1
|
Colognese BA, Argollo N. Zika virus infection and acute transverse myelitis: a comprehensive systematic review. Rev Inst Med Trop Sao Paulo 2024; 66:e66. [PMID: 39699422 DOI: 10.1590/s1678-9946202466066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 10/01/2024] [Indexed: 12/20/2024] Open
Abstract
The Zika virus (ZIKV) has been associated with several complications, including acute transverse myelitis (ATM), an acute inflammation of the spinal cord, with rapid development of motor, sensory and dysautonomic symptoms. It is a rare disease, and its clinical features, as well as differences in relation to idiopathic ATMs, are still not completely known. The objective of this paper is to review the literature in search of clinical features and complementary exams of ATM post-ZIKV infection, alone or in association with other neurological conditions (mixed diseases), as well as its treatments and prognoses. The search was made on 5 databases, using the PRISMA methodology (Preferred Reporting Items for Systematic Reviews and Meta-Analyses). Nine articles were selected (total of 20 subjects), which were divided between isolated ATM and mixed neurological syndromes with ATM. The study found a predominance of individuals aged 20 to 30. Among the six subjects in the mixed group, three were over 50 years old. The median prodromal period was 2 days for the mixed diseases group and 7 days for the isolated ATM group. Some individuals in the isolated ATM group exhibited signs of dysautonomia, such as syncope, postural lability, and arrhythmia. The mixed group had a higher incidence of coinfections, with 4 cases compared to 1 case in the isolated ATM group. Over 50% of the individuals had moderate to moderately severe disability. These findings suggest that severe conditions may progress to significant sequelae, highlighting the need for prompt diagnosis and treatment, particularly during endemic periods.
Collapse
Affiliation(s)
| | - Nayara Argollo
- Universidade Federal da Bahia, Hospital Universitário Professor Edgard Santos, Salvador, Bahia, Brazil
| |
Collapse
|
2
|
Pastor AF, Mahaney SM, Garcia J, Morales M, Quintanilla O, Arriaga MA, Thomas JM, VandeBerg JL. The Laboratory Opossum ( Monodelphis domestica) Is a Unique Model for Research on Zika Virus: Robust Immune Response, Widespread Dissemination, and Long-Term Persistence. Viruses 2024; 16:1847. [PMID: 39772157 PMCID: PMC11680235 DOI: 10.3390/v16121847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
The Zika virus (ZIKV) epidemic elicited a rapid commitment to the development of animal models for ZIKV research. Non-human primates (NHPs) and mice have made significant contributions to this research, but NHPs are expensive, have a long gestation period, and are available only in small numbers; non-genetically modified mice are resistant to infection. To address these deficiencies, we have established the laboratory opossum, Monodelphis domestica, as a small animal model that complements the mouse and monkey models. We developed and validated an indirect ELISA for measuring antibodies to ZIKV in opossums, as well as an immunohistochemistry (IHC) method to detect ZIKV NS1 protein in tissue samples. Opossum pups inoculated intracerebrally as embryos, juveniles inoculated by several routes, and mothers that cannibalized inoculated pups became persistently infected with ZIKV. The virus spread to multiple organs and persisted for up to 38 weeks (the latest endpoint of the experiments). A robust humoral immune response was mounted, and high titers of antibodies also persisted for 38 weeks. The results establish M. domestica as a natural, non-genetically modified animal model in which ZIKV persists long-term after experimental exposure and as a unique animal model for research on the immune response to ZIKV.
Collapse
Affiliation(s)
- André Filipe Pastor
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA; (S.M.M.); (M.A.A.); (J.M.T.III)
- Center for Vector-Borne Disease, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA
- Sertão Pernambucano Federal Institute of Education, Science, and Technology, Floresta 56400000, Pernambuco, Brazil
| | - Susan M. Mahaney
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA; (S.M.M.); (M.A.A.); (J.M.T.III)
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA
| | - Juan Garcia
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA (O.Q.)
| | - Marisol Morales
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA (O.Q.)
| | - Oscar Quintanilla
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA (O.Q.)
| | - Marco A. Arriaga
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA; (S.M.M.); (M.A.A.); (J.M.T.III)
| | - John M. Thomas
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA; (S.M.M.); (M.A.A.); (J.M.T.III)
- Center for Vector-Borne Disease, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA (O.Q.)
| | - John L. VandeBerg
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA; (S.M.M.); (M.A.A.); (J.M.T.III)
- Center for Vector-Borne Disease, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Edinburg/Harlingen/Brownsville, McAllen, TX 78520, USA
| |
Collapse
|
3
|
Wang S, Li W, Wang Z, Yang W, Li E, Xia X, Yan F, Chiu S. Emerging and reemerging infectious diseases: global trends and new strategies for their prevention and control. Signal Transduct Target Ther 2024; 9:223. [PMID: 39256346 PMCID: PMC11412324 DOI: 10.1038/s41392-024-01917-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/13/2024] [Accepted: 07/05/2024] [Indexed: 09/12/2024] Open
Abstract
To adequately prepare for potential hazards caused by emerging and reemerging infectious diseases, the WHO has issued a list of high-priority pathogens that are likely to cause future outbreaks and for which research and development (R&D) efforts are dedicated, known as paramount R&D blueprints. Within R&D efforts, the goal is to obtain effective prophylactic and therapeutic approaches, which depends on a comprehensive knowledge of the etiology, epidemiology, and pathogenesis of these diseases. In this process, the accessibility of animal models is a priority bottleneck because it plays a key role in bridging the gap between in-depth understanding and control efforts for infectious diseases. Here, we reviewed preclinical animal models for high priority disease in terms of their ability to simulate human infections, including both natural susceptibility models, artificially engineered models, and surrogate models. In addition, we have thoroughly reviewed the current landscape of vaccines, antibodies, and small molecule drugs, particularly hopeful candidates in the advanced stages of these infectious diseases. More importantly, focusing on global trends and novel technologies, several aspects of the prevention and control of infectious disease were discussed in detail, including but not limited to gaps in currently available animal models and medical responses, better immune correlates of protection established in animal models and humans, further understanding of disease mechanisms, and the role of artificial intelligence in guiding or supplementing the development of animal models, vaccines, and drugs. Overall, this review described pioneering approaches and sophisticated techniques involved in the study of the epidemiology, pathogenesis, prevention, and clinical theatment of WHO high-priority pathogens and proposed potential directions. Technological advances in these aspects would consolidate the line of defense, thus ensuring a timely response to WHO high priority pathogens.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Wujian Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Zhenshan Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin, China
| | - Wanying Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Entao Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China.
| | - Sandra Chiu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China.
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China.
- Department of Laboratory Medicine, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
4
|
Ai X, Yu H, Cai Y, Guan Y. Interactions Between Extracellular Vesicles and Autophagy in Neuroimmune Disorders. Neurosci Bull 2024; 40:992-1006. [PMID: 38421513 PMCID: PMC11251008 DOI: 10.1007/s12264-024-01183-5] [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: 05/29/2023] [Accepted: 11/15/2023] [Indexed: 03/02/2024] Open
Abstract
Neuroimmune disorders, such as multiple sclerosis, neuromyelitis optica spectrum disorder, myasthenia gravis, and Guillain-Barré syndrome, are characterized by the dysfunction of both the immune system and the nervous system. Increasing evidence suggests that extracellular vesicles and autophagy are closely associated with the pathogenesis of these disorders. In this review, we summarize the current understanding of the interactions between extracellular vesicles and autophagy in neuroimmune disorders and discuss their potential diagnostic and therapeutic applications. Here we highlight the need for further research to fully understand the mechanisms underlying these disorders, and to develop new diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Xiwen Ai
- Department of Neurology, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, 200127, China
| | - Haojun Yu
- Department of Neurology, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, 200127, China
| | - Yu Cai
- Department of Neurology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA.
| | - Yangtai Guan
- Department of Neurology, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, 200127, China.
| |
Collapse
|
5
|
Rengifo AC, Rivera J, Álvarez-Díaz DA, Naizaque J, Santamaria G, Corchuelo S, Gómez CY, Torres-Fernández O. Morphological and Molecular Changes in the Cortex and Cerebellum of Immunocompetent Mice Infected with Zika Virus. Viruses 2023; 15:1632. [PMID: 37631975 PMCID: PMC10458311 DOI: 10.3390/v15081632] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
Zika virus (ZIKV) disease continues to be a threat to public health, and it is estimated that millions of people have been infected and that there have been more cases of serious complications than those already reported. Despite many studies on the pathogenesis of ZIKV, several of the genes involved in the malformations associated with viral infection are still unknown. In this work, the morphological and molecular changes in the cortex and cerebellum of mice infected with ZIKV were evaluated. Neonatal BALB/c mice were inoculated with ZIKV intraperitoneally, and the respective controls were inoculated with a solution devoid of the virus. At day 10 postinoculation, the mice were euthanized to measure the expression of the markers involved in cortical and cerebellar neurodevelopment. The infected mice presented morphological changes accompanied by calcifications, as well as a decrease in most of the markers evaluated in the cortex and cerebellum. The modifications found could be predictive of astrocytosis, dendritic pathology, alterations in the regulation systems of neuronal excitation and inhibition, and premature maturation, conditions previously described in other models of ZIKV infection and microcephaly.
Collapse
Affiliation(s)
- Aura Caterine Rengifo
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| | - Jorge Rivera
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| | - Diego Alejandro Álvarez-Díaz
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
- Genómica de Microorganismos Emergentes, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia
| | - Julián Naizaque
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| | - Gerardo Santamaria
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| | - Sheryll Corchuelo
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| | - Claudia Yadira Gómez
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| | - Orlando Torres-Fernández
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| |
Collapse
|
6
|
Schmitt K, Curlin JZ, Remling-Mulder L, Aboellail T, Akkina R. Zika virus induced microcephaly and aberrant hematopoietic cell differentiation modeled in novel neonatal humanized mice. Front Immunol 2023; 14:1060959. [PMID: 36825016 PMCID: PMC9941325 DOI: 10.3389/fimmu.2023.1060959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 01/26/2023] [Indexed: 02/10/2023] Open
Abstract
Introduction Immunocompetent and immunocompromised murine models have been instrumental in answering important questions regarding ZIKV pathogenesis and vertical transmission. However, mimicking human congenital zika syndrome (CZS) characteristics in these murine models has been less than optimal and does not address the potential viral effects on the human immune system. Methods Here, we utilized neonatal humanized Rag2-/-γc-/- mice to model CZS and evaluate the potential viral effects on the differentiation of human hematopoietic stem cells in vivo. Newborn Rag2-/-γc-/- mice were engrafted with ZIKV-infected hematopoietic stem cells (HSC) and monitored for symptoms and lesions. Results Within 13 days, mice displayed outward clinical symptoms that encompassed stunted growth, hunched posture, ruffled fur, and ocular defects. Striking gross pathologies in the brain and visceral organs were noted. Our results also confirmed that ZIKV actively infected human CD34+ hematopoietic stem cells and restricted the development of terminally differentiated B cells. Histologically, there was multifocal mineralization in several different regions of the brain together with ZIKV antigen co-localization. Diffuse necrosis of pyramidal neurons was seen with collapse of the hippocampal formation. Discussion Overall, this model recapitulated ZIKV microcephaly and CZS together with viral adverse effects on the human immune cell ontogeny thus providing a unique in vivo model to assess the efficacy of novel therapeutics and immune interventions.
Collapse
|
7
|
Hageman G, Nihom J. Fetuses and infants with Amyoplasia congenita in congenital Zika syndrome: The evidence of a viral cause. A narrative review of 144 cases. Eur J Paediatr Neurol 2023; 42:1-14. [PMID: 36442412 DOI: 10.1016/j.ejpn.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 10/09/2022] [Accepted: 11/01/2022] [Indexed: 11/11/2022]
Abstract
OBJECTIVES Amyoplasia congenita is the most frequent type of arthrogryposis causing fetal hypokinesia, leading to congenital contractures at birth. The pathogenesis is thought to be impaired blood circulation to the fetus early in pregnancy, with hypotension and hypoxia damaging the anterior horn cells. In animal studies however a prenatal infection with a poliomyelitis-like viral agent was demonstrated. Congenital Zika virus syndrome (CZVS) has recently been described in infants with severe microcephaly, and in 10-25% of cases arthrogryposis. METHODS A search in PubMed for CZVS yielded 124 studies. After a selection for arthrogryposis, 35 papers were included, describing 144 cases. The studies were divided into two categories. 1) Those (87 cases) focussing on imaging or histological data of congenital brain defects, contained insufficient information to link arthrogryposis specifically to lesions of the brain or spinal motor neuron. 2) In the other 57 cases detailed clinical data could be linked to neurophysiological, imaging or histological data. RESULTS In category 1 the most frequent brain abnormalities in imaging studies were ventriculomegaly, calcifications (subcortical, basal ganglia, cerebellum), hypoplasia of the brainstem and cerebellum, atrophy of the cerebral cortex, migration disorders and corpus callosum anomalies. In category 2, in 38 of 57 cases clinical data were indicative of Amyoplasia congenita. This diagnosis was confirmed by electromyographic findings (13 cases), by MRI (37 cases) or histology (12 cases) of the spinal cord. The latter showed small or absent lateral corticospinal tracts, and cell loss and degeneration of motor neuron cells. Zika virus-proteins and flavivirus-like particles were detected in cytoplasm of spinal neurons. CONCLUSION The phenotype of arthrogryposis in CZVS is consistent with Amyoplasia congenita. These findings warrant search for an intrauterine infection with any neurotropic viral agent with affinity to spinal motor neurons in neonates with Amyoplasia.
Collapse
Affiliation(s)
- G Hageman
- Department of Neurology, Medical Spectrum Twente, Hospital Enschede, the Netherlands.
| | - J Nihom
- Department of Neurology, Medical Spectrum Twente, Hospital Enschede, the Netherlands
| |
Collapse
|
8
|
Li S, Armstrong N, Zhao H, Cruz-cosme R, Yang H, Zhong C, Fu W, Wang W, Yang D, Xia N, Cheng T, Tang Q. Zika Virus Infection Downregulates Connexin 43, Disrupts the Cardiomyocyte Gap Junctions and Induces Heart Diseases in A129 Mice. J Virol 2022; 96:e0137322. [PMID: 36226984 PMCID: PMC9645212 DOI: 10.1128/jvi.01373-22] [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: 09/04/2022] [Accepted: 09/21/2022] [Indexed: 11/20/2022] Open
Abstract
Zika virus (ZIKV) is transmitted mostly via mosquito bites and no vaccine is available, so it may reemerge. We and others previously demonstrated that neonatal infection of ZIKV results in heart failure and can be fatal. Animal models implicated ZIKV involvement in viral heart diseases. It is unknown whether and how ZIKV causes heart failure in adults. Herein, we studied the effects of ZIKV infection on the heart function of adult A129 mice. First, we found that ZIKV productively infects the rat-, mouse-, or human-originated heart cell lines and caused ubiquitination-mediated degradation of and distortive effects on connexin 43 (Cx43) protein that is important for communications between cardiomyocytes. Second, ZIKV infection caused 100% death of the A129 mice with decreasing body weight, worsening health score, shrugging fur, and paralysis. The viral replication was detected in multiple organs. In searching for the viral effects on heart of the A129 mice, we found that ZIKV infection resulted in the increase of cardiac muscle enzymes, implicating a viral acute myocardial injury. ZIKV-caused heart injury was also demonstrated by electrocardiogram (ECG) showing widened and fragmented QRS waves, prolonged PR interval, and slower heart rate. The intercalated disc (ICD) between two cardiomyocytes was destroyed, as shown by the electronic microscopy, and the Cx43 distribution in the ICDs was less organized in the ZIKV-infected mice compared to that in the phosphate-buffered saline (PBS)-treated mice. Consistently, ZIKV productively infected the heart of A129 mice and decreased Cx43 protein. Therefore, we demonstrated that ZIKV infection caused heart failure, which might lead to fatal sequelae in ZIKV-infected A129 mice. IMPORTANCE Zika virus (ZIKV) is a teratogen causing devastating sequelae to the newborns who suffer a congenital ZIKV infection while it brings about only mild symptoms to the health-competent older children or adults. Mouse models have played an important role in mechanistic and pathogenic studies of ZIKV. In this study, we employed 3 to 4 week-old A129 mice for ZIKV infection. RT-qPCR assays discovered that ZIKV replicated in multiple organs, including the heart. As a result of ZIKV infection, the A129 mice experienced weight loss, health score worsening, paralysis, and deaths. We revealed that the ZIKV infection caused abnormal electrocardiogram presentations, increased cardiac muscle enzymes, downregulated Cx43, and destroyed the gap junction and the intercalated disc between the cardiomyocytes, implicating that ZIKV may cause an acute myocardial injury in A129 mice. Therefore, our data imply that ZIKV infection may jeopardize the immunocompromised population with a severe clinical consequence, such as heart defect.
Collapse
Affiliation(s)
- Shuxuan Li
- School of Medicine, Henan University of Chinese Medicine, Zhengzhou, P.R. China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, P.R. China
| | - Najealicka Armstrong
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| | - Huan Zhao
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, P.R. China
| | - Ruth Cruz-cosme
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| | - Hongwei Yang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, P.R. China
| | - Chunlian Zhong
- School of Material and Chemical Engineering, Minjiang University, Fuzhou, P.R. China
| | - Wenkun Fu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, P.R. China
| | - Wei Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, P.R. China
| | - Decheng Yang
- Centre for Heart Lung Innovation - St. Paul’s Hospital, Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, P.R. China
| | - Tong Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, P.R. China
| | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| |
Collapse
|
9
|
Marshall EM, Koopmans MPG, Rockx B. A Journey to the Central Nervous System: Routes of Flaviviral Neuroinvasion in Human Disease. Viruses 2022; 14:2096. [PMID: 36298652 PMCID: PMC9611789 DOI: 10.3390/v14102096] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 11/17/2022] Open
Abstract
Many arboviruses, including viruses of the Flavivirus genera, are known to cause severe neurological disease in humans, often with long-lasting, debilitating sequalae in surviving patients. These emerging pathogens impact millions of people worldwide, yet still relatively little is known about the exact mechanisms by which they gain access to the human central nervous system. This review focusses on potential haematogenous and transneural routes of neuroinvasion employed by flaviviruses and identifies numerous gaps in knowledge, especially regarding lesser-studied interfaces of possible invasion such as the blood-cerebrospinal fluid barrier, and novel routes such as the gut-brain axis. The complex balance of pro-inflammatory and antiviral immune responses to viral neuroinvasion and pathology is also discussed, especially in the context of the hypothesised Trojan horse mechanism of neuroinvasion. A greater understanding of the routes and mechanisms of arboviral neuroinvasion, and how they differ between viruses, will aid in predictive assessments of the neuroinvasive potential of new and emerging arboviruses, and may provide opportunity for attenuation, development of novel intervention strategies and rational vaccine design for highly neurovirulent arboviruses.
Collapse
Affiliation(s)
| | | | - Barry Rockx
- Department of Viroscience, Erasmus University Medical Center, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
10
|
Microglia activate early anti-viral responses upon HSV-1 entry into the brain to counteract development of encephalitis-like disease in mice. J Virol 2022; 96:e0131121. [PMID: 35045263 DOI: 10.1128/jvi.01311-21] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Spread of herpes simplex virus 1 (HSV1) from the periphery to the central nervous system (CNS) can lead to extensive infection and pathological inflammation in the brain, causing herpes simplex encephalitis (HSE). It has been shown that microglia, the CNS-resident macrophages, are involved in early sensing of HSV1 and an induction of antiviral responses. In addition, infiltration of peripheral immune cells may contribute to control of viral infection. In this study, we tested the effect of microglia depletion in a mouse model of HSE. Increased viral titers and increased disease severity were observed in microglia-depleted mice. The effect of microglia depletion was more pronounced in wild-type than in cGas-/- mice, revealing that this immune sensor contributes to the antiviral activity of microglia. Importantly, microglia depletion led to reduced production of type I interferon (IFN), pro-inflammatory cytokines and chemokines at early time points after viral entry into the CNS. In line with this, in vitro experiments on murine primary CNS cells demonstrated microglial presence to be essential for IFN RNA induction, and control of HSV1 replication. However, the effect of microglia depletion on expression of IFNs, and inflammatory cytokines was restricted to early time point of HSV1 entry into the CNS. There was no major alteration of infiltration of CD45-positive cells in microglia-depleted mice. Collectively, our data demonstrate a key role for microglia in controlling HSV1 replication early after viral entry into the CNS and highlight the importance of a prompt antiviral innate response to reduce the risk of HSE development. Importance One of the most devastating and acute neurological conditions is encephalitis, i.e. inflammation of brain tissue. Herpes simplex virus 1 (HSV1) is a highly prevalent pathogen in humans, and the most frequent cause of viral sporadic encephalitis, called herpes simplex encephalitis (HSE). HSV1 has the ability to infect peripheral neurons and reach the central nervous system (CNS) of humans, where it can be detected by brain resident cells and infiltrating immune cells, leading to protective and damaging immune responses. In this study, we investigated the effects of a depletion of microglia, the main brain-resident immune cell type. For this purpose, we used a mouse model of HSE. We found that viral levels increased and disease symptoms worsened in microglia-depleted mice. In addition, mice lacking a major sensor of viral DNA, cGAS, manifested more pronounced disease than wild-type mice, highlighting the importance of this immune sensor in the activity of microglia. Evidently, microglia depletion led to a reduced production of many known antiviral factors, most notably type I interferon (IFN). The importance of microglia in the early control of HSV1 spread and the generation of antiviral responses is further demonstrated by experiments on murine mixed glial cell cultures. Interestingly, mice with microglia depletion exhibited an unaltered activation of antiviral responses and recruitment of immune cells from the periphery at later time points of infection, but this did not prevent the development of the disease. Overall, the data highlight the importance of a rapid activation of the host defense, with microglia playing a critical role in controlling HSV1 infection, which eventually prevents damage to neurons and brain tissue.
Collapse
|
11
|
Vue D, Tang Q. Zika Virus Overview: Transmission, Origin, Pathogenesis, Animal Model and Diagnosis. ZOONOSES (BURLINGTON, MASS.) 2021; 1:10.15212/zoonoses-2021-0017. [PMID: 34957474 PMCID: PMC8698461 DOI: 10.15212/zoonoses-2021-0017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Zika virus (ZIKV) was first discovered in 1947 in Uganda. ZIKV did not entice much attention until Brazil hosted the 2016 Summer Olympics Game, where ZIKV attracted a global audience. ZIKV is a flavivirus that can be transmitted chiefly through the biting of the mosquito or sexually or by breastfeeding at a lower scale. As time passed, the recent discovery of how the ZIKV causes congenital neurodevelopmental defects, including microcephaly, makes us reevaluate the importance of ZIKV interaction with centrosome organization because centrosome plays an important role in cell division. When the ZIKV disrupts centrosome organization and mitotic abnormalities, this will alter neural progenitor differentiation. Altering the neural progenitor differentiation will lead to cell cycle arrest, increase apoptosis, and inhibit the neural progenitor cell differentiation, as this can lead to abnormalities in neural cell development resulting in microcephaly. Understanding the importance of ZIKV infection throughout the years, this review article gives an overview of the history, transmission routes, pathogenesis, animal models, and diagnosis.
Collapse
Affiliation(s)
- Dallas Vue
- Department of Microbiology, Howard University College of Medicine, 520 W Street NW Washington, DC 20059, USA
| | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, 520 W Street NW Washington, DC 20059, USA
| |
Collapse
|
12
|
Ou TP, Auerswald H, In S, Peng B, Pang S, Boyer S, Choeung R, Dupont-Rouzeyrol M, Dussart P, Duong V. Replication Variance of African and Asian Lineage Zika Virus Strains in Different Cell Lines, Mosquitoes and Mice. Microorganisms 2021; 9:microorganisms9061250. [PMID: 34207488 PMCID: PMC8230095 DOI: 10.3390/microorganisms9061250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/23/2021] [Accepted: 05/25/2021] [Indexed: 11/20/2022] Open
Abstract
Since the epidemic in 2007, studies on vector competence for Zika virus (ZIKV) have intensified, showing that the transmission efficiency varies depending on the vector population, ZIKV strain, and dose of the infectious blood meal. In this study, we aimed to investigate the replication of African and Asian ZIKV strains in vitro and in vivo in order to reveal their phenotypic differences. In addition, we investigated the vector competence of Cambodian Aedes aegypti (Ae. aegypti) mosquitoes (urban and rural) for these ZIKV strains. We observed a significantly higher pathogenicity of the African ZIKV strain in vitro (in mosquito and mammalian cells), and in vivo in both Ae. aegypti and mice. Both mosquito populations were competent to transmit ZIKV as early as 7 days p.i., depending on the population and the ZIKV strain. Ae. aegypti from rural habitats showed significant higher transmission and survival rates than those from urban. We observed the highest transmission efficiency for the African ZIKV isolate (93.3% 14 days p.i.) and for the Cambodian ZIKV isolate (80% 14 days p.i.). Overall, our results highlight the phenotypic differences of the ZIKV lineages and the potential risk of ZIKV transmission by Ae. aegypti mosquitoes. Further investigations of Cambodian mosquito species and ZIKV specific surveillance in humans is necessary in order to improve the local risk assessment.
Collapse
Affiliation(s)
- Tey Putita Ou
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh 12156, Cambodia; (T.P.O.); (H.A.); (S.I.); (B.P.); (S.P.); (R.C.); (P.D.)
| | - Heidi Auerswald
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh 12156, Cambodia; (T.P.O.); (H.A.); (S.I.); (B.P.); (S.P.); (R.C.); (P.D.)
| | - Saraden In
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh 12156, Cambodia; (T.P.O.); (H.A.); (S.I.); (B.P.); (S.P.); (R.C.); (P.D.)
| | - Borin Peng
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh 12156, Cambodia; (T.P.O.); (H.A.); (S.I.); (B.P.); (S.P.); (R.C.); (P.D.)
| | - Senglong Pang
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh 12156, Cambodia; (T.P.O.); (H.A.); (S.I.); (B.P.); (S.P.); (R.C.); (P.D.)
| | - Sébastien Boyer
- Medical Entomology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh 12156, Cambodia;
| | - Rithy Choeung
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh 12156, Cambodia; (T.P.O.); (H.A.); (S.I.); (B.P.); (S.P.); (R.C.); (P.D.)
| | - Myrielle Dupont-Rouzeyrol
- URE Dengue and Arboviruses, Institut Pasteur in New Caledonia, Institut Pasteur International Network, Nouméa 98800, New Caledonia;
| | - Philippe Dussart
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh 12156, Cambodia; (T.P.O.); (H.A.); (S.I.); (B.P.); (S.P.); (R.C.); (P.D.)
| | - Veasna Duong
- Virology Unit, Institut Pasteur du Cambodge, Institut Pasteur International Network, Phnom Penh 12156, Cambodia; (T.P.O.); (H.A.); (S.I.); (B.P.); (S.P.); (R.C.); (P.D.)
- Correspondence:
| |
Collapse
|
13
|
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
|
14
|
Mwaliko C, Nyaruaba R, Zhao L, Atoni E, Karungu S, Mwau M, Lavillette D, Xia H, Yuan Z. Zika virus pathogenesis and current therapeutic advances. Pathog Glob Health 2021; 115:21-39. [PMID: 33191867 PMCID: PMC7850325 DOI: 10.1080/20477724.2020.1845005] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Zika virus (ZIKV) is an emerging arthropod-borne flavivirus that, upon infection, results in teratogenic effects and neurological disorders. ZIKV infections pose serious global public health concerns, prompting scientists to increase research on antivirals and vaccines against the virus. These efforts are still ongoing as the pathogenesis and immune evasion mechanisms of ZIKV have not yet been fully elaborated. Currently, no specific vaccines or drugs have been approved for ZIKV; however, some are undergoing clinical trials. Notably, several strategies have been used to develop antivirals, including drugs that target viral and host proteins. Additionally, drug repurposing is preferred since it is less costly and takes less time than other strategies because the drugs used have already been approved for human use. Likewise, different platforms have been evaluated for the design of vaccines, including DNA, mRNA, peptide, protein, viral vectors, virus-like particles (VLPSs), inactivated-virus, and live-attenuated virus vaccines. These vaccines have been shown to induce specific humoral and cellular immune responses and reduce viremia and viral RNA both in vitro and in vivo. Importantly, most of these vaccines have entered clinical trials. Understanding the viral disease mechanism will provide better strategies for developing therapeutic agents against ZIKV. This review provides a comprehensive summary of the viral pathogenesis of ZIKV and current advancements in the development of vaccines and drugs against this virus.
Collapse
Affiliation(s)
- Caroline Mwaliko
- Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China,International College, University of Chinese Academy of Sciences, Beijing, China,Microbiology, Sino-Africa Joint Research Center, Nairobi, Kenya
| | - Raphael Nyaruaba
- Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China,International College, University of Chinese Academy of Sciences, Beijing, China,Microbiology, Sino-Africa Joint Research Center, Nairobi, Kenya
| | - Lu Zhao
- Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China,International College, University of Chinese Academy of Sciences, Beijing, China
| | - Evans Atoni
- Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China,International College, University of Chinese Academy of Sciences, Beijing, China,Microbiology, Sino-Africa Joint Research Center, Nairobi, Kenya
| | - Samuel Karungu
- Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China,International College, University of Chinese Academy of Sciences, Beijing, China,Microbiology, Sino-Africa Joint Research Center, Nairobi, Kenya
| | - Matilu Mwau
- Center for Infectious and Parasitic Diseases Control Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Dimitri Lavillette
- CAS Key Laboratory of Molecular Virology & Immunology, Institut Pasteur of Shanghai, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Han Xia
- Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China,CONTACT Han Xia ; Zhiming Yuan Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Zhiming Yuan
- Key Laboratory of Special Pathogens and Biosafety, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
15
|
Saver AE, Crawford SA, Joyce JD, Bertke AS. Route of Infection Influences Zika Virus Shedding in a Guinea Pig Model. Cells 2019; 8:E1437. [PMID: 31739508 PMCID: PMC6912420 DOI: 10.3390/cells8111437] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/04/2019] [Accepted: 11/13/2019] [Indexed: 11/16/2022] Open
Abstract
Due to the recent epidemic of Zika virus (ZIKV) infection and resulting sequelae, as well as concerns about both the sexual and vertical transmission of the virus, renewed attention has been paid to the pathogenesis of this unique arbovirus. Numerous small animal models have been used in various ZIKV pathogenicity studies, however, they are often performed using immunodeficient or immunosuppressed animals, which may impact disease progression in a manner not relevant to immunocompetent humans. The use of immunocompetent animal models, such as macaques, is constrained by small sample sizes and the need for specialized equipment/staff. Here we report the establishment of ZIKV infection in an immunocompetent small animal model, the guinea pig, using both subcutaneous and vaginal routes of infection to mimic mosquito-borne and sexual transmission. Guinea pigs developed clinical signs consistent with mostly asymptomatic and mild disease observed in humans. We demonstrate that the route of infection does not significantly alter viral tissue tropism but does impact mucosal shedding mechanics. We also demonstrate persistent infection in sensory and autonomic ganglia, identifying a previously unrecognized niche of viral persistence that could contribute to viral shedding in secretions. We conclude that the guinea pig represents a useful and relevant model for ZIKV pathogenesis.
Collapse
Affiliation(s)
- Ashley E. Saver
- Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA; (A.E.S.); (S.A.C.)
| | - Stephanie A. Crawford
- Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA; (A.E.S.); (S.A.C.)
| | - Jonathan D. Joyce
- Department of Population Health Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA;
| | - Andrea S. Bertke
- Department of Population Health Sciences, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA;
| |
Collapse
|
16
|
Calderón-Peláez MA, Velandia-Romero ML, Bastidas-Legarda LY, Beltrán EO, Camacho-Ortega SJ, Castellanos JE. Dengue Virus Infection of Blood-Brain Barrier Cells: Consequences of Severe Disease. Front Microbiol 2019; 10:1435. [PMID: 31293558 PMCID: PMC6606788 DOI: 10.3389/fmicb.2019.01435] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 06/06/2019] [Indexed: 01/10/2023] Open
Abstract
More than 500 million people worldwide are infected each year by any of the four-dengue virus (DENV) serotypes. The clinical spectrum caused during these infections is wide and some patients may develop neurological alterations during or after the infection, which could be explained by the cryptic neurotropic and neurovirulent features of flaviviruses like DENV. Using in vivo and in vitro models, researchers have demonstrated that DENV can affect the cells from the blood-brain barrier (BBB) in several ways, which could result in brain tissue damage, neuronal loss, glial activation, tissue inflammation and hemorrhages. The latter suggests that BBB may be compromised during infection; however, it is not clear whether the damage is due to the infection per se or to the local and/or systemic inflammatory response established or activated by the BBB cells. Similarly, the kinetics and cascade of events that trigger tissue damage, and the cells that initiate it, are unknown. This review presents evidence of the BBB cell infection with DENV and the response established toward it by these cells; it also describes the consequences of this response on the nervous tissue, compares these evidence with the one reported with neurotropic viruses of the Flaviviridae family, and shows the complexity and unpredictability of dengue and the neurological alterations induced by it. Clinical evidence and in vitro and in vivo models suggest that this virus uses the bloodstream to enter nerve tissue where it infects the different cells of the neurovascular unit. Each of the cell populations respond individually and collectively and control infection and inflammation, in other cases this response exacerbates the damage leaving irreversible sequelae or causing death. This information will allow us to understand more about the complex disease known as dengue, and its impact on a specialized and delicate tissue like is the nervous tissue.
Collapse
|
17
|
Sherer ML, Khanal P, Talham G, Brannick EM, Parcells MS, Schwarz JM. Zika virus infection of pregnant rats and associated neurological consequences in the offspring. PLoS One 2019; 14:e0218539. [PMID: 31220154 PMCID: PMC6586346 DOI: 10.1371/journal.pone.0218539] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Accepted: 06/04/2019] [Indexed: 12/20/2022] Open
Abstract
Zika virus (ZIKV) is a mosquito-borne flavivirus associated with microcephaly and other neurological disorders in infants born to infected mothers. Despite being declared an international emergency by the World Health Organization, very little is known about the mechanisms of ZIKV pathogenesis or the long-term consequences of maternal ZIKV infection in the affected offspring, largely due to the lack of appropriate rodent models. To address this issue, our lab has developed a working model of prenatal ZIKV infection in rats. In this study, we infected immune competent pregnant female rats with 105-107 PFU of ZIKV (PRVABC59, Puerto Rico/Human/Dec 2015) in order to examine its pathogenesis in the dams and pups. We examined the febrile response and sickness behavior in the dams, in addition to neonatal mortality, microglia morphology, cortical organization, apoptosis, and brain region-specific volumes in the offspring. Here, we demonstrate that pregnant and non-pregnant female rats have a distinct febrile response to ZIKV infection. Moreover, prenatal ZIKV infection increased cell death and reduced tissue volume in the hippocampus and cortex in the neonatal offspring. For the first time, we demonstrate the efficacy and validity of an immunocompetent rat model for maternal ZIKV infection that results in significant brain malformations in the neonatal offspring.
Collapse
Affiliation(s)
- Morgan L. Sherer
- University of Delaware, Department of Psychological and Brain Sciences, Newark, Delaware, United States of America
| | - Pragyan Khanal
- University of Delaware, Department of Psychological and Brain Sciences, Newark, Delaware, United States of America
| | - Gwen Talham
- University of Delaware, Office of Laboratory Animal Medicine, Newark, Delaware, United States of America
| | - Erin M. Brannick
- University of Delaware, Department of Animal and Food Sciences, Newark, Delaware, United States of America
| | - Mark S. Parcells
- University of Delaware, Department of Animal and Food Sciences, Newark, Delaware, United States of America
| | - Jaclyn M. Schwarz
- University of Delaware, Department of Psychological and Brain Sciences, Newark, Delaware, United States of America
| |
Collapse
|
18
|
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: 2.2] [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
|
19
|
Zhang W, Tan YW, Yam WK, Tu H, Qiu L, Tan EK, Chu JJH, Zeng L. In utero infection of Zika virus leads to abnormal central nervous system development in mice. Sci Rep 2019; 9:7298. [PMID: 31086212 PMCID: PMC6513999 DOI: 10.1038/s41598-019-43303-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 04/16/2019] [Indexed: 12/14/2022] Open
Abstract
The World Health Organization has declared ZIKA virus (ZIKV) a global public health emergency, prompted by the association of ZIKV infections with severe brain abnormalities in the human fetus. ZIKV preferentially targets human neuronal precursor cells (NPCs) in both monolayer and cortical brain organoid culture systems and stunts their growth. Although ZIKV is well recognized to cause microcephaly, there is no systematic analysis to demonstrate the effect of ZIKV on central nervous system (CNS) development, including brain malformations and spinal cord dysfunction. Here, we conducted a longitudinal analysis to show that a novel mouse model (infected in utero and monitored after birth until adulthood) recapitulates the effects of ZIKV infection affecting neural stem cells fate and leads to a thinner cortex and a smaller brain. Furthermore, we demonstrate the effect of ZIKV on spinal cord function. Specifically, we found significant reductions in neuron numbers in the anterior horn of grey matter of the spinal cord and muscle dystrophy with a significant decrease in forepaw grip strength in the ZIKV group. Thus, the established mouse model of ZIKV infection leading to abnormal CNS development will help to further advance our understanding of the disease pathogenesis.
Collapse
Affiliation(s)
- Wei Zhang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Yong Wah Tan
- Collaborative Translation Unit for HFMD, Institute of Molecular and Cell Biology, Agency of Science, Technology & Research (A STAR), Singapore, 138673, Singapore
| | - Wan Keat Yam
- Collaborative Translation Unit for HFMD, Institute of Molecular and Cell Biology, Agency of Science, Technology & Research (A STAR), Singapore, 138673, Singapore
| | - Haitao Tu
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Lifeng Qiu
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Eng King Tan
- Research Department, National Neuroscience Institute, SGH Campus, Singapore, 169856, Singapore.,Department of Neurology, National Neuroscience Institute, SGH Campus, Singapore, 169856, Singapore.,Neuroscience & Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore, 169857, Singapore
| | - Justin Jang Hann Chu
- Collaborative Translation Unit for HFMD, Institute of Molecular and Cell Biology, Agency of Science, Technology & Research (A STAR), Singapore, 138673, Singapore.,Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Li Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, 308433, Singapore. .,Neuroscience & Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore, 169857, Singapore. .,Lee Kong Chian School of Medicine, Novena Campus, 11 Mandalay Road, Singapore, 308232, Singapore.
| |
Collapse
|
20
|
Mouse Strain and Sex-Dependent Differences in Long-term Behavioral Abnormalities and Neuropathologies after Developmental Zika Infection. J Neurosci 2019; 39:5393-5403. [PMID: 31085612 DOI: 10.1523/jneurosci.2666-18.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 03/25/2019] [Accepted: 04/14/2019] [Indexed: 12/13/2022] Open
Abstract
Exposure of the developing fetus to Zika virus (ZIKV) results in a set of brain abnormalities described as the congenital Zika syndrome. Although microcephaly is the most obvious outcome, neuropathologies, such as intracranial calcifications and polymicrogyria, can occur in the absence of microcephaly. Moreover, the full impact of exposure on motor, social, and cognitive skills during development remains uncharacterized. We examined the long-term neurobehavioral consequences of neonatal ZIKV exposure in four genetically divergent inbred mouse strains (C57BL/6J, 129S1/SvImJ, FVB/NJ, and DBA/2J). Male and female mice were infected on postnatal day 1, considered comparable with exposure late in the second trimester of humans. We demonstrate strain differences in early susceptibility to the virus and the time course of glial reaction in the brain. These changes were associated with strain- and sex-dependent differences in long-term behavioral abnormalities that include hyperactivity, impulsiveness, and motor incoordination. In addition, the adult brains of susceptible mice exhibited widespread calcifications that may underlie the behavioral deficits observed. Characterization of the neuropathological sequelae of developmental exposure to the Zika virus in different immunocompetent mouse strains provides a foundation for identifying genetic and immune factors that contribute to long-term neurobehavioral consequences in susceptible individuals.SIGNIFICANCE STATEMENT Developmental Zika virus (ZIKV) infection is now known to cause brain abnormalities in infants that do not display microcephaly at birth, and the full impact of these more subtle neuropathologies has yet to be determined. We demonstrate in a mouse model that long-lasting behavioral aberrations occur after developmental ZIKV exposure. We compare four divergent mouse strains and find that the effects of Zika infection differ greatly between strains, in terms of behavioral changes, sex differences, and the intracranial calcifications that develop in the brains of susceptible mice. These findings provide a foundation for identifying susceptibility factors that lead to the development of abnormal behaviors secondary to ZIKV infection early in life.
Collapse
|
21
|
Abstract
Zika virus (ZIKV) infection can result in serious consequences, including severe congenital manifestations, persistent infection in the testes, and neurologic sequelae. After a pandemic emergence, the virus has spread to much of North and South America and has been introduced to many countries outside of ZIKV-endemic areas as infected travelers return to their home countries. Rodent models have been important in gaining a better understanding of the wide range of disease etiologies associated with ZIKV infection and for the initial phase of developing countermeasures to prevent or treat viral infections. We discuss herein the advantages and disadvantages of small-animal models that have been developed to replicate various aspects of disease associated with ZIKV infection.
Collapse
Affiliation(s)
- Justin G Julander
- Institute for Antiviral Research, ADVS Department, Utah State University, Logan
| | | |
Collapse
|
22
|
Lei J, Vermillion MS, Jia B, Xie H, Xie L, McLane MW, Sheffield JS, Pekosz A, Brown A, Klein SL, Burd I. IL-1 receptor antagonist therapy mitigates placental dysfunction and perinatal injury following Zika virus infection. JCI Insight 2019; 4:122678. [PMID: 30944243 DOI: 10.1172/jci.insight.122678] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 02/14/2019] [Indexed: 12/25/2022] Open
Abstract
Zika virus (ZIKV) infection during pregnancy causes significant adverse sequelae in the developing fetus, and results in long-term structural and neurologic defects. Most preventive and therapeutic efforts have focused on the development of vaccines, antivirals, and antibodies. The placental immunologic response to ZIKV, however, has been largely overlooked as a target for therapeutic intervention. The placental inflammatory response, specifically IL-1β secretion and signaling, is induced by ZIKV infection and represents an environmental factor that is known to increase the risk of perinatal developmental abnormalities. We show in a mouse model that maternally administrated IL-1 receptor antagonist (IRA; Kineret, or anakinra), following ZIKV exposure, can preserve placental function (by improving trophoblast invasion and placental vasculature), increase fetal viability, and reduce neurobehavioral deficits in the offspring. We further demonstrate that while ZIKV RNA is highly detectable in placentas, it is not correlated with fetal viability. Beyond its effects in the placenta, we show that IL-1 blockade may also directly decrease fetal neuroinflammation by mitigating fetal microglial activation in a dose-dependent manner. Our studies distinguish the role of placental inflammation during ZIKV-infected pregnancies, and demonstrate that maternal IRA may attenuate fetal neuroinflammation and improve perinatal outcomes.
Collapse
Affiliation(s)
- Jun Lei
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Meghan S Vermillion
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.,Department of Molecular and Comparative Pathobiology
| | - Bei Jia
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Han Xie
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Li Xie
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael W McLane
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jeanne S Sheffield
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Amanda Brown
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA.,Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Irina Burd
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
23
|
Integrated MicroRNA and mRNA Profiling in Zika Virus-Infected Neurons. Viruses 2019; 11:v11020162. [PMID: 30781519 PMCID: PMC6410042 DOI: 10.3390/v11020162] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/08/2019] [Accepted: 02/14/2019] [Indexed: 12/13/2022] Open
Abstract
Zika virus (ZIKV) infections have caused a wide spectrum of neurological diseases, such as Guillain-Barré syndrome, myelitis, meningoencephalitis, and congenital microcephaly. No effective therapies currently exist for treating patients infected with ZIKV. MicroRNAs (miRNAs) are a group of small RNAs involved in the regulation of a wide variety of cellular and physiological processes. In this study, we analyzed digital miRNA and mRNA profiles in ZIKV-infected primary mouse neurons using the nCounter technology. A total of 599 miRNAs and 770 mRNAs were examined. We demonstrate that ZIKV infection causes global downregulation of miRNAs with only few upregulated miRNAs. ZIKV-modulated miRNAs including miR-155, miR-203, miR-29a, and miR-124-3p are known to play critical role in flavivirus infection, anti-viral immunity and brain injury. ZIKV infection also results in downregulation of miRNA processing enzymes. In contrast, ZIKV infection induces dramatic upregulation of anti-viral, inflammatory and apoptotic genes. Furthermore, our data demonstrate an inverse correlation between ZIKV-modulated miRNAs and target host mRNAs induced by ZIKV. Biofunctional analysis revealed that ZIKV-modulated miRNAs and mRNAs regulate the pathways related to neurological development and neuroinflammatory responses. Functional studies targeting specific miRNA are warranted to develop therapeutics for the management of ZIKV neurological disease.
Collapse
|
24
|
A 'Furry-Tale' of Zika Virus Infection: What Have We Learned from Animal Models? Viruses 2019; 11:v11010029. [PMID: 30621317 PMCID: PMC6356866 DOI: 10.3390/v11010029] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/21/2018] [Accepted: 01/04/2019] [Indexed: 01/07/2023] Open
Abstract
The worldwide attention that the Zika virus (ZIKV) attracted, following its declaration as a Public Health Emergency of International concern by WHO in 2016, has led to a large collective effort by the international scientific community to understand its biology. Despite the mild symptoms caused by ZIKV in most infected people, the virus displays a number of worrying features, such as its ability to cause transplacental infection, fetal abnormalities and vector independent transmission through body fluids. In addition, the virus has been associated with the induction of Guillain-Barre syndrome in a number of infected individuals. With travelling, the virus has spread outside the original ZIKV endemic areas making it imperative to find ways to control it. Thus far, the large number of animal models developed to study ZIKV pathogenesis have proven to be valuable tools in understanding how the virus replicates and manifests itself in the host, its tissue tropism and the type of immune responses it induces. Still, vital questions, such as the molecular mechanisms of ZIKV persistence and the long-term consequences of ZIKV infection in the developing brain, remain unanswered. Here, we reviewed and discussed the major and most recent findings coming from animal studies and their implications for a ZIKV vaccine design.
Collapse
|
25
|
Pena LJ, Miranda Guarines K, Duarte Silva AJ, Sales Leal LR, Mendes Félix D, Silva A, de Oliveira SA, Junqueira Ayres CF, Júnior AS, de Freitas AC. In vitro and in vivo models for studying Zika virus biology. J Gen Virol 2018; 99:1529-1550. [DOI: 10.1099/jgv.0.001153] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Lindomar José Pena
- 1Department of Virology, Aggeu Magalhaes Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife, Pernambuco, Brazil
| | - Klarissa Miranda Guarines
- 1Department of Virology, Aggeu Magalhaes Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife, Pernambuco, Brazil
| | - Anna Jéssica Duarte Silva
- 2Department of Genetics, Laboratory of Molecular Studies and Experimental Therapy (LEMTE), Center of Biological Sciences, Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
| | - Lígia Rosa Sales Leal
- 2Department of Genetics, Laboratory of Molecular Studies and Experimental Therapy (LEMTE), Center of Biological Sciences, Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
| | - Daniele Mendes Félix
- 1Department of Virology, Aggeu Magalhaes Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife, Pernambuco, Brazil
| | - Adalúcia Silva
- 1Department of Virology, Aggeu Magalhaes Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife, Pernambuco, Brazil
| | - Sheilla Andrade de Oliveira
- 3Department of Immunology, Aggeu Magalhaes Institute (IAM), Oswaldo Cruz Foundation (Fiocruz), Recife, Pernambuco, Brazil
| | | | - Abelardo Silva Júnior
- 5Department of Veterinary Medicine, Federal University of Viçosa (UFV), Viçosa, Minas Gerais, Brazil
| | - Antonio Carlos de Freitas
- 2Department of Genetics, Laboratory of Molecular Studies and Experimental Therapy (LEMTE), Center of Biological Sciences, Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
| |
Collapse
|
26
|
Mancera-Páez O, Román GC, Pardo-Turriago R, Rodríguez Y, Anaya JM. Concurrent Guillain-Barré syndrome, transverse myelitis and encephalitis post-Zika: A case report and review of the pathogenic role of multiple arboviral immunity. J Neurol Sci 2018; 395:47-53. [PMID: 30292020 DOI: 10.1016/j.jns.2018.09.028] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 09/23/2018] [Accepted: 09/25/2018] [Indexed: 12/15/2022]
Abstract
We review post-infectious and post-vaccination neurological syndromes involving peripheral and central nervous system (CNS) and report an illustrative case of simultaneous occurrence of Guillain-Barré syndrome (GBS), confirmed by nerve conduction velocities, plus MRI-demonstrated transverse myelitis (TM) and acute encephalitis [acute disseminated encephalomyelitis] (ADEM+GBS) affecting a 24-year-old woman from Cúcuta, Colombia, who developed acute Zika virus (ZIKV) infection confirmed by serum reverse transcriptase-polymerase chain reaction (RT-PCR) and convalescent ZIKV IgG antibodies. With intensive care treatment, respiratory support, steroids, and intravenous immunoglobulin (IVIg), patient survived with residual flaccid paraparesis. She had preexisting immunity against Chikungunya virus (CHIKV) and Dengue virus (DENV) acquired before the arrival of ZIKV in Colombia. From reports in the Caribbean, Central and South America we review 19 cases of ZIKV-associated TM, encephalitis and ADEM occurring after a mean latent period of 10.5 days (range 1-96) post-infection. Although GBS and ADEM are usually considered post-infectious and associated with development of antibodies against peripheral nerve and CNS epitopes, we postulate that our case of ADEM+GBS is para-infectious, induced by acute ZIKV neurotropism boosted by active immunity against other arboviruses. Animal models of ZIKV demonstrated strong viral neurotropism enhanced by passive immunity with antibodies against arboviruses such as West Nile virus, CHIKV, or DENV. These considerations are relevant to prevent potential ZIKV vaccine-induced reactions involving central and peripheral nervous system.
Collapse
Affiliation(s)
- Oscar Mancera-Páez
- Universidad Nacional de Colombia, Hospital Universitario Nacional, Faculty of Medicine, Department of Neurology, Bogotá, Colombia.; David Cabello International Alzheimer Disease Scholarship Fund, Houston Methodist Hospital, Houston, TX, USA..
| | - Gustavo C Román
- Department of Neurology, Methodist Neurological Institute and the Institute for Academic Medicine Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, USA; Weill Cornell Medical College, Department of Neurology, Cornell University, NY, New York, USA.
| | - Rodrigo Pardo-Turriago
- Universidad Nacional de Colombia, Hospital Universitario Nacional, Faculty of Medicine, Department of Neurology, Bogotá, Colombia..
| | - Yhojan Rodríguez
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia.
| | - Juan-Manuel Anaya
- Center for Autoimmune Diseases Research (CREA), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
27
|
Chen Y, Liu T, Zhang Z, Chen M, Rong L, Ma L, Yu B, Wu D, Zhang P, Zhu X, Huang X, Zhang H, Li YP. Novel genetically stable infectious clone for a Zika virus clinical isolate and identification of RNA elements essential for virus production. Virus Res 2018; 257:14-24. [PMID: 30144463 DOI: 10.1016/j.virusres.2018.08.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/18/2018] [Accepted: 08/20/2018] [Indexed: 01/19/2023]
Abstract
Zika virus (ZIKV) is an Aedes mosquitoes-transmitted flavivirus, and its infection may cause severe neurological diseases. A genetically stable infectious clone is essential for ZIKV research, however the toxicity and instability of the viral cDNA in bacteria potentially due to its bacterial promoter activity are major challenges. Here, we constructed a full-length cDNA clone for isolate ZG01 by introducing non-coding changes T1865C/A1868G to reduce the bacterial promoter activity. Wild-type and recombinant ZG01 were highly attenuated in Vero cells, thus we serially passaged wild-type ZG01 through neonatal mice and Vero cells to generate high-titer virus, from which four mutations (4m, C2178T/G2913A/T4991C/T10561C) were identified. Addition of 4m greatly enhanced the infectivity, as ZG01_4m released ZIKV of 107.0-107.5 plaque-forming unit (PFU)/ml in infected Vero and A549 cells. ZG01_4m resembled the infectivity of high-titer ZG01 in vitro and in vivo. Notably, ZG01_4m plasmid was genetically stable after multiple rounds of transformation-purification in bacteria. Using ZG01_4m, we identified a potential RNA-RNA interaction between 5'UTR and 3'UTR and demonstrated that the nucleotides involved were essential for ZIKV production. The genetically stable ZG01 cDNA clone provides a reliable tool for the study of this important virus, and the strategy used here is feasible for the development of reverse genetics systems for other ZIKV isolates and related flaviviruses.
Collapse
Affiliation(s)
- Yiyi Chen
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Ting Liu
- Department of Immunology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Zhenzhen Zhang
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Mingxiao Chen
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Liang Rong
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Ling Ma
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Bolan Yu
- Key Lab for Major Obstetric Diseases of Guangdong Province, Guangzhou Medical University, Guangzhou 510150, China
| | - De Wu
- Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511430, China
| | - Ping Zhang
- Department of Immunology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Xun Zhu
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Xi Huang
- The Fifth Affiliated Hospital of Sun Yat-sen University, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Hui Zhang
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yi-Ping Li
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-Sen University, Guangzhou 510080, China; Program in Pathobiology, The Fifth Affiliated Hospital and Zhongshan School of Medicine, Sun Yat-sen University, Guangdong, China.
| |
Collapse
|
28
|
Yun SI, Song BH, Frank JC, Julander JG, Olsen AL, Polejaeva IA, Davies CJ, White KL, Lee YM. Functional Genomics and Immunologic Tools: The Impact of Viral and Host Genetic Variations on the Outcome of Zika Virus Infection. Viruses 2018; 10:v10080422. [PMID: 30103523 PMCID: PMC6116225 DOI: 10.3390/v10080422] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 07/31/2018] [Accepted: 08/02/2018] [Indexed: 12/13/2022] Open
Abstract
Zika virus (ZIKV) causes no-to-mild symptoms or severe neurological disorders. To investigate the importance of viral and host genetic variations in determining ZIKV infection outcomes, we created three full-length infectious cDNA clones as bacterial artificial chromosomes for each of three spatiotemporally distinct and genetically divergent ZIKVs: MR-766 (Uganda, 1947), P6-740 (Malaysia, 1966), and PRVABC-59 (Puerto Rico, 2015). Using the three molecularly cloned ZIKVs, together with 13 ZIKV region-specific polyclonal antibodies covering nearly the entire viral protein-coding region, we made three conceptual advances: (i) We created a comprehensive genome-wide portrait of ZIKV gene products and their related species, with several previously undescribed gene products identified in the case of all three molecularly cloned ZIKVs. (ii) We found that ZIKV has a broad cell tropism in vitro, being capable of establishing productive infection in 16 of 17 animal cell lines from 12 different species, although its growth kinetics varied depending on both the specific virus strain and host cell line. More importantly, we identified one ZIKV-non-susceptible bovine cell line that has a block in viral entry but fully supports the subsequent post-entry steps. (iii) We showed that in mice, the three molecularly cloned ZIKVs differ in their neuropathogenicity, depending on the particular combination of viral and host genetic backgrounds, as well as in the presence or absence of type I/II interferon signaling. Overall, our findings demonstrate the impact of viral and host genetic variations on the replication kinetics and neuropathogenicity of ZIKV and provide multiple avenues for developing and testing medical countermeasures against ZIKV.
Collapse
Affiliation(s)
- Sang-Im Yun
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA.
| | - Byung-Hak Song
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA.
| | - Jordan C Frank
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA.
| | - Justin G Julander
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA.
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, USA.
| | - Aaron L Olsen
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA.
| | - Irina A Polejaeva
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA.
- Veterinary Diagnostics and Infectious Diseases, Utah Science Technology and Research, Utah State University, Logan, UT 84341, USA.
| | - Christopher J Davies
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA.
- Veterinary Diagnostics and Infectious Diseases, Utah Science Technology and Research, Utah State University, Logan, UT 84341, USA.
| | - Kenneth L White
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA.
- Veterinary Diagnostics and Infectious Diseases, Utah Science Technology and Research, Utah State University, Logan, UT 84341, USA.
| | - Young-Min Lee
- Department of Animal Dairy and Veterinary Sciences, College of Agriculture and Applied Sciences, Utah State University, Logan, UT 84322, USA.
- Veterinary Diagnostics and Infectious Diseases, Utah Science Technology and Research, Utah State University, Logan, UT 84341, USA.
| |
Collapse
|
29
|
Lebov JF, Brown LM, MacDonald PDM, Robertson K, Bowman NM, Hooper SR, Becker-Dreps S. Review: Evidence of Neurological Sequelae in Children With Acquired Zika Virus Infection. Pediatr Neurol 2018; 85:16-20. [PMID: 30343688 DOI: 10.1016/j.pediatrneurol.2018.03.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 03/01/2018] [Indexed: 12/30/2022]
Abstract
Limited information is available on health outcomes related to Zika virus infection acquired during childhood. Zika virus can cause severe central nervous system malformations in congenitally exposed fetuses and neonates. In vitro studies show the capacity of Zika virus to infect neural progenitor cells, induce central and peripheral neuronal cell deaths, and target different brain cells over the course of brain development. Studies of postnatally infected mice and nonhuman primates have detected degradation of neural cells and morphologic brain cell changes consistent with a broad neuroinflammatory response. In addition, case reports of central nervous system disease in adults and in adolescents secondary to Zika virus infection suggest that Zika virus may have a broader impact on neurological health beyond that observed in congenitally exposed newborns. Long-term neurological complications have been observed with other acquired flaviviral infections, with clinical symptoms manifesting for years after primary infection. The extent to which postnatal Zika virus infection in humans negatively affects the central and peripheral nervous systems and causes long-term neurological damage or cognitive effects is currently unknown. To better understand the potential for neurological sequelae associated with acquired Zika virus infection in children, we reviewed the biological, clinical, and epidemiologic literature and summarized the evidence for this link. First, we review biological mechanisms for neurological manifestations of Zika virus infection in experimental studies. Second, we review observational studies of congenital Zika virus infection and case studies and surveillance reports of neurological sequelae of Zika virus infection in adults and in children. Lastly, we discuss the challenges of conducting Zika virus-neurological sequela studies and future directions for pediatric Zika virus research.
Collapse
Affiliation(s)
- Jill F Lebov
- RTI International, Center for Applied Public Health Research, Durham, North Carolina.
| | - Linda M Brown
- RTI International, Center for Applied Public Health Research, Rockville, Maryland
| | - Pia D M MacDonald
- RTI International, Center for Applied Public Health Research, Berkeley, California
| | - Kevin Robertson
- Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Natalie M Bowman
- Division of Infectious Diseases, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Stephen R Hooper
- Department of Psychiatry, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Sylvia Becker-Dreps
- Department of Family Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
30
|
Julander JG, Siddharthan V, Park AH, Preston E, Mathur P, Bertolio M, Wang H, Zukor K, Van Wettere AJ, Sinex DG, Morrey JD. Consequences of in utero exposure to Zika virus in offspring of AG129 mice. Sci Rep 2018; 8:9384. [PMID: 29925850 PMCID: PMC6010449 DOI: 10.1038/s41598-018-27611-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 05/31/2018] [Indexed: 12/20/2022] Open
Abstract
Zika virus (ZIKV) can cause various diseases in offspring after congenital infection. The purpose of this study was to identify disease phenotypes in pups exposed to ZIKV in utero. Female interferon-α/β, -γ receptor knockout mice (AG129) were infected intraperitoneally with ZIKV 7.5 days' post coitus (dpc). Viral RNA, antigen and infectious virus were detected in some, but not all, maternal and fetal tissues at various times during gestation. Fetuses of infected dams had significant intrauterine growth restriction (IUGR), which was more pronounced as females neared parturition. Pups born to infected dams were significantly smaller and had significantly shortened skull lengths, as determined by measurement with a caliper and by micro-CT analysis, as compared with age-matched controls. Growth rates of exposed pups after birth, however, was similar to sham-exposed offspring. Viral RNA was detected in pups of infected dams after birth. A lower survival rate was observed in neonates exposed to ZIKV in utero. A mortality rate of over 50%, attributed to consequences of ZIKV infection, occurred after birth in pups born to infected dams. A transient hearing loss was observed in some animals exposed to virus in utero. No motor deficits or cognitive deficits were detected using running wheel or viral paresis scoring assays. Abnormalities in offspring included smaller size, shorter skull length and increased neonatal mortality, while the only functional deficit we could detect was a low incidence of transient hearing loss.
Collapse
Affiliation(s)
- Justin G Julander
- Institute for Antiviral Research, Animal, Dairy, and Veterinary Sciences Department, 5600 Old Main Hill, Utah State University, Logan, Utah, 84322-5600, USA.
| | - Venkatraman Siddharthan
- Institute for Antiviral Research, Animal, Dairy, and Veterinary Sciences Department, 5600 Old Main Hill, Utah State University, Logan, Utah, 84322-5600, USA
| | - Albert H Park
- Division of Otolaryngology- Head and Neck Surgery, University of Utah, 50 N Medical Dr., Salt Lake City, UT, USA
| | - Elizabeth Preston
- Audiology Division, Department of Communicative Disorders and Deaf Education, Utah State University, 2620 Old Main Hill, Logan, UT, 84322-2620, USA
| | - Pranav Mathur
- Department of Neurobiology and Anatomy, University of Utah, 1795 E South Campus Drive, Salt Lake City, UT, 84112, USA
| | - Michael Bertolio
- Institute for Antiviral Research, Animal, Dairy, and Veterinary Sciences Department, 5600 Old Main Hill, Utah State University, Logan, Utah, 84322-5600, USA
| | - Hong Wang
- Institute for Antiviral Research, Animal, Dairy, and Veterinary Sciences Department, 5600 Old Main Hill, Utah State University, Logan, Utah, 84322-5600, USA
| | - Katherine Zukor
- Institute for Antiviral Research, Animal, Dairy, and Veterinary Sciences Department, 5600 Old Main Hill, Utah State University, Logan, Utah, 84322-5600, USA
| | - Arnaud J Van Wettere
- Utah Veterinary Diagnostics Laboratory, Utah State University, 950 E 1400 N, Logan, UT, 84341, USA
| | - Donal G Sinex
- Department of Biology, Utah State University, 5305 Old Main Hill, Logan, UT, 84322-5305, USA
| | - John D Morrey
- Institute for Antiviral Research, Animal, Dairy, and Veterinary Sciences Department, 5600 Old Main Hill, Utah State University, Logan, Utah, 84322-5600, USA
| |
Collapse
|
31
|
Watanabe M, Buth JE, Vishlaghi N, de la Torre-Ubieta L, Taxidis J, Khakh BS, Coppola G, Pearson CA, Yamauchi K, Gong D, Dai X, Damoiseaux R, Aliyari R, Liebscher S, Schenke-Layland K, Caneda C, Huang EJ, Zhang Y, Cheng G, Geschwind DH, Golshani P, Sun R, Novitch BG. Self-Organized Cerebral Organoids with Human-Specific Features Predict Effective Drugs to Combat Zika Virus Infection. Cell Rep 2018; 21:517-532. [PMID: 29020636 DOI: 10.1016/j.celrep.2017.09.047] [Citation(s) in RCA: 289] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 08/01/2017] [Accepted: 09/14/2017] [Indexed: 12/21/2022] Open
Abstract
The human cerebral cortex possesses distinct structural and functional features that are not found in the lower species traditionally used to model brain development and disease. Accordingly, considerable attention has been placed on the development of methods to direct pluripotent stem cells to form human brain-like structures termed organoids. However, many organoid differentiation protocols are inefficient and display marked variability in their ability to recapitulate the three-dimensional architecture and course of neurogenesis in the developing human brain. Here, we describe optimized organoid culture methods that efficiently and reliably produce cortical and basal ganglia structures similar to those in the human fetal brain in vivo. Neurons within the organoids are functional and exhibit network-like activities. We further demonstrate the utility of this organoid system for modeling the teratogenic effects of Zika virus on the developing brain and identifying more susceptibility receptors and therapeutic compounds that can mitigate its destructive actions.
Collapse
Affiliation(s)
- Momoko Watanabe
- Department of Neurobiology, David Geffen School of Medicine at UCLA, 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; Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jessie E Buth
- Department of Neurobiology, David Geffen School of Medicine at UCLA, 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; Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Neda Vishlaghi
- Department of Neurobiology, David Geffen School of Medicine at UCLA, 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; Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Luis de la Torre-Ubieta
- Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Center for Autism Research and Treatment and Program in Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Neurology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jiannis Taxidis
- Department of Neurology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Baljit S Khakh
- Department of Neurobiology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Physiology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Giovanni Coppola
- Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Center for Autism Research and Treatment and Program in Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Neurology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Caroline A Pearson
- Department of Neurobiology, David Geffen School of Medicine at UCLA, 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; Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ken Yamauchi
- Department of Neurobiology, David Geffen School of Medicine at UCLA, 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; Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Danyang Gong
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Xinghong Dai
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Robert Damoiseaux
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Roghiyh Aliyari
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Simone Liebscher
- Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Katja Schenke-Layland
- Department of Cardiology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University Tübingen, 72076 Tübingen, Germany; Department of Cell and Tissue Engineering, Fraunhofer Institute for Interfacial Engineering and Biotechnology, 70569 Stuttgart, Germany
| | - Christine Caneda
- Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Eric J Huang
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ye Zhang
- Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Genhong Cheng
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Daniel H Geschwind
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Center for Autism Research and Treatment and Program in Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Neurology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Peyman Golshani
- Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA; Center for Autism Research and Treatment and Program in Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Neurology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at UCLA, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ren Sun
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Bennett G Novitch
- Department of Neurobiology, David Geffen School of Medicine at UCLA, 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; Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
32
|
Koppolu V, Shantha Raju T. Zika virus outbreak: a review of neurological complications, diagnosis, and treatment options. J Neurovirol 2018; 24:255-272. [PMID: 29441490 DOI: 10.1007/s13365-018-0614-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 01/18/2018] [Indexed: 02/07/2023]
Abstract
Zika virus (ZIKV) is an arbovirus transmitted mainly by mosquitos of Aedes species. The virus has emerged in recent years and spread throughout North and South Americas. The recent outbreak of ZIKV started in Brazil (2015) has resulted in infections surpassing a million mark. Contrary to the previous beliefs that Zika causes mildly symptomatic infections fever, headache, rash, arthralgia, and conjunctivitis, the recent outbreak associated ZIKV to serious neurological complications such as microcephaly, Guillain-Barré syndrome, and eye infections. The recent outbreak has resulted in an astonishing number of microcephaly cases in fetus and infants. Consequently, numerous studies were conducted using in vitro cell and in vivo animal models. These studies showed clear links between ZIKV infections and neurological abnormalities. Diagnosis methods based on nucleic acid and serological detection facilitated rapid and accurate identification of ZIKV infections. New transmission modalities such as sexual and transplacental transmission were uncovered. Given the seriousness of ZIKV infections, WHO declared the development of safe and effective vaccines and new antiviral drugs as an urgent global health priority. Rapid work in this direction has led to the identification of several vaccine and antiviral drug candidates. Here, we review the remarkable progress made in understanding the molecular links between ZIKV infections and neurological irregularities, new diagnosis methods, potential targets for antiviral drugs, and the current state of vaccine development.
Collapse
Affiliation(s)
- Veerendra Koppolu
- Global Bioassay Development and Quality, Biopharmaceutical Development, MedImmune, Gaithersburg, MD, USA
| | - T Shantha Raju
- Global Bioassay Development and Quality, Biopharmaceutical Development, MedImmune, Gaithersburg, MD, USA.
| |
Collapse
|
33
|
Ramalho FS, Yamamoto AY, da Silva LL, Figueiredo LTM, Rocha LB, Neder L, Teixeira SR, Apolinário LA, Ramalho LNZ, Silva DM, Coutinho CM, Melli PP, Augusto MJ, Santoro LB, Duarte G, Mussi-Pinhata MM. Congenital Zika Virus Infection Induces Severe Spinal Cord Injury. Clin Infect Dis 2018; 65:687-690. [PMID: 28444144 DOI: 10.1093/cid/cix374] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/18/2017] [Indexed: 01/01/2023] Open
Abstract
We report 2 fatal cases of congenital Zika virus (ZIKV) infection. Brain anomalies, including atrophy of the cerebral cortex and brainstem, and cerebellar aplasia were observed. The spinal cord showed architectural distortion, severe neuronal loss, and microcalcifications. The ZIKV proteins and flavivirus-like particles were detected in cytoplasm of spinal neurons, and spinal cord samples were positive for ZIKV RNA.
Collapse
Affiliation(s)
- Fernando S Ramalho
- Department of Pathology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Ribeirão Preto, Brazil
| | - Aparecida Y Yamamoto
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Luis L da Silva
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Luiz T M Figueiredo
- Department of Medicine, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Lenaldo B Rocha
- Institute of Biological and Natural Sciences, Federal University of Triângulo Mineiro, Uberaba
| | - Luciano Neder
- Department of Pathology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Ribeirão Preto, Brazil
| | - Sara R Teixeira
- Department of Medicine, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Letícia A Apolinário
- Department of Pathology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Ribeirão Preto, Brazil
| | - Leandra N Z Ramalho
- Department of Pathology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Ribeirão Preto, Brazil
| | - Deisy M Silva
- Department of Pathology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Ribeirão Preto, Brazil
| | - Conrado M Coutinho
- Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Patrícia P Melli
- Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Marlei J Augusto
- Department of Pathology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Ribeirão Preto, Brazil
| | - Ligia B Santoro
- Department of Pathology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes 3900, Ribeirão Preto, Brazil
| | - Geraldo Duarte
- Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Marisa M Mussi-Pinhata
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
34
|
Araújo PSRD, Silva Júnior MLDM, Tenório M, Santos FGTD. Co-infection ZIKV and HSV-1 associated with meningoencephalitis: Case report and literature review. J Infect Public Health 2018; 12:97-100. [PMID: 29703710 DOI: 10.1016/j.jiph.2018.04.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 03/15/2018] [Accepted: 04/08/2018] [Indexed: 10/17/2022] Open
Abstract
A man, 26years-old, presented fever, mental confusion and a progressively worsening headache 6days prior to admission. The CSF study was suggestive of meningoencephalitis, the PCR study revealed presence of HSV-1 and ZIKV, while other immunology tests were negative. ZIKV was also identified in serum. The MRI showed temporal lobe hyper-intensity in FLAIR-weight sequence with areas of contrast enhancement and the electroencephalogram showed slow wave activity in such region. Patient was treated with acyclovir and supportive measures and had good clinical outcome at evaluation after 6 months. Neurological spectrum of ZIKV manifestations is wide, but meningoencephalitis is not frequent. Co-infection HSV-1 plus ZIKV was not yet related in humans, but there is increased cellular damage caused by association of ZIKV and herpes virus family infection. ZIKV may facilitate infection or recrudescence by other viruses or cause concurrently neuronal injury by direct or indirect mechanisms. We suggest that clinicians attempt new manifestations related to ZIKV and include this agent in differential diagnosis of neurological diseases even when other agents were identified.
Collapse
Affiliation(s)
- Paulo S Ramos de Araújo
- Instituto Aggeu Magalhaes, Fiocruz, Recife, Pernambuco, Brazil; Universidade Federal de Pernambuco, Departamento de Medicina Tropical, Recife, Pernambuco, Brazil
| | | | - Marli Tenório
- Instituto Aggeu Magalhaes, Fiocruz, Recife, Pernambuco, Brazil
| | | |
Collapse
|
35
|
Nico D, Conde L, Rivera-Correa JL, Vasconcelos-Dos-Santos A, Mesentier-Louro L, Freire-de-Lima L, Arruda MB, Freire-de-Lima CG, Ferreira ODC, Lopes Moreira ME, Zin AA, Vasconcelos ZFM, Otero RM, Palatnik-de-Sousa CB, Tanuri A, Todeschini AR, Savino W, Rodriguez A, Morrot A. Prevalence of IgG Autoantibodies against GD3 Ganglioside in Acute Zika Virus Infection. Front Med (Lausanne) 2018; 5:25. [PMID: 29594116 PMCID: PMC5854646 DOI: 10.3389/fmed.2018.00025] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 01/25/2018] [Indexed: 01/28/2023] Open
Abstract
Zika virus (ZIKV) disease has become a global health emergency with devastating effects on public health. Recent evidences implicate the virus as an emergent neuropathological agent promoting serious pathologies of the human nervous system, that include destructive and malformation consequences such as development of ocular and fetal brain lesions, microcephaly in neonates, and Guillain–Barré syndrome (GBS) in adults. These neurological disorders of both central and peripheral nervous systems are thought to be associated to the neurotropic properties of the virus that has ability to infect neural stem cells as well as peripheral neurons, a hallmark of its pathogenicity. The presence of autoantibodies against gangliosides plays a pivotal role in the etiogenesis of GBS and a variety of neurological disorders. Gangliosides are a class of galactose-containing cerebrosides mainly expressed in nervous system tissues playing a critical role in the physiology of neural cells and neurogenesis. Herein, our findings indicate that patients at acute phase of ZIKV infection without any neurological signs show increased levels of IgG autoantibody against GD3 gangliosides, a class of glycolipid found to be highly expressed in neural stem cell acting in the maintenance of their self-renewal cellular capacity. It is possible that a pathological threshold of these antibodies is only acquired in secondary or subsequent infections. In the light of these evidences, we propose that the target of GD3 by autoimmune responses may possibly has an effect in the neuropathy and neurogenesis disorder seen during ZIKV infection.
Collapse
Affiliation(s)
- Dirlei Nico
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana Conde
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Juan L Rivera-Correa
- Department of Microbiology, New York University School of Medicine, New York, NY, United States
| | | | - Louise Mesentier-Louro
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo Freire-de-Lima
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | - Maria Elisabeth Lopes Moreira
- Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, Unidade de Pesquisa Clínica, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Andrea Araújo Zin
- Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, Unidade de Pesquisa Clínica, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Zilton Farias Meira Vasconcelos
- Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, Unidade de Pesquisa Clínica, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Rosalia Mendez Otero
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Amilcar Tanuri
- Departamento de Genética, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Adriane Regina Todeschini
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Wilson Savino
- Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Rio de Janeiro, Brazil
| | - Ana Rodriguez
- Department of Microbiology, New York University School of Medicine, New York, NY, United States
| | - Alexandre Morrot
- Faculdade de Medicina, Centro de Pesquisas em Tuberculose, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Instituto Oswaldo Cruz, Laboratório de Imunopatologia, FIOCRUZ, Rio de Janeiro, Brazil
| |
Collapse
|
36
|
Abstract
Despite being discovered approximately 70 years ago, Zika virus (ZIKV) has received little attention, until the occurrence of alarming epidemics in the Pacific Islands and Latin America between 2013 and 2016. These series of outbreaks resulted in crippling neurological complications in adults, and congenital deformities in new-borns. The dire outcomes marked ZIKV as a re-emerging pathogen of public health concern. Over a period of two years, extensive studies have been conducted to understand different aspects of ZIKV from pathogen biology to infection, including the immune response during virus-host interplay in established animal models, as well as potential therapeutics against ZIKV infection. The vast diversity of novel findings has added value to ZIKV research, and a strategic consolidation is crucial to encompass the latest advances and developments, as well as missing pieces of the puzzle. This review thus aims to provide a concise yet extensive update on current ZIKV studies.
Collapse
Affiliation(s)
- Cheryl Yi-Pin Lee
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| | - Lisa F P Ng
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Institute of Infection and Global Health, University of Liverpool, UK.
| |
Collapse
|
37
|
Zika virus-induced acute myelitis and motor deficits in adult interferon αβ/γ receptor knockout mice. J Neurovirol 2018; 24:273-290. [PMID: 29476408 PMCID: PMC5992253 DOI: 10.1007/s13365-017-0595-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/11/2017] [Accepted: 10/23/2017] [Indexed: 12/22/2022]
Abstract
Zika virus (ZIKV) has received widespread attention because of its effect on the developing fetus. It is becoming apparent, however, that severe neurological sequelae, such as Guillian-Barrë syndrome (GBS), myelitis, encephalitis, and seizures can occur after infection of adults. This study demonstrates that a contemporary strain of ZIKV can widely infect astrocytes and neurons in the brain and spinal cord of adult, interferon α/β receptor knockout mice (AG129 strain) and cause progressive hindlimb paralysis, as well as severe seizure-like activity during the acute phase of disease. The severity of hindlimb motor deficits correlated with increased numbers of ZIKV-infected lumbosacral spinal motor neurons and decreased numbers of spinal motor neurons. Electrophysiological compound muscle action potential (CMAP) amplitudes in response to stimulation of the lumbosacral spinal cord were reduced when obvious motor deficits were present. ZIKV immunoreactivity was high, intense, and obvious in tissue sections of the brain and spinal cord. Infection in the brain and spinal cord was also associated with astrogliosis as well as T cell and neutrophil infiltration. CMAP and histological analysis indicated that peripheral nerve and muscle functions were intact. Consequently, motor deficits in these circumstances appear to be primarily due to myelitis and possibly encephalitis as opposed to a peripheral neuropathy or a GBS-like syndrome. Thus, acute ZIKV infection of adult AG129 mice may be a useful model for ZIKV-induced myelitis, encephalitis, and seizure activity.
Collapse
|
38
|
Zika Virus Fatally Infects Wild Type Neonatal Mice and Replicates in Central Nervous System. Viruses 2018; 10:v10010049. [PMID: 29361773 PMCID: PMC5795462 DOI: 10.3390/v10010049] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 01/17/2018] [Accepted: 01/18/2018] [Indexed: 12/30/2022] Open
Abstract
Zika virus (ZIKV) has been defined as a teratogenic pathogen behind the increased number of cases of microcephaly in French Polynesia, Brazil, Puerto Rico, and other South American countries. Experimental studies using animal models have achieved tremendous insight into understanding the viral pathogenesis, transmission, teratogenic mechanisms, and virus-host interactions. However, the animals used in published investigations are mostly interferon (IFN)-compromised, either genetically or via antibody treatment. Herein, we studied ZIKV infection in IFN-competent mice using African (MR766) and Asian strains (PRVABC59 and SZ-WIV01). After testing four different species of mice, we found that BALB/c neonatal mice were resistant to ZIKV infection, that Kunming, ICR and C57BL/6 neonatal mice were fatally susceptible to ZIKV infection, and that the fatality of C57BL/6 neonates from 1 to 3 days old were in a viral dose-dependent manner. The size and weight of the brain were significantly reduced, and the ZIKV-infected mice showed neuronal symptoms such as hind-limb paralysis, tremor, and poor balance during walking. Pathologic and immunofluorescent experiments revealed that ZIKV infected different areas of the central nervous system (CNS) including gray matter, hippocampus, cerebral cortex, and spinal cord, but not olfactory bulb. Interestingly, ZIKV replicated in multiple organs and resulted in pathogenesis in liver and testis, implying that ZIKV infection may engender a high health risk in neonates by postnatal infection. In summary, we investigated ZIKV pathogenesis using an animal model that is not IFN-compromised.
Collapse
|
39
|
Winkler CW, Peterson KE. Using immunocompromised mice to identify mechanisms of Zika virus transmission and pathogenesis. Immunology 2018; 153:443-454. [PMID: 29266213 DOI: 10.1111/imm.12883] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 12/13/2017] [Accepted: 12/14/2017] [Indexed: 12/11/2022] Open
Abstract
Zika virus (ZIKV) is responsible for a recent global epidemic that has been associated with congenital brain malformations in fetuses and with Guillain-Barré syndrome in adults. Within the last 2 years, a major effort has been made to develop murine models to study the mechanism of viral transmission, pathogenesis and the host immune response. Here, we discuss the findings from these models regarding the role that the innate and adaptive immune responses have in controlling ZIKV infection and pathogenesis. Additionally, we examine how innate and adaptive immune responses influence sexual and vertical transmission of ZIKV infection as well as how these responses can influence the ability of ZIKV to cross the placenta and to induce damage in the developing brain.
Collapse
Affiliation(s)
- Clayton W Winkler
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT, USA
| | - Karin E Peterson
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, MT, USA
| |
Collapse
|
40
|
Husstedt IW, Maschke M, Eggers C, Neuen-Jacob E, Arendt G. [Zika virus infection and the nervous system]. DER NERVENARZT 2018; 89:136-143. [PMID: 29318332 DOI: 10.1007/s00115-017-0472-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Zika virus is an arbovirus from the family of flaviviruses, which is transmitted by the mosquito Aedes aegyptii and also by the Asian mosquito Aedes albopticus. The largest observed Zika virus epidemic is currently taking place in North and South America, in the Caribbean, southern USA and Southeast Asia. In most cases the infection is an unspecific, acute, febrile disease. Neurological manifestations consist mainly of microcephaly in newborns and Guillain-Barré syndrome but other rare manifestations have also become known in the meantime, such as meningoencephalitis and myelitis. Therefore, the Zika virus, similar to other flaviviruses, has neuropathogenic properties. In particular, the drastic increase in microcephaly cases in Brazil has induced great research activities. The virus is transmitted perinatally and can be detected in the amniotic fluid, placenta and brain tissue of the newborn. Vaccination or a causal therapy does not yet exist. The significant increase in Guillain-Barré syndrome induced by the Zika virus was observed during earlier outbreaks. In the meantime, scientifically clear connections between a Zika virus infection and these neurological manifestations have been shown. Long-term studies and animal models should be used for a better understanding of the pathomechanisms of this disease.
Collapse
Affiliation(s)
- I W Husstedt
- Klinik Maria Frieden und Medizinische Fakultät, Westfälische Wilhelms-Universität Münster, Am Krankenhaus 1, 48291, Telgte, Deutschland.
| | - M Maschke
- Klinik für Neurologie, Krankenhaus der Barmherzigen Brüder, Trier, Deutschland
| | - C Eggers
- Krankenhaus der Barmherzigen Brüder, Linz, Österreich
| | - E Neuen-Jacob
- Institut für Neuropathologie, Universitätsklinikum Düsseldorf, Düsseldorf, Deutschland
| | - G Arendt
- Klinik für Neurologie, Universitätsklinikum Düsseldorf, Düsseldorf, Deutschland
| |
Collapse
|
41
|
Liu J, Li Q, Li X, Qiu Z, Li A, Liang W, Chen H, Cai X, Chen X, Duan X, Li J, Wu W, Xu M, Mao Y, Chen H, Li J, Gu W, Li H. Zika Virus Envelope Protein induces G2/M Cell Cycle Arrest and Apoptosis via an Intrinsic Cell Death Signaling Pathway in Neuroendocrine PC12 Cells. Int J Biol Sci 2018; 14:1099-1108. [PMID: 29989100 PMCID: PMC6036729 DOI: 10.7150/ijbs.26400] [Citation(s) in RCA: 30] [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: 03/31/2018] [Accepted: 05/13/2018] [Indexed: 11/29/2022] Open
Abstract
Clinical evidence suggests that there exists a strong correlation between Zika virus (ZIKV) infection and abnormal development of the nervous system. However, the underlying mechanisms remain to be elusive. In this study, recombinant lentiviral vectors coding for ZIKV structural proteins and truncations (prM-Env, M-Env and Env) were transduced into PC12 cells. Envelope (Env) overexpression induced significant inhibition of proliferation and triggered G2/M cell cycle arrest and apoptosis in PC12 cells. Flow cytometry and western blot analysis showed that the apoptosis was associated with up-regulation of both p53 and p21Cip1/Waf1 and down-regulation of cyclin B1. Presence of aberrant nuclei clusters were confirmed by immunofluorescence staining analysis. The data indicate that overexpression of prM-Env, M-Env or Env led to apoptosis via an intrinsic cell death signaling pathway that is dependent on the activation of caspase-9 and caspase-3 and accompanied by an increased ratio of Bax to Bcl-2 in transduced PC12 cells. In summary, our results suggest that ZIKV Env protein causes apoptosis in PC12 cells via an intrinsic cell death signaling pathway, which may contribute to ZIKV-induced abnormal development of the nervous system.
Collapse
Affiliation(s)
- Jun Liu
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Institute of Comparative Medicine and Center of Laboratory Animals, Southern Medical University, Guangzhou, China
- Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Qingqing Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Xiaoxin Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Zhenzhen Qiu
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Guangzhou Bioneeds Biotechnology Co., Ltd, Guangzhou, China
| | - Andrew Li
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, USA
| | - Wenhan Liang
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Huangyao Chen
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Xiangsheng Cai
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Clinical Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Xinglu Chen
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Clinical Laboratory, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Xinyue Duan
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Jingjing Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Wangsheng Wu
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- Animal Science and Technology College, Jilin Agricultural University, Changchun, China
| | - Mingyu Xu
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yingying Mao
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Huiying Chen
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Jinlong Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Weiwang Gu
- Institute of Comparative Medicine and Center of Laboratory Animals, Southern Medical University, Guangzhou, China
- Pearl Lab Animal Science and Technology Co. Ltd, Dongguang, China
- School of Chemical and Environmental Engineering, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute, Jiangmen, China
- ✉ Corresponding authors: Weiwang Gu, MS, Institute of Comparative Medicine and Center of Laboratory Animals, Southern Medical University, 1023 South Shatai Road, Guangzhou, Guangdong 510515, China. Phone: 86-20-61648043; Fax: 86-20-61648043; E-mail: ; Hongwei Li, Ph.D., School of Laboratory Medicine and Biotechnology, Southern Medical University, 1023 South Shatai Road, Guangzhou, Guangdong 510515, China. Phone: 86-20-61648555; Fax: 86-20-61648555; E-mail:
| | - Hongwei Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
- ✉ Corresponding authors: Weiwang Gu, MS, Institute of Comparative Medicine and Center of Laboratory Animals, Southern Medical University, 1023 South Shatai Road, Guangzhou, Guangdong 510515, China. Phone: 86-20-61648043; Fax: 86-20-61648043; E-mail: ; Hongwei Li, Ph.D., School of Laboratory Medicine and Biotechnology, Southern Medical University, 1023 South Shatai Road, Guangzhou, Guangdong 510515, China. Phone: 86-20-61648555; Fax: 86-20-61648555; E-mail:
| |
Collapse
|
42
|
Krause KK, Azouz F, Shin OS, Kumar M. Understanding the Pathogenesis of Zika Virus Infection Using Animal Models. Immune Netw 2017; 17:287-297. [PMID: 29093650 PMCID: PMC5662778 DOI: 10.4110/in.2017.17.5.287] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 10/06/2017] [Accepted: 10/09/2017] [Indexed: 12/20/2022] Open
Abstract
Zika virus (ZIKV) is a member of Flaviviridae family that has emerged as a pathogen of significant public health importance. The rapid expansion of ZIKV in the South and Central America has recently gained medical attention emphasizing the capacity of ZIKV to spread to non-endemic regions. ZIKV infection during pregnancy has been demonstrated to cause microcephaly and other fetal developmental abnormalities. An increased incidence of Guillain-Barre syndrome, an immune mediated neuropathy of the peripheral nervous system, has also been reported in ZIKV-infected patients in French Polynesia and Brazil. No effective therapies currently exist for treating patients infected with ZIKV. Despite the relatively short time interval, an intensive effort by the global scientific community has resulted in development of animal models to study multiple aspects of ZIKV biology. Several animal models have been established to investigate pathogenesis of ZIKV in adults, pregnant mothers, and developing fetuses. Here we review the remarkable progress of newly developed small and large animal models for understanding ZIKV pathogenesis.
Collapse
Affiliation(s)
- Keeton K Krause
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, Pacific Center for Emerging Infectious Diseases Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA
| | - Francine Azouz
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, Pacific Center for Emerging Infectious Diseases Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA
| | - Ok Sarah Shin
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Korea
| | - Mukesh Kumar
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, Pacific Center for Emerging Infectious Diseases Research, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA
| |
Collapse
|
43
|
Ferreira AC, Zaverucha-do-Valle C, Reis PA, Barbosa-Lima G, Vieira YR, Mattos M, Silva PDP, Sacramento C, de Castro Faria Neto HC, Campanati L, Tanuri A, Brüning K, Bozza FA, Bozza PT, Souza TML. Sofosbuvir protects Zika virus-infected mice from mortality, preventing short- and long-term sequelae. Sci Rep 2017; 7:9409. [PMID: 28842610 PMCID: PMC5573375 DOI: 10.1038/s41598-017-09797-8] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 07/28/2017] [Indexed: 01/04/2023] Open
Abstract
Zika virus (ZIKV) causes significant public health concerns because of its association with congenital malformations, neurological disorders in adults, and, more recently, death. Considering the necessity to mitigate ZIKV-associated diseases, antiviral interventions are an urgent necessity. Sofosbuvir, a drug in clinical use against hepatitis C virus (HCV), is among the FDA-approved substances endowed with anti-ZIKV activity. In this work, we further investigated the in vivo activity of sofosbuvir against ZIKV. Neonatal Swiss mice were infected with ZIKV (2 × 107 PFU) and treated with sofosbuvir at 20 mg/kg/day, a concentration compatible with pre-clinical development of this drug. We found that sofosbuvir reduced acute levels of ZIKV from 60 to 90% in different anatomical compartments, such as the blood plasma, spleen, kidney, and brain. Early treatment with sofosbuvir doubled the percentage and time of survival of ZIKV-infected animals. Sofosbuvir also prevented the acute neuromotor impairment triggered by ZIKV. In the long-term behavioural analysis of ZIKV-associated sequelae, sofosbuvir prevented loss of hippocampal- and amygdala-dependent memory. Our results indicate that sofosbuvir inhibits ZIKV replication in vivo, which is consistent with the prospective necessity of antiviral drugs to treat ZIKV-infected individuals.
Collapse
Affiliation(s)
- André C Ferreira
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Camila Zaverucha-do-Valle
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- Instituto Nacional de Infectologia (INI), Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Patrícia A Reis
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | | | | | - Mayara Mattos
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Priscila de Paiva Silva
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Carolina Sacramento
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Hugo C de Castro Faria Neto
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Loraine Campanati
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Amilcar Tanuri
- Instituto de Biologia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Karin Brüning
- BMK Consortium: Blanver Farmoquímica Ltda; Microbiológica Química e Farmacêutica Ltda, Karin Bruning & Cia. Ltda, Taboão da Serra, SP, Brazil
| | - Fernando A Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
- Instituto Nacional de Infectologia (INI), Fiocruz, Rio de Janeiro, RJ, Brazil
| | - Patrícia T Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil
| | - Thiago Moreno L Souza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, RJ, Brazil.
- Instituto Nacional de Infectologia (INI), Fiocruz, Rio de Janeiro, RJ, Brazil.
- National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Fiocruz, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
44
|
Saiz JC, Martín-Acebes MA, Bueno-Marí R, Salomón OD, Villamil-Jiménez LC, Heukelbach J, Alencar CH, Armstrong PK, Ortiga-Carvalho TM, Mendez-Otero R, Rosado-de-Castro PH, Pimentel-Coelho PM. Zika Virus: What Have We Learnt Since the Start of the Recent Epidemic? Front Microbiol 2017; 8:1554. [PMID: 28878742 PMCID: PMC5572254 DOI: 10.3389/fmicb.2017.01554] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 07/31/2017] [Indexed: 01/03/2023] Open
Abstract
Zika is a viral disease transmitted mainly by mosquitoes of the genus Aedes. In recent years, it has expanded geographically, changing from an endemic mosquito-borne disease across equatorial Asia and Africa, to an epidemic disease causing large outbreaks in several areas of the world. With the recent Zika virus (ZIKV) outbreaks in the Americas, the disease has become a focus of attention of public health agencies and of the international research community, especially due to an association with neurological disorders in adults and to the severe neurological and ophthalmological abnormalities found in fetuses and newborns of mothers exposed to ZIKV during pregnancy. A large number of studies have been published in the last 3 years, revealing the structure of the virus, how it is transmitted and how it affects human cells. Many different animal models have been developed, which recapitulate several features of ZIKV disease and its neurological consequences. Moreover, several vaccine candidates are now in active preclinical development, and three of them have already entered phase I clinical trials. Likewise, many different compounds targeting viral and cellular components are being tested in in vitro and in experimental animal models. This review aims to discuss the current state of this rapidly growing literature from a multidisciplinary perspective, as well as to present an overview of the public health response to Zika and of the perspectives for the prevention and treatment of this disease.
Collapse
Affiliation(s)
- Juan-Carlos Saiz
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y AlimentariaMadrid, Spain
| | - Miguel A. Martín-Acebes
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y AlimentariaMadrid, Spain
| | - Rubén Bueno-Marí
- Departamento de Investigación y Desarrollo (I+D), Laboratorios LokímicaValencia, Spain
| | | | | | - Jorg Heukelbach
- Department of Community Health, School of Medicine, Federal University of CearáFortaleza, Brazil
- College of Public Health, Medical and Veterinary Sciences, Division of Tropical Health and Medicine, James Cook University, TownsvilleQLD, Australia
| | - Carlos H. Alencar
- Department of Community Health, School of Medicine, Federal University of CearáFortaleza, Brazil
| | - Paul K. Armstrong
- Communicable Disease Control Directorate, Western Australia Department of Health, PerthWA, Australia
| | - Tania M. Ortiga-Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Rosalia Mendez-Otero
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| | - Paulo H. Rosado-de-Castro
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
- Instituto D’Or de Pesquisa e EnsinoRio de Janeiro, Brazil
| | - Pedro M. Pimentel-Coelho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de JaneiroRio de Janeiro, Brazil
| |
Collapse
|
45
|
Cumberworth SL, Barrie JA, Cunningham ME, de Figueiredo DPG, Schultz V, Wilder-Smith AJ, Brennan B, Pena LJ, Freitas de Oliveira França R, Linington C, Barnett SC, Willison HJ, Kohl A, Edgar JM. Zika virus tropism and interactions in myelinating neural cell cultures: CNS cells and myelin are preferentially affected. Acta Neuropathol Commun 2017; 5:50. [PMID: 28645311 PMCID: PMC5481922 DOI: 10.1186/s40478-017-0450-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 06/02/2017] [Indexed: 12/02/2022] Open
Abstract
The recent global outbreak of Zika virus (ZIKV) infection has been linked to severe neurological disorders affecting the peripheral and central nervous systems (PNS and CNS, respectively). The pathobiology underlying these diverse clinical phenotypes are the subject of intense research; however, even the principal neural cell types vulnerable to productive Zika infection remain poorly characterised. Here we used CNS and PNS myelinating cultures from wild type and Ifnar1 knockout mice to examine neuronal and glial tropism and short-term consequences of direct infection with a Brazilian variant of ZIKV. Cell cultures were infected pre- or post-myelination for various intervals, then stained with cell-type and ZIKV-specific antibodies. In bypassing systemic immunity using ex vivo culture, and the type I interferon response in Ifnar1 deficient cells, we were able to evaluate the intrinsic infectivity of neural cells. Through systematic quantification of ZIKV infected cells in myelinating cultures, we found that ZIKV infection is enhanced in the absence of the type I interferon responses and that CNS cells are considerably more susceptible to infection than PNS cells. In particular, we demonstrate that CNS axons and myelinating oligodendrocytes are especially vulnerable to injury. These results have implications for understanding the pathobiology of neurological symptoms associated with ZIKV infection. Furthermore, we provide a quantifiable ex vivo infection model that can be used for fundamental and therapeutic studies on viral neuroinvasion and its consequences.
Collapse
Affiliation(s)
| | - Jennifer A Barrie
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, G12 8TA, Glasgow, Scotland, UK
| | - Madeleine E Cunningham
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, G12 8TA, Glasgow, Scotland, UK
| | - Daniely Paulino Gomes de Figueiredo
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, G12 8TA, Glasgow, Scotland, UK
| | - Verena Schultz
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, G12 8TA, Glasgow, Scotland, UK
| | - Adrian J Wilder-Smith
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, G12 8TA, Glasgow, Scotland, UK
| | - Benjamin Brennan
- MRC-University of Glasgow Centre for Virus Research, G61 1QH, Glasgow, Scotland, UK
| | - Lindomar J Pena
- Oswaldo Cruz Foundation/Aggeu Magalhães Institute, Department of Virology, UFPE Campus-Cidade Universitária, Recife/PE, Brazil
| | | | - Christopher Linington
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, G12 8TA, Glasgow, Scotland, UK
| | - Susan C Barnett
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, G12 8TA, Glasgow, Scotland, UK
| | - Hugh J Willison
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, G12 8TA, Glasgow, Scotland, UK
| | - Alain Kohl
- MRC-University of Glasgow Centre for Virus Research, G61 1QH, Glasgow, Scotland, UK.
| | - Julia M Edgar
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, G12 8TA, Glasgow, Scotland, UK.
- Department of Neurogenetics, Max Planck Institute for Experimental Medicine, Hermann-Rein-Strasse 3, 37075, Goettingen, Germany.
| |
Collapse
|
46
|
Abstract
Zika virus (ZIKV) is an emerging mosquito-transmitted flavivirus that now causes epidemics affecting millions of people on multiple continents. The virus has received global attention because of some of its unusual epidemiological and clinical features, including persistent infection in the male reproductive tract and sexual transmission, an ability to cross the placenta during pregnancy and infect the developing fetus to cause congenital malformations, and its association with Guillain-Barré syndrome in adults. This past year has witnessed an intensive effort by the global scientific community to understand the biology of ZIKV and to develop pathogenesis models for the rapid testing of possible countermeasures. Here, we review the recent advances in and utility and limitations of newly developed mouse and nonhuman primate models of ZIKV infection and pathogenesis.
Collapse
Affiliation(s)
- Thomas E Morrison
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Michael S Diamond
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|