1
|
Tisoncik-Go J, Lewis TB, Whitmore LS, Voss K, Niemeyer S, Dai J, Kim P, Hubbell K, Iwayama N, Ahrens C, Wangari S, Murnane R, Edlefsen PT, Guerriero KA, Gale M, Fuller DH, O’Connor MA. Persistent innate immune dysfunction and ZIKV replication in the gastrointestinal tract during SIV infection in pigtail macaques. Front Immunol 2025; 16:1535807. [PMID: 40103823 PMCID: PMC11913663 DOI: 10.3389/fimmu.2025.1535807] [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: 11/27/2024] [Accepted: 02/03/2025] [Indexed: 03/20/2025] Open
Abstract
Mosquito-borne flaviviruses, including dengue (DENV) and Zika (ZIKV) viruses, have caused widespread epidemics in areas with high HIV prevalence, partly due to the expanded geographic range of arthropod vectors. Despite the occurrence of large flavivirus outbreaks in areas with high HIV prevalence, little is known about the effects of flavivirus infection in people living with HIV (PLWH). Here, we use a pigtail macaque model of HIV/AIDS to investigate the impact of simian immunodeficiency virus (SIV)-induced immunosuppression on ZIKV replication and pathogenesis. During acute SIV infection, peripheral ZIKV cellular targets expanded and innate immune activation increased. In vitro, peripheral blood mononuclear cells (PBMC) from SIV infected pigtail macaques were less permissive to ZIKV infection. In vivo, ZIKV viremia was delayed and ZIKV was more persistent in the gastrointestinal tissues of SIV-ZIKV co-infected animals. This persistence was associated with changes in innate cellular (monocytes, neutrophils) recruitment to the blood and tissues, reduced anti-ZIKV immunity, and sustained expression of peripheral inflammatory and innate immune genes. Collectively, these findings uniquely suggest that untreated SIV infection may promote inflammatory cellular innate responses and create a state of persistent immune activation that contributes to prolonged ZIKV viremia and persistence in the gastrointestinal tract. Furthermore, these results suggest that PLWH and other immunocompromised individuals could be at higher risk for prolonged ZIKV infection, potentially extending the window of ZIKV transmission. These insights highlight the importance of including PLWH in strategies for deploying vaccines and treatments against ZIKV.
Collapse
Affiliation(s)
- Jennifer Tisoncik-Go
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
- Department of Immunology, University of Washington, Seattle, WA, United States
- Center for Innate Immunity and Immune Disease (CIIID), University of Washington, Seattle, WA, United States
| | - Thomas B. Lewis
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
- Department of Microbiology, University of Washington, Seattle, WA, United States
| | - Leanne S. Whitmore
- Department of Immunology, University of Washington, Seattle, WA, United States
| | - Kathleen Voss
- Department of Immunology, University of Washington, Seattle, WA, United States
| | - Skyler Niemeyer
- Department of Microbiology, University of Washington, Seattle, WA, United States
| | - Jin Dai
- Department of Immunology, University of Washington, Seattle, WA, United States
| | - Paul Kim
- Department of Microbiology, University of Washington, Seattle, WA, United States
| | - Kai Hubbell
- Department of Microbiology, University of Washington, Seattle, WA, United States
| | - Naoto Iwayama
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Chul Ahrens
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Solomon Wangari
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Robert Murnane
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Paul T. Edlefsen
- Biostatistics Bioinformatics and Epidemiology (BBE), Program of the Vaccine and Infectious Disease (ViDD) Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Kathryn A. Guerriero
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Michael Gale
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
- Department of Immunology, University of Washington, Seattle, WA, United States
- Center for Innate Immunity and Immune Disease (CIIID), University of Washington, Seattle, WA, United States
- Department of Microbiology and Immunology, Institute on Infectious Diseases, University of Minnesota, Minneapolis, MN, United States
| | - Deborah H. Fuller
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
- Department of Microbiology, University of Washington, Seattle, WA, United States
| | - Megan A. O’Connor
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
- Department of Microbiology, University of Washington, Seattle, WA, United States
| |
Collapse
|
2
|
Tisoncik-Go J, Lewis TB, Whitmore LS, Voss K, Niemeyer S, Dai J, Kim P, Hubbell K, Iwayama N, Ahrens C, Wangari S, Murnane R, Edlefsen PT, Guerriero KA, Gale M, Fuller DH, O'Connor MA. Chronic innate immune impairment and ZIKV persistence in the gastrointestinal tract during SIV infection in pigtail macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.23.609309. [PMID: 39229223 PMCID: PMC11370579 DOI: 10.1101/2024.08.23.609309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Mosquito borne flaviviruses, including dengue (DENV) and Zika (ZIKV) viruses, have caused global epidemics in areas with high HIV prevalence due to the expanded geographic range of arthropod vectors. Despite the occurrence of large flavivirus outbreaks in countries with high HIV prevalence, there is little knowledge regarding the effects of flavivirus infection in people living with HIV (PLWH). Here, we use a pigtail macaque model of HIV/AIDS to investigate the impact of simian immunodeficiency virus (SIV)-induced immunosuppression on ZIKV replication and pathogenesis. Early acute SIV infection induced expansion of peripheral ZIKV cellular targets and increased innate immune activation and peripheral blood mononuclear cells (PBMC) from SIV infected macaques were less permissive to ZIKV infection in vitro. In SIV-ZIKV co-infected animals, we found increased persistence of ZIKV in the periphery and tissues corresponding to alterations in innate cellular (monocytes, neutrophils) recruitment to the blood and tissues, decreased anti-ZIKV immunity, and chronic peripheral inflammatory and innate immune gene expression. Collectively, these findings suggest that untreated SIV infection may impair cellular innate responses and create an environment of chronic immune activation that promotes prolonged ZIKV viremia and persistence in the gastrointestinal tract. These results suggest that PLWH or other immunocompromised individuals could be at a higher risk for chronic ZIKV replication, which in turn could increase the timeframe of ZIKV transmission. Thus, PLWH are important populations to target during the deployment of vaccine and treatment strategies against ZIKV.
Collapse
Affiliation(s)
- Jennifer Tisoncik-Go
- Washington National Primate Research Center, University of Washington (Seattle, Washington)
- Department of Immunology, University of Washington (Seattle, Washington)
- Center for Innate Immunity and Immune Disease (CIIID), University of Washington (Seattle, Washington)
| | - Thomas B Lewis
- Washington National Primate Research Center, University of Washington (Seattle, Washington)
- Department of Microbiology, University of Washington (Seattle, Washington)
| | - Leanne S Whitmore
- Department of Immunology, University of Washington (Seattle, Washington)
| | - Kathleen Voss
- Washington National Primate Research Center, University of Washington (Seattle, Washington)
- Department of Immunology, University of Washington (Seattle, Washington)
| | - Skyler Niemeyer
- Department of Microbiology, University of Washington (Seattle, Washington)
| | - Jin Dai
- Department of Immunology, University of Washington (Seattle, Washington)
| | - Paul Kim
- Department of Microbiology, University of Washington (Seattle, Washington)
| | - Kai Hubbell
- Department of Microbiology, University of Washington (Seattle, Washington)
| | - Naoto Iwayama
- Washington National Primate Research Center, University of Washington (Seattle, Washington)
| | - Chul Ahrens
- Washington National Primate Research Center, University of Washington (Seattle, Washington)
| | - Solomon Wangari
- Washington National Primate Research Center, University of Washington (Seattle, Washington)
| | - Robert Murnane
- Washington National Primate Research Center, University of Washington (Seattle, Washington)
| | | | - Kathryn A Guerriero
- Washington National Primate Research Center, University of Washington (Seattle, Washington)
| | - Michael Gale
- Washington National Primate Research Center, University of Washington (Seattle, Washington)
- Department of Immunology, University of Washington (Seattle, Washington)
- Center for Innate Immunity and Immune Disease (CIIID), University of Washington (Seattle, Washington)
- Department of Global Health, University of Washington (Seattle, Washington)
| | - Deborah H Fuller
- Washington National Primate Research Center, University of Washington (Seattle, Washington)
- Department of Microbiology, University of Washington (Seattle, Washington)
| | - Megan A O'Connor
- Washington National Primate Research Center, University of Washington (Seattle, Washington)
- Department of Microbiology, University of Washington (Seattle, Washington)
| |
Collapse
|
3
|
Azamor T, Cunha DP, Nobre Pires KS, Lira Tanabe EL, Melgaço JG, Vieira da Silva AM, Ribeiro-Alves M, Calvo TL, Tubarão LN, da Silva J, Fernandes CB, Fonseca de Souza A, Torrentes de Carvalho A, Avvad-Portari E, da Cunha Guida L, Gomes L, Lopes Moreira ME, Dinis Ano Bom AP, Cristina da Costa Neves P, Missailidis S, Vasconcelos Z, Borbely AU, Moraes MO. Decidual production of interferon lambda in response to ZIKV persistence: Clinical evidence and in vitro modelling. Heliyon 2024; 10:e30613. [PMID: 38737240 PMCID: PMC11087979 DOI: 10.1016/j.heliyon.2024.e30613] [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: 12/20/2023] [Revised: 04/30/2024] [Accepted: 04/30/2024] [Indexed: 05/14/2024] Open
Abstract
Zika virus (ZIKV) infections during pregnancy can result in Congenital Zika Syndrome (CZS), a range of severe neurological outcomes in fetuses that primarily occur during early gestational stages possibly due to placental damage. Although some placentas can maintain ZIKV persistence for weeks or months after the initial infection and diagnosis, the impact of this viral persistence is still unknown. Here, we aimed to investigate the immunological repercussion of ZIKV persistence in term placentas. As such, term placentas from 64 pregnant women diagnosed with Zika in different gestational periods were analyzed by ZIKV RT-qPCR, examination of decidua and placental villous histopathology, and expression of inflammation-related genes and IFNL1-4. Subsequently, we explored primary cultures of term decidual Extravillous Trophoblasts (EVTs) and Term Chorionic Villi (TCV) explants, as in vitro models to access the immunological consequences of placental ZIKV infection. Placenta from CZS cases presented low IFNL1-4 expression, evidencing the critical protective role of theses cytokines in the clinical outcome. Term placentas cleared for ZIKV showed increased levels of IFNL1, 3, and 4, whether viral persistence was related with a proinflammatory profile. Conversely, upon ZIKV persistence placentas with decidual inflammation showed high IFNL1-4 levels. In vitro experiments showed that term EVTs are more permissive, and secreted higher levels of IFN-α2 and IFN-λ1 compared to TCV explants. The results suggest that, upon ZIKV persistence, the maternal-skewed decidua contributes to placental inflammatory and antiviral signature, through chronic deciduitis and IFNL upregulation. Although further studies are needed to elucidate the mechanisms underlying the decidual responses against ZIKV. Hence, this study presents unique insights and valuable in vitro models for evaluating the immunological landscape of placentas upon ZIKV persistence.
Collapse
|
4
|
Hale GL. Flaviviruses and the Traveler: Around the World and to Your Stage. A Review of West Nile, Yellow Fever, Dengue, and Zika Viruses for the Practicing Pathologist. Mod Pathol 2023; 36:100188. [PMID: 37059228 DOI: 10.1016/j.modpat.2023.100188] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/02/2023] [Accepted: 04/05/2023] [Indexed: 04/16/2023]
Abstract
Flaviviruses are a genus of single-stranded RNA viruses that impose an important and growing burden to human health. There are over 3 billion individuals living in areas where flaviviruses are endemic. Flaviviruses and their arthropod vectors (which include mosquitoes and ticks) take advantage of global travel to expand their distribution and cause severe disease in humans, and they can be grouped according to their vector and pathogenicity. The mosquito-borne flaviviruses cause a spectrum of diseases from encephalitis to hepatitis and vascular shock syndrome, congenital abnormalities, and fetal death. Neurotropic infections such as Zika virus and West Nile virus cross the blood-brain barrier and infect neurons and other cells, leading to meningoencephalitis. In the hemorrhagic fever clade, there are yellow fever virus, the prototypical hemorrhagic fever virus that infects hepatocytes, and dengue virus, which infects cells of the reticuloendothelial system and can lead to a dramatic plasma cell leakage and shock syndrome. Zika virus also causes congenital infections and fetal death and is the first and only example of a teratogenic arbovirus in humans. Diagnostic testing for flaviviruses broadly includes the detection of viral RNA in serum (particularly within the first 10 days of symptoms), viral isolation by cell culture (rarely performed due to complexity and biosafety concerns), and histopathologic evaluation with immunohistochemistry and molecular testing on formalin-fixed paraffin-embedded tissue blocks. This review focuses on 4 mosquito-borne flaviviruses-West Nile, yellow fever, dengue, and Zika virus-and discusses the mechanisms of transmission, the role of travel in geographic distribution and epidemic emergence, and the clinical and histopathologic features of each. Finally, prevention strategies such as vector control and vaccination are discussed.
Collapse
Affiliation(s)
- Gillian L Hale
- Department of Pathology, University of Utah, Salt Lake City, Utah.
| |
Collapse
|
5
|
Gouveia de Melo ACM, van der Linden V, Serpa SC, Rolim Filho EL, Lins OG. Electromyography in Congenital Zika Syndrome. J Clin Neurophysiol 2023; 40:350-354. [PMID: 34510092 DOI: 10.1097/wnp.0000000000000893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
INTRODUCTION Congenital Zika virus syndrome is a distinct pattern of birth defects in fetuses infected by the Zika virus. It presents a broad clinical spectrum that includes occurrences of microcephaly, hypertonia, dysphagia, hyperexcitability, seizures, and arthrogryposis. Imaging findings show neuronal migration disorders. METHODOLOGY Case reports have suggested that arthrogryposis has a neurogenic cause. We analyzed needle electromyography and nerve conduction examinations on 77 patients aged 2-24 months presenting highly probable congenital Zika virus syndrome, with or without arthrogryposis. RESULTS All those with arthrogryposis presented with chronic muscle denervation in the electromyography examination. Similarly, children with single or reversible joint abnormalities at birth showed the same findings. Denervation in the paravertebral musculature was found in all of the children with diaphragmatic paralysis or thoracic deformities. CONCLUSIONS We propose that congenital contractures associated with congenital Zika virus syndrome are caused by the malformation of upper and lower motor neurons during embryogenesis.
Collapse
Affiliation(s)
| | | | - Sarah C Serpa
- Pediatric Neurology Service, Hospital Jorge de Medeiros, Recife, Brazil
| | - Epitácio L Rolim Filho
- Pediatric Orthopedics Service, Association for Assistance of Children with Deficiencies (AACD), Recife, Brazil
- Department of Orthopedics, Medical School, Federal University of Pernambuco, Recife, Brazil; and
| | - Otávio G Lins
- Department of Neurology, Medical School, Federal University of Pernambuco, Recife, Brazil
| |
Collapse
|
6
|
Matuck B, Ferraz da Silva LF, Warner BM, Byrd KM. The need for integrated research autopsies in the era of precision oral medicine. J Am Dent Assoc 2023; 154:194-205. [PMID: 36710158 PMCID: PMC9974796 DOI: 10.1016/j.adaj.2022.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/18/2022] [Accepted: 11/20/2022] [Indexed: 01/29/2023]
Abstract
BACKGROUND Autopsy has benefited the practice of medicine for centuries; however, its use to advance the practice of oral health care is relatively limited. In the era of precision oral medicine, the research autopsy is poised to play an important role in understanding oral-systemic health, including infectious disease, autoimmunity, craniofacial genetics, and cancer. TYPES OF STUDIES REVIEWED The authors reviewed relevant articles that used medical and dental research autopsies to summarize the advantages of minimally invasive autopsies of dental, oral, and craniofacial tissues and to outline practices for supporting research autopsies of the oral and craniofacial complex. RESULTS The authors provide a historical summary of research autopsy in dentistry and provide a perspective on the value of autopsies for high-resolution multiomic studies to benefit precision oral medicine. As the promise of high-resolution multiomics is being realized, there is a need to integrate the oral and craniofacial complex into the practice of autopsy in medicine. Furthermore, the collaboration of autopsy centers with researchers will accelerate the understanding of dental, oral, and craniofacial tissues as part of the whole body. CONCLUSIONS Autopsies must integrate oral and craniofacial tissues as part of biobanking procedures. As new technologies allow for high-resolution, multimodal phenotyping of human samples, using optimized sampling procedures will allow for unprecedented understanding of common and rare dental, oral, and craniofacial diseases in the future. PRACTICAL IMPLICATIONS The COVID-19 pandemic highlighted the oral cavity as a site for viral infection and transmission potential; this was only discovered via clinical autopsies. The realization of the integrated autopsy's value in full body health initiatives will benefit patients across the globe.
Collapse
Affiliation(s)
- Bruno Matuck
- Department of Pathology, School of Medicine University of São Paulo, São Paulo, Brazil
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Blake M. Warner
- Salivary Disorders Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Kevin Matthew Byrd
- Salivary Disorders Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lab of Oral & Craniofacial Innovation (LOCI), Department of Innovation and Technology Research, ADA Science & Research Institute, Gaithersburg, MD, USA
| |
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
|
Azamor T, Cunha DP, da Silva AMV, Bezerra OCDL, Ribeiro-Alves M, Calvo TL, Kehdy FDSG, Manta FDN, Pinto TGDT, Ferreira LP, Portari EA, Guida LDC, Gomes L, Moreira MEL, de Carvalho EF, Cardoso CC, Muller M, Ano Bom APD, Neves PCDC, Vasconcelos Z, Moraes MO. Congenital Zika Syndrome Is Associated With Interferon Alfa Receptor 1. Front Immunol 2021; 12:764746. [PMID: 34899713 PMCID: PMC8657619 DOI: 10.3389/fimmu.2021.764746] [Citation(s) in RCA: 8] [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: 08/25/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
Host factors that influence Congenital Zika Syndrome (CZS) outcome remain elusive. Interferons have been reported as the main antiviral factor in Zika and other flavivirus infections. Here, we accessed samples from 153 pregnant women (77 without and 76 with CZS) and 143 newborns (77 without and 66 with CZS) exposed to ZIKV conducted a case-control study to verify whether interferon alfa receptor 1 (IFNAR1) and interferon lambda 2 and 4 (IFNL2/4) single nucleotide polymorphisms (SNPs) contribute to CZS outcome, and characterized placenta gene expression profile at term. Newborns carrying CG/CC genotypes of rs2257167 in IFNAR1 presented higher risk of developing CZS (OR=3.41; IC=1.35-8.60; Pcorrected=0.032). No association between IFNL SNPs and CZS was observed. Placenta from CZS cases displayed lower levels of IFNL2 and ISG15 along with higher IFIT5. The rs2257167 CG/CC placentas also demonstrated high levels of IFIT5 and inflammation-related genes. We found CZS to be related with exacerbated type I IFN and insufficient type III IFN in placenta at term, forming an unbalanced response modulated by the IFNAR1 rs2257167 genotype. Despite of the low sample size se findings shed light on the host-pathogen interaction focusing on the genetically regulated type I/type III IFN axis that could lead to better management of Zika and other TORCH (Toxoplasma, Others, Rubella, Cytomegalovirus, Herpes) congenital infections.
Collapse
Affiliation(s)
- Tamiris Azamor
- Laboratório de Hanseníase, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
- Vice-Diretoria de Desenvolvimento Tecnológico, Instituto de Tecnologia em Imunobiológicos, Fiocruz, Rio de Janeiro, Brazil
| | - Daniela Prado Cunha
- Unidade de Pesquisa Clínica, Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, Fiocruz, Rio de Janeiro, Brazil
| | - Andréa Marques Vieira da Silva
- Vice-Diretoria de Desenvolvimento Tecnológico, Instituto de Tecnologia em Imunobiológicos, Fiocruz, Rio de Janeiro, Brazil
| | | | - Marcelo Ribeiro-Alves
- Laboratório de Pesquisa Clínica em DST/AIDS, Instituto Nacional de Infectologia, Fiocruz, Rio de Janeiro, Brazil
| | - Thyago Leal Calvo
- Laboratório de Hanseníase, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | | | | | | | | | - Elyzabeth Avvad Portari
- Unidade de Pesquisa Clínica, Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, Fiocruz, Rio de Janeiro, Brazil
| | - Letícia da Cunha Guida
- Unidade de Pesquisa Clínica, Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, Fiocruz, Rio de Janeiro, Brazil
| | - Leonardo Gomes
- Unidade de Pesquisa Clínica, Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, Fiocruz, Rio de Janeiro, Brazil
| | - Maria Elisabeth Lopes Moreira
- Unidade de Pesquisa Clínica, Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, Fiocruz, Rio de Janeiro, Brazil
| | | | - Cynthia Chester Cardoso
- Laboratório de Virologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo Muller
- Vice-Diretoria de Desenvolvimento Tecnológico, Instituto de Tecnologia em Imunobiológicos, Fiocruz, Rio de Janeiro, Brazil
| | - Ana Paula Dinis Ano Bom
- Vice-Diretoria de Desenvolvimento Tecnológico, Instituto de Tecnologia em Imunobiológicos, Fiocruz, Rio de Janeiro, Brazil
| | | | - Zilton Vasconcelos
- Unidade de Pesquisa Clínica, Instituto Nacional de Saúde da Mulher, da Criança e do Adolescente Fernandes Figueira, Fiocruz, Rio de Janeiro, Brazil
| | - Milton Ozório Moraes
- Laboratório de Hanseníase, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| |
Collapse
|
9
|
Embryonic Stage of Congenital Zika Virus Infection Determines Fetal and Postnatal Outcomes in Mice. Viruses 2021; 13:v13091807. [PMID: 34578389 PMCID: PMC8473443 DOI: 10.3390/v13091807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/01/2021] [Accepted: 09/08/2021] [Indexed: 01/29/2023] Open
Abstract
Zika virus (ZIKV) infection during pregnancy causes a wide spectrum of congenital abnormalities and postnatal developmental sequelae such as fetal loss, intrauterine growth restriction (IUGR), microcephaly, or motor and neurodevelopmental disorders. Here, we investigated whether a mouse pregnancy model recapitulated a wide range of symptoms after congenital ZIKV infection, and whether the embryonic age of congenital infection changed the fetal or postnatal outcomes. Infection with ZIKV strain PRVABC59 from embryonic day 6.5 (E6.5) to E8.5, corresponding to the mid-first trimester in humans, caused fetal death, fetal resorption, or severe IUGR, whereas infection from E9.5 to E14.5, corresponding to the late-first to second trimester in humans, caused stillbirth, neonatal death, microcephaly, and postnatal growth deficiency. Furthermore, 4-week-old offspring born to dams infected at E12.5 showed abnormalities in neuropsychiatric state, motor behavior, autonomic function, or reflex and sensory function. Thus, our model recapitulated the multiple symptoms seen in human cases, and the embryonic age of congenital infection was one of the determinant factors of offspring outcomes in mice. Furthermore, maternal neutralizing antibodies protected the offspring from neonatal death after congenital infection at E9.5, suggesting that neonatal death in our model could serve as criteria for screening of vaccine candidates.
Collapse
|
10
|
Li M, Brokaw A, Furuta AM, Coler B, Obregon-Perko V, Chahroudi A, Wang HY, Permar SR, Hotchkiss CE, Golos TG, Rajagopal L, Adams Waldorf KM. Non-human Primate Models to Investigate Mechanisms of Infection-Associated Fetal and Pediatric Injury, Teratogenesis and Stillbirth. Front Genet 2021; 12:680342. [PMID: 34290739 PMCID: PMC8287178 DOI: 10.3389/fgene.2021.680342] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 05/25/2021] [Indexed: 12/25/2022] Open
Abstract
A wide array of pathogens has the potential to injure the fetus and induce teratogenesis, the process by which mutations in fetal somatic cells lead to congenital malformations. Rubella virus was the first infectious disease to be linked to congenital malformations due to an infection in pregnancy, which can include congenital cataracts, microcephaly, hearing impairment and congenital heart disease. Currently, human cytomegalovirus (HCMV) is the leading infectious cause of congenital malformations globally, affecting 1 in every 200 infants. However, our knowledge of teratogenic viruses and pathogens is far from complete. New emerging infectious diseases may induce teratogenesis, similar to Zika virus (ZIKV) that caused a global pandemic in 2016-2017; thousands of neonates were born with congenital microcephaly due to ZIKV exposure in utero, which also included a spectrum of injuries to the brain, eyes and spinal cord. In addition to congenital anomalies, permanent injury to fetal and neonatal organs, preterm birth, stillbirth and spontaneous abortion are known consequences of a broader group of infectious diseases including group B streptococcus (GBS), Listeria monocytogenes, Influenza A virus (IAV), and Human Immunodeficiency Virus (HIV). Animal models are crucial for determining the mechanism of how these various infectious diseases induce teratogenesis or organ injury, as well as testing novel therapeutics for fetal or neonatal protection. Other mammalian models differ in many respects from human pregnancy including placentation, labor physiology, reproductive tract anatomy, timeline of fetal development and reproductive toxicology. In contrast, non-human primates (NHP) most closely resemble human pregnancy and exhibit key similarities that make them ideal for research to discover the mechanisms of injury and for testing vaccines and therapeutics to prevent teratogenesis, fetal and neonatal injury and adverse pregnancy outcomes (e.g., stillbirth or spontaneous abortion). In this review, we emphasize key contributions of the NHP model pre-clinical research for ZIKV, HCMV, HIV, IAV, L. monocytogenes, Ureaplasma species, and GBS. This work represents the foundation for development and testing of preventative and therapeutic strategies to inhibit infectious injury of human fetuses and neonates.
Collapse
Affiliation(s)
- Miranda Li
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States
- Department of Biological Sciences, Columbia University, New York, NY, United States
| | - Alyssa Brokaw
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Anna M. Furuta
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Brahm Coler
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Veronica Obregon-Perko
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta and Emory University, Atlanta, GA, United States
| | - Hsuan-Yuan Wang
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Charlotte E. Hotchkiss
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Thaddeus G. Golos
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Lakshmi Rajagopal
- Department of Global Health, University of Washington, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Kristina M. Adams Waldorf
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
11
|
Zika Virus Induces an Atypical Tripartite Unfolded Protein Response with Sustained Sensor and Transient Effector Activation and a Blunted BiP Response. mSphere 2021; 6:e0036121. [PMID: 34106769 PMCID: PMC8265652 DOI: 10.1128/msphere.00361-21] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
To study how the Zika virus (ZIKV) interacts with the host unfolded protein response (UPR), we undertook a kinetics study. We show that ZIKV infection triggers an atypical tripartite UPR in A549 cells involving transient activation of the effectors X-box-binding protein 1, activating transcription factor 4 (ATF4), CCAAT enhancer-binding protein-homologous protein, and growth arrest and DNA damage-inducible protein 34 during early infection and sustained activation of all three UPR sensors: RNA-activated protein kinase-like endoplasmic reticulum-resident kinase (PERK), inositol-requiring kinase-1α (IRE1α), and ATF6. Sustained phosphorylation of the eukaryotic translation initiation factor 2α and rRNA degradation coincide with host translational shutoff, cell lysis, and virus release during late infection. We show a blunted response of the master negative regulator, the immunoglobulin heavy-chain-binding protein (BiP), by chemical UPR inducers, and we show that ZIKV suppresses BiP transcription and translation, suggesting that it may be necessary to blunt the BiP response to sustain UPR sensor activation. The PERK inhibitor GSK2606414 alone has no effects but synergizes with the ATF6 inhibitor Ceapin-A7 to inhibit early and late infection, whereas Ceapin-A7 alone inhibits late infection. Likewise, 4-phenylbutyric acid inhibits ZIKV replication by attenuating the PERK and ATF6 pathways and potentiating the IRE1α pathway, suggesting that ZIKV infection is differentially and temporally regulated by different UPR arms. ZIKV infection is inhibited by pretreatment of chemical UPR inducers but is refractory to the inhibitory activity of chemical inducers once infection has been established, suggesting that ZIKV has anti-UPR mechanisms that may be able to modulate and co-opt the UPR in its life cycle. IMPORTANCE The Zika virus originates from Africa and Asia but is emerging in other parts of the world. It usually causes an asymptomatic or mild, acute infection but can cause serious neurological complications, such as microcephaly and Guillain-Barré syndromes. Therefore, there is a pressing need for an antiviral. Viruses are obligative parasites and are dependent on the hosts for their propagation. As a result, we can target viruses by targeting host dependency. The host unfolded protein response is a cellular homeostatic response to stresses but can also be triggered by virus infections. We show here that Zika virus infection can cause stress and trigger the unfolded protein response. The Zika virus is able to manipulate, subvert, and co-opt the host unfolded protein response to aid its own replication. Understanding host dependency is important in the quest of a new class of antivirals called host-targeting agents.
Collapse
|
12
|
Nascimento-Carvalho GC, Nascimento-Carvalho EC, Ramos CL, Vilas-Boas AL, Moreno-Carvalho OA, Vinhaes CL, Barreto-Duarte B, Queiroz ATL, Andrade BB, Nascimento-Carvalho CM. Zika-exposed microcephalic neonates exhibit higher degree of inflammatory imbalance in cerebrospinal fluid. Sci Rep 2021; 11:8474. [PMID: 33875756 PMCID: PMC8055905 DOI: 10.1038/s41598-021-87895-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 04/05/2021] [Indexed: 11/09/2022] Open
Abstract
Not every neonate with congenital Zika virus (ZIKV) infection (CZI) is born with microcephaly. We compared inflammation mediators in CSF (cerebrospinal fluid obtained from lumbar puncture) between ZIKV-exposed neonates with/without microcephaly (cases) and controls. In Brazil, in the same laboratory, we identified 14 ZIKV-exposed neonates during the ZIKV epidemic (2015-2016), 7(50%) with and 7(50%) without microcephaly, without any other congenital infection, and 14 neonates (2017-2018) eligible to be controls and to match cases. 29 inflammation mediators were measured using Luminex immunoassay and multidimensional analyses were employed. Neonates with ZIKV-associated microcephaly presented substantially higher degree of inflammatory perturbation, associated with uncoupled inflammatory response and decreased correlations between concentrations of inflammatory biomarkers. The groups of microcephalic and non-microcephalic ZIKV-exposed neonates were distinguished from the control group (area under curve [AUC] = 1; P < 0.0001). Between controls and those non-microcephalic exposed to ZIKV, IL-1β, IL-3, IL-4, IL-7 and EOTAXIN were the top CSF markers. By comparing the microcephalic cases with controls, the top discriminant scores were for IL-1β, IL-3, EOTAXIN and IL-12p70. The degree of inflammatory imbalance may be associated with microcephaly in CZI and it may aid additional investigations in experimental pre-clinical models testing immune modulators in preventing extensive damage of the Central Nervous System.
Collapse
Affiliation(s)
| | | | - Clara L Ramos
- Bahiana Foundation for Science Development, Bahiana School of Medicine, Salvador, Bahia, 40290-000, Brazil
| | - Ana-Luisa Vilas-Boas
- Bahiana Foundation for Science Development, Bahiana School of Medicine, Salvador, Bahia, 40290-000, Brazil
| | | | - Caian L Vinhaes
- Bahiana Foundation for Science Development, Bahiana School of Medicine, Salvador, Bahia, 40290-000, Brazil
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia, 40296-710, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Bahia, 40296-710, Brazil
| | - Beatriz Barreto-Duarte
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia, 40296-710, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Bahia, 40296-710, Brazil
- University Salvador (UNIFACS), Laureate Universities, Salvador, Bahia, 41820-021, Brazil
| | - Artur T L Queiroz
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia, 40296-710, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Bahia, 40296-710, Brazil
| | - Bruno B Andrade
- Bahiana Foundation for Science Development, Bahiana School of Medicine, Salvador, Bahia, 40290-000, Brazil
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation, Salvador, Bahia, 40296-710, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Bahia, 40296-710, Brazil
- University Salvador (UNIFACS), Laureate Universities, Salvador, Bahia, 41820-021, Brazil
- School of Medicine, Faculdade de Tecnologia e Ciências (Uni-FTC), Salvador, Bahia, 41741-590, Brazil
- Wellcome Centre for Infectious Disease Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, 7700, South Africa
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | | |
Collapse
|
13
|
Freitas DA, Souza-Santos R, Carvalho LMA, Barros WB, Neves LM, Brasil P, Wakimoto MD. Congenital Zika syndrome: A systematic review. PLoS One 2020; 15:e0242367. [PMID: 33320867 PMCID: PMC7737899 DOI: 10.1371/journal.pone.0242367] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 10/31/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The signs and symptoms of Zika virus infection are usually mild and self-limited. However, the disease has been linked to neurological complications such as Guillain-Barré syndrome and peripheral nerve involvement, and also to abortion and fetal deaths due to vertical transmission, resulting in various congenital malformations in newborns, including microcephaly. This review aimed to describe the o signs and symptoms that characterize the congenital Zika syndrome. METHODS AND FINDINGS A systematic review was performed with a protocol and described according to the recommendations of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses statement. The search strategy yielded 2,048 studies. After the exclusion of duplicates and application of inclusion criteria, 46 studies were included. The main signs and symptoms associated with the congenital Zika syndrome were microcephaly, parenchymal or cerebellar calcifications, ventriculomegaly, central nervous system hypoplasia or atrophy, arthrogryposis, ocular findings in the posterior and anterior segments, abnormal visual function and low birthweight for gestational age. CONCLUSIONS Zika virus infection during pregnancy can cause a series of changes in the growth and development of children, while impacting the healthcare system due to the severity of cases. Our findings outline the disease profile in newborns and infants and may contribute to the development and updating of more specific clinical protocols.
Collapse
Affiliation(s)
- Danielle A. Freitas
- National School of Public Health, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Reinaldo Souza-Santos
- National School of Public Health, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Liege M. A. Carvalho
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Wagner B. Barros
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Luiza M. Neves
- Fernandes Figueira Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Patrícia Brasil
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Mayumi D. Wakimoto
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
14
|
Animal models of congenital zika syndrome provide mechanistic insight into viral pathogenesis during pregnancy. PLoS Negl Trop Dis 2020; 14:e0008707. [PMID: 33091001 PMCID: PMC7580937 DOI: 10.1371/journal.pntd.0008707] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
In utero Zika virus (ZIKV; family Flaviviridae) infection causes a distinct pattern of birth defects and disabilities in the developing fetus and neonate that has been termed congenital zika syndrome (CZS). Over 8,000 children were affected by the 2016 to 2017 ZIKV outbreak in the Americas, many of whom developed CZS as a result of in utero exposure. To date, there is no consensus about how ZIKV causes CZS; animal models, however, are providing mechanistic insights. Using nonhuman primates, immunocompromised mice, immunocompetent mice, and other animal models (e.g., pigs, sheep, guinea pigs, and hamsters), studies are showing that maternal immunological responses, placental infection and inflammation, as well as viral genetic factors play significant roles in predicting the downstream consequences of in utero ZIKV infection on the development of CZS in offspring. There are thousands of children suffering from adverse consequences of CZS. Therefore, the animal models developed to study ZIKV-induced adverse outcomes in offspring could provide mechanistic insights into how other viruses, including influenza and hepatitis C viruses, impact placental viability and fetal growth to cause long-term adverse outcomes in an effort to identify therapeutic treatments.
Collapse
|
15
|
Rossi F, Josey B, Sayitoglu EC, Potens R, Sultu T, Duru AD, Beljanski V. Characterization of zika virus infection of human fetal cardiac mesenchymal stromal cells. PLoS One 2020; 15:e0239238. [PMID: 32941515 PMCID: PMC7498051 DOI: 10.1371/journal.pone.0239238] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 09/01/2020] [Indexed: 12/16/2022] Open
Abstract
Zika virus (ZIKV) is a single-stranded RNA virus belonging to the family Flaviviridae. ZIKV predominantly enters cells using the TAM-family protein tyrosine kinase receptor AXL, which is expressed on a range of cell types, including neural progenitor cells, keratinocytes, dendritic cells, and osteoblasts. ZIKV infections have been associated with fetal brain damage, which prompted the World Health Organization to declare a public health emergency in 2016. ZIKV infection has also been linked to birth defects in other organs. Several studies have reported congenital heart defects (CHD) in ZIKV infected infants and cardiovascular complications in adults infected with ZIKV. To develop a better understanding of potential causes for these pathologies at a cellular level, we characterized ZIKV infection of human fetal cardiac mesenchymal stromal cells (fcMSCs), a cell type that is known to contribute to both embryological development as well as adult cardiac physiology. Total RNA, supernatants, and/or cells were collected at various time points post-infection to evaluate ZIKV replication, cell death, and antiviral responses. We found that ZIKV productively infected fcMSCs with peak (~70%) viral mRNA detected at 48 h. Use of an antibody blocking the AXL receptor decreased ZIKV infection (by ~50%), indicating that the receptor is responsible to a large extent for viral entry into the cell. ZIKV also altered protein expression of several mesenchymal cell markers, which suggests that ZIKV could affect fcMSCs’ differentiation process. Gene expression analysis of fcMSCs exposed to ZIKV at 6, 12, and 24 h post-infection revealed up-regulation of genes/pathways associated with interferon-stimulated antiviral responses. Stimulation of TLR3 (using poly I:C) or TLR7 (using Imiquimod) prior to ZIKV infection suppressed viral replication in a dose-dependent manner. Overall, fcMSCs can be a target for ZIKV infection, potentially resulting in CHD during embryological development and/or cardiovascular issues in ZIKV infected adults.
Collapse
Affiliation(s)
- Fiorella Rossi
- NSU Cell Therapy Institute, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States of America
| | - Benjamin Josey
- NSU Cell Therapy Institute, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States of America
| | - Ece Canan Sayitoglu
- NSU Cell Therapy Institute, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States of America
| | - Renee Potens
- NSU Cell Therapy Institute, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States of America
| | - Tolga Sultu
- Department of Molecular Biology and Genetics, Bogaziçi University, Istanbul, Turkey
| | - Adil Doganay Duru
- NSU Cell Therapy Institute, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States of America
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Vladimir Beljanski
- NSU Cell Therapy Institute, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States of America
- * E-mail:
| |
Collapse
|
16
|
Sousa IBAD, Souza C, Barbosa MDS, Croda JHR, Gonçalves CCM, Bernardes SS, Marchioro SB. Gestational outcomes in women infected by Zika virus during pregnancy in Mato Grosso do Sul, Brazil: A cross-sectional study. Int J Infect Dis 2020; 98:359-365. [PMID: 32619757 DOI: 10.1016/j.ijid.2020.06.084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 06/23/2020] [Accepted: 06/24/2020] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVES This study aimed to describe the demographic and clinical parameters of women infected by Zika virus who had infants with stigmata of Congenital Zika Syndrome (CZS) versus those who had normal-appearing infants at birth, thereby providing further details on the clinical caveats of neonatal ZIKV infection. METHODOLOGY This cross-sectional study was performed in the state of Mato Grosso do Sul, Central-West region of Brazil, and included 117 mother-infant pairs who were interviewed and 120 gestational outcomes. All mothers had laboratory confirmation by qRT-PCR of ZIKV infection during pregnancy. RESULTS The prevalence of congenital abnormalities related to ZIKV was 2.69 cases per 10,000 live births during this period. Exanthem was the main clinical finding, observed in 92.5% of the mothers in this study. Regarding the timing of ZIKV infection, the first trimester was the most frequent time of infection among mothers of infants with CZS (54.55%) (p=0.0007). The case fatality rate was 5% (n=6). Among the 23 children who were classified as having CZS, 13 (56.52%) of them presented with microcephaly. Only 13 (56.52%) children with CZS were tested by qRT-PCR for ZIKV infection at birth, five (38%) were positive. CONCLUSIONS This study highlights the congenital alterations of ZIKV infection during pregnancy in an epidemic burst, demonstrating that the alterations found in other studies are similar to the present research.
Collapse
Affiliation(s)
| | - Cristina Souza
- Faculty of Health Sciences, School of Medicine, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil
| | | | - Julio Henrique Rosa Croda
- Faculty of Health Sciences, Federal University of Grande Dourados, Dourados, Mato Grosso do Sul, Brazil; Faculty of Health Sciences, School of Medicine, Federal University of Mato Grosso do Sul, Campo Grande, Mato Grosso do Sul, Brazil; Oswaldo Cruz Foundation, Campo Grande, Mato Grosso do Sul, Brazil
| | | | - Sara Santos Bernardes
- Faculty of Health Sciences, Federal University of Grande Dourados, Dourados, Mato Grosso do Sul, Brazil; Laboratory of Tissue Microenviroment, Department of General Pathology, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Silvana Beutinger Marchioro
- Faculty of Health Sciences, Federal University of Grande Dourados, Dourados, Mato Grosso do Sul, Brazil; Institute of Health Sciences, Immunology Laboratory Federal University of Bahia, Salvador, Bahia, Brazil.
| |
Collapse
|
17
|
Valdespino-Vázquez MY, Sevilla-Reyes EE, Lira R, Yocupicio-Monroy M, Piten-Isidro E, Boukadida C, Hernández-Pando R, Soriano-Jimenez JD, Herrera-Salazar A, Figueroa-Damián R, Reyes-Terán G, Zamora-Escudero R, Cardona-Pérez JA, Maldonado-Rodríguez A, Moreno-Verduzco ER, Torres-Flores JM. Congenital Zika Syndrome and Extra-Central Nervous System Detection of Zika Virus in a Pre-term Newborn in Mexico. Clin Infect Dis 2020; 68:903-912. [PMID: 30188990 PMCID: PMC6399440 DOI: 10.1093/cid/ciy616] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 07/30/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND During pregnancy, the Zika virus (ZIKV) replicates in the placenta and central nervous system (CNS) of infected fetuses; nevertheless, the ability of ZIKV to replicate in other fetal tissues has not been extensively characterized. METHODS We researched whether dissemination of congenitally-acquired ZIKV outside the CNS exists by searching for the accumulation of the viral envelope protein, ZIKV ribonucleic acid (RNA), and infectious viral particles in different organs of a deceased newborn with Congenital Zika Syndrome. A real-time qualitative polymerase chain reaction (qPCR) was used to detect ZIKV RNA in the brain, thymus, lungs, kidneys, adrenal glands, spleen, liver, and small intestine. The same tissues were analyzed by indirect immunofluorescence and immunoperoxidase assays using the monoclonal antibody 4G2 to detect ZIKV envelope antigens. Isolation of infectious ZIKV in a cell culture was carried out using brain and kidney samples. RESULTS A postmortem, virological analysis of multiple organs, such as the kidneys (epithelial cells in the renal tubules), lungs (bronchial epithelia), thymus (epithelial cells inside the Hassall's corpuscles), and brain (neurons, ependymal cells, and macrophages) revealed the presence of ZIKV RNA and envelope antigens. Other tissues of the deceased newborn tested positive by qPCR for Epstein-Barr virus and human herpesvirus 6, including the brain cortex (Epstein-Barr) and the thymus, kidneys, and adrenal glands (human herpesvirus 6). The kidneys were identified as a significant niche for viral replication, given that infectious particles were successfully isolated from renal tissues. CONCLUSIONS Our findings demonstrate the ability of congenitally-acquired ZIKV to produce disseminated infections and the viral tropism towards epithelial cells.
Collapse
Affiliation(s)
| | - Edgar E Sevilla-Reyes
- Departamento de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias, Ciudad de Mexico, México
| | - Rosalia Lira
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, Unidad Médica de Alta Especialidad Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de Mexico, México
| | - Martha Yocupicio-Monroy
- Universidad Autónoma de la Ciudad de México, Posgrado en Ciencias Genómicas, Ciudad de Mexico, México
| | - Elvira Piten-Isidro
- Departamento de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias, Ciudad de Mexico, México
| | - Celia Boukadida
- Departamento de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias, Ciudad de Mexico, México
| | - Rogelio Hernández-Pando
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de Mexico, México
| | | | | | | | - Gustavo Reyes-Terán
- Departamento de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias, Ciudad de Mexico, México
| | | | | | - Angélica Maldonado-Rodríguez
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, Unidad Médica de Alta Especialidad Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de Mexico, México
| | - Elsa Romelia Moreno-Verduzco
- Subdirección de Servicios Auxiliares de Diagnóstico, Instituto Nacional de Perinatología, Ciudad de Mexico, México
| | - Jesús Miguel Torres-Flores
- Laboratorio de Virología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de Mexico, México
| |
Collapse
|
18
|
Steinbach RJ, Haese NN, Smith JL, Colgin LMA, MacAllister RP, Greene JM, Parkins CJ, Kempton JB, Porsov E, Wang X, Renner LM, McGill TJ, Dozier BL, Kreklywich CN, Andoh TF, Grafe MR, Pecoraro HL, Hodge T, Friedman RM, Houser LA, Morgan TK, Stenzel P, Lindner JR, Schelonka RL, Sacha JB, Roberts VHJ, Neuringer M, Brigande JV, Kroenke CD, Frias AE, Lewis AD, Kelleher MA, Hirsch AJ, Streblow DN. A neonatal nonhuman primate model of gestational Zika virus infection with evidence of microencephaly, seizures and cardiomyopathy. PLoS One 2020; 15:e0227676. [PMID: 31935257 PMCID: PMC6959612 DOI: 10.1371/journal.pone.0227676] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/23/2019] [Indexed: 12/17/2022] Open
Abstract
Zika virus infection during pregnancy is associated with miscarriage and with a broad spectrum of fetal and neonatal developmental abnormalities collectively known as congenital Zika syndrome (CZS). Symptomology of CZS includes malformations of the brain and skull, neurodevelopmental delay, seizures, joint contractures, hearing loss and visual impairment. Previous studies of Zika virus in pregnant rhesus macaques (Macaca mulatta) have described injury to the developing fetus and pregnancy loss, but neonatal outcomes following fetal Zika virus exposure have yet to be characterized in nonhuman primates. Herein we describe the presentation of rhesus macaque neonates with a spectrum of clinical outcomes, including one infant with CZS-like symptoms including cardiomyopathy, motor delay and seizure activity following maternal infection with Zika virus during the first trimester of pregnancy. Further characterization of this neonatal nonhuman primate model of gestational Zika virus infection will provide opportunities to evaluate the efficacy of pre- and postnatal therapeutics for gestational Zika virus infection and CZS.
Collapse
Affiliation(s)
- Rosemary J. Steinbach
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Nicole N. Haese
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
- Division of Pathobiology & Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Jessica L. Smith
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Lois M. A. Colgin
- Division of Comparative Medicine, Pathology Services Unit, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Rhonda P. MacAllister
- Division of Comparative Medicine, Clinical Medicine Unit, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Justin M. Greene
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
- Division of Pathobiology & Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Christopher J. Parkins
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - J. Beth Kempton
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Edward Porsov
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Xiaojie Wang
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Lauren M. Renner
- Department of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Trevor J. McGill
- Department of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Brandy L. Dozier
- Division of Comparative Medicine, Clinical Medicine Unit, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Craig N. Kreklywich
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Takeshi F. Andoh
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Marjorie R. Grafe
- Department of Pathology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Heidi L. Pecoraro
- Veterinary Diagnostic Services Department, North Dakota State University, Fargo, North Dakota, United States of America
| | - Travis Hodge
- Division of Comparative Medicine, Time Mated Breeding Services Unit, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Robert M. Friedman
- Department of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Lisa A. Houser
- Division of Comparative Medicine, Behavioral Services Unit, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Terry K. Morgan
- Department of Pathology, Oregon Health & Science University, Portland, Oregon, United States of America
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Peter Stenzel
- Department of Pathology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Jonathan R. Lindner
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Robert L. Schelonka
- Division of Neonatology, Department of Pediatrics, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Jonah B. Sacha
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
- Division of Pathobiology & Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Victoria H. J. Roberts
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Martha Neuringer
- Department of Neuroscience, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - John V. Brigande
- Department of Otolaryngology, Oregon Hearing Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Christopher D. Kroenke
- Advanced Imaging Research Center, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Antonio E. Frias
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Anne D. Lewis
- Division of Comparative Medicine, Pathology Services Unit, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Meredith A. Kelleher
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Alec J. Hirsch
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
- Division of Pathobiology & Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| | - Daniel Neal Streblow
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
- Division of Pathobiology & Immunology, Oregon National Primate Research Center, Beaverton, Oregon, United States of America
| |
Collapse
|
19
|
Endy TP. Viral Febrile Illnesses and Emerging Pathogens. HUNTER'S TROPICAL MEDICINE AND EMERGING INFECTIOUS DISEASES 2020. [PMCID: PMC7151808 DOI: 10.1016/b978-0-323-55512-8.00036-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Emerging or emerged diseases and viral pathogens are responsible historically and currently for large epidemics, global pandemics, and significant morbidity and mortality. Our civilization will continue to face the emergence of new pathogens and viruses: viruses will continue to evolve and adapt to new environments at a high rate; our population continues to grow through birth rate, land development, and migration; climate change will continue to increase the vector burden and spread and change the migratory pattern of animals; and our societal mobility will continue to increase through rapid transportation. The clinical evaluation of the febrile patient with a potential emerging viral pathogen involves documenting the likelihood for an infection by a detailed travel history, calculation of an incubation time by exposure, and an understanding of the disease progression though the clinical illness, which drives the differential diagnosis and the type of diagnostics ordered. Ultimately, the proper identification and diagnosis of a patient with a viral febrile illness due to an emerging pathogen will elicit the appropriate precautions to protect health care providers and communities, deliver appropriate therapeutic interventions, and initiate a targeted public health response. The majority of emerging diseases are caused by viruses, with many that are transmitted by insect vectors or are zoonotic. RNA viruses in particular have high mutation rates and can evolve rapidly in new and changing environments. This, in combination with societal factors, climate change, and rapid travel, has increased the number of epidemics from emerging pathogens in the last several decades. Understanding the travel history, incubation time of potential viruses, and the clinical presentation by illness day is essential in making the right diagnosis and identifying the infecting virus.
Collapse
|
20
|
Sutarjono B. Can We Better Understand How Zika Leads to Microcephaly? A Systematic Review of the Effects of the Zika Virus on Human Brain Organoids. J Infect Dis 2019; 219:734-745. [PMID: 30256965 DOI: 10.1093/infdis/jiy572] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Accepted: 09/22/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The innovative human brain organoid model represents a unique opportunity to better understand the genesis of congenital brain abnormalities, particularly microcephaly, caused by Zika virus (ZIKV) infection during early pregnancy. METHODS A systematic review was conducted to investigate how ZIKV leads to microcephaly in a novel experimental model that mimics early brain development. Studies were gathered by searching MEDLINE/Pubmed, LILACS, and LiSSa for reports on effects of ZIKV infection on human brain organoids. From 146 identified papers, 13 articles were selected for review. RESULTS This review found that ZIKV of African, Latin American, and Asian lineages caused productive replication after 72 hours, preferentially infected neural progenitor cells over mature neurons, reduced both cell populations, and caused premature differentiation. Limited data involving only African and Latin American lineages showed a reduction in populations of proliferating cells and intermediate cells, and overall decreased viability. Furthermore, all 3 lineages caused heightened apoptosis and reduced organoid size. CONCLUSIONS This review concludes that, in organoids, ZIKV causes productive replication, infects neural progenitor cells over mature neurons, decreases both populations, causes premature differentiation, induces apoptosis, and reduces size.
Collapse
Affiliation(s)
- Bayu Sutarjono
- Saba University School of Medicine, Devens, Massachusetts
| |
Collapse
|
21
|
Zika virus and microcephaly in Southeast Asia: A cause for concern? J Infect Public Health 2019; 13:11-15. [PMID: 31669035 DOI: 10.1016/j.jiph.2019.09.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 08/30/2019] [Accepted: 09/18/2019] [Indexed: 11/21/2022] Open
Abstract
Zika virus (ZIKV), a mosquito transmitted virus in the family Flaviviridae, genus Flavivirus, recently emerged to cause infections in more than 70 countries and territories around the world. While human infection is normally asymptomatic, it can also result in a mild febrile disease similar to dengue fever. However, when a pregnant woman is infected, ZIKV can cause fetal abnormalities including microcephaly. Evidence has suggested that ZIKV has circulated in Southeast Asia for more than a decade and yet cases of ZIKV associated microcephaly remain sparsely documented. This review seeks to collate the information currently existing on ZIKV associated microcephaly in Southeast Asia, and assess the potential future risk posed by this virus.
Collapse
|
22
|
Abstract
Zika virus (ZIKV) is an arthropod-borne virus that belongs to the Flaviviridae family. Although most cases are mild or go undetected, rare severe neurologic effects, including congenital ZIKV syndrome (CZS) and Guillain-Barré syndrome, have been identified. The serious neurologic complications associated with ZIKV prompted the declaration of the public health emergency of international concern by the World Health Organization. Overall, transmission occurred throughout South and Central America as well as the Caribbean, affecting 48 countries and territories from March 2015 to March 2017. Long-term management of CZS requires a comprehensive combination of supportive services throughout early development.
Collapse
Affiliation(s)
- Savina Reid
- Department of Neurology, Columbia University Medical Center, Milstein Hospital, 177 Fort Washington Avenue, 8GS-300, New York, NY 10032, USA
| | - Kathryn Rimmer
- Department of Neurology, Columbia University Medical Center, Milstein Hospital, 177 Fort Washington Avenue, 8GS-300, New York, NY 10032, USA
| | - Kiran Thakur
- Division of Critical Care and Hospitalist Neurology, Department of Neurology, Columbia University Medical Center, Milstein Hospital, 177 Fort Washington Avenue, 8GS-300, New York, NY 10032, USA.
| |
Collapse
|
23
|
Wahl A, De C, Abad Fernandez M, Lenarcic EM, Xu Y, Cockrell AS, Cleary RA, Johnson CE, Schramm NJ, Rank LM, Newsome IG, Vincent HA, Sanders W, Aguilera-Sandoval CR, Boone A, Hildebrand WH, Dayton PA, Baric RS, Pickles RJ, Braunstein M, Moorman NJ, Goonetilleke N, Victor Garcia J. Precision mouse models with expanded tropism for human pathogens. Nat Biotechnol 2019; 37:1163-1173. [PMID: 31451733 PMCID: PMC6776695 DOI: 10.1038/s41587-019-0225-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 07/12/2019] [Indexed: 12/12/2022]
Abstract
A major limitation of current humanized mouse models is that they primarily enable the analysis of human-specific pathogens that infect hematopoietic cells. However, most human pathogens target other cell types, including epithelial, endothelial and mesenchymal cells. Here, we show that implantation of human lung tissue, which contains up to 40 cell types, including nonhematopoietic cells, into immunodeficient mice (lung-only mice) resulted in the development of a highly vascularized lung implant. We demonstrate that emerging and clinically relevant human pathogens such as Middle East respiratory syndrome coronavirus, Zika virus, respiratory syncytial virus and cytomegalovirus replicate in vivo in these lung implants. When incorporated into bone marrow/liver/thymus humanized mice, lung implants are repopulated with autologous human hematopoietic cells. We show robust antigen-specific humoral and T-cell responses following cytomegalovirus infection that control virus replication. Lung-only mice and bone marrow/liver/thymus-lung humanized mice substantially increase the number of human pathogens that can be studied in vivo, facilitating the in vivo testing of therapeutics. Implantation of lung tissue into humanized mice enables in vivo study of the human immune response to pathogens.
Collapse
Affiliation(s)
- Angela Wahl
- Division of Infectious Diseases, International Center for the Advancement of Translational Science, Center for AIDS Research, University of North Carolina, School of Medicine, Chapel Hill, NC, USA.
| | - Chandrav De
- Division of Infectious Diseases, International Center for the Advancement of Translational Science, Center for AIDS Research, University of North Carolina, School of Medicine, Chapel Hill, NC, USA
| | - Maria Abad Fernandez
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Erik M Lenarcic
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Yinyan Xu
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Adam S Cockrell
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Rachel A Cleary
- Division of Infectious Diseases, International Center for the Advancement of Translational Science, Center for AIDS Research, University of North Carolina, School of Medicine, Chapel Hill, NC, USA
| | - Claire E Johnson
- Division of Infectious Diseases, International Center for the Advancement of Translational Science, Center for AIDS Research, University of North Carolina, School of Medicine, Chapel Hill, NC, USA
| | - Nathaniel J Schramm
- Division of Infectious Diseases, International Center for the Advancement of Translational Science, Center for AIDS Research, University of North Carolina, School of Medicine, Chapel Hill, NC, USA
| | - Laura M Rank
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Isabel G Newsome
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC, USA
| | - Heather A Vincent
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Wes Sanders
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Christian R Aguilera-Sandoval
- Division of Infectious Diseases, International Center for the Advancement of Translational Science, Center for AIDS Research, University of North Carolina, School of Medicine, Chapel Hill, NC, USA.,BD Life Sciences, San Jose, CA, USA
| | - Allison Boone
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.,Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, USA
| | - William H Hildebrand
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Paul A Dayton
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC, USA
| | - Ralph S Baric
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.,Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Raymond J Pickles
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.,Marsico Lung Institute, University of North Carolina, Chapel Hill, NC, USA
| | - Miriam Braunstein
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA
| | - Nathaniel J Moorman
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Nilu Goonetilleke
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, USA.,UNC HIV Cure Center, University of North Carolina, Chapel Hill, NC, USA
| | - J Victor Garcia
- Division of Infectious Diseases, International Center for the Advancement of Translational Science, Center for AIDS Research, University of North Carolina, School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
24
|
Counotte MJ, Meili KW, Taghavi K, Calvet G, Sejvar J, Low N. Zika virus infection as a cause of congenital brain abnormalities and Guillain-Barré syndrome: A living systematic review. F1000Res 2019; 8:1433. [PMID: 31754425 PMCID: PMC6852328 DOI: 10.12688/f1000research.19918.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/01/2019] [Indexed: 01/10/2023] Open
Abstract
Background: The Zika virus (ZIKV) caused a large outbreak in the Americas leading to the declaration of a Public Health Emergency of International Concern in February 2016. A causal relation between infection and adverse congenital outcomes such as microcephaly was declared by the World Health Organization (WHO) informed by a systematic review structured according to a framework of ten dimensions of causality, based on the work of Bradford Hill. Subsequently, the evidence has continued to accumulate, which we incorporate in regular updates of the original work, rendering it a living systematic review. Methods: We present an update of our living systematic review on the causal relation between ZIKV infection and adverse congenital outcomes and between ZIKV and GBS for four dimensions of causality: strength of association, dose-response, specificity, and consistency. We assess the evidence published between January 18, 2017 and July 1, 2019. Results: We found that the strength of association between ZIKV infection and adverse outcomes from case-control studies differs according to whether exposure to ZIKV is assessed in the mother (OR 3.8, 95% CI: 1.7-8.7, I
2=19.8%) or the foetus/infant (OR 37.4, 95% CI: 11.0-127.1, I
2=0%). In cohort studies, the risk of congenital abnormalities was 3.5 times higher after ZIKV infection (95% CI: 0.9-13.5, I
2=0%). The strength of association between ZIKV infection and GBS was higher in studies that enrolled controls from hospital (OR: 55.8, 95% CI: 17.2-181.7, I
2=0%) than in studies that enrolled controls at random from the same community or household (OR: 2.0, 95% CI: 0.8-5.4, I
2=74.6%). In case-control studies, selection of controls from hospitals could have biased results. Conclusions: The conclusions that ZIKV infection causes adverse congenital outcomes and GBS are reinforced with the evidence published between January 18, 2017 and July 1, 2019.
Collapse
Affiliation(s)
| | - Kaspar Walter Meili
- Institute of Social and Preventive Medicine, University Bern, Bern, Switzerland
| | - Katayoun Taghavi
- Institute of Social and Preventive Medicine, University Bern, Bern, Switzerland
| | - Guilherme Calvet
- Acute Febrile Illnesses Laboratory, Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - James Sejvar
- Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Nicola Low
- Institute of Social and Preventive Medicine, University Bern, Bern, Switzerland
| |
Collapse
|
25
|
Werner SL, Banda BK, Burnsides CL, Stuber AJ. Zoonosis: Update on Existing and Emerging Vector-Borne Illnesses in the USA. CURRENT EMERGENCY AND HOSPITAL MEDICINE REPORTS 2019; 7:91-106. [PMID: 32288973 PMCID: PMC7102350 DOI: 10.1007/s40138-019-00189-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW This review describes mosquito- and tick-borne diseases found in the Western Hemisphere. It focuses on emerging diseases and recent geographic shifts in the presence of disease vectors. RECENT FINDINGS Mosquito and tick vectors have become more widespread as environmental conditions have become more favorable. Zika recently has emerged as a concern for fetal anomalies. West Nile Virus has become widespread. Lyme disease and other tick-borne diseases are more prevalent in areas previously inhospitable to these ticks. SUMMARY Healthcare providers must consider the possibility of mosquito- and tick-borne diseases in broader geographic areas and council patients traveling to endemic areas on precautions against these diseases. Treatment for suspected cases of serious tick-borne illnesses should not be delayed pending culture results.
Collapse
Affiliation(s)
- Sandra Lee Werner
- MetroHealth/Cleveland Clinic/CWRU Emergency Medicine Residency Program, Department of Emergency Medicine, MetroHealth Medical Center, 2500 MetroHealth Drive, Cleveland, OH 44109 USA
| | - Bhanu Kirthi Banda
- MetroHealth/Cleveland Clinic/CWRU Emergency Medicine Residency Program, Department of Emergency Medicine, MetroHealth Medical Center, 2500 MetroHealth Drive, Cleveland, OH 44109 USA
| | - Christopher Lee Burnsides
- MetroHealth/Cleveland Clinic/CWRU Emergency Medicine Residency Program, Department of Emergency Medicine, MetroHealth Medical Center, 2500 MetroHealth Drive, Cleveland, OH 44109 USA
| | - Alexander James Stuber
- MetroHealth/Cleveland Clinic/CWRU Emergency Medicine Residency Program, Department of Emergency Medicine, MetroHealth Medical Center, 2500 MetroHealth Drive, Cleveland, OH 44109 USA
| |
Collapse
|
26
|
Chimelli L, Moura Pone S, Avvad-Portari E, Farias Meira Vasconcelos Z, Araújo Zin A, Prado Cunha D, Raposo Thompson N, Lopes Moreira ME, Wiley CA, da Silva Pone MV. Persistence of Zika Virus After Birth: Clinical, Virological, Neuroimaging, and Neuropathological Documentation in a 5-Month Infant With Congenital Zika Syndrome. J Neuropathol Exp Neurol 2019; 77:193-198. [PMID: 29346650 DOI: 10.1093/jnen/nlx116] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
During the Zika epidemic in Brazil, a baby was born at term with microcephaly and arthrogryposis. The mother had Zika symptoms at 10 weeks of gestation. At 17 weeks, ultrasound showed cerebral malformation and ventriculomegaly. At 24 weeks, the amniotic fluid contained ZIKV RNA and at birth, placenta and maternal blood were also positive using RT-qPCR. At birth the baby urine contained ZIKV RNA, whereas CSF at birth and urine at 17 days did not. Seizures started at 6 days. EEG was abnormal and CT scan showed cerebral atrophy, calcifications, lissencephaly, ventriculomegaly, and cerebellar hypoplasia. Bacterial sepsis at 2 months was treated. A sudden increase in head circumference occurred at 4 months necessitating ventricle-peritoneal shunt placement. At 5 months, the infant died with sepsis due to bacterial meningitis. Neuropathological findings were as severe as some of those found in neonates who died soon after birth, including hydrocephalus, destructive lesions/calcification, gliosis, abnormal neuronal migration, dysmaturation of nerve cells, hypomyelination, loss of descending axons, and spinal motor neurons. ZIKV RNA was detected only in frozen brain tissue using RT-qPCR, but infected cells were not detected by in situ hybridization. Progressive gliosis and microgliosis in the midbrain may have contributed to aqueduct compression and subsequent hydrocephalus. The etiology of progressive disease after in utero infection is not clear and requires investigation.
Collapse
Affiliation(s)
- Leila Chimelli
- Laboratory of Neuropathology, State Institute of Brain and UFRJ
| | | | | | | | | | | | | | | | - Clayton A Wiley
- National Institute of Women, Children and Adolescents Health Fernandes Figueira (IFF) - Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil; and Division of Neuropathology, UPMC Presbiterian Hospital, Pittsburgh, Pennsylvania
| | | |
Collapse
|
27
|
Zhang Z, Sun M, Deng J, Yu J, Yang X, Zhao W, Chen G, Wang P. Zika Virus Induced More Severe Inflammatory Response Than Dengue Virus in Chicken Embryonic Livers. Front Microbiol 2019; 10:1127. [PMID: 31191474 PMCID: PMC6540742 DOI: 10.3389/fmicb.2019.01127] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 05/03/2019] [Indexed: 12/22/2022] Open
Abstract
Dengue (DENV) and Zika virus (ZIKV) are important flaviviruses in tropical and subtropical regions, causing severe Dengue Hemorrhagic Fever (DHF)/Dengue Shock Syndrome (DSS) and microcephaly, respectively. The infection of both viruses during pregnancy were reported with adverse fetal outcomes. To investigate the effects of ZIKV and DENV infections on fetal development, we established an infection model in chicken embryos. Compared with DENV-2, the infection of ZIKV significantly retarded the development of chicken embryos. High viral loads of both DENV-2 and ZIKV was detected in brain, eye and heart 7 and 11 days post-infection, respectively. Interestingly, only ZIKV but not DENV-2 was detected in the liver. Even both of them induced apparent liver inflammation, ZIKV infection showed a more severe inflammatory response than DENV-2 infection based on the inflammation scores and the gene expression levels of IL-1β, TNF, IL-6, and TGFβ-2 in liver. Our results demonstrated that ZIKV induced more severe inflammatory response in chicken embryo liver compared to DENV-2, which might partially attribute to viral replication in liver cells. Clinicians should be aware of the potential liver injury associated with ZIKV infection in patients, especially in perinatal fetuses.
Collapse
Affiliation(s)
- Zongyi Zhang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Jinan University, Guangzhou, China
| | - Menghan Sun
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Jinan University, Guangzhou, China
| | - Jieping Deng
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Jinan University, Guangzhou, China
| | - Jianhai Yu
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xuesong Yang
- Division of Histology and Embryology, Key Laboratory for Regenerative Medicine of the Ministry of Education, School of Basic Medical Sciences, Jinan University, Guangzhou, China
| | - Wei Zhao
- Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Guobing Chen
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Jinan University, Guangzhou, China
| | - Pengcheng Wang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Jinan University, Guangzhou, China
| |
Collapse
|
28
|
Rajapakse NS, Ellsworth K, Liesman RM, Ho ML, Henry N, Theel ES, Wallace A, Alvino ACI, Medeiros de Mello L, Meneses J. Unilateral Phrenic Nerve Palsy in Infants with Congenital Zika Syndrome. Emerg Infect Dis 2019; 24. [PMID: 30016248 PMCID: PMC6056128 DOI: 10.3201/eid2408.180057] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
This case series of right unilateral diaphragmatic paralysis suggests peripheral nervous system involvement. Since the first identification of neonatal microcephaly cases associated with congenital Zika virus infection in Brazil in 2015, a distinctive constellation of clinical features of congenital Zika syndrome has been described. Fetal brain disruption sequence is hypothesized to underlie the devastating effects of the virus on the central nervous system. However, little is known about the effects of congenital Zika virus infection on the peripheral nervous system. We describe a series of 4 cases of right unilateral diaphragmatic paralysis in infants with congenital Zika syndrome suggesting peripheral nervous system involvement and Zika virus as a unique congenital infectious cause of this finding. All the patients described also had arthrogryposis (including talipes equinovarus) and died from complications related to progressive respiratory failure.
Collapse
|
29
|
Mohr EL. Modeling Zika Virus-Associated Birth Defects in Nonhuman Primates. J Pediatric Infect Dis Soc 2018; 7:S60-S66. [PMID: 30590626 PMCID: PMC8506225 DOI: 10.1093/jpids/piy120] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/31/2018] [Indexed: 01/22/2023]
Abstract
In utero infection with Zika virus (ZIKV) during pregnancy can lead to the development of birth defects and postnatal deficits. A nonhuman primate (NHP) model of congenital ZIKV infection can help fill the gaps in knowledge where tissue studies are required to define viral pathogenesis and identify targets for therapeutic intervention. This model system has already identified critical features of ZIKV pathogenesis in congenital infection. Before translating these NHP studies to human clinical trials, we must understand the similarities and differences between human and NHP fetal immune system development, neural development, and infant assessment tools. Because of the overall similarity between fetal and infant development in humans and NHPs, this NHP model can complement human clinical trials by defining immune correlates of protection and evaluating therapeutic interventions.
Collapse
Affiliation(s)
- Emma L Mohr
- Department of Pediatrics, University of Wisconsin-Madison
| |
Collapse
|
30
|
O'Connor MA, Tisoncik-Go J, Lewis TB, Miller CJ, Bratt D, Moats CR, Edlefsen PT, Smedley J, Klatt NR, Gale M, Fuller DH. Early cellular innate immune responses drive Zika viral persistence and tissue tropism in pigtail macaques. Nat Commun 2018; 9:3371. [PMID: 30135445 PMCID: PMC6105614 DOI: 10.1038/s41467-018-05826-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 07/24/2018] [Indexed: 12/22/2022] Open
Abstract
The immunological and virological events that contribute to the establishment of Zika virus (ZIKV) infection in humans are unclear. Here, we show that robust cellular innate immune responses arising early in the blood and tissues in response to ZIKV infection are significantly stronger in males and correlate with increased viral persistence. In particular, early peripheral blood recruitment of plasmacytoid dendritic cells and higher production of monocyte chemoattractant protein (MCP-1) correspond with greater viral persistence and tissue dissemination. We also identify non-classical monocytes as primary in vivo targets of ZIKV infection in the blood and peripheral lymph node. These results demonstrate the potential differences in ZIKV pathogenesis between males and females and a key role for early cellular innate immune responses in the blood in viral dissemination and ZIKV pathogenesis.
Collapse
Affiliation(s)
- Megan A O'Connor
- Department of Microbiology, University of Washington, Seattle, 98195, WA, USA
- Washington National Primate Research Center, Seattle, 98121, WA, USA
| | - Jennifer Tisoncik-Go
- Department of Immunology, University of Washington, Seattle, 98109, WA, USA
- Center for Innate Immunity and Immune Disease (CIIID), University of Washington, Seattle, 98109, WA, USA
| | - Thomas B Lewis
- Department of Microbiology, University of Washington, Seattle, 98195, WA, USA
- Washington National Primate Research Center, Seattle, 98121, WA, USA
| | - Charlene J Miller
- Department of Pharmaceutics, University of Washington, Seattle, 98195, WA, USA
- Department of Pediatrics, University of Miami, Miami, 33136, FL, USA
| | - Debra Bratt
- Washington National Primate Research Center, Seattle, 98121, WA, USA
| | - Cassie R Moats
- Washington National Primate Research Center, Seattle, 98121, WA, USA
- Oregon National Primate Research Center, Hillsboro, 97006, OR, USA
| | - Paul T Edlefsen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, 98109, WA, USA
| | - Jeremy Smedley
- Washington National Primate Research Center, Seattle, 98121, WA, USA
- Oregon National Primate Research Center, Hillsboro, 97006, OR, USA
| | - Nichole R Klatt
- Washington National Primate Research Center, Seattle, 98121, WA, USA
- Department of Pharmaceutics, University of Washington, Seattle, 98195, WA, USA
- Department of Pediatrics, University of Miami, Miami, 33136, FL, USA
| | - Michael Gale
- Department of Immunology, University of Washington, Seattle, 98109, WA, USA
- Center for Innate Immunity and Immune Disease (CIIID), University of Washington, Seattle, 98109, WA, USA
| | - Deborah Heydenburg Fuller
- Department of Microbiology, University of Washington, Seattle, 98195, WA, USA.
- Washington National Primate Research Center, Seattle, 98121, WA, USA.
| |
Collapse
|
31
|
Rajapakse NS, Ellsworth K, Liesman RM, Ho ML, Henry N, Theel ES, Wallace A, Alvino ACI, Medeiros de Mello L, Meneses J. Unilateral Phrenic Nerve Palsy in Infants with Congenital Zika Syndrome. Emerg Infect Dis 2018. [DOI: 10.3201/eid2408180057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
32
|
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
|
33
|
Zika Virus Infection during Pregnancy; Maternofetal Risk Assessment, Transmission, Complications, and Management: A Review of the Literature. ARCHIVES OF CLINICAL INFECTIOUS DISEASES 2018. [DOI: 10.5812/archcid.12848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
34
|
Rua R, McGavern DB. Advances in Meningeal Immunity. Trends Mol Med 2018; 24:542-559. [PMID: 29731353 PMCID: PMC6044730 DOI: 10.1016/j.molmed.2018.04.003] [Citation(s) in RCA: 187] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/08/2018] [Accepted: 04/09/2018] [Indexed: 12/26/2022]
Abstract
The central nervous system (CNS) is an immunologically specialized tissue protected by a blood-brain barrier. The CNS parenchyma is enveloped by a series of overlapping membranes that are collectively referred to as the meninges. The meninges provide an additional CNS barrier, harbor a diverse array of resident immune cells, and serve as a crucial interface with the periphery. Recent studies have significantly advanced our understanding of meningeal immunity, demonstrating how a complex immune landscape influences CNS functions under steady-state and inflammatory conditions. The location and activation state of meningeal immune cells can profoundly influence CNS homeostasis and contribute to neurological disorders, but these cells are also well equipped to protect the CNS from pathogens. In this review, we discuss advances in our understanding of the meningeal immune repertoire and provide insights into how this CNS barrier operates immunologically under conditions ranging from neurocognition to inflammatory diseases.
Collapse
Affiliation(s)
- Rejane Rua
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Dorian B McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
35
|
Genetics and mechanisms leading to human cortical malformations. Semin Cell Dev Biol 2018; 76:33-75. [DOI: 10.1016/j.semcdb.2017.09.031] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 09/21/2017] [Accepted: 09/21/2017] [Indexed: 02/06/2023]
|
36
|
Martinot AJ, Abbink P, Afacan O, Prohl AK, Bronson R, Hecht JL, Borducchi EN, Larocca RA, Peterson RL, Rinaldi W, Ferguson M, Didier PJ, Weiss D, Lewis MG, De La Barrera RA, Yang E, Warfield SK, Barouch DH. Fetal Neuropathology in Zika Virus-Infected Pregnant Female Rhesus Monkeys. Cell 2018; 173:1111-1122.e10. [PMID: 29606355 DOI: 10.1016/j.cell.2018.03.019] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 03/02/2018] [Accepted: 03/07/2018] [Indexed: 01/13/2023]
Abstract
The development of interventions to prevent congenital Zika syndrome (CZS) has been limited by the lack of an established nonhuman primate model. Here we show that infection of female rhesus monkeys early in pregnancy with Zika virus (ZIKV) recapitulates many features of CZS in humans. We infected 9 pregnant monkeys with ZIKV, 6 early in pregnancy (weeks 6-7 of gestation) and 3 later in pregnancy (weeks 12-14 of gestation), and compared findings with uninfected controls. 100% (6 of 6) of monkeys infected early in pregnancy exhibited prolonged maternal viremia and fetal neuropathology, including fetal loss, smaller brain size, and histopathologic brain lesions, including microcalcifications, hemorrhage, necrosis, vasculitis, gliosis, and apoptosis of neuroprogenitor cells. High-resolution MRI demonstrated concordant lesions indicative of deep gray matter injury. We also observed spinal, ocular, and neuromuscular pathology. Our data show that vascular compromise and neuroprogenitor cell dysfunction are hallmarks of CZS pathogenesis, suggesting novel strategies to prevent and to treat this disease.
Collapse
Affiliation(s)
- Amanda J Martinot
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Peter Abbink
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Onur Afacan
- Computational Radiology Laboratory, Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Anna K Prohl
- Computational Radiology Laboratory, Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Jonathan L Hecht
- Division of Anatomic Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Erica N Borducchi
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Rafael A Larocca
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Rebecca L Peterson
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | | | | | - Peter J Didier
- Tulane National Primate Research Center, Tulane University, Covington, LA 70433, USA
| | | | | | | | - Edward Yang
- Computational Radiology Laboratory, Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Simon K Warfield
- Computational Radiology Laboratory, Department of Radiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA.
| |
Collapse
|
37
|
Ramos CL, Moreno-Carvalho OA, Nascimento-Carvalho CM. Cerebrospinal fluid aspects of neonates with or without microcephaly born to mothers with gestational Zika virus clinical symptoms. J Infect 2018; 76:563-569. [PMID: 29432825 DOI: 10.1016/j.jinf.2018.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 01/26/2018] [Accepted: 02/03/2018] [Indexed: 11/24/2022]
Affiliation(s)
- Clara L Ramos
- Bahiana School of Medicine, Bahiana Foundation for Science Development, Salvador, 40295-050, Brazil.
| | | | - Cristiana M Nascimento-Carvalho
- Post-graduation Program in Health Sciences and Department of Paediatrics, Federal University of Bahia School of Medicine, Salvador, 40025-010, Brazil; Post-graduation Program in Health Sciences and Department of Paediatrics, Federal University of Bahia School of Medicine, Salvador, 40025-010, BrazilBrazilian Council for Scientific and Technological Development, Brazilia, Brazil
| |
Collapse
|
38
|
Zika virus infection in pregnant rhesus macaques causes placental dysfunction and immunopathology. Nat Commun 2018; 9:263. [PMID: 29343712 PMCID: PMC5772047 DOI: 10.1038/s41467-017-02499-9] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 12/04/2017] [Indexed: 12/13/2022] Open
Abstract
Zika virus (ZIKV) infection during pregnancy leads to an increased risk of fetal growth restriction and fetal central nervous system malformations, which are outcomes broadly referred to as the Congenital Zika Syndrome (CZS). Here we infect pregnant rhesus macaques and investigate the impact of persistent ZIKV infection on uteroplacental pathology, blood flow, and fetal growth and development. Despite seemingly normal fetal growth and persistent fetal-placenta-maternal infection, advanced non-invasive in vivo imaging studies reveal dramatic effects on placental oxygen reserve accompanied by significantly decreased oxygen permeability of the placental villi. The observation of abnormal oxygen transport within the placenta appears to be a consequence of uterine vasculitis and placental villous damage in ZIKV cases. In addition, we demonstrate a robust maternal-placental-fetal inflammatory response following ZIKV infection. This animal model reveals a potential relationship between ZIKV infection and uteroplacental pathology that appears to affect oxygen delivery to the fetus during development. Zika virus infection during pregnancy can result in birth defects, but underlying pathogenesis at the maternal-fetal interface is unclear. Here, the authors use non-invasive in vivo imaging of Zika-infected rhesus macaques and show that infection results in abnormal oxygen transport across the placenta.
Collapse
|
39
|
Chimelli L, Avvad-Portari E. Congenital Zika virus infection: a neuropathological review. Childs Nerv Syst 2018; 34:95-99. [PMID: 29167994 DOI: 10.1007/s00381-017-3651-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 10/27/2017] [Indexed: 11/27/2022]
Abstract
BACKGROUND A relationship between Zika virus (ZikV) infection in pregnancy and the occurrence of microcephaly was established during the Zika outbreak in Brazil (2015-2016). Neuropathological findings in congenital Zika syndrome helped to understand its pathogenetic mechanisms. RESULTS The most relevant postmortem findings in the central nervous system (CNS) of fetuses and neonates infected with ZikV early in gestation are microcephaly with ex-vacuo ventriculomegaly and large head circumference associated with obstructive hydrocephalus due to severe midbrain and aqueduct distortion. Babies with severe brain lesions are born with arthrogryposis. Histologically, there is extensive destruction of the hemispheric parenchyma, calcifications, various disturbances of neuronal migration, reactive gliosis, microglial hyperplasia and occasional perivascular cuffs of lymphocytes, also in the meninges. Hypoplastic lesions secondary to the lack of descending nerve fibers include small basis pontis, pyramids and spinal corticospinal tracts. Cerebellar hypoplasia is also common. Severe nerve motor nerve cell loss is observed in the anterior horn of the spinal cord. CONCLUSION A spectrum of neuropathological changes, from severe microcephaly to obstructive hydrocephalus was observed. The severity of the lesions is directly related to the gestational age, the most severe occurring when the mother is infected in the first trimester. Infection of progenitor cells at the germinal matrix was demonstrated. The lack of spinal motor neurons is responsible for fetal acynesia and consequent arthrogryposis.
Collapse
Affiliation(s)
- L Chimelli
- Laboratory of Neuropathology, State Institute of Brain Paulo Niemeyer and Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - E Avvad-Portari
- Fernandes Figueira Institute-Oswaldo Cruz Foundation (Fiocruz) and State University of Rio de Janeiro (UERJ), Rio de Janeiro, Brazil.
| |
Collapse
|
40
|
Mittal R, Nguyen D, Debs LH, Patel AP, Liu G, Jhaveri VM, S. Kay SI, Mittal J, Bandstra ES, Younis RT, Chapagain P, Jayaweera DT, Liu XZ. Zika Virus: An Emerging Global Health Threat. Front Cell Infect Microbiol 2017; 7:486. [PMID: 29276699 PMCID: PMC5727043 DOI: 10.3389/fcimb.2017.00486] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/13/2017] [Indexed: 01/07/2023] Open
Abstract
Zika virus (ZIKV) is an emerging healthcare threat. The presence of the mosquito Aedes species across South and Central America in combination with complementary climates have incited an epidemic of locally transmitted cases of ZIKV infection in Brazil. As one of the most significant current public health concerns in the Americas, ZIKV epidemic has been a cause of alarm due to its known and unknown complications. At this point, there has been a clear association between ZIKV infection and severe clinical manifestations in both adults and neonates, including but not limited to neurological deficits such as Guillain-Barré syndrome (GBS) and microcephaly, respectively. The gravity of the fetal anomalies linked to ZIKV vertical transmission from the mother has prompted a discussion on whether to include ZIKV as a formal member of the TORCH [Toxoplasma gondii, other, rubella virus, cytomegalovirus (CMV), and herpes] family of pathogens known to breach placental barriers and cause congenital disease in the fetus. The mechanisms of these complex phenotypes have yet to be fully described. As such, diagnostic tools are limited and no effective modalities are available to treat ZIKV. This article will review the recent advancements in understanding the pathogenesis of ZIKV infection as well as diagnostic tests available to detect the infection. Due to the increase in incidence of ZIKV infections, there is an immediate need to develop new diagnostic tools and novel preventive as well as therapeutic modalities based on understanding the molecular mechanisms underlying the disease.
Collapse
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, United States,*Correspondence: Rahul Mittal
| | - Desiree Nguyen
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Luca H. Debs
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Amit P. Patel
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - George Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Vasanti M. Jhaveri
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Sae-In S. Kay
- Department of Surgery, Division of Otorhinolaryngology, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Jeenu Mittal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Emmalee S. Bandstra
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Ramzi T. Younis
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, United States,Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, United States,Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Prem Chapagain
- Department of Physics and Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| | - Dushyantha T. Jayaweera
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Xue Zhong Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, FL, United States,Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, United States,Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, United States,Xue Zhong Liu
| |
Collapse
|
41
|
Antiviral CD8 T cells induce Zika-virus-associated paralysis in mice. Nat Microbiol 2017; 3:141-147. [PMID: 29158604 PMCID: PMC5780207 DOI: 10.1038/s41564-017-0060-z] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 10/11/2017] [Indexed: 02/07/2023]
Abstract
Zika virus (ZIKV) is an emerging, mosquito-borne RNA virus. The rapid spread of ZIKV within the Americas has unveiled microcephaly 1 and Guillain-Barré syndrome2,3 as ZIKV-associated neurological complications. Recent reports have also indicated other neurological manifestations to be associated with ZIKV, including myelitis 4 , meningoencephalitis 5 and fatal encephalitis 6 . Here, we investigate the neuropathogenesis of ZIKV infection in type I interferon receptor IFNAR knockout (Ifnar1 -/- ) mice, an infection model that exhibits high viral burden within the central nervous system. We show that systemic spread of ZIKV from the site of infection to the brain requires Ifnar1 deficiency in the haematopoietic compartment. However, spread of ZIKV within the central nervous system is supported by Ifnar1-deficient non-haematopoietic cells. Within this context, ZIKV infection of astrocytes results in breakdown of the blood-brain barrier and a large influx of CD8+ effector T cells. We also find that antiviral activity of CD8+ T cells within the brain markedly limits ZIKV infection of neurons, but, as a consequence, instigates ZIKV-associated paralysis. Taken together, our study uncovers mechanisms underlying ZIKV neuropathogenesis within a susceptible mouse model and suggests blood-brain barrier breakdown and T-cell-mediated neuropathology as potential underpinnings of ZIKV-associated neurological complications in humans.
Collapse
|
42
|
Cicuto Ferreira Rocha NA, de Campos AC, Cicuto Ferreira Rocha F, Pereira Dos Santos Silva F. Microcephaly and Zika virus: Neuroradiological aspects, clinical findings and a proposed framework for early evaluation of child development. Infant Behav Dev 2017; 49:70-82. [PMID: 28755567 DOI: 10.1016/j.infbeh.2017.07.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 05/30/2017] [Accepted: 07/14/2017] [Indexed: 10/19/2022]
Abstract
BACKGROUND AND AIMS As the recent outbreak of microcephaly cases caused by Zika virus has been declared a global health emergency, providing assessment guidelines for multidisciplinary teams providing early developmental screening and stimulation to infants with microcephaly is much needed. Thus, the aim of this manuscript is to provide an overview on what is known about neuroradiological aspects and clinical findings in infants with microcephaly caused by Zika virus and to propose a framework for early evaluation of child development. METHODS The keywords "Zika virus" and "microcephaly" were searched in PubMed database for articles published from incept to May 2017. These texts were reviewed, and the ones addressing neuroradiological and clinical findings in infants were selected. Recommendations for early assessment were made based on the International Classification of Functionality Disability and Health (ICF) model. OUTCOMES AND RESULTS The database search yielded 599 publications and 36 were selected. The studies detected microcephaly with diffuse brain malformations and calcifications, ventriculomegaly, optic nerve hypoplasia, macular atrophy, cataracts, impaired visual and hearing function, arthrogryposis, spasticity, hyperreflexia, irritability, tremors, and seizures, but very little is known about early development. Early assessments were described based on the ICF domains (Body Function and Structures, Activities and Participation and Contextual factors). CONCLUSION AND IMPLICATIONS Studies published showed abnormal brain, optic, neurologic and orthopedic findings, but very little is known about other aspects of functioning in infants with microcephaly caused by Zika virus. The biopsychosocial model based on the ICF paradigm provides an adequate framework to describe the condition of the infant with microcephaly receiving rehabilitative efforts to minimize disability. Efforts towards early identification of developmental delays should be taken within the first six months of life.
Collapse
Affiliation(s)
- Nelci Adriana Cicuto Ferreira Rocha
- Department of Physical Therapy, Child Development Analysis Laboratory (LADI), Federal University of São Carlos (UFSCar), Rodovia Washington Luis, km 235, São Carlos, SP, 13565-905, Brazil.
| | - Ana Carolina de Campos
- Department of Physical Therapy, Child Development Analysis Laboratory (LADI), Federal University of São Carlos (UFSCar), Rodovia Washington Luis, km 235, São Carlos, SP, 13565-905, Brazil
| | - Fellipe Cicuto Ferreira Rocha
- Medical School, Pontifical Catholic University of Campinas (PUC-Campinas), Av. John Boyd Dunlop, s/n - Jardim Ipaussurama, Campinas, SP, 13060-904, Brazil
| | - Fernanda Pereira Dos Santos Silva
- Department of Physical Therapy, Child Development Analysis Laboratory (LADI), Federal University of São Carlos (UFSCar), Rodovia Washington Luis, km 235, São Carlos, SP, 13565-905, Brazil
| |
Collapse
|