Unit (C.L., A.M.S., L.Q.), Neurology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Spain; Centro para la Investigación Biomédica en red en Enfermedades Raras-(CIBERER) Madrid (C.L., A.M.S., L.Q.), Spain; Institute of Infection (D.S.G., S.K.H., H.J.W.), Immunity & Inflammation, University of Glasgow, University Place, UK; Target Discovery Institute (G.B., R.F.), NDM Research Building, University of Oxford, Old Road Campus, UK; Wellcome Centre for Human Genetics (W.D., G.R.S., J.M.), Nuffield Department of Medicine, University of Oxford, UK; Center for Autoimmune Diseases Research (CREA) (C.R.-S., J.-M.A.), Universidad del Rosario, Bogotá, Colombia; Departamento de Medicina (A.K.F., C.M.R.-V.), Universidad del Norte, Barranquilla, Colombia; Grupo de Epidemiología y Salud Poblacional (GESP) (L.O.,), School of Public Health, Universidad del Valle, Cali, Colombia; Department of Microbiology (B.P.), School of Basic Sciences, Universidad del Valle, Cali, Colombia; Dengue Hemorrhagic Fever Research Unit (J.M.), Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol Univeristy, Bangkok, Thailand; Department of Neurology (C.A.P.), Johns Hopkins University School of Medicine, Baltimore, MD; and LifeFactors (J.-M.A.), Rionegro, Colombia; Division of Emerging Infectious Disease (W.D.), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Claudia M Romero-Vivas
- From the Nuffield Department of Clinical Neurosciences (A.J.D., S.R.), University of Oxford, John Radcliffe Hospital, UK; Neuromuscular Diseases Unit (C.L., A.M.S., L.Q.), Neurology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Spain; Centro para la Investigación Biomédica en red en Enfermedades Raras-(CIBERER) Madrid (C.L., A.M.S., L.Q.), Spain; Institute of Infection (D.S.G., S.K.H., H.J.W.), Immunity & Inflammation, University of Glasgow, University Place, UK; Target Discovery Institute (G.B., R.F.), NDM Research Building, University of Oxford, Old Road Campus, UK; Wellcome Centre for Human Genetics (W.D., G.R.S., J.M.), Nuffield Department of Medicine, University of Oxford, UK; Center for Autoimmune Diseases Research (CREA) (C.R.-S., J.-M.A.), Universidad del Rosario, Bogotá, Colombia; Departamento de Medicina (A.K.F., C.M.R.-V.), Universidad del Norte, Barranquilla, Colombia; Grupo de Epidemiología y Salud Poblacional (GESP) (L.O.,), School of Public Health, Universidad del Valle, Cali, Colombia; Department of Microbiology (B.P.), School of Basic Sciences, Universidad del Valle, Cali, Colombia; Dengue Hemorrhagic Fever Research Unit (J.M.), Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol Univeristy, Bangkok, Thailand; Department of Neurology (C.A.P.), Johns Hopkins University School of Medicine, Baltimore, MD; and LifeFactors (J.-M.A.), Rionegro, Colombia; Division of Emerging Infectious Disease (W.D.), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Lyda Osorio
- From the Nuffield Department of Clinical Neurosciences (A.J.D., S.R.), University of Oxford, John Radcliffe Hospital, UK; Neuromuscular Diseases Unit (C.L., A.M.S., L.Q.), Neurology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Spain; Centro para la Investigación Biomédica en red en Enfermedades Raras-(CIBERER) Madrid (C.L., A.M.S., L.Q.), Spain; Institute of Infection (D.S.G., S.K.H., H.J.W.), Immunity & Inflammation, University of Glasgow, University Place, UK; Target Discovery Institute (G.B., R.F.), NDM Research Building, University of Oxford, Old Road Campus, UK; Wellcome Centre for Human Genetics (W.D., G.R.S., J.M.), Nuffield Department of Medicine, University of Oxford, UK; Center for Autoimmune Diseases Research (CREA) (C.R.-S., J.-M.A.), Universidad del Rosario, Bogotá, Colombia; Departamento de Medicina (A.K.F., C.M.R.-V.), Universidad del Norte, Barranquilla, Colombia; Grupo de Epidemiología y Salud Poblacional (GESP) (L.O.,), School of Public Health, Universidad del Valle, Cali, Colombia; Department of Microbiology (B.P.), School of Basic Sciences, Universidad del Valle, Cali, Colombia; Dengue Hemorrhagic Fever Research Unit (J.M.), Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol Univeristy, Bangkok, Thailand; Department of Neurology (C.A.P.), Johns Hopkins University School of Medicine, Baltimore, MD; and LifeFactors (J.-M.A.), Rionegro, Colombia; Division of Emerging Infectious Disease (W.D.), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Beatriz Parra
- From the Nuffield Department of Clinical Neurosciences (A.J.D., S.R.), University of Oxford, John Radcliffe Hospital, UK; Neuromuscular Diseases Unit (C.L., A.M.S., L.Q.), Neurology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Spain; Centro para la Investigación Biomédica en red en Enfermedades Raras-(CIBERER) Madrid (C.L., A.M.S., L.Q.), Spain; Institute of Infection (D.S.G., S.K.H., H.J.W.), Immunity & Inflammation, University of Glasgow, University Place, UK; Target Discovery Institute (G.B., R.F.), NDM Research Building, University of Oxford, Old Road Campus, UK; Wellcome Centre for Human Genetics (W.D., G.R.S., J.M.), Nuffield Department of Medicine, University of Oxford, UK; Center for Autoimmune Diseases Research (CREA) (C.R.-S., J.-M.A.), Universidad del Rosario, Bogotá, Colombia; Departamento de Medicina (A.K.F., C.M.R.-V.), Universidad del Norte, Barranquilla, Colombia; Grupo de Epidemiología y Salud Poblacional (GESP) (L.O.,), School of Public Health, Universidad del Valle, Cali, Colombia; Department of Microbiology (B.P.), School of Basic Sciences, Universidad del Valle, Cali, Colombia; Dengue Hemorrhagic Fever Research Unit (J.M.), Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol Univeristy, Bangkok, Thailand; Department of Neurology (C.A.P.), Johns Hopkins University School of Medicine, Baltimore, MD; and LifeFactors (J.-M.A.), Rionegro, Colombia; Division of Emerging Infectious Disease (W.D.), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Gavin R Screaton
- From the Nuffield Department of Clinical Neurosciences (A.J.D., S.R.), University of Oxford, John Radcliffe Hospital, UK; Neuromuscular Diseases Unit (C.L., A.M.S., L.Q.), Neurology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Spain; Centro para la Investigación Biomédica en red en Enfermedades Raras-(CIBERER) Madrid (C.L., A.M.S., L.Q.), Spain; Institute of Infection (D.S.G., S.K.H., H.J.W.), Immunity & Inflammation, University of Glasgow, University Place, UK; Target Discovery Institute (G.B., R.F.), NDM Research Building, University of Oxford, Old Road Campus, UK; Wellcome Centre for Human Genetics (W.D., G.R.S., J.M.), Nuffield Department of Medicine, University of Oxford, UK; Center for Autoimmune Diseases Research (CREA) (C.R.-S., J.-M.A.), Universidad del Rosario, Bogotá, Colombia; Departamento de Medicina (A.K.F., C.M.R.-V.), Universidad del Norte, Barranquilla, Colombia; Grupo de Epidemiología y Salud Poblacional (GESP) (L.O.,), School of Public Health, Universidad del Valle, Cali, Colombia; Department of Microbiology (B.P.), School of Basic Sciences, Universidad del Valle, Cali, Colombia; Dengue Hemorrhagic Fever Research Unit (J.M.), Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol Univeristy, Bangkok, Thailand; Department of Neurology (C.A.P.), Johns Hopkins University School of Medicine, Baltimore, MD; and LifeFactors (J.-M.A.), Rionegro, Colombia; Division of Emerging Infectious Disease (W.D.), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Juthathip Mongkolsapaya
- From the Nuffield Department of Clinical Neurosciences (A.J.D., S.R.), University of Oxford, John Radcliffe Hospital, UK; Neuromuscular Diseases Unit (C.L., A.M.S., L.Q.), Neurology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Spain; Centro para la Investigación Biomédica en red en Enfermedades Raras-(CIBERER) Madrid (C.L., A.M.S., L.Q.), Spain; Institute of Infection (D.S.G., S.K.H., H.J.W.), Immunity & Inflammation, University of Glasgow, University Place, UK; Target Discovery Institute (G.B., R.F.), NDM Research Building, University of Oxford, Old Road Campus, UK; Wellcome Centre for Human Genetics (W.D., G.R.S., J.M.), Nuffield Department of Medicine, University of Oxford, UK; Center for Autoimmune Diseases Research (CREA) (C.R.-S., J.-M.A.), Universidad del Rosario, Bogotá, Colombia; Departamento de Medicina (A.K.F., C.M.R.-V.), Universidad del Norte, Barranquilla, Colombia; Grupo de Epidemiología y Salud Poblacional (GESP) (L.O.,), School of Public Health, Universidad del Valle, Cali, Colombia; Department of Microbiology (B.P.), School of Basic Sciences, Universidad del Valle, Cali, Colombia; Dengue Hemorrhagic Fever Research Unit (J.M.), Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol Univeristy, Bangkok, Thailand; Department of Neurology (C.A.P.), Johns Hopkins University School of Medicine, Baltimore, MD; and LifeFactors (J.-M.A.), Rionegro, Colombia; Division of Emerging Infectious Disease (W.D.), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Roman Fischer
- From the Nuffield Department of Clinical Neurosciences (A.J.D., S.R.), University of Oxford, John Radcliffe Hospital, UK; Neuromuscular Diseases Unit (C.L., A.M.S., L.Q.), Neurology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Spain; Centro para la Investigación Biomédica en red en Enfermedades Raras-(CIBERER) Madrid (C.L., A.M.S., L.Q.), Spain; Institute of Infection (D.S.G., S.K.H., H.J.W.), Immunity & Inflammation, University of Glasgow, University Place, UK; Target Discovery Institute (G.B., R.F.), NDM Research Building, University of Oxford, Old Road Campus, UK; Wellcome Centre for Human Genetics (W.D., G.R.S., J.M.), Nuffield Department of Medicine, University of Oxford, UK; Center for Autoimmune Diseases Research (CREA) (C.R.-S., J.-M.A.), Universidad del Rosario, Bogotá, Colombia; Departamento de Medicina (A.K.F., C.M.R.-V.), Universidad del Norte, Barranquilla, Colombia; Grupo de Epidemiología y Salud Poblacional (GESP) (L.O.,), School of Public Health, Universidad del Valle, Cali, Colombia; Department of Microbiology (B.P.), School of Basic Sciences, Universidad del Valle, Cali, Colombia; Dengue Hemorrhagic Fever Research Unit (J.M.), Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol Univeristy, Bangkok, Thailand; Department of Neurology (C.A.P.), Johns Hopkins University School of Medicine, Baltimore, MD; and LifeFactors (J.-M.A.), Rionegro, Colombia; Division of Emerging Infectious Disease (W.D.), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Carlos A Pardo
- From the Nuffield Department of Clinical Neurosciences (A.J.D., S.R.), University of Oxford, John Radcliffe Hospital, UK; Neuromuscular Diseases Unit (C.L., A.M.S., L.Q.), Neurology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Spain; Centro para la Investigación Biomédica en red en Enfermedades Raras-(CIBERER) Madrid (C.L., A.M.S., L.Q.), Spain; Institute of Infection (D.S.G., S.K.H., H.J.W.), Immunity & Inflammation, University of Glasgow, University Place, UK; Target Discovery Institute (G.B., R.F.), NDM Research Building, University of Oxford, Old Road Campus, UK; Wellcome Centre for Human Genetics (W.D., G.R.S., J.M.), Nuffield Department of Medicine, University of Oxford, UK; Center for Autoimmune Diseases Research (CREA) (C.R.-S., J.-M.A.), Universidad del Rosario, Bogotá, Colombia; Departamento de Medicina (A.K.F., C.M.R.-V.), Universidad del Norte, Barranquilla, Colombia; Grupo de Epidemiología y Salud Poblacional (GESP) (L.O.,), School of Public Health, Universidad del Valle, Cali, Colombia; Department of Microbiology (B.P.), School of Basic Sciences, Universidad del Valle, Cali, Colombia; Dengue Hemorrhagic Fever Research Unit (J.M.), Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol Univeristy, Bangkok, Thailand; Department of Neurology (C.A.P.), Johns Hopkins University School of Medicine, Baltimore, MD; and LifeFactors (J.-M.A.), Rionegro, Colombia; Division of Emerging Infectious Disease (W.D.), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Susan K Halstead
- From the Nuffield Department of Clinical Neurosciences (A.J.D., S.R.), University of Oxford, John Radcliffe Hospital, UK; Neuromuscular Diseases Unit (C.L., A.M.S., L.Q.), Neurology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Spain; Centro para la Investigación Biomédica en red en Enfermedades Raras-(CIBERER) Madrid (C.L., A.M.S., L.Q.), Spain; Institute of Infection (D.S.G., S.K.H., H.J.W.), Immunity & Inflammation, University of Glasgow, University Place, UK; Target Discovery Institute (G.B., R.F.), NDM Research Building, University of Oxford, Old Road Campus, UK; Wellcome Centre for Human Genetics (W.D., G.R.S., J.M.), Nuffield Department of Medicine, University of Oxford, UK; Center for Autoimmune Diseases Research (CREA) (C.R.-S., J.-M.A.), Universidad del Rosario, Bogotá, Colombia; Departamento de Medicina (A.K.F., C.M.R.-V.), Universidad del Norte, Barranquilla, Colombia; Grupo de Epidemiología y Salud Poblacional (GESP) (L.O.,), School of Public Health, Universidad del Valle, Cali, Colombia; Department of Microbiology (B.P.), School of Basic Sciences, Universidad del Valle, Cali, Colombia; Dengue Hemorrhagic Fever Research Unit (J.M.), Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol Univeristy, Bangkok, Thailand; Department of Neurology (C.A.P.), Johns Hopkins University School of Medicine, Baltimore, MD; and LifeFactors (J.-M.A.), Rionegro, Colombia; Division of Emerging Infectious Disease (W.D.), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Hugh J Willison
- From the Nuffield Department of Clinical Neurosciences (A.J.D., S.R.), University of Oxford, John Radcliffe Hospital, UK; Neuromuscular Diseases Unit (C.L., A.M.S., L.Q.), Neurology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Spain; Centro para la Investigación Biomédica en red en Enfermedades Raras-(CIBERER) Madrid (C.L., A.M.S., L.Q.), Spain; Institute of Infection (D.S.G., S.K.H., H.J.W.), Immunity & Inflammation, University of Glasgow, University Place, UK; Target Discovery Institute (G.B., R.F.), NDM Research Building, University of Oxford, Old Road Campus, UK; Wellcome Centre for Human Genetics (W.D., G.R.S., J.M.), Nuffield Department of Medicine, University of Oxford, UK; Center for Autoimmune Diseases Research (CREA) (C.R.-S., J.-M.A.), Universidad del Rosario, Bogotá, Colombia; Departamento de Medicina (A.K.F., C.M.R.-V.), Universidad del Norte, Barranquilla, Colombia; Grupo de Epidemiología y Salud Poblacional (GESP) (L.O.,), School of Public Health, Universidad del Valle, Cali, Colombia; Department of Microbiology (B.P.), School of Basic Sciences, Universidad del Valle, Cali, Colombia; Dengue Hemorrhagic Fever Research Unit (J.M.), Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol Univeristy, Bangkok, Thailand; Department of Neurology (C.A.P.), Johns Hopkins University School of Medicine, Baltimore, MD; and LifeFactors (J.-M.A.), Rionegro, Colombia; Division of Emerging Infectious Disease (W.D.), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Luis Querol
- From the Nuffield Department of Clinical Neurosciences (A.J.D., S.R.), University of Oxford, John Radcliffe Hospital, UK; Neuromuscular Diseases Unit (C.L., A.M.S., L.Q.), Neurology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Spain; Centro para la Investigación Biomédica en red en Enfermedades Raras-(CIBERER) Madrid (C.L., A.M.S., L.Q.), Spain; Institute of Infection (D.S.G., S.K.H., H.J.W.), Immunity & Inflammation, University of Glasgow, University Place, UK; Target Discovery Institute (G.B., R.F.), NDM Research Building, University of Oxford, Old Road Campus, UK; Wellcome Centre for Human Genetics (W.D., G.R.S., J.M.), Nuffield Department of Medicine, University of Oxford, UK; Center for Autoimmune Diseases Research (CREA) (C.R.-S., J.-M.A.), Universidad del Rosario, Bogotá, Colombia; Departamento de Medicina (A.K.F., C.M.R.-V.), Universidad del Norte, Barranquilla, Colombia; Grupo de Epidemiología y Salud Poblacional (GESP) (L.O.,), School of Public Health, Universidad del Valle, Cali, Colombia; Department of Microbiology (B.P.), School of Basic Sciences, Universidad del Valle, Cali, Colombia; Dengue Hemorrhagic Fever Research Unit (J.M.), Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol Univeristy, Bangkok, Thailand; Department of Neurology (C.A.P.), Johns Hopkins University School of Medicine, Baltimore, MD; and LifeFactors (J.-M.A.), Rionegro, Colombia; Division of Emerging Infectious Disease (W.D.), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Simon Rinaldi
- From the Nuffield Department of Clinical Neurosciences (A.J.D., S.R.), University of Oxford, John Radcliffe Hospital, UK; Neuromuscular Diseases Unit (C.L., A.M.S., L.Q.), Neurology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Spain; Centro para la Investigación Biomédica en red en Enfermedades Raras-(CIBERER) Madrid (C.L., A.M.S., L.Q.), Spain; Institute of Infection (D.S.G., S.K.H., H.J.W.), Immunity & Inflammation, University of Glasgow, University Place, UK; Target Discovery Institute (G.B., R.F.), NDM Research Building, University of Oxford, Old Road Campus, UK; Wellcome Centre for Human Genetics (W.D., G.R.S., J.M.), Nuffield Department of Medicine, University of Oxford, UK; Center for Autoimmune Diseases Research (CREA) (C.R.-S., J.-M.A.), Universidad del Rosario, Bogotá, Colombia; Departamento de Medicina (A.K.F., C.M.R.-V.), Universidad del Norte, Barranquilla, Colombia; Grupo de Epidemiología y Salud Poblacional (GESP) (L.O.,), School of Public Health, Universidad del Valle, Cali, Colombia; Department of Microbiology (B.P.), School of Basic Sciences, Universidad del Valle, Cali, Colombia; Dengue Hemorrhagic Fever Research Unit (J.M.), Office for Research and Development, Siriraj Hospital, Faculty of Medicine, Mahidol Univeristy, Bangkok, Thailand; Department of Neurology (C.A.P.), Johns Hopkins University School of Medicine, Baltimore, MD; and LifeFactors (J.-M.A.), Rionegro, Colombia; Division of Emerging Infectious Disease (W.D.), Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
19
|
Shang J, Li C, Jin Z, Zu S, Chen S, Chen J, Chen Z, Tang H, Qin CF, Ye Q, Wu A. Immune profiles in mouse brain and testes infected by Zika virus with variable pathogenicity. Front Cell Infect Microbiol 2022; 12:948980. [PMID: 35992167 PMCID: PMC9385972 DOI: 10.3389/fcimb.2022.948980] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
The Zika virus is responsible for neurological diseases such as microcephaly, Guillain-Barré syndrome, neuropathy, and myelitis in human adults and children. Previous studies have shown that the Zika virus can infect nerve progenitor cells and interfere with neural development. However, it is unclear how the immune system responds to infection with Zika viruses with variable pathogenicity. Here, we used two Zika strains with relatively different pathogenicity, the Asian ancestral strain CAM/2010 and the America pandemic strain GZ01/2016, to infect the brains of mice. We found that both strains elicited a strong immune response. Notably, the strain with relatively high pathogenicity, GZ01/2016, caused more intense immune regulation, with stronger CD8+ T cell and macrophage activation at 14 days post infection (dpi), as well as a greater immune gene disturbance. Notably, several TNF family genes were upregulated at 14 dpi, including Tnfrsf9, Tnfsf13, Tnfrsf8, Cd40, and Tnfsf10. It was notable that GZ01/2016 could maintain the survival of nerve cells at 7dpi but caused neurological disorders at 14dpi. These results indicate that Zika viruses with high pathogenicity may induce sustained activation of the immune system leading to nerve tissue damage.
Collapse
Affiliation(s)
- Jingzhe Shang
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Suzhou Institute of Systems Medicine, Suzhou, China
| | - Chunfeng Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, United States
| | - Zhujia Jin
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Suzhou Institute of Systems Medicine, Suzhou, China
| | - Shulong Zu
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Suzhou Institute of Systems Medicine, Suzhou, China
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
- Center for Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Songjie Chen
- Departments of Genetics, School of Medicine, Stanford University, Stanford, CA, United States
| | - Junlan Chen
- State Key Laboratory Breeding Base of Basic Science of Stomatology, Ministry of Education, Hospital of Stomatology, Faculty of Medical Sciences, Wuhan University, Wuhan, China
| | - Ziyi Chen
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Suzhou Institute of Systems Medicine, Suzhou, China
| | - Hua Tang
- Institute of Immunology, Shandong First Medical University, Tai’an, China
| | - Cheng-Feng Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Qing Ye
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
- *Correspondence: Qing Ye, ; Aiping Wu,
| | - Aiping Wu
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Suzhou Institute of Systems Medicine, Suzhou, China
- *Correspondence: Qing Ye, ; Aiping Wu,
| |
Collapse
|
20
|
Reagin KL, Funk KE. The role of antiviral CD8 + T cells in cognitive impairment. Curr Opin Neurobiol 2022; 76:102603. [PMID: 35810534 DOI: 10.1016/j.conb.2022.102603] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 11/16/2022]
Abstract
The impact of the immune system on the etiopathogenesis of neurodegenerative diseases, including Alzheimer's disease, is a rapidly growing area of investigation. Evidence from human patients and animal models implicates neurotropic viral infections, and specifically the antiviral immune response of brain-infiltrating CD8+ T cells, as potential drivers of disease pathology. While infiltration and retention of CD8+ T cells within the brain following viral infection is associated with improved survival, CD8+ T cells also contribute to neuronal death and gliosis which underlie cognitive impairment in several disease models. Here we review the role of antiviral CD8+ T cells as potential mediators of cognitive impairment and highlight the mechanisms by which brain-resident CD8+ T cells may contribute to neurodegenerative disease pathology.
Collapse
Affiliation(s)
- Katie L Reagin
- Department of Biological Sciences, University of North Carolina, Charlotte, NC, USA
| | - Kristen E Funk
- Department of Biological Sciences, University of North Carolina, Charlotte, NC, USA.
| |
Collapse
|
21
|
Fikatas A, Dehairs J, Noppen S, Doijen J, Vanderhoydonc F, Meyen E, Swinnen JV, Pannecouque C, Schols D. Deciphering the Role of Extracellular Vesicles Derived from ZIKV-Infected hcMEC/D3 Cells on the Blood-Brain Barrier System. Viruses 2021; 13:v13122363. [PMID: 34960632 PMCID: PMC8708812 DOI: 10.3390/v13122363] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/12/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022] Open
Abstract
To date, no vaccines or antivirals are available against Zika virus (ZIKV). In addition, the mechanisms underlying ZIKV-associated pathogenesis of the central nervous system (CNS) are largely unexplored. Getting more insight into the cellular pathways that ZIKV recruits to facilitate infection of susceptible cells will be crucial for establishing an effective treatment strategy. In general, cells secrete a number of vesicles, known as extracellular vesicles (EVs), in response to viral infections. These EVs serve as intercellular communicators. Here, we investigated the role of EVs derived from ZIKV-infected human brain microvascular endothelial cells on the blood–brain barrier (BBB) system. We demonstrated that ZIKV-infected EVs (IEVs) can incorporate viral components, including ZIKV RNA, NS1, and E-protein, and further transfer them to several types of CNS cells. Using label-free impedance-based biosensing, we observed that ZIKV and IEVs can temporally disturb the monolayer integrity of BBB-mimicking cells, possibly by inducing structural rearrangements of the adherent protein VE-cadherin (immunofluorescence staining). Finally, differences in the lipidomic profile between EVs and their parental cells possibly suggest a preferential sorting mechanism of specific lipid species into the vesicles. To conclude, these data suggest that IEVs could be postulated as vehicles (Trojan horse) for ZIKV transmission via the BBB.
Collapse
Affiliation(s)
- Antonios Fikatas
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (A.F.); (S.N.); (J.D.); (E.M.); (C.P.)
| | - Jonas Dehairs
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (J.D.); (F.V.); (J.V.S.)
| | - Sam Noppen
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (A.F.); (S.N.); (J.D.); (E.M.); (C.P.)
| | - Jordi Doijen
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (A.F.); (S.N.); (J.D.); (E.M.); (C.P.)
| | - Frank Vanderhoydonc
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (J.D.); (F.V.); (J.V.S.)
| | - Eef Meyen
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (A.F.); (S.N.); (J.D.); (E.M.); (C.P.)
| | - Johannes V. Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (J.D.); (F.V.); (J.V.S.)
| | - Christophe Pannecouque
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (A.F.); (S.N.); (J.D.); (E.M.); (C.P.)
| | - Dominique Schols
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (A.F.); (S.N.); (J.D.); (E.M.); (C.P.)
- Correspondence: ; Tel.: +32-16-32-19-98
| |
Collapse
|
22
|
Abstract
Zika virus (ZIKV) infection became a worldwide concern due to its correlation with the development of microcephaly and other neurological disorders. ZIKV neurotropism is well characterized, but the role of peripheral viral amplification to brain infection remains unknown. Here, we found that ZIKV replicates in human primary skeletal muscle myoblasts, impairing its differentiation into myotubes but not interfering with the integrity of the already-formed muscle fibers. Using mouse models, we showed ZIKV tropism to muscle tissue either during embryogenesis after maternal transmission or when infection occurred after birth. Interestingly, ZIKV replication in the mouse skeletal muscle started immediately after ZIKV inoculation, preceding viral RNA detection in the brain and causing no disruption to the integrity of the blood brain barrier, and remained active for more than 2 weeks, whereas replication in the spleen and liver were not sustained over time. In addition, ZIKV infection of the skeletal muscle induces necrotic lesions, inflammation, and fiber atrophy. We also found a reduction in the expression of regulatory myogenic factors that are essential for muscle repair after injury. Taken together, our results indicate that the skeletal muscle is an early site of viral amplification and lesion that may result in late consequences in muscle development after ZIKV infection. IMPORTANCE Zika Virus (ZIKV) neurotropism and its deleterious effects on central nervous system have been well characterized. However, investigations of the initial replication sites for the establishment of infection and viral spread to neural tissues remain underexplored. A complete description of the range of ZIKV-induced lesions and others factors that can influence the severity of the disease is necessary to prevent ZIKV’s deleterious effects. ZIKV has been shown to access the central nervous system without significantly affecting blood-brain barrier permeability. Here, we demonstrated that skeletal muscle is an earlier site of ZIKV replication, contributing to the increase of peripheral ZIKV load. ZIKV replication in muscle promotes necrotic lesions and inflammation and also impairs myogenesis. Overall, our findings showed that skeletal muscle is involved in pathogenesis and opens new fields in the investigation of the long-term consequences of early infection.
Collapse
|
23
|
Wong FC, Ye L, Demir IE, Kahlert C. Schwann cell-derived exosomes: Janus-faced mediators of regeneration and disease. Glia 2021; 70:20-34. [PMID: 34519370 DOI: 10.1002/glia.24087] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 12/20/2022]
Abstract
The phenotypic plasticity of Schwann cells (SCs) has contributed to the regenerative potential of the peripheral nervous system (PNS), but also pathological processes. This double-sided effect has led to an increasing attention to the role of extracellular vesicles (EVs) or exosomes in SCs to examine the intercellular communication between SCs and their surroundings. Here, we first describe the current knowledge of SC and EV biology, which forms the basis for the updates on advances in SC-derived exosomes research. We seek to explore in-depth the exosome-mediated molecular mechanisms involved in the regulation of SCs and their microenvironment. This review concludes with potential applications of SC-derived exosomes as delivery vehicles for therapeutics and biomarkers. The goal of this review is to emphasize the crucial role of SC-derived exosomes in the functional integration of the PNS, highlighting an emerging area in which there is much to explore and re-explore.
Collapse
Affiliation(s)
- Fang Cheng Wong
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Linhan Ye
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany.,Germany German Cancer Consortium (DKTK), Partner Site, Munich, Germany.,CRC 1321 Modelling and Targeting Pancreatic Cancer, Munich, Germany
| | - Ihsan Ekin Demir
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany.,Germany German Cancer Consortium (DKTK), Partner Site, Munich, Germany.,Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey.,CRC 1321 Modelling and Targeting Pancreatic Cancer, Munich, Germany.,Else Kröner Clinician Scientist Professor for "Translational Pancreatic Surgery
| | - Christoph Kahlert
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT/UCC), Dresden, Germany
| |
Collapse
|
24
|
Monticolo F, Chiusano ML. Computational Approaches for Cancer-Fighting: From Gene Expression to Functional Foods. Cancers (Basel) 2021; 13:4207. [PMID: 34439361 PMCID: PMC8393935 DOI: 10.3390/cancers13164207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/13/2021] [Accepted: 08/17/2021] [Indexed: 01/22/2023] Open
Abstract
It is today widely accepted that a healthy diet is very useful to prevent the risk for cancer or its deleterious effects. Nutrigenomics studies are therefore taking place with the aim to test the effects of nutrients at molecular level and contribute to the search for anti-cancer treatments. These efforts are expanding the precious source of information necessary for the selection of natural compounds useful for the design of novel drugs or functional foods. Here we present a computational study to select new candidate compounds that could play a role in cancer prevention and care. Starting from a dataset of genes that are co-expressed in programmed cell death experiments, we investigated on nutrigenomics treatments inducing apoptosis, and searched for compounds that determine the same expression pattern. Subsequently, we selected cancer types where the genes showed an opposite expression pattern and we confirmed that the apoptotic/nutrigenomics expression trend had a significant positive survival in cancer-affected patients. Furthermore, we considered the functional interactors of the genes as defined by public protein-protein interaction data, and inferred on their involvement in cancers and/or in programmed cell death. We identified 7 genes and, from available nutrigenomics experiments, 6 compounds effective on their expression. These 6 compounds were exploited to identify, by ligand-based virtual screening, additional molecules with similar structure. We checked for ADME criteria and selected 23 natural compounds representing suitable candidates for further testing their efficacy in apoptosis induction. Due to their presence in natural resources, novel drugs and/or the design of functional foods are conceivable from the presented results.
Collapse
Affiliation(s)
| | - Maria Luisa Chiusano
- Department of Agricultural Sciences, Università degli Studi di Napoli Federico II, Via Università 100, 80055 Portici, Italy;
| |
Collapse
|
25
|
Kousa YA, Hossain RA. Causes of Phenotypic Variability and Disabilities after Prenatal Viral Infections. Trop Med Infect Dis 2021; 6:tropicalmed6020095. [PMID: 34205913 PMCID: PMC8293342 DOI: 10.3390/tropicalmed6020095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/25/2021] [Accepted: 05/29/2021] [Indexed: 12/14/2022] Open
Abstract
Prenatal viral infection can lead to a spectrum of neurodevelopmental disabilities or fetal demise. These can include microencephaly, global developmental delay, intellectual disability, refractory epilepsy, deafness, retinal defects, and cortical-visual impairment. Each of these clinical conditions can occur on a semi-quantitative to continuous spectrum, from mild to severe disease, and often as a collective of phenotypes. Such serious outcomes result from viruses’ overlapping neuropathology and hosts’ common neuronal and gene regulatory response to infections. The etiology of variability in clinical outcomes is not yet clear, but it may be related to viral, host, vector, and/or environmental risk and protective factors that likely interact in multiple ways. In this perspective of the literature, we work toward understanding the causes of phenotypic variability after prenatal viral infections by highlighting key aspects of the viral lifecycle that can affect human disease, with special attention to the 2015 Zika pandemic. Therefore, this work offers important insights into how viral infections and environmental teratogens affect the prenatal brain, toward our ultimate goal of preventing neurodevelopmental disabilities.
Collapse
Affiliation(s)
- Youssef A. Kousa
- Division of Neurology, Children’s National Hospital, Washington, DC 20010, USA
- Department of Genomics and Precision Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
- Correspondence:
| | - Reafa A. Hossain
- Structural Virology Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institute of Health, Bethesda, MD 20892, USA;
| |
Collapse
|
26
|
Chen W, Foo SS, Hong E, Wu C, Lee WS, Lee SA, Evseenko D, Lopes Moreira ME, García-Sastre A, Cheng G, Nielsen-Saines K, Brasil P, Avvad-Portari E, Jung JU. Zika virus NS3 protease induces bone morphogenetic protein-dependent brain calcification in human fetuses. Nat Microbiol 2021; 6:455-466. [PMID: 33510473 PMCID: PMC8012254 DOI: 10.1038/s41564-020-00850-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 12/11/2020] [Indexed: 01/29/2023]
Abstract
The most frequent fetal birth defect associated with prenatal Zika virus (ZIKV) infection is brain calcification, which in turn may potentially affect neurological development in infants. Understanding the mechanism could inform the development of potential therapies against prenatal ZIKV brain calcification. In perivascular cells, bone morphogenetic protein (BMP) is an osteogenic factor that undergoes maturation to activate osteogenesis and calcification. Here, we show that ZIKV infection of cultivated primary human brain pericytes triggers BMP2 maturation, leading to osteogenic gene expression and calcification. We observed extensive calcification near ZIKV+ pericytes of fetal human brain specimens and in vertically transmitted ZIKV+ human signal transducer and activator of transcription 2-knockin mouse pup brains. ZIKV infection of primary pericytes stimulated BMP2 maturation, inducing osteogenic gene expression and calcification that were completely blocked by anti-BMP2/4 neutralizing antibody. Not only did ZIKV NS3 expression alone induce BMP2 maturation, osteogenic gene expression and calcification, but purified NS3 protease also effectively cleaved pro-BMP2 in vitro to generate biologically active mature BMP2. These findings highlight ZIKV-induced calcification where the NS3 protease subverts the BMP2-mediated osteogenic signalling pathway to trigger brain calcification.
Collapse
Affiliation(s)
- Weiqiang Chen
- Department of Cancer Biology and Global Center for Pathogens Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Suan-Sin Foo
- Department of Cancer Biology and Global Center for Pathogens Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Eunjin Hong
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Christine Wu
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Wai-Suet Lee
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Shin-Ae Lee
- Department of Cancer Biology and Global Center for Pathogens Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Denis Evseenko
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Maria Elisabeth Lopes Moreira
- Clinical Research Unit, Fernandes Figueira Institute-FioCruz, Avenida Rui Barbosa, 716, Flamengo, Rio De Janeiro, RJ CEP 22250-020, Brazil
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA;,Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA;,Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA;,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Genhong Cheng
- Department of Microbiology and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Marion Davies Children’s Health Center, 10833 LeConte Avenue, Los Angeles, CA 90095, USA
| | - Karin Nielsen-Saines
- Division of Pediatric Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Marion Davies Children’s Health Center, 10833 LeConte Avenue, Los Angeles, CA 90095, USA
| | - Patrícia Brasil
- Laboratório de Pesquisa Clínica em Doenças Febris Agudas, Instituto Nacional de Infectologia Evandro Chagas, FioCruz, 4365 Avenida Brasil, Rio de Janeiro – RJ, 21040-360, Brazil
| | - Elyzabeth Avvad-Portari
- Department of Pathology, Fernandes Figueira Institute-FioCruz, Avenida Rui Barbosa, 716, Flamengo, Rio De Janeiro, RJ CEP 22250-020, Brazil
| | - Jae U. Jung
- Department of Cancer Biology and Global Center for Pathogens Research and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA;,Correspondence: (Jae U. Jung, PhD)
| |
Collapse
|
27
|
Bhagat R, Kaur G, Seth P. Molecular mechanisms of zika virus pathogenesis: An update. Indian J Med Res 2021; 154:433-445. [PMID: 35345069 PMCID: PMC9131805 DOI: 10.4103/ijmr.ijmr_169_20] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Indexed: 01/04/2023] Open
Abstract
Zika virus (ZIKV), member of the family Flaviviridae belonging to genus Flavivirus, is an arthropod-borne virus. The ZIKV is known to cause severe congenital birth defects in neonates. Due to a large number of worldwide outbreaks and associated neurological complications with ZIKV, a public health emergency was declared by the World Health Organization on February 1, 2016. The virus exhibits neurotropism and has a specific propensity towards neural precursor cells of the developing brain. In utero ZIKV infection causes massive cell death in the developing brain resulting in various motor and cognitive disabilities in newborns. The virus modulates cell machinery at several levels to replicate itself and inhibits toll like receptors-3 signalling, deregulates microRNA circuitry and induces a chronic inflammatory response in affected cells. Several significant advances have been made to understand the mechanisms of neuropathogenesis, its prevention and treatment. The current review provides an update on cellular and molecular mechanisms of ZIKV-induced alterations in the function of various brain cells.
Collapse
Affiliation(s)
- Reshma Bhagat
- Department of Cellular & Molecular Neuroscience, National Brain Research Centre, Manesar, Gurgaon, India
- Department of Genetics, Washington University in Saint Louis, Missouri, United States of America
| | - Guneet Kaur
- Department of Cellular & Molecular Neuroscience, National Brain Research Centre, Manesar, Gurgaon, India
| | - Pankaj Seth
- Department of Cellular & Molecular Neuroscience, National Brain Research Centre, Manesar, Gurgaon, India
| |
Collapse
|
28
|
Zimmer A, Youngblood A, Adnane A, Miller BJ, Goldsmith DR. Prenatal exposure to viral infection and neuropsychiatric disorders in offspring: A review of the literature and recommendations for the COVID-19 pandemic. Brain Behav Immun 2021; 91:756-770. [PMID: 33152446 PMCID: PMC7759331 DOI: 10.1016/j.bbi.2020.10.024] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/21/2020] [Accepted: 10/27/2020] [Indexed: 02/08/2023] Open
Abstract
The SARS-CoV-2 virus has emerged as a striking 21st century pandemic. Communities across the globe have experienced significant infection rates and widespread psychosocial stress and trauma, leading to calls for increased allocation of resources for mental health screening and treatment. In addition to the burden of psychosocial stress, there is increasing evidence of direct viral neuroinvasion of the central nervous system through physical contact with the nasal mucosa. In a parallel fashion, there is a significant body of ongoing research related to the risk of in utero viral transmission and the resulting neurodevelopmental impact in the fetus. Aberrant neurodevelopment secondary to viral transmission has previously been related to the later development of psychosis, schizophrenia, and schizophrenia spectrum disorders, generating the hypothesis that this population of individuals exposed to SARS-CoV-2 may see an increased incidence in future decades. We discuss the current understanding of the possible neurotropism and vertical transmission of SARS-CoV-2, and relate this to the history of viral pandemics to better understand the relationship of viral infection, aberrant immune response and neurodevelopment, and the risk for schizophrenia disorder.
Collapse
Affiliation(s)
- Anna Zimmer
- Emory University School of Medicine, Atlanta, GA, USA
| | | | - Adam Adnane
- Emory University School of Medicine, Atlanta, GA, USA
| | - Brian J Miller
- Department of Psychiatry and Health Behavior, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - David R Goldsmith
- Emory University School of Medicine, Atlanta, GA, USA; Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA.
| |
Collapse
|
29
|
Puthanveetil P, Kong X, Bräse S, Voros G, Peer WA. Transcriptome analysis of two structurally related flavonoids; Apigenin and Chrysin revealed hypocholesterolemic and ketogenic effects in mouse embryonic fibroblasts. Eur J Pharmacol 2020; 893:173804. [PMID: 33347826 DOI: 10.1016/j.ejphar.2020.173804] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/02/2020] [Accepted: 12/10/2020] [Indexed: 12/14/2022]
Abstract
There is no known single therapeutic drug for treating hypercholesterolemia that comes with negligible systemic side effects. In the current study, using next generation RNA sequencing approach in mouse embryonic fibroblasts we discovered that two structurally related flavonoid compounds. Apigenin and Chrysin exhibited moderate blocking ability of multiple transcripts that regulate rate limiting enzymes in the cholesterol biosynthesis pathway. The observed decrease in cholesterol biosynthesis pathway correlated well with an increase in transcripts involved in generation and trafficking of ketone bodies as evident by the upregulation of Bdh1 and Slc16a6 transcripts. The hypocholesterolemic potential of Apigenin and Chrysin at higher concentrations along with their ability to generate ketogenic substrate especially during embryonic stage is useful or detrimental for embryonic health is not clear and still debatable. Our study will serve as a steppingstone to further the investigation in whole animal studies and also in translating this knowledge to human studies.
Collapse
Affiliation(s)
- Prasanth Puthanveetil
- Department of Pharmacology, College of Graduate Studies, Midwestern University, Downers Grove, IL, USA.
| | - Xiaoli Kong
- Department of Mathematics and Statistics, Loyola University Chicago, Chicago, IL, USA.
| | - Stefan Bräse
- Institute of Biological and Chemical Systems (IBCS), Karlsruhe Institute of Technology (KIT), Hermann von Helmholtz Platz 1, 76344, Eggenstein Leopoldshafen, Germany.
| | - Gabor Voros
- Department of Cardiovascular Diseases, University Hospital Gasthuisberg, Catholic University Leuven, Herestraat 49, 3000, Leuven, Belgium.
| | - Wendy Ann Peer
- Department of Environmental Science and Technology, College of Agricultural and Natural Resources, University of Maryland, MD, USA.
| |
Collapse
|
30
|
Yang S, Gorshkov K, Lee EM, Xu M, Cheng YS, Sun N, Soheilian F, de Val N, Ming G, Song H, Tang H, Zheng W. Zika Virus-Induced Neuronal Apoptosis via Increased Mitochondrial Fragmentation. Front Microbiol 2020; 11:598203. [PMID: 33424801 PMCID: PMC7785723 DOI: 10.3389/fmicb.2020.598203] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 12/02/2020] [Indexed: 12/19/2022] Open
Abstract
The 2015 to 2016 outbreak of Zika virus (ZIKV) infections in the Americas coincided with a dramatic increase in neurodevelopmental abnormalities, including fetal microcephaly, in newborns born to infected women. In this study, we observed mitochondrial fragmentation and disrupted mitochondrial membrane potential after 24 h of ZIKV infection in human neural stem cells and the SNB-19 glioblastoma cell line. The severity of these changes correlated with the amount of ZIKV proteins expressed in infected cells. ZIKV infection also decreased the levels of mitofusin 2, which modulates mitochondria fusion. Mitochondrial division inhibitor 1 (Mdivi-1), a small molecule inhibiting mitochondria fission, ameliorated mitochondria disruptions and reduced cell death in ZIKV-infected cells. Collectively, this study suggests that abnormal mitochondrial fragmentation contributes to ZIKV-induced neuronal cell death; rebalancing mitochondrial dynamics of fission-fusion could be a therapeutic strategy for drug development to treat ZIKV-mediated neuronal apoptosis.
Collapse
Affiliation(s)
- Shu Yang
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| | - Kirill Gorshkov
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| | - Emily M Lee
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| | - Miao Xu
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| | - Yu-Shan Cheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| | - Nuo Sun
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Ferri Soheilian
- Electron Microscopy Laboratory, National Cancer Institute, Center for Cancer Research, Leidos Biomedical Research, Frederick National Laboratory, Frederick, MD, United States
| | - Natalia de Val
- Electron Microscopy Laboratory, National Cancer Institute, Center for Cancer Research, Leidos Biomedical Research, Frederick National Laboratory, Frederick, MD, United States
| | - Guoli Ming
- Department of Neuroscience, Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, PA, United States
| | - Hongjun Song
- Department of Neuroscience, Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, PA, United States
| | - Hengli Tang
- Department of Biological Science, Florida State University, Tallahassee, FL, United States
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
31
|
Monticolo F, Palomba E, Chiusano ML. Identification of Novel Potential Genes Involved in Cancer by Integrated Comparative Analyses. Int J Mol Sci 2020; 21:ijms21249560. [PMID: 33334055 PMCID: PMC7765469 DOI: 10.3390/ijms21249560] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/30/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
The main hallmarks of cancer diseases are the evasion of programmed cell death, uncontrolled cell division, and the ability to invade adjacent tissues. The explosion of omics technologies offers challenging opportunities to identify molecular agents and processes that may play relevant roles in cancer. They can support comparative investigations, in one or multiple experiments, exploiting evidence from one or multiple species. Here, we analyzed gene expression data from induction of programmed cell death and stress response in Homo sapiens and compared the results with Saccharomyces cerevisiae gene expression during the response to cell death. The aim was to identify conserved candidate genes associated with Homo sapiens cell death, favored by crosslinks based on orthology relationships between the two species. We identified differentially-expressed genes, pathways that are significantly dysregulated across treatments, and characterized genes among those involved in induced cell death. We investigated on co-expression patterns and identified novel genes that were not expected to be associated with death pathways, that have a conserved pattern of expression between the two species. Finally, we analyzed the resulting list by HumanNet and identified new genes predicted to be involved in cancer. The data integration and the comparative approach between distantly-related reference species that were here exploited pave the way to novel discoveries in cancer therapy and also contribute to detect conserved genes potentially involved in programmed cell death.
Collapse
Affiliation(s)
- Francesco Monticolo
- Department of Agricultural Sciences, Università Degli Studi di Napoli Federico II, 80055 Naples, Italy;
| | - Emanuela Palomba
- Department of RIMAR, Stazione Zoologica “Anton Dohrn”, 80122 Naples, Italy;
| | - Maria Luisa Chiusano
- Department of Agricultural Sciences, Università Degli Studi di Napoli Federico II, 80055 Naples, Italy;
- Department of RIMAR, Stazione Zoologica “Anton Dohrn”, 80122 Naples, Italy;
- Correspondence:
| |
Collapse
|
32
|
Souza GAPD, Salvador EA, de Oliveira FR, Cotta Malaquias LC, Abrahão JS, Leomil Coelho LF. An in silico integrative protocol for identifying key genes and pathways useful to understand emerging virus disease pathogenesis. Virus Res 2020; 284:197986. [PMID: 32339536 DOI: 10.1016/j.virusres.2020.197986] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/21/2020] [Accepted: 04/21/2020] [Indexed: 12/31/2022]
Abstract
The pathogenesis of an emerging virus disease is a difficult task due to lack of scientific data about the emerging virus during outbreak threats. Several biological aspects should be studied faster, such as virus replication and dissemination, immune responses to this emerging virus on susceptible host and specially the virus pathogenesis. Integrative in silico transcriptome analysis is a promising approach for understanding biological events in complex diseases. In this study, we propose an in silico protocol for identifying key genes and pathways useful to understand emerging virus disease pathogenesis. To validate our protocol, the emerging arbovirus Zika virus (ZIKV) was chosen as a target micro-organism. First, an integrative transcriptome data from neural cells infected with ZIKV was used to identify shared differentially expressed genes (DEGs). The DEGs were used to identify the potential candidate genes and pathways in ZIKV pathogenesis through gene enrichment analysis and protein‑protein interaction network construction. Thirty DEGs (24 upregulated and 6 downregulated) were identified in all ZIKV-infected cells, primarily associated with endoplasmic reticulum stress and DNA replication pathways. Some of these genes and pathways had biological functions linked to neurogenesis and/or apoptosis, confirming the potential of this protocol to find key genes and pathways involved on disease pathogenesis. Moreover, the proposed in silico protocol performed anintegrated analysis that is able to predict and identify putative biomarkers from different transcriptome data. These biomarkers could be useful to understand virus disease pathogenesis and also help the identification of candidate antiviral drugs.
Collapse
Affiliation(s)
- Gabriel Augusto Pires de Souza
- Laboratório de Vacinas, Instituto de Ciências Biomédicas, Departamento de Microbiologia e Imunologia, Universidade Federal de Alfenas, Alfenas, Minas Gerais, Brazil; Laboratório de Vírus, Instituto de Ciências Biológicas, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ezequiel Aparecido Salvador
- Laboratório de Vacinas, Instituto de Ciências Biomédicas, Departamento de Microbiologia e Imunologia, Universidade Federal de Alfenas, Alfenas, Minas Gerais, Brazil
| | - Fernanda Roza de Oliveira
- Laboratório de Vacinas, Instituto de Ciências Biomédicas, Departamento de Microbiologia e Imunologia, Universidade Federal de Alfenas, Alfenas, Minas Gerais, Brazil
| | - Luiz Cosme Cotta Malaquias
- Laboratório de Vacinas, Instituto de Ciências Biomédicas, Departamento de Microbiologia e Imunologia, Universidade Federal de Alfenas, Alfenas, Minas Gerais, Brazil
| | - Jonatas Santos Abrahão
- Laboratório de Vírus, Instituto de Ciências Biológicas, Departamento de Microbiologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luiz Felipe Leomil Coelho
- Laboratório de Vacinas, Instituto de Ciências Biomédicas, Departamento de Microbiologia e Imunologia, Universidade Federal de Alfenas, Alfenas, Minas Gerais, Brazil.
| |
Collapse
|
33
|
Zika virus infection differentially affects genome-wide transcription in neuronal cells and myeloid dendritic cells. PLoS One 2020; 15:e0231049. [PMID: 32287277 PMCID: PMC7156053 DOI: 10.1371/journal.pone.0231049] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 03/15/2020] [Indexed: 11/19/2022] Open
Abstract
Zika virus (ZIKV) is an emerging mosquito-borne flavivirus that has attracted global attention and international awareness. ZIKV infection exhibits mild symptoms including fever and pains; however, ZIKV has recently been shown to be related to increased birth defects, including microcephaly, in infants. In addition, ZIKV is related to the onset of neurological disorders, such as a type of paralysis similar to Guillain-Barré syndrome. However, the mechanisms through which ZIKV affect neuronal cells and myeloid dendritic cells and how ZIKV avoids host immunity are unclear. Accordingly, in this study, we analyzed RNA sequencing data from ZIKV-infected neuronal cells and myeloid dendritic cells by comparative network analyses using protein-protein interaction information. Comparative network analysis revealed major genes showing differential changes in the peripheral neurons, neural crest cells, and myeloid dendritic cells after ZIKV infection. The genes were related to DNA repair systems and prolactin signaling as well as the interferon signaling, neuroinflammation, and cell cycle pathways. These pathways were interconnected by the interaction of proteins in the pathway and significantly regulated by ZIKV infection in neuronal cells and myeloid dendritic cells. Our analysis showed that neuronal cell damage occurred through up-regulation of neuroinflammation and down-regulation of the DNA repair system, but not in myeloid dendritic cells. Interestingly, immune escape by ZIKV infection could be caused by downregulation of prolactin signaling including IRS2, PIK3C3, JAK3, STAT3, and IRF1 as well as mitochondria dysfunction and oxidative phosphorylation in myeloid dendritic cells. These findings provide insight into the mechanisms of ZIKV infection in the host and the association of ZIKV with neurological and immunological symptoms, which may facilitate the development of therapeutic agents and vaccines.
Collapse
|
34
|
Zhu Z, Mesci P, Bernatchez JA, Gimple RC, Wang X, Schafer ST, Wettersten HI, Beck S, Clark AE, Wu Q, Prager BC, Kim LJY, Dhanwani R, Sharma S, Garancher A, Weis SM, Mack SC, Negraes PD, Trujillo CA, Penalva LO, Feng J, Lan Z, Zhang R, Wessel AW, Dhawan S, Diamond MS, Chen CC, Wechsler-Reya RJ, Gage FH, Hu H, Siqueira-Neto JL, Muotri AR, Cheresh DA, Rich JN. Zika Virus Targets Glioblastoma Stem Cells through a SOX2-Integrin α vβ 5 Axis. Cell Stem Cell 2020; 26:187-204.e10. [PMID: 31956038 DOI: 10.1016/j.stem.2019.11.016] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 07/10/2019] [Accepted: 11/22/2019] [Indexed: 12/12/2022]
Abstract
Zika virus (ZIKV) causes microcephaly by killing neural precursor cells (NPCs) and other brain cells. ZIKV also displays therapeutic oncolytic activity against glioblastoma (GBM) stem cells (GSCs). Here we demonstrate that ZIKV preferentially infected and killed GSCs and stem-like cells in medulloblastoma and ependymoma in a SOX2-dependent manner. Targeting SOX2 severely attenuated ZIKV infection, in contrast to AXL. As mechanisms of SOX2-mediated ZIKV infection, we identified inverse expression of antiviral interferon response genes (ISGs) and positive correlation with integrin αv (ITGAV). ZIKV infection was disrupted by genetic targeting of ITGAV or its binding partner ITGB5 and by an antibody specific for integrin αvβ5. ZIKV selectively eliminated GSCs from species-matched human mature cerebral organoids and GBM surgical specimens, which was reversed by integrin αvβ5 inhibition. Collectively, our studies identify integrin αvβ5 as a functional cancer stem cell marker essential for GBM maintenance and ZIKV infection, providing potential brain tumor therapy.
Collapse
Affiliation(s)
- Zhe Zhu
- Department of Medicine, Division of Regenerative Medicine, University of California School of Medicine, San Diego, La Jolla, CA 92037, USA; Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA 92037, USA
| | - Pinar Mesci
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA 92037, USA; Department of Pediatrics, Rady Children's Hospital San Diego, School of Medicine, University of California, San Diego, La Jolla, CA 92307, USA; Department of Cellular and Molecular Medicine, Stem Cell Program, School of Medicine, University of California, San Diego, La Jolla, CA 92307, USA
| | - Jean A Bernatchez
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92037, USA
| | - Ryan C Gimple
- Department of Medicine, Division of Regenerative Medicine, University of California School of Medicine, San Diego, La Jolla, CA 92037, USA; Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA 92037, USA; Case Western Reserve University Medical Scientist Training Program, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Xiuxing Wang
- Department of Medicine, Division of Regenerative Medicine, University of California School of Medicine, San Diego, La Jolla, CA 92037, USA; Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA 92037, USA
| | - Simon T Schafer
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Hiromi I Wettersten
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA 92037, USA; Department of Pathology, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92037, USA
| | - Sungjun Beck
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92037, USA
| | - Alex E Clark
- Department of Cellular and Molecular Medicine and Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92037, USA
| | - Qiulian Wu
- Department of Medicine, Division of Regenerative Medicine, University of California School of Medicine, San Diego, La Jolla, CA 92037, USA; Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA 92037, USA
| | - Briana C Prager
- Department of Medicine, Division of Regenerative Medicine, University of California School of Medicine, San Diego, La Jolla, CA 92037, USA; Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA 92037, USA; Case Western Reserve University Medical Scientist Training Program, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Leo J Y Kim
- Department of Medicine, Division of Regenerative Medicine, University of California School of Medicine, San Diego, La Jolla, CA 92037, USA; Case Western Reserve University Medical Scientist Training Program, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Rekha Dhanwani
- La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Sonia Sharma
- La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Alexandra Garancher
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Sara M Weis
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA 92037, USA; Department of Pathology, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92037, USA
| | - Stephen C Mack
- Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX 77030, USA
| | - Priscilla D Negraes
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA 92037, USA; Department of Pediatrics, Rady Children's Hospital San Diego, School of Medicine, University of California, San Diego, La Jolla, CA 92307, USA
| | - Cleber A Trujillo
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA 92037, USA; Department of Pediatrics, Rady Children's Hospital San Diego, School of Medicine, University of California, San Diego, La Jolla, CA 92307, USA
| | - Luiz O Penalva
- Children's Cancer Research Institute - UTHSCSA, San Antonio, TX 78229, USA
| | - Jing Feng
- Department of Anesthesiology, Center for the Study of Itch, Washington University School of Medicine in St. Louis, St. Louis, MO 63130, USA
| | - Zhou Lan
- Department of Anesthesiology, Center for the Study of Itch, Washington University School of Medicine in St. Louis, St. Louis, MO 63130, USA
| | - Rong Zhang
- Departments of Medicine, Molecular Microbiology, Pathology, and Immunology and The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Alex W Wessel
- Departments of Medicine, Molecular Microbiology, Pathology, and Immunology and The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Sanjay Dhawan
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
| | - Michael S Diamond
- Departments of Medicine, Molecular Microbiology, Pathology, and Immunology and The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Clark C Chen
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
| | - Robert J Wechsler-Reya
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Fred H Gage
- Laboratory of Genetics, Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Hongzhen Hu
- Department of Anesthesiology, Center for the Study of Itch, Washington University School of Medicine in St. Louis, St. Louis, MO 63130, USA
| | - Jair L Siqueira-Neto
- Department of Cellular and Molecular Medicine, Stem Cell Program, School of Medicine, University of California, San Diego, La Jolla, CA 92307, USA.
| | - Alysson R Muotri
- Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA 92037, USA; Department of Pediatrics, Rady Children's Hospital San Diego, School of Medicine, University of California, San Diego, La Jolla, CA 92307, USA.
| | - David A Cheresh
- Department of Pathology, Moores Cancer Center, University of California, San Diego, La Jolla, CA 92037, USA.
| | - Jeremy N Rich
- Department of Medicine, Division of Regenerative Medicine, University of California School of Medicine, San Diego, La Jolla, CA 92037, USA; Sanford Consortium for Regenerative Medicine, 2880 Torrey Pines Scenic Drive, La Jolla, CA 92037, USA; Department of Neurosciences, University of California, San Diego, School of Medicine, La Jolla, CA 92037, USA.
| |
Collapse
|
35
|
Zimmerman MG, Wrammert J, Suthar MS. Cross-Reactive Antibodies during Zika Virus Infection: Protection, Pathogenesis, and Placental Seeding. Cell Host Microbe 2020; 27:14-24. [PMID: 31917957 PMCID: PMC7802743 DOI: 10.1016/j.chom.2019.12.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Humoral immunity is an essential component of the protective immune response to flavivirus infection. Typically, primary infection generates a robust neutralizing antibody response that mediates viral control and protection. It is becoming increasingly apparent that secondary infection with a closely related flavivirus strain can result in immunological cross-reactivity; however, the consequences to infection outcome remain controversial. Since its introduction to Brazil in 2015, Zika virus (ZIKV) has caused an epidemic of fetal congenital malformations within the Americas. Because ZIKV is a mosquito-borne flavivirus with a high degree of sequence and structural homology to Dengue virus (DENV), the role of immunological cross-reactivity in ZIKV and DENV infections has become a great concern. In this review, we highlight contemporary findings that implicate a role for flavivirus antibodies in mediating protection, contributing to pathogenesis, and seeding the human placenta.
Collapse
Affiliation(s)
- Matthew G Zimmerman
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA
| | - Jens Wrammert
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA
| | - Mehul S Suthar
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Vaccine Center, Yerkes National Primate Research Center, Atlanta, GA 30329, USA.
| |
Collapse
|
36
|
Association between Zika virus and future neurological diseases. J Neurol Sci 2019; 409:116617. [PMID: 31835212 DOI: 10.1016/j.jns.2019.116617] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/22/2019] [Accepted: 12/04/2019] [Indexed: 12/28/2022]
|
37
|
Nem de Oliveira Souza I, Frost PS, França JV, Nascimento-Viana JB, Neris RLS, Freitas L, Pinheiro DJLL, Nogueira CO, Neves G, Chimelli L, De Felice FG, Cavalheiro ÉA, Ferreira ST, Assunção-Miranda I, Figueiredo CP, Da Poian AT, Clarke JR. Acute and chronic neurological consequences of early-life Zika virus infection in mice. Sci Transl Med 2019; 10:10/444/eaar2749. [PMID: 29875203 DOI: 10.1126/scitranslmed.aar2749] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 05/17/2018] [Indexed: 12/13/2022]
Abstract
Although congenital Zika virus (ZIKV) exposure has been associated with microcephaly and other neurodevelopmental disorders, long-term consequences of perinatal infection are largely unknown. We evaluated short- and long-term neuropathological and behavioral consequences of neonatal ZIKV infection in mice. ZIKV showed brain tropism, causing postnatal-onset microcephaly and several behavioral deficits in adulthood. During the acute phase of infection, mice developed frequent seizures, which were reduced by tumor necrosis factor-α (TNF-α) inhibition. During adulthood, ZIKV replication persisted in neonatally infected mice, and the animals showed increased susceptibility to chemically induced seizures, neurodegeneration, and brain calcifications. Altogether, the results show that neonatal ZIKV infection has long-term neuropathological and behavioral complications in mice and suggest that early inhibition of TNF-α-mediated neuroinflammation might be an effective therapeutic strategy to prevent the development of chronic neurological abnormalities.
Collapse
Affiliation(s)
| | - Paula S Frost
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil.,Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Julia V França
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | | | - Rômulo L S Neris
- Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Leandro Freitas
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, SP 04023-062, Brazil
| | - Daniel J L L Pinheiro
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, SP 04023-062, Brazil
| | - Clara O Nogueira
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Gilda Neves
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Leila Chimelli
- Laboratory of Neuropathology, State Institute of Brain Paulo Niemeyer, Rio de Janeiro, Brazil
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil.,Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Ésper A Cavalheiro
- Department of Neurology and Neurosurgery, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, SP 04023-062, Brazil
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil.,Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil
| | - Iranaia Assunção-Miranda
- Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil.
| | - Claudia P Figueiredo
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil.
| | - Andrea T Da Poian
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil.
| | - Julia R Clarke
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21944-590, Brazil.
| |
Collapse
|
38
|
Xu G, Li S, Liu X, Gao P, Chen X, Wang H, Zhang M, Yang Y, Gao GF, Zhang F. PARP-1 mediated cell death is directly activated by ZIKV infection. Virology 2019; 537:254-262. [PMID: 31539774 DOI: 10.1016/j.virol.2019.08.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 08/03/2019] [Accepted: 08/23/2019] [Indexed: 02/06/2023]
Abstract
Zika virus (ZIKV) has emerged as a severe health threat due to its association with microcephaly. It has been reported that the strong cytopathic effects, including cell-cycle arrest and cell death are responsible for the nervous system disease. However, the mechanisms by which ZIKV infection induced cell death were largely unknown. Here, we reported that cell death is readily detected after ZIKV infection as indicated by PI staining and the reduction of cell viability. Importantly, cell death can be induced by overexpression of ZIKV NS3 protein alone but not the other non-structure proteins. Mass spectrometry analysis revealed that NS3 bond to and activated PARP-1. In agreement with these observations, we found that PARP-1 was massively activated during ZIKV infection and the intracellular ATP and NAD+ concentrations rapidly declined. Finally, PARP-1 knockdown simultaneously restrained ZIKV infection-induced cell death and ablated host restriction of virus infection. Our finding indicates that PARP-1 activation is an important cellular event during ZIKV infection, which contributes to the cell death.
Collapse
Affiliation(s)
- Gang Xu
- Guangdong Key Laboratory for Diagnosis and Treatment of Emerging Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, 518112, China.
| | - Shihua Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101, China
| | - Xinyuan Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ping Gao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101, China
| | - Xiaotong Chen
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101, China
| | - Haiyan Wang
- Guangdong Key Laboratory for Diagnosis and Treatment of Emerging Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, 518112, China
| | - Mingxia Zhang
- Guangdong Key Laboratory for Diagnosis and Treatment of Emerging Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, 518112, China
| | - Yang Yang
- Guangdong Key Laboratory for Diagnosis and Treatment of Emerging Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, 518112, China
| | - George Fu Gao
- Guangdong Key Laboratory for Diagnosis and Treatment of Emerging Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, 518112, China; CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101, China
| | - Fuping Zhang
- Guangdong Key Laboratory for Diagnosis and Treatment of Emerging Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, 518112, China; CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, 100101, China.
| |
Collapse
|
39
|
Abstract
This review discusses the current state of the viral metabolism field and gaps in knowledge that will be important for future studies to investigate. We discuss metabolic rewiring caused by viruses, the influence of oncogenic viruses on host cell metabolism, and the use of viruses as guides to identify critical metabolic nodes for cancer anabolism. We also discuss the need for more mechanistic studies identifying viral proteins responsible for metabolic hijacking and for in vivo studies of viral-induced metabolic rewiring. Improved technologies for detailed metabolic measurements and genetic manipulation will lead to important discoveries over the next decade.
Collapse
Affiliation(s)
- Shivani K Thaker
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA
| | - James Ch'ng
- Department of Pediatrics, Division of Hematology/Oncology, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Heather R Christofk
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, 90095, USA.
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, 90095, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA, 90095, USA.
| |
Collapse
|
40
|
Chesnut M, Muñoz LS, Harris G, Freeman D, Gama L, Pardo CA, Pamies D. In vitro and in silico Models to Study Mosquito-Borne Flavivirus Neuropathogenesis, Prevention, and Treatment. Front Cell Infect Microbiol 2019; 9:223. [PMID: 31338335 PMCID: PMC6629778 DOI: 10.3389/fcimb.2019.00223] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/11/2019] [Indexed: 01/07/2023] Open
Abstract
Mosquito-borne flaviviruses can cause disease in the nervous system, resulting in a significant burden of morbidity and mortality. Disease models are necessary to understand neuropathogenesis and identify potential therapeutics and vaccines. Non-human primates have been used extensively but present major challenges. Advances have also been made toward the development of humanized mouse models, but these models still do not fully represent human pathophysiology. Recent developments in stem cell technology and cell culture techniques have allowed the development of more physiologically relevant human cell-based models. In silico modeling has also allowed researchers to identify and predict transmission patterns and discover potential vaccine and therapeutic candidates. This review summarizes the research on in vitro and in silico models used to study three mosquito-borne flaviviruses that cause neurological disease in humans: West Nile, Dengue, and Zika. We also propose a roadmap for 21st century research on mosquito-borne flavivirus neuropathogenesis, prevention, and treatment.
Collapse
Affiliation(s)
- Megan Chesnut
- Center for Alternatives to Animal Testing, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Laura S. Muñoz
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Neuroviruses Emerging in the Americas Study, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Georgina Harris
- Center for Alternatives to Animal Testing, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Dana Freeman
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Lucio Gama
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Carlos A. Pardo
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Neuroviruses Emerging in the Americas Study, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - David Pamies
- Center for Alternatives to Animal Testing, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
41
|
Abstract
In 2015, public awareness of Zika virus (ZIKV) rose in response to alarming statistics of infants with microcephaly being born to women who were infected with the virus during pregnancy, triggering global concern over these potentially devastating consequences. Although we have discovered a great deal about the genome and pathogenesis of this reemergent flavivirus since this recent outbreak, we still have much more to learn, including the nature of the virus-host interactions and mechanisms that determine its tropism and pathogenicity in the nervous system, which are in turn shaped by the continual evolution of the virus. Inevitably, we will find out more about the potential long-term effects of ZIKV exposure on the nervous system from ongoing longitudinal studies. Integrating clinical and epidemiological data with a wider range of animal and human cell culture models will be critical to understanding the pathogenetic mechanisms and developing more specific antiviral compounds and vaccines.
Collapse
Affiliation(s)
- Kimberly M Christian
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
- Department of Developmental and Cell Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Institute for Epigenetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
- Department of Developmental and Cell Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
42
|
Zhou W, Woodson M, Sherman MB, Neelakanta G, Sultana H. Exosomes mediate Zika virus transmission through SMPD3 neutral Sphingomyelinase in cortical neurons. Emerg Microbes Infect 2019; 8:307-326. [PMID: 30866785 PMCID: PMC6455149 DOI: 10.1080/22221751.2019.1578188] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The harmful effects of ZIKA virus (ZIKV) infection are reflected by severe neurological manifestations such as microcephaly in neonates and other complications associated with Guillain-Barré syndrome in adults. The transmission dynamics of ZIKV in or between neurons, or within the developing brains of the foetuses are not fully understood. Using primary cultures of murine cortical neurons, we show that ZIKV uses exosomes as mediators of viral transmission between neurons. Cryo-electron microscopy showed heterogeneous population of neuronal exosomes with a size range of 30–200 nm. Increased production of exosomes from neuronal cells was noted upon ZIKV infection. Neuronal exosomes contained both ZIKV viral RNA and protein(s) that were highly infectious to naïve cells. RNaseA and neutralizing antibodies treatment studies suggest the presence of viral RNA/proteins inside exosomes. Exosomes derived from time- and dose-dependent incubations showed increasing viral loads suggesting higher packaging and delivery of ZIKV RNA and proteins. Furthermore, we noted that ZIKV induced both activity and gene expression of neutral Sphingomyelinase (nSMase)-2/SMPD3, an important molecule that regulates production and release of exosomes. Silencing of SMPD3 in neurons resulted in reduced viral burden and transmission through exosomes. Treatment with SMPD3 specific inhibitor GW4869, significantly reduced ZIKV loads in both cortical neurons and in exosomes derived from these neuronal cells. Taken together, our results suggest that ZIKV modulates SMPD3 activity in cortical neurons for its infection and transmission through exosomes perhaps leading to severe neuronal death that may result in neurological manifestations such as microcephaly in the developing embryonic brains.
Collapse
Affiliation(s)
- Wenshuo Zhou
- a Department of Biological Sciences, Center for Molecular Medicine , Old Dominion University , Norfolk , VA , USA
| | - Michael Woodson
- b Department of Biochemistry and Molecular Biology , University of Texas Medical Branch , Galveston , TX , USA
| | - Michael B Sherman
- b Department of Biochemistry and Molecular Biology , University of Texas Medical Branch , Galveston , TX , USA.,c Sealy Center for Structural Biology and Molecular Biophysics , University of Texas Medical Branch , Galveston , TX , USA
| | - Girish Neelakanta
- a Department of Biological Sciences, Center for Molecular Medicine , Old Dominion University , Norfolk , VA , USA
| | - Hameeda Sultana
- a Department of Biological Sciences, Center for Molecular Medicine , Old Dominion University , Norfolk , VA , USA.,d Department of Medicine, Division of Infectious Diseases and International Health , University of Virginia School of Medicine , Charlottesville , VA , USA
| |
Collapse
|
43
|
Xu D, Li C, Qin CF, Xu Z. Update on the Animal Models and Underlying Mechanisms for ZIKV-Induced Microcephaly. Annu Rev Virol 2019; 6:459-479. [PMID: 31206355 DOI: 10.1146/annurev-virology-092818-015740] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The circulation of Zika virus (ZIKV) in nearly 80 countries and territories poses a significant global threat to public health. ZIKV is causally linked to severe developmental defects in the brain, recognized as congenital Zika syndrome (CZS), which includes microcephaly and other serious congenital neurological complications. Since the World Health Organization declared the ZIKV outbreak a public health emergency of international concern, remarkable progress has been made in the generation of different ZIKV infection animal models to gain insight into cellular targets and pathogenesis and to explore the associated underlying mechanisms. Here we focus on summarizing our current understanding of the effects of ZIKV on mammalian brain development in different developmental stages and discuss the potential underlying mechanisms of ZIKV-induced CZS, as well as future perspectives.
Collapse
Affiliation(s)
- Dan Xu
- College of Biological Science and Engineering, Institute of Life Sciences, Fuzhou University, Fuzhou 350108, China;
| | - Cui Li
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China;
| | - Cheng-Feng Qin
- Department of Virology, State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China;
| | - Zhiheng Xu
- State Key Laboratory of Molecular Developmental Biology, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; .,Parkinson's Disease Center, Beijing Institute for Brain Disorders, Beijing 100069, China
| |
Collapse
|
44
|
ZIKV Strains Differentially Affect Survival of Human Fetal Astrocytes versus Neurons and Traffic of ZIKV-Laden Endocytotic Compartments. Sci Rep 2019; 9:8069. [PMID: 31147629 PMCID: PMC6542792 DOI: 10.1038/s41598-019-44559-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 04/23/2019] [Indexed: 01/05/2023] Open
Abstract
Malformations of the fetal CNS, known as microcephaly, have been linked to Zika virus (ZIKV) infection. Here, the responses of mammalian and mosquito cell lines, in addition to primary human fetal astrocytes and neurons were studied following infection by ZIKV strains Brazil 2016 (ZIKV-BR), French Polynesia 2013 (ZIKV-FP), and Uganda #976 1947 (ZIKV-UG). Viral production, cell viability, infectivity rate, and mobility of endocytotic ZIKV-laden vesicles were compared. All cell types (SK-N-SH, Vero E6, C6/36, human fetal astrocytes and human fetal neurons) released productive virus. Among primary cells, astrocytes were more susceptible to ZIKV infection than neurons, released more progeny virus and tolerated higher virus loads than neurons. In general, the infection rate of ZIKV-UG strain was the highest. All ZIKV strains elicited differences in trafficking of ZIKV-laden endocytotic vesicles in the majority of cell types, including astrocytes and neurons, except in mosquito cells, where ZIKV infection failed to induce cell death. These results represent a thorough screening of cell viability, infection and production of three ZIKV strains in five different cell types and demonstrate that ZIKV affects vesicle mobility in all but mosquito cells.
Collapse
|
45
|
Thaker SK, Chapa T, Garcia G, Gong D, Schmid EW, Arumugaswami V, Sun R, Christofk HR. Differential Metabolic Reprogramming by Zika Virus Promotes Cell Death in Human versus Mosquito Cells. Cell Metab 2019; 29:1206-1216.e4. [PMID: 30827860 PMCID: PMC6818653 DOI: 10.1016/j.cmet.2019.01.024] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 12/04/2018] [Accepted: 01/30/2019] [Indexed: 12/11/2022]
Abstract
Zika virus is a pathogen that poses serious consequences, including congenital microcephaly. Although many viruses reprogram host cell metabolism, whether Zika virus alters cellular metabolism and the functional consequences of Zika-induced metabolic changes remain unknown. Here, we show that Zika virus infection differentially reprograms glucose metabolism in human versus C6/36 mosquito cells by increasing glucose use in the tricarboxylic acid cycle in human cells versus increasing glucose use in the pentose phosphate pathway in mosquito cells. Infection of human cells selectively depletes nucleotide triphosphate levels, leading to elevated AMP/ATP ratios, AMP-activated protein kinase (AMPK) phosphorylation, and caspase-mediated cell death. AMPK is also phosphorylated in Zika virus-infected mouse brain. Inhibiting AMPK in human cells decreases Zika virus-mediated cell death, whereas activating AMPK in mosquito cells promotes Zika virus-mediated cell death. These findings suggest that the differential metabolic reprogramming during Zika virus infection of human versus mosquito cells determines whether cell death occurs.
Collapse
Affiliation(s)
- Shivani K Thaker
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Travis Chapa
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Gustavo Garcia
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Danyang Gong
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ernst W Schmid
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90098, USA
| | - Ren Sun
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; UCLA Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Bioengineering, Samueli School of Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Heather R Christofk
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; UCLA Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
46
|
Affiliation(s)
- Themasap A Khan
- Department of Psychiatry and Behavioral Sciences.,Graduate Program in Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | | |
Collapse
|
47
|
Abstract
BACKGROUND The flaviviridae family comprises single-stranded RNA viruses that enter cells via clathrin-mediated pH-dependent endocytosis. Although the initial events of the virus entry have been already identified, data regarding intracellular virus trafficking and delivery to the replication site are limited. The purpose of this study was to map the transport route of Zika virus and to identify the fusion site within the endosomal compartment. METHODS Tracking of viral particles in the cell was carried out with confocal microscopy. Immunostaining of two structural proteins of Zika virus enabled precise mapping of the route of the ribonucleocapsid and the envelope and, consequently, mapping the fusion site in the endosomal compartment. The results were verified using RNAi silencing and chemical inhibitors. RESULTS After endocytic internalization, Zika virus is trafficked through the endosomal compartment to fuse in late endosomes. Inhibition of endosome acidification using bafilomycin A1 hampers the infection, as the fusion is inhibited; instead, the virus is transported to late compartments where it undergoes proteolytic degradation. The degradation products are ejected from the cell via slow recycling vesicles. Surprisingly, NH4Cl, which is also believed to block endosome acidification, shows a very different mode of action. In the presence of this basic compound, the endocytic hub is reprogrammed. Zika virus-containing vesicles never reach the late stage, but are rapidly trafficked to the plasma membrane via a fast recycling pathway after the clathrin-mediated endocytosis. Further, we also noted that, similarly as other members of the flaviviridae family, Zika virus undergoes furin- or furin-like-dependent activation during late steps of infection, while serine or cysteine proteases are not required for Zika virus maturation or entry. CONCLUSIONS Zika virus fusion occurs in late endosomes and is pH-dependent. These results broaden our understanding of Zika virus intracellular trafficking and may in future allow for development of novel treatment strategies. Further, we identified a novel mode of action for agents commonly used in studies of virus entry. Schematic representation of differences in ZIKV trafficking in the presence of Baf A1 and NH4Cl.
Collapse
|
48
|
Muñoz LS, Parra B, Pardo CA. Neurological Implications of Zika Virus Infection in Adults. J Infect Dis 2019; 216:S897-S905. [PMID: 29267923 DOI: 10.1093/infdis/jix511] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The 2015-2016 epidemic of Zika virus (ZIKV) in the Americas and the Caribbean was associated with an unprecedented burden of neurological disease among adults. Clinically, Guillain-Barre syndrome (GBS) predominated among regions affected by the ZIKV epidemic, but the spectrum of neurological disease in the adults appears broader as cases of encephalopathy, encephalitis, meningitis, myelitis, and seizures have also been reported. A para-infectious temporal profile of ZIKV-associated GBS (ZIKV-GBS) has been described in clinical studies, which may suggest a direct viral neuropathic effect. However, ZIKV neuropathogenesis has not yet been fully understood. Mechanisms for ZIKV-GBS and other neurological syndromes have been hypothesized, such as adaptive viral genetic changes, immunological interactions with other circulating flaviviruses, and host and factors. This review summarizes the current evidence on ZIKV-associated neurological complications in the adults.
Collapse
Affiliation(s)
- Laura S Muñoz
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Beatriz Parra
- Department of Microbiology, Universidad del Valle School of Medicine, Cali, Colombia
| | - Carlos A Pardo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | |
Collapse
|
49
|
Kim J, Koo BK, Yoon KJ. Modeling Host-Virus Interactions in Viral Infectious Diseases Using Stem-Cell-Derived Systems and CRISPR/Cas9 Technology. Viruses 2019; 11:v11020124. [PMID: 30704043 PMCID: PMC6409779 DOI: 10.3390/v11020124] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/14/2019] [Accepted: 01/23/2019] [Indexed: 02/06/2023] Open
Abstract
Pathologies induced by viral infections have undergone extensive study, with traditional model systems such as two-dimensional (2D) cell cultures and in vivo mouse models contributing greatly to our understanding of host-virus interactions. However, the technical limitations inherent in these systems have constrained efforts to more fully understand such interactions, leading to a search for alternative in vitro systems that accurately recreate in vivo physiology in order to advance the study of viral pathogenesis. Over the last decade, there have been significant technological advances that have allowed researchers to more accurately model the host environment when modeling viral pathogenesis in vitro, including induced pluripotent stem cells (iPSCs), adult stem-cell-derived organoid culture systems and CRISPR/Cas9-mediated genome editing. Such technological breakthroughs have ushered in a new era in the field of viral pathogenesis, where previously challenging questions have begun to be tackled. These include genome-wide analysis of host-virus crosstalk, identification of host factors critical for viral pathogenesis, and the study of viral pathogens that previously lacked a suitable platform, e.g., noroviruses, rotaviruses, enteroviruses, adenoviruses, and Zika virus. In this review, we will discuss recent advances in the study of viral pathogenesis and host-virus crosstalk arising from the use of iPSC, organoid, and CRISPR/Cas9 technologies.
Collapse
Affiliation(s)
- Jihoon Kim
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria.
| | - Bon-Kyoung Koo
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Dr. Bohr-Gasse 3, 1030 Vienna, Austria.
| | - Ki-Jun Yoon
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea.
| |
Collapse
|
50
|
Zika virus induces abnormal cranial osteogenesis by negatively affecting cranial neural crest development. INFECTION GENETICS AND EVOLUTION 2019; 69:176-189. [PMID: 30665021 DOI: 10.1016/j.meegid.2019.01.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/17/2019] [Accepted: 01/18/2019] [Indexed: 12/20/2022]
Abstract
Zika virus (ZIKV) infection during gestation is deemed to be coupled to birth defects through direct impairment of the nervous system during neurogenesis. However, in this study, our data showed that ZIKV infection dramatically suppressed cranial osteogenesis, shown by Safranin O/Fast Green and alizarin red staining, in chick embryos, which provides another possibility that craniofacial bone malformation caused by ZIKV may be a major cause of ZIKV-mediated birth defects. By immunofluorescent staining and electron microcopy, we confirmed ZIKV infection in chick embryo neural tubes and sites of neural crest. Next, in vivo (chick embryos) and in vitro [primary culture of neural crest cells (NCC)] ZIKV and HNK-1 double immunofluorescent staining demonstrated that ZIKV infection inhibited the production of migratory NCC. The reduction of both AP-2α- and Pax7-positive NCC in HH10 chick embryos infected by ZIKV confirmed that abnormal development of cranial NCC also occurred in the migratory process. Whole mount in situ hybridization demonstrated that cadherin 6B expression was elevated and Slug, FoxD3, and BMP4/Msx1 expressions decreased in ZIKV-infected HH10 chick embryos, implying that epithelial-mesenchymal transition (EMT) of neural crest production was blocked by ZIKV infection. Moreover, in vivo and in vitro pHIS3 and Pax7 double immunofluorescent staining showed that NCC proliferation was repressed by ZIKV infection. C-caspase-3 and AP-2α double immunofluorescent staining in HH10 chick embryos and western blotting showed that NCC apoptosis increased following ZIKV infection. Finally, electron microscopy showed multiple autophagosomes in ZIKV-infected embryos, and western blot and LC3B immunofluorescent staining demonstrated that autophagy-related genes were activated by ZIKV infection. Taken together, our data first showed that ZIKV infection during embryogenesis could interfere with cranial neural crest development, which in turn causes aberrant cranial osteogenesis. Our results provided new insights into brain malformations induced by ZIKV infection.
Collapse
|