1
|
Grijpink LCM, van der Valk WH, van Beelen ESA, de Groot JCMJ, Locher H, Vossen ACTM. Cytomegalovirus host receptor expression in the human fetal inner ear. PLoS One 2025; 20:e0320605. [PMID: 40163451 PMCID: PMC11957294 DOI: 10.1371/journal.pone.0320605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/20/2025] [Indexed: 04/02/2025] Open
Abstract
Fetal infection with human cytomegalovirus (hCMV) can cause sensorineural hearing loss and vestibular impairment, yet its pathogenesis remains unclear. This study aims to identify potential target cell types of hCMV in the human fetal inner ear. Viral particles use several envelope glycoproteins to enter target cells, including the pentameric complex, the trimeric complex and glycoprotein B. Platelet-derived growth factor receptor alpha (PDGFRA) serves as the receptor in fibroblasts, neuropilin-2 (NRP2) in epithelial, endothelial and dendritic cells as well as in leukocytes. Upon binding of these glycoproteins, glycoprotein B initiates membrane fusion which is proposed to be mediated by EGFR. When and where these proteins are expressed in the fetal inner ear during development is unknown. To address this, expression patterns of PDGFRA, NRP2 and EGFR were investigated in human fetal inner ear tissue using single-nucleus RNA sequencing data (first trimester: N = 2) and immunohistochemistry (first trimester: N = 6, second trimester: N = 5). PDGFRA gene and protein expression was detected in mesenchymal cells, NRP2 protein expression in epithelial cells and endothelial cells, and EGFR gene and protein expression in both epithelial cells and mesenchymal cells. Notably, all three receptors were present in tissue from the first and second trimesters. In conclusion, hCMV host receptors PDGFRA, NRP2 and EGFR are expressed in mesenchymal, epithelial and endothelial cells within the cochlea and vestibular organs during the first and second trimesters. These cell types may serve as targets for hCMV infection of the fetal inner ear.
Collapse
Affiliation(s)
- Lucia C. M. Grijpink
- Leiden University Center for Infectious Diseases (LUCID), Medical Microbiology and Infection Control, Leiden University Medical Center, Leiden, The Netherlands
| | - Wouter H. van der Valk
- Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Edward S. A. van Beelen
- Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - John C. M. J. de Groot
- Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Heiko Locher
- Department of Otorhinolaryngology and Head & Neck Surgery, Leiden University Medical Center, Leiden, The Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden University Medical Center, Leiden, The Netherlands
| | - Ann C. T. M. Vossen
- Leiden University Center for Infectious Diseases (LUCID), Medical Microbiology and Infection Control, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
2
|
Wang Z, Zhang D, Shui M, Ho IW, Kou WS, Wei J, Wan JB, Wang R, Zhang QW. Investigation of the whitening activity of ginsenosides from Panax notoginseng and optimization of the dosage form. J Ginseng Res 2024; 48:543-551. [PMID: 39583176 PMCID: PMC11583341 DOI: 10.1016/j.jgr.2023.12.005] [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: 12/16/2022] [Revised: 12/14/2023] [Accepted: 12/27/2023] [Indexed: 11/26/2024] Open
Abstract
Background Ginsenoside, as an active ingredient in traditional Chinese medicine, has been widely used for skin whitening for several years. Recent research has found that Panax notoginseng has a higher content of ginsenosides compared with the Panax ginseng. Those ginsenosides have promising potential to be developed as skin whitening agents. Methods We selected five dammarane ginsenosides isolated from P. notoginseng and their mixtures to investigate the skin lightning activity. Zebrafish embryo model was used for initial screening of the whitening activity. Subsequently, the whitening effect of components was examined and compared via testing the inhibition of melanin and activity of tyrosinase in B16 cells treated with these components. Molecular docking was also applied to investigate the interactions between ginsenosides and tyrosinase. Finally, the most effective saponins were selected for dosage form optimization and the whitening effect of saponin-loaded ethosomes was further demonstrated on the C57BL/6 mouse model. Results Experimental results showed that the protopanaxtriol saponins (PTS) were the most potent saponins with a decent safety profile, and the molecule docking results demonstrated that PTS had strong inhibitory ability to tyrosinase. PTS was successfully encapsulated into ethosomes with an encapsulation efficiency of 93%. The PTS ethosome gel could effectively inhibit the melanin production caused by UVB tanning on the back skin of mice. Conclusion The PTS ethosome gel provides an effective and safe formulation of PTS to whiten the UVB-tanned skin in vivo and could be used as a potential skin whitening agent in the future.
Collapse
Affiliation(s)
- Zeyu Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Daiyan Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Mingju Shui
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Ian Wa Ho
- Pui Chng Middle School (Macau), Macao SAR, China
- Macau Society of Supramolecular Chemistry and Biomaterials, Macao SAR, China
| | - Weng Si Kou
- Pui Chng Middle School (Macau), Macao SAR, China
- Macau Society of Supramolecular Chemistry and Biomaterials, Macao SAR, China
| | - Jianwen Wei
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
- Macau Society of Supramolecular Chemistry and Biomaterials, Macao SAR, China
| | - Jian-Bo Wan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Ruibing Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Qing-Wen Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| |
Collapse
|
3
|
Characterization of the Second Apoptosis Inhibitor Encoded by Guinea Pig Cytomegalovirus. J Virol 2022; 96:e0162222. [PMID: 36472439 PMCID: PMC9769370 DOI: 10.1128/jvi.01622-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Despite the usefulness of guinea pig cytomegalovirus (GPCMV) for studies on congenital CMV infection, its viral mechanisms for the evasion of host defense strategies have not been fully elucidated. We reported previously that GPCMV gp38.1 functions as a viral mitochondria-localized inhibitor of apoptosis-like function, and its weak activity suggested the presence of an additional inhibitory molecule(s). Here, we identified gp38.3-2, a 42-amino-acid (aa) reading frame embedded within the gp38.3 gene that encodes a positional homolog of murine CMV (MCMV) m41. Characterization of gp38.3-2 resulted in the following findings: (i) the aa sequence of gp38.3-2 shows some similarity to that of MCMV m41.1, a viral inhibitor of oligomerization of a member of Bcl-2 family protein BAK, but there is no correspondence in their predicted secondary structures; (ii) gp38.3-2, but not gp38.3, showed inhibitory activities against staurosporine-induced apoptosis; (iii) three-dimensional protein complex prediction suggests that the N-terminal α-helix of gp38.3-2 interacts with residues in the BH3 and BH1 motifs of BAK, and analysis of gp38.3-2 and BAK mutants supported this model; (iv) guinea pig fibroblast cells infected with gp38.3-2-deficient GPCMV strain Δ38.3-2 died earlier than cells infected with rescued strain r38.3-2, resulting in lower yields of Δ38.3-2; (v) Δ38.3-2 exhibited a partial but significant decrease in monocyte and macrophage infection in comparison with r38.3-2; and, however, (vi) little difference in the viral infection of guinea pigs was observed between these two strains. Therefore, we hypothesize that gp38.3-2 contributes little to the evasion of host defense mechanisms under the experimental conditions used. IMPORTANCE Although GPCMV provides a useful animal model for studies on the pathogenesis of congenital CMV infection and the development of CMV vaccine strategies, our understanding of the viral mechanisms by which it evades apoptosis of infected cells has been limited in comparison with those of murine and human CMVs. Here, we report a second GPCMV apoptosis inhibitor (42 amino acids in length) that interacts with BAK, a Bcl-2 family proapoptotic protein. Three-dimensional structural prediction indicated a unique BAK recognition by gp38.3-2 via the BH3 and BH1 motif sequences. Our findings suggest the potential development of BH3 mimetics that can regulate inhibition or induction of apoptosis based on short ~40-amino-acid peptide molecules as with GPCMV.
Collapse
|
4
|
Lo TH, Lin PH, Hsu WC, Tsao PN, Liu TC, Yang TH, Hsu CJ, Huang LM, Lu CY, Wu CC. Prognostic determinants of hearing outcomes in children with congenital cytomegalovirus infection. Sci Rep 2022; 12:5219. [PMID: 35338167 PMCID: PMC8956567 DOI: 10.1038/s41598-022-08392-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 02/24/2022] [Indexed: 12/20/2022] Open
Abstract
Congenital cytomegalovirus (cCMV) infection is the most prevalent cause of non-genetic sensorineural hearing loss (SNHL) in children. However, the prognostic determinants of SNHL remain unclear. Children with cCMV infection in a tertiary hospital were enrolled. The presence of cCMV-related symptoms at birth, the newborn hearing screening (NHS) results, and the blood viral loads were ascertained. Audiologic outcomes and initial blood viral loads were compared between different groups. Of the 39 children enrolled, 16 developed SNHL. SNHL developed in 60% of children who were initially symptomatic, and in 34.5% of those who were initially asymptomatic with normal hearing or isolated hearing loss, respectively. Failuire in NHS was a reliable tool for early detection of SNHL. The initial viral loads were higher in children who were symptomatic at birth, those who failed NHS, and those who developed SNHL. We observed SNHL deterioration in a patient after CMV DNAemia clearance was achieved, and in another patient with the flare-up of viral load. The presence of cCMV-related symptoms at birth, failure in NHS, and blood viral load might be the prognostic factors for hearing outcomes. Regular audiologic examinations are necessary in all children with cCMV infection even after CMV DNAemia clearance.
Collapse
Affiliation(s)
- Ta-Hsuan Lo
- Department of Otolaryngology, National Taiwan University Hospital, Address: 7, Chung-Shan S. Rd., 10002, Taipei, Taiwan, ROC.,Department of Otolaryngology, National Taiwan University Biomedical Park Hospital, Hsinchu, Taiwan
| | - Pei-Hsuan Lin
- Department of Otolaryngology, National Taiwan University Hospital Yunlin Branch, Yunlin, Taiwan
| | - Wei-Chung Hsu
- Department of Otolaryngology, National Taiwan University Hospital, Address: 7, Chung-Shan S. Rd., 10002, Taipei, Taiwan, ROC
| | - Po-Nien Tsao
- Department of Pediatrics, National Taiwan University Hospital, Address: 7, Chung-Shan S. Rd., 10002, Taipei, Taiwan, ROC
| | - Tien-Chen Liu
- Department of Otolaryngology, National Taiwan University Hospital, Address: 7, Chung-Shan S. Rd., 10002, Taipei, Taiwan, ROC
| | - Tzong-Hann Yang
- Department of Otorhinolaryngology, Taipei City Hospital, Taipei, Taiwan
| | - Chuan-Jen Hsu
- Department of Otolaryngology, Taichung Tzu-Chi Hospital, Taichung, Taiwan
| | - Li-Min Huang
- Department of Pediatrics, National Taiwan University Hospital, Address: 7, Chung-Shan S. Rd., 10002, Taipei, Taiwan, ROC
| | - Chun-Yi Lu
- Department of Pediatrics, National Taiwan University Hospital, Address: 7, Chung-Shan S. Rd., 10002, Taipei, Taiwan, ROC.
| | - Chen-Chi Wu
- Department of Otolaryngology, National Taiwan University Hospital, Address: 7, Chung-Shan S. Rd., 10002, Taipei, Taiwan, ROC. .,Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan. .,Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan. .,Department of Medical Research, National Taiwan University Biomedical Park Hospital, Hsinchu, Taiwan.
| |
Collapse
|
5
|
Zhou YP, Mei MJ, Wang XZ, Huang SN, Chen L, Zhang M, Li XY, Qin HB, Dong X, Cheng S, Wen L, Yang B, An XF, He AD, Zhang B, Zeng WB, Li XJ, Lu Y, Li HC, Li H, Zou WG, Redwood AJ, Rayner S, Cheng H, McVoy MA, Tang Q, Britt WJ, Zhou X, Jiang X, Luo MH. A congenital CMV infection model for follow-up studies of neurodevelopmental disorders, neuroimaging abnormalities, and treatment. JCI Insight 2022; 7:152551. [PMID: 35014624 PMCID: PMC8765053 DOI: 10.1172/jci.insight.152551] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/23/2021] [Indexed: 12/28/2022] Open
Abstract
Congenital cytomegalovirus (cCMV) infection is the leading infectious cause of neurodevelopmental disorders. However, the neuropathogenesis remains largely elusive due to a lack of informative animal models. In this study, we developed a congenital murine CMV (cMCMV) infection mouse model with high survival rate and long survival period that allowed long-term follow-up study of neurodevelopmental disorders. This model involves in utero intracranial injection and mimics many reported clinical manifestations of cCMV infection in infants, including growth restriction, hearing loss, and impaired cognitive and learning-memory abilities. We observed that abnormalities in MRI/CT neuroimaging were consistent with brain hemorrhage and loss of brain parenchyma, which was confirmed by pathological analysis. Neuropathological findings included ventriculomegaly and cortical atrophy associated with impaired proliferation and migration of neural progenitor cells in the developing brain at both embryonic and postnatal stages. Robust inflammatory responses during infection were shown by elevated inflammatory cytokine levels, leukocyte infiltration, and activation of microglia and astrocytes in the brain. Pathological analyses and CT neuroimaging revealed brain calcifications induced by cMCMV infection and cell death via pyroptosis. Furthermore, antiviral treatment with ganciclovir significantly improved neurological functions and mitigated brain damage as shown by CT neuroimaging. These results demonstrate that this model is suitable for investigation of mechanisms of infection-induced brain damage and long-term studies of neurodevelopmental disorders, including the development of interventions to limit CNS damage associated with cCMV infection.
Collapse
Affiliation(s)
- Yue-Peng Zhou
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Meng-Jie Mei
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xian-Zhang Wang
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Sheng-Nan Huang
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Lin Chen
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Ming Zhang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Xin-Yan Li
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Hai-Bin Qin
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiao Dong
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Shuang Cheng
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Le Wen
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,The Joint Center of Translational Precision Medicine, Guangzhou Institute of Pediatrics, Guangzhou Women and Children Medical Center, Guangzhou, China
| | - Bo Yang
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,The Joint Center of Translational Precision Medicine, Guangzhou Institute of Pediatrics, Guangzhou Women and Children Medical Center, Guangzhou, China
| | - Xue-Fang An
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Ao-Di He
- The Joint Center of Translational Precision Medicine, Guangzhou Institute of Pediatrics, Guangzhou Women and Children Medical Center, Guangzhou, China
| | - Bing Zhang
- The Joint Center of Translational Precision Medicine, Guangzhou Institute of Pediatrics, Guangzhou Women and Children Medical Center, Guangzhou, China
| | - Wen-Bo Zeng
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Xiao-Jun Li
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Youming Lu
- The Joint Center of Translational Precision Medicine, Guangzhou Institute of Pediatrics, Guangzhou Women and Children Medical Center, Guangzhou, China
| | - Hong-Chuang Li
- University of Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Haidong Li
- University of Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Wei-Guo Zou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Alec J. Redwood
- The Institute for Respiratory Health, University of Western Australia, Crawley, Western Australia, Australia
| | - Simon Rayner
- Department of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway.,Hybrid Technology Hub — Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Han Cheng
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Michael A. McVoy
- Department of Pediatrics, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| | - William J. Britt
- Department of Pediatrics, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Xin Zhou
- University of Chinese Academy of Sciences, Beijing, China.,Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy of Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, China
| | - Xuan Jiang
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,The Joint Center of Translational Precision Medicine, Guangzhou Institute of Pediatrics, Guangzhou Women and Children Medical Center, Guangzhou, China
| | - Min-Hua Luo
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China.,The Joint Center of Translational Precision Medicine, Guangzhou Institute of Pediatrics, Guangzhou Women and Children Medical Center, Guangzhou, China.,Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Majima R, Koshizuka T, Inoue N. The Guinea pig cytomegalovirus GP119.1 gene encodes an IgG-binding glycoprotein that is incorporated into the virion. Microbiol Immunol 2021; 65:28-39. [PMID: 33616978 DOI: 10.1111/1348-0421.12867] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 11/10/2020] [Accepted: 12/05/2020] [Indexed: 11/29/2022]
Abstract
Cytomegaloviruses (CMVs) encode various immunoevasins, including viral receptors for the Fc domain of host IgG (vFcγR), to evade host immune responses. Although guinea pig CMV (GPCMV) provides a useful animal model for congenital CMV infection, the GPCMV genes encoding such receptors have not yet been characterized. In this study, we analyzed a locus that may encode gene products for the GPCMV immune evasion mechanisms and identified the following. (a) RACE analyses identified four transcripts in the GP117 to GP122 locus. One of the transcripts contained the GP119.1 ORF, which has weak homologies with human CMV UL119/UL118 encoding a viral FcγR and with guinea pig FcγR. (b) A transient transfection assay with plasmids expressing EGFP-tagged GP119.1 or its mutated forms identified its true translational initiation site, localization mainly in the endoplasmic reticulum, and N-glycosylation. (c) Importantly, GP119.1 bound to guinea pig IgG or the IgG-Fc fragment. (d) GP119.1 is present in the virion with a molecular mass of 15 and 23~30 kDa, and a portion of the GP119.1 products are N-glycosylated. (e) GP119.1 was dispensable for viral growth on guinea pig fibroblasts and epithelial cells in vitro. Taken together, our findings indicate that GP119.1 is an IgG-Fc binding glycoprotein incorporated into the virion, and this finding warrants further studies on the functions of GP119.1 in animal models.
Collapse
Affiliation(s)
- Ryuichi Majima
- Microbiology and Immunology, Gifu Pharmaceutical University, Japan
| | - Tetsuo Koshizuka
- Microbiology and Immunology, Gifu Pharmaceutical University, Japan
| | - Naoki Inoue
- Microbiology and Immunology, Gifu Pharmaceutical University, Japan
| |
Collapse
|
7
|
Noguchi K, Majima R, Takahashi K, Iwase Y, Yamada S, Satoh K, Koshizuka T, Inoue N. Identification and functional analyses of a cell-death inhibitor encoded by guinea pig cytomegalovirus gp38.1 in cell culture and in animals. J Gen Virol 2020; 101:1270-1279. [PMID: 32915127 DOI: 10.1099/jgv.0.001493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cytomegaloviruses (CMVs) employ an array of strategies designed to interfere with host defence responses against pathogens. Studies on such evasion mechanisms are important for understanding the pathogenesis of CMV diseases. Although guinea pig CMV (GPCMV) provides a useful animal model for congenital CMV infection, its evasion strategies are not fully elucidated. Here, we analysed a genome locus that may encode gene products for the GPCMV evasion mechanisms and found the following. (1) RACE analyses identified five transcripts in the GP38-gp38.4 locus, one of which was a spliced product encoding gp38.1. Similarities in the splicing pattern and gene position of gp38.1 to human CMV UL37 and its exon 1 encoding vMIA (viral mitochondria-localized inhibitor of apoptosis) suggest that the gp38.1 gene encodes an apoptosis inhibitor. (2) In a transient transfection assay, gp38.1 localized in the mitochondria and relocated BAX from the cytoplasm to the mitochondria, although its co-localization with BAK was not evident. Further, the expression of gp38.1 partially reduced staurosporine-induced apoptosis. (3) GPCMV defective in the gp38.1 ORF (Δ38.1) and the virus that rescues the defect (r38.1) were generated. Guinea pig fibroblast cells infected with Δ38.1 died earlier than r38.1-infected cells, which resulted in the lower yields of Δ38.1. (4) In animals, viral loads in the spleens of r38.1-infected guinea pigs were higher than those in the spleens of Δ38.1-infected animals. In conclusion, although GPCMV gp38.1 exerts a vMIA-like function, its inhibitory effect was not robust, suggesting the presence of additional inhibitory molecule(s), such as a BAK-specific inhibitor.
Collapse
Affiliation(s)
- Kazuma Noguchi
- Present address: Kaken Pharmaceutical, Tokyo, Japan
- Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| | - Ryuichi Majima
- Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| | - Keita Takahashi
- Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| | - Yoshihiko Iwase
- Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| | - Souichi Yamada
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Keisuke Satoh
- Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| | - Tetsuo Koshizuka
- Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| | - Naoki Inoue
- Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
8
|
Okumura M, Matsuura-Miura M, Makino R, Miura T, Noguchi K, Majima R, Koshizuka T, Inoue N. Enhancement of guinea pig cytomegalovirus infection by two endogenously expressed components of the pentameric glycoprotein complex in epithelial cells. Sci Rep 2020; 10:8530. [PMID: 32444790 PMCID: PMC7244513 DOI: 10.1038/s41598-020-65545-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 05/06/2020] [Indexed: 11/09/2022] Open
Abstract
A better understanding of the mechanisms underlying cell tropisms and the efficiency of viral infection is critical for the development of vaccines and antiviral drugs for viral diseases. In this study, we worked on the entry mechanisms of guinea pig cytomegalovirus and found that endogenous expression of a combination of two components (GP131 and GP133) of the pentameric glycoprotein complex, which is required for non-fibroblast cell tropisms, enhanced viral infection more than 10-fold. In addition, D138A alteration in GP131 increased this enhancement by an additional 10-fold. Although differences in the efficiency of viral infection among various cell types are usually explained by differences in viral entry or traffic processes, our experimental evidences dismissed such possibilities. Instead, our findings that i) endogenous expression of GP131 and GP133 after nuclear delivery of viral DNA still enhanced infection and ii) an HDAC inhibitor overcame the need of the endogenous expression led us to hypothesize a novel mechanism that controls the efficiency of viral infection through the activation of gene expression from viral DNA delivered to the nuclei. Further studies of this unexpected phenomena warrant to understand novel but also general mechanisms for cell tropisms of viral infection and determinants that control infection efficiency.
Collapse
Affiliation(s)
- Misaki Okumura
- Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| | - Miku Matsuura-Miura
- Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan.,Akashi City Hall, Hyogo, Japan
| | - Reina Makino
- Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| | - Takuya Miura
- Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan.,JCR Pharmaceuticals Co., Ltd., Hyogo, Japan
| | - Kazuma Noguchi
- Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| | - Ryuichi Majima
- Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| | - Tetsuo Koshizuka
- Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| | - Naoki Inoue
- Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan.
| |
Collapse
|
9
|
Marsico C, Aban I, Kuo H, James SH, Sanchez PJ, Ahmed A, Arav-Boger R, Michaels MG, Ashouri N, Englund JA, Estrada B, Jacobs RF, Romero JR, Sood SK, Whitworth S, Jester PM, Whitley RJ, Kimberlin DW. Blood Viral Load in Symptomatic Congenital Cytomegalovirus Infection. J Infect Dis 2020; 219:1398-1406. [PMID: 30535363 DOI: 10.1093/infdis/jiy695] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/03/2018] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Viral loads (VLs) frequently are followed during treatment of symptomatic congenital cytomegalovirus disease, but their predictive value is unclear. METHODS Post hoc analysis of 2 antiviral studies was performed. Seventy-three subjects were treated for 6 weeks and 47 subjects were treated for 6 months. Whole blood VL was determined by real-time polymerase chain reaction before and during therapy. RESULTS Higher baseline VL was associated with central nervous system involvement (3.82 log, range 1-5.65 vs 3.32 log, range 1-5.36; P = .001), thrombocytopenia (3.68 log, range 1-5.65 vs 3.43 log, range 1-5.36; P = .03), and transaminitis at presentation (3.73 log, range 1-5.60 vs 3.39 log, range 1-5.65; P = .009), but with overlap in the amount of virus detected between groups. In subjects treated for 6 months, lower VL at presentation correlated with better hearing outcomes at 12 months, but VL breakpoints predictive of hearing loss were not identified. Sustained viral suppression during 6 months of therapy correlated with better hearing outcomes at 6, 12, and 24 months (P = .01, P = .0007, P = .04), but a majority without viral suppression still had improved hearing. CONCLUSIONS In infants with symptomatic congenital cytomegalovirus disease, higher whole blood VL before initiation of antiviral therapy has no clinically meaningful predictive value for long-term outcomes.
Collapse
Affiliation(s)
- Concetta Marsico
- Neonatology Unit, Department of Medical and Surgical Sciences, University of Bologna, Italy
| | - Immaculada Aban
- Department of Biostatistics, Division of Infectious Diseases, University of Alabama at Birmingham
| | - Huichien Kuo
- Department of Biostatistics, Division of Infectious Diseases, University of Alabama at Birmingham
| | - Scott H James
- Department of Pediatrics, Division of Infectious Diseases, University of Alabama at Birmingham
| | - Pablo J Sanchez
- Department of Pediatrics, Divisions of Pediatric Infectious Diseases and Neonatology, Nationwide Children's Hospital - Ohio State University College of Medicine, Columbus
| | - Amina Ahmed
- Department of Pediatrics, Carolinas Medical Center, Charlotte, North Carolina
| | | | - Marian G Michaels
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pennsylvania
| | - Negar Ashouri
- Infectious Diseases, CHOC Children's Hospital, Orange, California
| | | | | | | | | | - Sunil K Sood
- Steven and Alexandra Cohen Children's Medical Center, New Hyde Park, New York
| | | | - Penelope M Jester
- Department of Pediatrics, Division of Infectious Diseases, University of Alabama at Birmingham
| | - Richard J Whitley
- Department of Pediatrics, Division of Infectious Diseases, University of Alabama at Birmingham
| | - David W Kimberlin
- Department of Pediatrics, Division of Infectious Diseases, University of Alabama at Birmingham
| | | |
Collapse
|
10
|
Tsuprun V, Keskin N, Schleiss MR, Schachern P, Cureoglu S. Cytomegalovirus-induced pathology in human temporal bones with congenital and acquired infection. Am J Otolaryngol 2019; 40:102270. [PMID: 31402062 DOI: 10.1016/j.amjoto.2019.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/31/2019] [Accepted: 08/02/2019] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Publications on histopathology of human temporal bones with cytomegalovirus (CMV) infection are limited. We aim to determine histopathology of the inner ears and the middle ears in human temporal bones with congenital and acquired CMV infections. METHODS Temporal bones from 2 infants with congenital and 2 adults with acquired CMV infection were evaluated by light microscopy. RESULTS Two infants with congenital CMV infection showed striking pathological changes in the inner ear. There was a hypervascularization of the stria vascularis in the cochlea of the first infant, but no obvious loss of outer and inner hair cells was seen in the organ of Corti. However, cytomegalic cells and a loss of outer hair cells were found in the cochlea of the second infant. The vestibular organs of both infants showed cytomegalic cells, mostly located on dark cells. There was a loss of type I and type II hair cells in the macula of the saccule and utricle. Loss of hair cells and degeneration of nerve fibers was also seen in the semicircular canals. Both infants with congenital infection showed abundant inflammatory cells and fibrous structures in the middle ear cavity. No evidence of cytomegalic cells and hair cell loss was found in the cochlea or vestibular labyrinth in acquired CMV infection. CONCLUSIONS In two infants with congenital CMV infection, the cochlea, vestibule, and middle ear were highly affected. Temporal bones of adult donors with acquired viral infection showed histological findings similar to donors of the same age without ear disease.
Collapse
|
11
|
Prediction of the Outcome of Cochlear Implantation in the Patients with Congenital Cytomegalovirus Infection based on Magnetic Resonance Imaging Characteristics. J Clin Med 2019; 8:jcm8020136. [PMID: 30682778 PMCID: PMC6406882 DOI: 10.3390/jcm8020136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/14/2019] [Accepted: 01/21/2019] [Indexed: 12/03/2022] Open
Abstract
The goal of this study was to elucidate radiologic biomarker that can predict the outcome of cochlear implantation (CI) in congenital cytomegalovirus (cCMV) related deafness. A retrospective survey of speech perception after CI and an evaluation of brain magnetic resonance imaging (MRI) findings were performed in 10 cochlear implantees with cCMV-related prelingual deafness. Specifically, a special attention was paid to the degree of white matter (WM) abnormality shown in brain MRI, which was used to divide our cohort into two groups: The mild and severe pathology groups. Age-matched prelingual deaf patients with idiopathic sensorineural hearing loss were selected as controls. Subjects in mild pathology groups showed higher a Category of Auditory Performance (CAP) score (5.2 ± 0.8) than those with severe pathologies (3.4 ± 1.5) (P = 0.041). Importantly, speech performance from subjects with mild pathology was comparable to that of the control group (mean CAP score of 5.2 ± 0.8 vs. 5.1 ± 1.2) (P = 0.898). Mild pathologies related to the limited WM lesion in MRI not accompanied by severe MRI pathologies, such as diffuse WM abnormality, myelination delay, ventriculomegaly, migration abnormality, and cerebellar hypoplasia, can be tolerated and do not adversely affect the CI outcome in cCMV deafness.
Collapse
|
12
|
Roles of GP33, a guinea pig cytomegalovirus-encoded G protein-coupled receptor homolog, in cellular signaling, viral growth and inflammation in vitro and in vivo. PLoS Pathog 2018; 14:e1007487. [PMID: 30571759 PMCID: PMC6319746 DOI: 10.1371/journal.ppat.1007487] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 01/04/2019] [Accepted: 11/27/2018] [Indexed: 11/23/2022] Open
Abstract
Cytomegaloviruses (CMVs) encode cellular homologs to evade host immune functions. In this study, we analyzed the roles of GP33, a guinea pig CMV (GPCMV)-encoded G protein-coupled receptor (GPCR) homolog, in cellular signaling, viral growth and pathogenesis. The cDNA structure of GP33 was determined by RACE. The effects of GP33 on some signaling pathways were analyzed in transient transfection assays. The redET two-step recombination system for a BAC containing the GPCMV genome was used to construct a mutant GPCMV containing an early stop codon in the GP33 gene (Δ33) and a rescued GPCMV (r33). We found the following: 1) GP33 activated the CRE- and NFAT-, but not the NFκB-mediated signaling pathway. 2) GP33 was dispensable for infection in tissue cultures and in normal animals. 3) In pregnant animals, viral loads of r33 in the livers, lungs, spleens, and placentas at 6 days post-infection were higher than those of Δ33, although the viruses were cleared by 3 weeks post-infection. 4) The presence of GP33 was associated with frequent lesions, including alveolar hemorrhage in the lungs, and inflammation in the lungs, livers, and spleens of the dams. Our findings suggest that GP33 has critical roles in the pathogenesis of GPCMV during pregnancy. We hypothesize that GP33-mediated signaling activates cytokine secretion from the infected cells, which results in inflammation in some of the maternal organs and the placentas. Alternatively, GP33 may facilitate transient inflammation that is induced by the chemokine network specific to the pregnancy. Cytomegalovirus (CMV) is a major pathogen that causes congenital diseases, including birth defects and developmental abnormalities in newborns. Better understanding of the immune evasion mechanisms may open the way to the development of new types of live attenuated vaccines for congenital CMV infection. In contrast to murine and rat CMVs, guinea pig CMV (GPCMV) causes infection in utero, which makes GPCMV animal models a useful tool for understanding the pathogenesis of congenital infection and evaluation of vaccine strategies. By constructing a GPCMV mutant lacking GP33, a viral G protein-coupled receptor homolog, this study found that GP33 was involved in the induction of significant inflammatory responses in pregnant but not in normal animals. As GP33 activated the NFAT- and CRE-, but not the NFκB-signal pathway, it is plausible that GP33 enhanced cytokine expression, which results in pathogenic outcomes in the maternal organs and placentas.
Collapse
|
13
|
Diamond DJ, LaRosa C, Chiuppesi F, Contreras H, Dadwal S, Wussow F, Bautista S, Nakamura R, Zaia JA. A fifty-year odyssey: prospects for a cytomegalovirus vaccine in transplant and congenital infection. Expert Rev Vaccines 2018; 17:889-911. [PMID: 30246580 PMCID: PMC6343505 DOI: 10.1080/14760584.2018.1526085] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/17/2018] [Indexed: 02/08/2023]
Abstract
INTRODUCTION It has been almost fifty years since the Towne strain was used by Plotkin and collaborators as the first vaccine candidate for cytomegalovirus (CMV). While that approach showed partial efficacy, there have been a multitude of challenges to improve on the promise of a CMV vaccine. Efforts have been dichotomized into a therapeutic vaccine for patients with CMV-infected allografts, either stem cells or solid organ, and a prophylactic vaccine for congenital infection. AREAS COVERED This review will evaluate research prospects for a therapeutic vaccine for transplant recipients that recognizes CMV utilizing primarily T cell responses. Similarly, we will provide an extensive discussion on attempts to develop a vaccine to prevent the manifestations of congenital infection, based on eliciting a humoral anti-CMV protective response. The review will also describe newer developments that have upended the efforts toward such a vaccine through the discovery of a second pathway of CMV infection that utilizes an alternative receptor for entry using a series of antigens that have been determined to be important for prevention of infection. EXPERT COMMENTARY There is a concerted effort to unify separate therapeutic and prophylactic vaccine strategies into a single delivery agent that would be effective for both transplant-related and congenital infection.
Collapse
Affiliation(s)
- Don J. Diamond
- Department of Experimental Therapeutics, Beckman Research
Institute of City of Hope, Duarte, CA
| | - Corinna LaRosa
- Department of Experimental Therapeutics, Beckman Research
Institute of City of Hope, Duarte, CA
| | - Flavia Chiuppesi
- Department of Experimental Therapeutics, Beckman Research
Institute of City of Hope, Duarte, CA
| | - Heidi Contreras
- Department of Experimental Therapeutics, Beckman Research
Institute of City of Hope, Duarte, CA
| | - Sanjeet Dadwal
- Department of Medical Specialties, City of Hope National
Medical Center, Duarte, CA
| | - Felix Wussow
- Department of Experimental Therapeutics, Beckman Research
Institute of City of Hope, Duarte, CA
| | - Supriya Bautista
- Department of Experimental Therapeutics, Beckman Research
Institute of City of Hope, Duarte, CA
| | - Ryotaro Nakamura
- Department of Hematology & Hematopoetic Cell
Transplantation, City of Hope National Medical Center, Duarte, CA
| | - John A. Zaia
- Center for Gene Therapy, Hematological Malignancy and Stem
Cell Transplantation Institute, City of Hope, Duarte, CA
| |
Collapse
|
14
|
Miura T, Makino R, Yamada K, Matsuura M, Okumura M, Yamada S, Watanabe S, Inoue N. Differences in the effects of mutations in GP131, a guinea pig cytomegalovirus homologue of pentameric complex component UL130, on macrophage and epithelial cell infection. J Gen Virol 2018; 99:1425-1431. [PMID: 30113297 DOI: 10.1099/jgv.0.001137] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
As congenital cytomegalovirus (CMV) infection is the major cause of developmental abnormalities in children, the development of effective vaccines is critical to public health. Recent studies have demonstrated that the pentameric complex (Pentamer) of glycoproteins, which is required for human CMV infection of endothelial and epithelial cells, could be a potent vaccine antigen. As guinea pig CMV (GPCMV) infects congenitally and encodes homologues of all Pentamer components, GPCMV models are considered to be useful for the development of vaccine strategies. Here, to clarify the precise requirement of GP131, one of the GPCMV Pentamer components, for the infection of epithelial cells and macrophages, we prepared several mutants with a charged amino acid-to-alanine alteration in GP131 and found some differences in the effects of the mutations on the infection of the two cell types, suggesting the existence of cell type-dependent recognition or function of Pentamer in GPCMV infection.
Collapse
Affiliation(s)
- Takuya Miura
- 1Laboratory of Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan.,†Present address: JCR Pharmaceuticals, Hyogo, Japan
| | - Reina Makino
- 1Laboratory of Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| | - Kouhei Yamada
- 1Laboratory of Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan.,‡Present address: Nagoya City Hall, Aichi, Japan
| | - Miku Matsuura
- 1Laboratory of Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan.,§Present address: Hanshin Dispensing Pharmacy, Hyogo, Japan
| | - Misaki Okumura
- 1Laboratory of Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| | - Souichi Yamada
- 2Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Shinji Watanabe
- 1Laboratory of Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan.,¶Present address: Eiken Chemical, Tochigi, Japan
| | - Naoki Inoue
- 1Laboratory of Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
15
|
Abstract
The development of a cytomegalovirus (CMV) vaccine has become a top priority due to its potential cost-effectiveness and associated public health benefits. However, there are a number of challenges facing vaccine development including the following: (1) CMV has many mechanisms for evading immune responses , and natural immunity is not perfect, (2) the immune correlates for protection are unclear, (3) a narrow range of CMV hosts limits the value of animal models, and (4) the placenta is a specialized organ formed transiently and its immunological status changes with time. In spite of these limitations, several types of CMV vaccine candidate, including live-attenuated, DISC , subunit, DNA, vectored, and peptide vaccines, have been developed or are currently under development. The recognition of the pentameric complex as the major neutralization target and identification of various strategies to block viral immune response evasion mechanisms have opened new avenues to CMV vaccine development. Here, we discuss the immune correlates for protection, the characteristics of the various vaccine candidates and their clinical trials, and the relevant animal models.
Collapse
|
16
|
Itell HL, Nelson CS, Martinez DR, Permar SR. Maternal immune correlates of protection against placental transmission of cytomegalovirus. Placenta 2017; 60 Suppl 1:S73-S79. [PMID: 28456432 PMCID: PMC5650553 DOI: 10.1016/j.placenta.2017.04.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 04/07/2017] [Accepted: 04/13/2017] [Indexed: 02/02/2023]
Abstract
Human cytomegalovirus (HCMV) is the most common congenitally transmitted pathogen worldwide, impacting an estimated 1 million newborns annually. In a subset of infected infants, congenital HCMV causes severe, long-lasting sequelae, including deafness, microcephaly, neurodevelopmental delay, and even death. Accordingly, a maternal vaccine to prevent congenital HCMV infection continues to be a top public health priority. Nevertheless, all vaccines tested to date have failed to meet clinical trial endpoints. Maternal immunity provides partial protection against congenital HCMV transmission, as vertical transmission from seropositive mothers is relatively rare. Therefore, an understanding of the maternal immune correlates of protection against HCMV congenital infection will be critical to inform design of an efficacious maternal vaccine. This review summarizes our understanding of the innate and adaptive immune correlates of protection against congenital transmission of HCMV, and discusses the advantages and applications of a novel nonhuman primate model of congenital CMV transmission to aid in rational vaccine design and evaluation.
Collapse
Affiliation(s)
- Hannah L Itell
- Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Cody S Nelson
- Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - David R Martinez
- Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Sallie R Permar
- Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA; Department of Pediatrics, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
17
|
An Ex vivo culture model for placental cytomegalovirus infection using slices of Guinea pig placental tissue. Placenta 2015; 37:85-8. [PMID: 26625961 DOI: 10.1016/j.placenta.2015.10.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 10/15/2015] [Accepted: 10/22/2015] [Indexed: 01/01/2023]
Abstract
Congenital infection with human cytomegalovirus (CMV) through the placenta is one of the major causes of birth and developmental abnormalities. Guinea pig CMV (GPCMV) causes in utero infection, which makes its animal models useful for studies on congenital diseases. Here, we established an ex vivo culture method for tissue slices prepared from guinea pig placentas and demonstrated that viral spread in the model resembles those in the placenta of GPCMV-infected animals and that the infection is independent of the pentameric glycoprotein complex for endothelial/epithelial cell tropism. Thus, this model affords a useful tool for pathobiological studies on CMV placental infection.
Collapse
|
18
|
Ikuta K, Ogawa H, Hashimoto H, Okano W, Tani A, Sato E, Kosugi I, Kobayashi T, Omori K, Suzutani T. Restricted infection of murine cytomegalovirus (MCMV) in neonatal mice with MCMV-induced sensorineural hearing loss. J Clin Virol 2015. [PMID: 26209396 DOI: 10.1016/j.jcv.2015.06.083] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND Congenital infection with human Cytomegalovirus (HCMV) is known to be a causative agent of sensorineural hearing loss (SNHL). OBJECTIVES To clarify the nongenetic etiology of SNHL by identifying the Cytomegalovirus (CMV)-infected region in the cochleae. STUDY DESIGN We established an animal model of SNHL by injecting neonatal Balb/c mice with intracerebral murine Cytomegalovirus (MCMV) within 24h after delivery. RESULTS At 3 weeks of age, unilateral and bilateral SNHL were observed in 24% (5/21) and 29% (6/21) of the mice, respectively. SNHL thereafter progressed, with 79% of mice developing bilateral SNHL by 6 weeks of age. MCMV antigens and DNA were detected in the spiral ganglion, and cells surrounding the meninges and scala tympani at 1 week of age. However, both MCMV antigens and DNA had completely disappeared by 2 weeks of age. It is possible that the MCMV reached the spiral ganglion via cerebrospinal fluid as the result of meningitis, as the stria vascularis was found to be MCMV antigen negative. Myosin VI expression in the outer hair cells was lost at 3 weeks of age. MCMV and myosin VI expression disappeared before and during SNHL progression, respectively. CONCLUSIONS There was a definite lag time between the period in which MCMV antigens/DNA-positive cells were observed and that in which SNHL developed and myosin VI-negative hair cells were observed. Further study is needed to explore the role of MCMV in the loss of myosin VI expression in the outer hair cells.
Collapse
Affiliation(s)
- Kazufumi Ikuta
- Department of Microbiology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Hiroshi Ogawa
- Department of Otolaryngology, Fukushima Medical University School of Medicine, Fukushima, Japan; Department of Otolaryngology, Aizu Medical Center, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Hiromi Hashimoto
- Department of Microbiology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Wataru Okano
- Department of Otolaryngology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Akiko Tani
- Department of Otolaryngology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Etsuko Sato
- Department of Otolaryngology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Isao Kosugi
- Department of Pathology II, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Takahiro Kobayashi
- Department of Microbiology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Koichi Omori
- Department of Otolaryngology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Tatsuo Suzutani
- Department of Microbiology, Fukushima Medical University School of Medicine, Fukushima, Japan
| |
Collapse
|
19
|
Bradford RD, Yoo YG, Golemac M, Pugel EP, Jonjic S, Britt WJ. Murine CMV-induced hearing loss is associated with inner ear inflammation and loss of spiral ganglia neurons. PLoS Pathog 2015; 11:e1004774. [PMID: 25875183 PMCID: PMC4395355 DOI: 10.1371/journal.ppat.1004774] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 03/02/2015] [Indexed: 01/22/2023] Open
Abstract
Congenital human cytomegalovirus (HCMV) occurs in 0.5–1% of live births and approximately 10% of infected infants develop hearing loss. The mechanism(s) of hearing loss remain unknown. We developed a murine model of CMV induced hearing loss in which murine cytomegalovirus (MCMV) infection of newborn mice leads to hematogenous spread of virus to the inner ear, induction of inflammatory responses, and hearing loss. Characteristics of the hearing loss described in infants with congenital HCMV infection were observed including, delayed onset, progressive hearing loss, and unilateral hearing loss in this model and, these characteristics were viral inoculum dependent. Viral antigens were present in the inner ear as were CD3+ mononuclear cells in the spiral ganglion and stria vascularis. Spiral ganglion neuron density was decreased after infection, thus providing a mechanism for hearing loss. The lack of significant inner ear histopathology and persistence of inflammation in cochlea of mice with hearing loss raised the possibility that inflammation was a major component of the mechanism(s) of hearing loss in MCMV infected mice. Congenital infection with human cytomegalovirus (HCMV) is the most common viral infection of the fetus and occurs in 0.5–2.0% of all live births in most regions in the world. Infection of the fetus can result in a spectrum of end-organ disease, including long term damage to the central nervous system (CNS). Although less than 10% of infected infants exhibit clinical evidence of end-organ disease, up to 10% of the total number of infected infants develop hearing loss. Mechanisms of disease leading to hearing loss are poorly understood because of the limited availability of pathological specimens and accessibility of the inner ear. Existing small animal models fail to recapitulate many features of this infection of the inner ear. In this report we describe a mouse model in which newborn animals infected peripherally with murine CMV develop hearing loss following hematogenous spread of virus to the inner ear. Hearing loss occurs in 30–50% of animals and characteristics of hearing loss in infants with congenital HCMV infection, including delayed onset of hearing loss, progressive hearing loss, and unilateral hearing loss were present in infected mice. Our findings suggest that host derived inflammatory responses and not direct virus-mediated cytopathology are responsible for hearing loss. Findings from this study provide insight into potential mechanisms of hearing loss in infants with congenital HCMV infection.
Collapse
Affiliation(s)
- Russell D. Bradford
- Department of Pediatrics, University of Alabama School of Medicine, Birmingham, Alabama, United States of America
| | - Young-Gun Yoo
- Department of Pediatrics, University of Alabama School of Medicine, Birmingham, Alabama, United States of America
| | - Mijo Golemac
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Ester Pernjak Pugel
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Stipan Jonjic
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - William J. Britt
- Department of Pediatrics, University of Alabama School of Medicine, Birmingham, Alabama, United States of America
- Department of Microbiology, University of Alabama School of Medicine, Birmingham, Alabama, United States of America
- Department of Neurobiology, University of Alabama School of Medicine, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
20
|
Weisblum Y, Panet A, Haimov-Kochman R, Wolf DG. Models of vertical cytomegalovirus (CMV) transmission and pathogenesis. Semin Immunopathol 2014; 36:615-25. [PMID: 25291972 DOI: 10.1007/s00281-014-0449-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 09/29/2014] [Indexed: 02/04/2023]
Abstract
Despite the considerable clinical impact of congenital human cytomegalovirus (HCMV) infection, the mechanisms of maternal-fetal transmission and the resultant placental and fetal damage are largely unknown. Here, we discuss animal models for the evaluation of CMV vaccines and virus-induced pathology and particularly explore surrogate human models for HCMV transmission and pathogenesis in the maternal-fetal interface. Studies in floating and anchoring placental villi and more recently, ex vivo modeling of HCMV infection in integral human decidual tissues, provide unique insights into patterns of viral tropism, spread, and injury, defining the outcome of congenital infection, and the effect of potential antiviral interventions.
Collapse
Affiliation(s)
- Yiska Weisblum
- Clinical Virology Unit, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | | | | | | |
Collapse
|
21
|
Li X, Shi X, Qiao Y, Xu K, Zeng L, Wang C, Xu Z, Niu H. Observation of permeability of blood-labyrinth barrier during cytomegalovirus-induced hearing loss. Int J Pediatr Otorhinolaryngol 2014; 78:995-9. [PMID: 24814236 DOI: 10.1016/j.ijporl.2014.03.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 03/17/2014] [Accepted: 03/18/2014] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Congenital cytomegalovirus (CMV) infection is the most common infectious cause of sensorineural hearing loss in children. This study aims to investigate the pathogenesis CMV-induced hearing loss from the view of integrity of blood-labyrinth-barrier (BLB). METHODS Newborn BALB/c mice were randomly divided into three groups (n=22, respectively): CMV group, control group and normal group. The CMV group and control group were intracerebrally injected with equal volume (15 μl) of murine CMV (MCMV; 10(4)IU/0.1 ml) and PBS, respectively, and normal group did not receive any treatment. After three weeks, auditory-evoked brainstem response was assessed, and permeability of BLB was evaluated by Evans blue method. Means between groups were compared using t-test. RESULTS We observed that mice injected with MCMV had a hearing loss and it was connected with the permeability changes of BLB. Besides, using hematoxylin-eosin staining, we noticed hyperaemia in stria vascularis and spiral ligament and bleeding in scala vestibule and scala tympani in CMV group. CONCLUSION All these data indicated the possible association between CMV-induced hearing loss and BLB dysfunction with the characteristics of inflammation. Our data provide a possible path to investigate the mechanism of CMV-induced hearing damage.
Collapse
Affiliation(s)
- Xuanyi Li
- Department of Otolaryngology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou 221006, China
| | - Xi Shi
- Institute of Audiology and Speech Science of Xuzhou Medical College, Xuzhou 221006, China
| | - Yuehua Qiao
- Department of Otolaryngology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou 221006, China; Institute of Audiology and Speech Science of Xuzhou Medical College, Xuzhou 221006, China.
| | - Kailin Xu
- Laboratory of Transplantation Immunity of Xuzhou Medical College, Xuzhou 221006, China
| | - Lingyu Zeng
- Laboratory of Transplantation Immunity of Xuzhou Medical College, Xuzhou 221006, China
| | - Caiji Wang
- Department of Otolaryngology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou 221006, China
| | - Zhou Xu
- School of Basic Education Science of Xuzhou Medical College, Xuzhou 221006, China
| | - Haichen Niu
- Institute of Audiology and Speech Science of Xuzhou Medical College, Xuzhou 221006, China
| |
Collapse
|
22
|
Yamada S, Fukuchi S, Hashimoto K, Fukui Y, Tsuda M, Kataoka M, Katano H, Inoue N. Guinea pig cytomegalovirus GP129/131/133, homologues of human cytomegalovirus UL128/130/131A, are necessary for infection of monocytes and macrophages. J Gen Virol 2014; 95:1376-1382. [DOI: 10.1099/vir.0.064527-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The GP129, GP131 and GP133 genes of guinea pig cytomegalovirus (GPCMV) are homologues of human cytomegalovirus UL128, UL130 and UL131A, respectively, which are essential for infection of endothelial and epithelial cells, and for viral transmission to leukocytes. Our previous study demonstrated that a GPCMV strain lacking the 1.6 kb locus that contains the GP129, GP131 and GP133 genes had a growth defect in animals. Here, we demonstrated that the WT strain, but not the 1.6 kb-deleted strain, formed capsids in macrophages prepared from the peritoneal fluid. To understand the mechanism, we prepared GPCMV strains defective in each of GP129, GP131 and GP133, and found that they were all essential for the infection of peritoneal, splenic and PBMC-derived macrophages/monocytes, and for expression of immediate-early antigens in the macrophages/monocytes, although they were dispensable for infection of fibroblasts. Monocyte/macrophage tropism could be one of the important determinants for viral dissemination in vivo.
Collapse
Affiliation(s)
- Souichi Yamada
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Saki Fukuchi
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Kaede Hashimoto
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yoshiko Fukui
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Mihoko Tsuda
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Michiyo Kataoka
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Harutaka Katano
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Naoki Inoue
- Department of Microbiology and Immunology, Gifu Pharmaceutical University, Gifu, Japan
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
23
|
Auerbach MR, Yan D, Vij R, Hongo JA, Nakamura G, Vernes JM, Meng YG, Lein S, Chan P, Ross J, Carano R, Deng R, Lewin-Koh N, Xu M, Feierbach B. A neutralizing anti-gH/gL monoclonal antibody is protective in the guinea pig model of congenital CMV infection. PLoS Pathog 2014; 10:e1004060. [PMID: 24722349 PMCID: PMC3983071 DOI: 10.1371/journal.ppat.1004060] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 02/25/2014] [Indexed: 11/29/2022] Open
Abstract
Human cytomegalovirus (HCMV) is the most common cause of congenital virus infection. Congenital HCMV infection occurs in 0.2–1% of all births, and causes birth defects and developmental abnormalities, including sensorineural hearing loss and developmental delay. Several key studies have established the guinea pig as a tractable model for the study of congenital HCMV infection and have shown that polyclonal antibodies can be protective [1]–[3]. In this study, we demonstrate that an anti-guinea pig CMV (GPCMV) glycoprotein H/glycoprotein L neutralizing monoclonal antibody protects against fetal infection and loss in the guinea pig. Furthermore, we have delineated the kinetics of GPCMV congenital infection, from maternal infection (salivary glands, seroconversion, placenta) to fetal infection (fetus and amniotic fluid). Our studies support the hypothesis that a neutralizing monoclonal antibody targeting an envelope GPCMV glycoprotein can protect the fetus from infection and may shed light on the therapeutic intervention of HCMV congenital infection in humans. Human cytomegalovirus (HCMV) is the most common cause of congenital virus infection and causes developmental abnormalities, including hearing loss and developmental delay. Although there is no therapy for congenital HCMV disease, there is evidence from both human and animal studies that antibodies can have efficacy in this setting. Such studies have focused exclusively on polyclonal antibodies, in which the targets of protective antibodies are unknown. Guinea pigs have been used as a model of human maternal fetal transmission of infection because of similarities in placental anatomy between human and guinea pig. Furthermore, guinea pig CMV (GPCMV) has been demonstrated to cross the placenta and cause fetal infection and loss, similar to the effects of infection with HCMV. However, the kinetics of maternal and fetal infection in this model has not been carefully investigated. In this work, we have delineated the kinetics of maternal to fetal infection and found that congenital infection is rapid following maternal infection. Importantly, we demonstrate that a monoclonal antibody against a protein critical for viral entry protects pregnant guinea pigs against fetal infection. Thus, our studies may be informative for development of a therapeutic intervention to treat congenital HCMV infection in humans.
Collapse
Affiliation(s)
- Marcy R. Auerbach
- Department of Infectious Diseases, Genentech, South San Francisco, California, United States of America
| | - Donghong Yan
- Department of Translational Immunology, Genentech, South San Francisco, California, United States of America
| | - Rajesh Vij
- Department of Antibody Engineering, Genentech, South San Francisco, California, United States of America
| | - Jo-Anne Hongo
- Department of Antibody Engineering, Genentech, South San Francisco, California, United States of America
| | - Gerald Nakamura
- Department of Antibody Engineering, Genentech, South San Francisco, California, United States of America
| | - Jean-Michel Vernes
- Department of Biochemical and Cellular Pharmacology, Genentech, South San Francisco, California, United States of America
| | - Y. Gloria Meng
- Department of Biochemical and Cellular Pharmacology, Genentech, South San Francisco, California, United States of America
| | - Samantha Lein
- Department of Antibody Engineering, Genentech, South San Francisco, California, United States of America
| | - Pamela Chan
- Department of Biochemical and Cellular Pharmacology, Genentech, South San Francisco, California, United States of America
| | - Jed Ross
- Department of Biomedical Imaging, Genentech, South San Francisco, California, United States of America
| | - Richard Carano
- Department of Biomedical Imaging, Genentech, South San Francisco, California, United States of America
| | - Rong Deng
- Department of Clinical Pharmacology, Genentech, South San Francisco, California, United States of America
| | - Nicholas Lewin-Koh
- Department of Biostatistics, Genentech, South San Francisco, California, United States of America
| | - Min Xu
- Department of Translational Immunology, Genentech, South San Francisco, California, United States of America
| | - Becket Feierbach
- Department of Infectious Diseases, Genentech, South San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
24
|
Schleiss MR. Developing a Vaccine against Congenital Cytomegalovirus (CMV) Infection: What Have We Learned from Animal Models? Where Should We Go Next? Future Virol 2013; 8:1161-1182. [PMID: 24523827 DOI: 10.2217/fvl.13.106] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Congenital human cytomegalovirus (HCMV) infection can lead to long-term neurodevelopmental sequelae, including mental retardation and sensorineural hearing loss. Unfortunately, CMVs are highly adapted to their specific species, precluding the evaluation of HCMV vaccines in animal models prior to clinical trials. Several species-specific CMVs have been characterized and developed in models of pathogenesis and vaccine-mediated protection against disease. These include the murine CMV (MCMV), the porcine CMV (PCMV), the rhesus macaque CMV (RhCMV), the rat CMV (RCMV), and the guinea pig CMV (GPCMV). Because of the propensity of the GPCMV to cross the placenta, infecting the fetus in utero, it has emerged as a model of particular interest in studying vaccine-mediated protection of the fetus. In this paper, a review of these various models, with particular emphasis on the value of the model in the testing and evaluation of vaccines against congenital CMV, is provided. Recent exciting developments and advances in these various models are summarized, and recommendations offered for high-priority areas for future study.
Collapse
Affiliation(s)
- Mark R Schleiss
- University of Minnesota Medical School Center for Infectious Diseases and Microbiology Translational Research Department of Pediatrics Division of Pediatric Infectious Diseases and Immunology 2001 6 Street SE Minneapolis, MN 55455-3007
| |
Collapse
|
25
|
Peng LH, Liu S, Xu SY, Chen L, Shan YH, Wei W, Liang WQ, Gao JQ. Inhibitory effects of salidroside and paeonol on tyrosinase activity and melanin synthesis in mouse B16F10 melanoma cells and ultraviolet B-induced pigmentation in guinea pig skin. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2013; 20:1082-1087. [PMID: 23746955 DOI: 10.1016/j.phymed.2013.04.015] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 04/05/2013] [Accepted: 04/21/2013] [Indexed: 06/02/2023]
Abstract
Salidroside, the major active component of Rhodiola rosea, a herb with antioxidant, free radical scavenging and tyrosinase inhibitory effects, has been recently reported in protecting the kerationcytes from the UV radiation, suggesting the potential of this component in depigmentation. Paeonol is isolated from Moutan Cortex Radicis with anti-inflammation/microbial activities, was reported to induce the down-regulation of microphthalmia-associated transcription factor and subsequently tyrosinase. To testify the potential of these compounds as melanin formation inhibitors for hyperpigmentation therapy, the influence of salidroside and paeonol on pigmentation was investigated. With arbutin as a positive control, salidroside and paeonol were evaluated for their inhibitory effect on the cell viability, tyrosinase activity and melanin synthesis in B16F10 melanoma cells, as well as their effects in UVB-induced hyperpigmentation in brown guinea pig skins. It was demonstrated that the significant inhibition of salidroside (33.0%) and paeonol (22.2-30.9%) on the tyrosinase activity is slightly lower than that of arbutin (18.4-44.7%). However, salidroside exhibited the dose-dependent inhibition (30.6-42.0%) in melanin synthesis at a low concentration of 100 μM, paeonol and arbutin expressed inhibition rates of 27.4-37.2% and 25.8-45.6% within 500-1000 μM. The in vivo topical application of these compounds was demonstrated to obviously decrease the hyperpigmentation on UVB stimulated guinea pig skin. This study provided the original evidence for the salidroside and paeonol as therapeutic agents for pigmentation disorder and skin lightening, with further clinical investigation of these compounds in the field of depigmentation was suggested.
Collapse
Affiliation(s)
- Li-Hua Peng
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, PR China
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Llorente AM, Castillo CL. Congenital cytomegalovirus infection in fraternal twins: a longitudinal case study examining neurocognitive and neurobehavioral correlates. APPLIED NEUROPSYCHOLOGY-CHILD 2013; 1:63-73. [PMID: 23428280 DOI: 10.1080/21622965.2012.677605] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Cytomegalovirus (CMV) is the most ubiquitous member of the herpes virus family and is the leading cause of congenital (vertical) infection in newborns (Fowler, Stagno, & Pass, 2003; Llorente, Steigmeyer, Cooper, Rivers, & Gazley, 2011; Noyola et al., 2000; Steigmeyer & Llorente, 2010). CMV is related to the group of viruses capable of causing more pernicious infectious diseases, such as chicken pox (Santos de Barona, 1998). Although the virus generally remains dormant, individuals whose symptoms are clinically apparent often are dramatically affected. Common symptomatic characteristics of the virus include microcephaly, jaundice, liver-spleen infections, pneumonia, cardiac anomalies, chorioretinitis, vision loss, sensory-neural hearing loss, mental retardation, and mononucleosis (Demmler, 1991; Kashden, Frison, Fowler, Pass, & Boll, 1998; Noyola et al., 2000; Pass, 2005; Santos de Barona). The prognosis of individuals with CMV is highly variable, and the prognosis of individuals with congenital CMV can usually be determined based on the extent of infection at birth. The purpose of this investigation is to present longitudinal results of neuropsychological evaluation of two dizygotic twin sets (one twin of each set is asymptomatic CMV-positive and the other is uninfected) who were reared in the same environment. In addition, the present findings are discussed within the context of emerging murine and other animal analogues of CMV as well as within the extant CMV literature.
Collapse
Affiliation(s)
- Antolin M Llorente
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21209, USA.
| | | |
Collapse
|
27
|
Hashimoto K, Yamada S, Katano H, Fukuchi S, Sato Y, Kato M, Yamaguchi T, Moriishi K, Inoue N. Effects of immunization of pregnant guinea pigs with guinea pig cytomegalovirus glycoprotein B on viral spread in the placenta. Vaccine 2013; 31:3199-205. [PMID: 23684839 DOI: 10.1016/j.vaccine.2013.04.078] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 04/24/2013] [Indexed: 11/28/2022]
Abstract
BACKGROUND Cytomegalovirus (CMV) is the most common cause of congenital virus infection. Infection of guinea pigs with guinea pig CMV (GPCMV) can provide a useful model for the analysis of its pathogenesis as well as for the evaluation of vaccines. Although glycoprotein B (gB) vaccines have been reported to reduce the incidence and mortality of congenital infection in human clinical trials and guinea pig animal models, the mechanisms of protection remain unclear. METHODS To understand the gB vaccine protection mechanisms, we analyzed the spread of challenged viruses in the placentas and fetuses of guinea pig dams immunized with recombinant adenoviruses expressing GPCMV gB and β-galactosidase, rAd-gB and rAd-LacZ, respectively. RESULTS Mean body weight of the fetuses in the dams immunized with rAd-LacZ followed by GPCMV challenge 3 weeks after immunization was 78% of that observed for dams immunized with rAd-gB. Under conditions in which congenital infection occurred in 75% of fetuses in rAd-LacZ-immunized dams, only 13% of fetuses in rAd-gB-immunized dams were congenitally infected. The placentas were infected less frequently in the gB-immunized animals. In the placentas of the rAd-LacZ- and rAd-gB-immunized animals, CMV early antigens were detected mainly in the spongiotrophoblast layer. Focal localization of viral antigens in the spongiotrophoblast layer suggests cell-to-cell viral spread in the placenta. In spite of a similar level of antibodies against gB and avidity indices among fetuses in each gB-immunized dam, congenital infection was sometimes observed in a littermate fetus. In such infected fetuses, CMV spread to most organs. CONCLUSIONS Our results suggest that antibodies against gB protected against infection mainly at the interface of the placenta rather than from the placenta to the fetus. The development of strategies to block cell-to-cell viral spread in the placenta is, therefore, required for effective protection against congenital CMV infection.
Collapse
Affiliation(s)
- Kaede Hashimoto
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Characterization of the guinea pig CMV gH/gL/GP129/GP131/GP133 complex in infection and spread. Virology 2013; 441:75-84. [PMID: 23562482 DOI: 10.1016/j.virol.2013.03.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 02/15/2013] [Accepted: 03/13/2013] [Indexed: 11/20/2022]
Abstract
In human cytomegalovirus (HCMV), the UL128-131A locus plays an essential role in cellular tropism and spread. Here, we report the complete annotation of the GP129-133 locus from guinea pig cytomegalovirus (GPCMV) and the discovery of the UL131A homolog, named GP133. We have found that similar to HCMV the GP129-133 proteins form a pentamer complex with the GPCMV glycoproteins gH and gL. In addition, we find that the GP129-133 proteins play a critical role in entry as the GP129-133 deletion mutant shows a defect in both endothelial and fibroblast cell entry. Although the GP129-133 deletion strain can propagate in vitro, we find that the deletion fails to spread in vivo. Interestingly, the wildtype strain can spontaneously give rise to the GP129-133 deletion strain during in vivo spread, suggesting genetic instability at this locus.
Collapse
|
29
|
Kato K, Otake H, Tagaya M, Takahashi Y, Ito Y, Hama A, Muramatsu H, Kojima S, Naganawa S, Nakashima T. Progressive hearing loss following acquired cytomegalovirus infection in an immunocompromised child. Am J Otolaryngol 2013; 34:89-92. [PMID: 23084431 DOI: 10.1016/j.amjoto.2012.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 09/04/2012] [Indexed: 10/27/2022]
Abstract
We report a rare case of progressive hearing loss after acquired CMV infection in a child with Langerhans cell histiocytosis (LCH). A 5-month-old female was diagnosed as having LCH. When she was 14 months old, she received an unrelated donor umbilical cord blood transfusion for the treatment of intractable LCH. CMV infection was confirmed after the blood transfusion. Because her own umbilical cord had no CMV, the CMV infection was not congenital. When she was 7 years old, mixed hearing loss was noted with bilateral otitis media with effusion. After that time, the sensorineural hearing loss progressed to bilateral profound hearing loss over 3 years. Three-dimensional fluid-attenuated inversion recovery magnetic resonance imaging with gadolinium contrast enhancement revealed a high intensity area in the inner ear that suggested bilateral labyrinthitis. This case demonstrates the possibility that, under the immunodeficiency, the acquired CMV infection causes progressive sensorineural hearing loss.
Collapse
|
30
|
de Vries JJC, Vesseur A, Rotteveel LJC, Korver AMH, Rusman LG, Wessels E, Kroes ACM, Mylanus EA, Oudesluys-Murphy AM, Frijns JHM, Vossen ACTM. Cytomegalovirus DNA detection in dried blood spots and perilymphatic fluids from pediatric and adult cochlear implant recipients with prelingual deafness. J Clin Virol 2012; 56:113-7. [PMID: 23141930 DOI: 10.1016/j.jcv.2012.10.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 10/17/2012] [Indexed: 12/01/2022]
Abstract
BACKGROUND Congenital cytomegalovirus (CMV) infection is the leading cause of non-genetic congenital hearing loss. The contribution of congenital CMV to prelingual deafness and the pathophysiology is largely unknown. OBJECTIVE (1) To analyze the prevalence of congenital CMV among cochlear implant (CI) recipients with prelingual deafness. (2) To genotype CMV present in dried blood spots (DBS) and in the inner ear years after birth. STUDY DESIGN Children and adults with prelingual deafness who received a CI in 2010-2011 were included prospectively. Perilymphatic fluids were collected during CI surgery and, in the pediatric cases, DBS were retrieved for CMV DNA detection. Furthermore, a cohort of children with prelingual deafness who received a CI between 2003 and 2008 were included retrospectively. CMV detection in DBS and perilymph was followed by gB and gH genotyping. RESULTS Seventysix pediatric CI recipients were included. Seventy DBS were tested for CMV DNA, resulting in a prevalence of congenital CMV of 14% (10/70). Perilymphatic fluid was available from 29 pediatric CI recipients. One perilymph fluid, of a 21-month old girl with congenital CMV, asymptomatic at birth, was CMV DNA positive. The CMV strain in the perilymph was genotypically identical to the strain present in her DBS (gB1/gH2). Perilymph samples from 21 adult CI recipients were CMV DNA negative. CONCLUSIONS Our study stresses the important contribution of congenital CMV among pediatric CI recipients. Furthermore, our genotyping data support the hypothesis that CMV-related hearing loss is associated with ongoing viral replication in the inner ear up to years after birth.
Collapse
Affiliation(s)
- Jutte J C de Vries
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Inner ear lesions in congenital cytomegalovirus infection of human fetuses. Acta Neuropathol 2011; 122:763-74. [PMID: 22033878 DOI: 10.1007/s00401-011-0895-y] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 10/12/2011] [Accepted: 10/13/2011] [Indexed: 10/16/2022]
Abstract
Congenital cytomegalovirus (CMV) infection is the leading cause of non-hereditary congenital sensorineural hearing loss (SNHL). The natural course and the pathophysiology of inner ear lesions during human fetal CMV infection have not yet been reported. Inner ear lesions were investigated in six CMV-infected fetuses aged 19-35 postconceptional weeks and correlated with central nervous system (CNS) lesions. All the fetuses had high viral loads in the amniotic fluid and severe visceral and CNS lesions visible by ultrasound. Diffuse lesions consisting of both cytomegalic cells containing inclusion bodies and inflammation were found within all studied structures including the inner ear, brain, other organs, and placenta, suggesting hematogenous dissemination. Cochlear infection was consistently present and predominated in the stria vascularis (5/6), whereas the supporting cells in the organ of Corti were less often involved (2/6). Vestibular infection, found in 4/6 cases, was florid; the non-sensory epithelia, including the dark cells, were extensively infected. The endolymphatic sac was infected in 1 of 3 cases. The severity of inner ear infection was correlated with the CNS lesions, confirming the neurotropism of CMV. This study documenting infection of the structures involved in endolymph secretion and potassium homeostasis in fetuses with high amniotic fluid viral loads suggests that potassium dysregulation in the endolymphatic compartment of the inner ear may lead to secondary degeneration of the sensory structures. In addition, the occurrence of SNHL depends on the intensity and duration of the viral infection and inflammation.
Collapse
|
32
|
Abstract
Human cytomegalovirus (HCMV) is a ubiquitous beta human herpesvirus type 5. Compared to other human herpesviruses, HCMV is the largest, with a genome of approximately 235 kb containing approximately 250 ORFs with the potential to encode proteins. Usually, HCMV asymptomatically infects the host during childhood, and establishes life-long latency. The infection is life-threatening for infants and immunocompromised individuals, because of direct cytopathicity by viral replication, causing systemic organ injuries. Intrauterine infection occasionally causes microcephaly, sensorineural hearing loss and mental retardation. HCMV genome contains a number of accessory genes. Most of them are engaged in immune evasion or inhibition of cell death, possibly, resulting in a symbiosis between virus and host. CD34-positive myeloid progenitor cells are considered as a site of latency. However, the molecular mechanisms by which HCMV establishes and maintains latency and reactivates remain poorly understood. Recently in Japan, the decline of maternal HCMV seropositivity may increase the risk of intrauterine infection. It needs to immediately establish the protection against transplacental HCMV infection, such as a new type of neutralizing antibody or vaccine, which effectively interferes viral entry specific to endothelial and epithelial cells. Furthermore, HCMV infection might be considered as the most important factor for driving immune senescence in the elderly.
Collapse
|
33
|
Schachtele SJ, Mutnal MB, Schleiss MR, Lokensgard JR. Cytomegalovirus-induced sensorineural hearing loss with persistent cochlear inflammation in neonatal mice. J Neurovirol 2011; 17:201-11. [PMID: 21416394 PMCID: PMC3098308 DOI: 10.1007/s13365-011-0024-7] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Revised: 02/25/2011] [Accepted: 03/02/2011] [Indexed: 12/20/2022]
Abstract
Congenital cytomegalovirus (CMV) infection is the leading cause of sensorineural hearing loss (SNHL) in children. During murine (M)CMV-induced encephalitis, the immune response is important for both the control of viral dissemination and the clearance of virus from the brain. While the importance of CMV-induced SNHL has been described, the mechanisms surrounding its pathogenesis and the role of inflammatory responses remain unclear. This study presents a neonatal mouse model of profound SNHL in which MCMV preferentially infected both cochlear perilymphatic epithelial cells and spiral ganglion neurons. Interestingly, MCMV infection induced cochlear hair cell death by 21 days post-infection, despite a clear lack of direct infection of hair cells and the complete clearance of the virus from the cochlea by 14 dpi. Flow cytometric, immunohistochemical, and quantitative PCR analysis of MCMV-infected cochlea revealed a robust and chronic inflammatory response, including a prolonged increase in reactive oxygen species production by infiltrating macrophages. These data support a pivotal role for inflammation during MCMV-induced SNHL.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Antigens, CD/analysis
- Child
- Cloning, Molecular
- Cochlea/immunology
- Cochlea/pathology
- Cochlea/virology
- Cytomegalovirus/genetics
- Cytomegalovirus/growth & development
- Cytomegalovirus/immunology
- Cytomegalovirus Infections/complications
- Cytomegalovirus Infections/immunology
- Cytomegalovirus Infections/pathology
- Cytomegalovirus Infections/virology
- Disease Models, Animal
- Epithelial Cells/pathology
- Epithelial Cells/virology
- Escherichia coli
- Evoked Potentials, Auditory, Brain Stem
- Female
- Flow Cytometry
- Hearing Loss, Sensorineural/etiology
- Hearing Loss, Sensorineural/immunology
- Hearing Loss, Sensorineural/pathology
- Hearing Loss, Sensorineural/virology
- Humans
- Immunohistochemistry
- Inflammation/immunology
- Inflammation/pathology
- Inflammation/virology
- Lymphocytes/immunology
- Lymphocytes/virology
- Macrophages/immunology
- Macrophages/virology
- Mice
- Muromegalovirus/genetics
- Muromegalovirus/growth & development
- Muromegalovirus/immunology
- Neurons/pathology
- Neurons/virology
- Polymerase Chain Reaction
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
Collapse
Affiliation(s)
- Scott J. Schachtele
- Center for Infectious Diseases and Microbiology Translational Research, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Manohar B. Mutnal
- Center for Infectious Diseases and Microbiology Translational Research, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Mark R. Schleiss
- Center for Infectious Diseases and Microbiology Translational Research, Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - James R. Lokensgard
- Center for Infectious Diseases and Microbiology Translational Research, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
34
|
Juanjuan C, Yan F, Li C, Haizhi L, Ling W, Xinrong W, Juan X, Tao L, Zongzhi Y, Suhua C. Murine model for congenital CMV infection and hearing impairment. Virol J 2011; 8:70. [PMID: 21320351 PMCID: PMC3045346 DOI: 10.1186/1743-422x-8-70] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Accepted: 02/15/2011] [Indexed: 11/28/2022] Open
Abstract
Background Congenital cytomegalovirus (CMV) infection is the leading cause of sensorineural hearing loss (SNHL), and SNHL is the most frequent sequela of congenital CMV infection. But the pathogenic mechanism remains unknown, and there is no ideal CMV intrauterine infection animal model to study the mechanisms by which SNHL develops. Methods We established the congenital murine cytomegalovirus (MCMV) infection model by directly injecting the virus into the placenta on day 12.5 of gestation. Then, we observed the development and the MCMV congenital infection rate of the fetuses on the day they were born. Furthermore, we detected the auditory functions, the conditions of the MCMV infection, and the histological change of the inner ears of 28-day-old and 70-day-old offspring. Results Both the fetal loss rate and the teratism rate of offspring whose placentas were inoculated with MCMV increased, and their body length, head circumference, and weight decreased. The hearing level of offspring both decreased at both 28- and 70-days post birth; the 70-day-old mice developed lower hearing levels than did the 28-day old mice. No significant inflammatory changes in the cochleae of the mice were observed. MCMV DNA signals were mainly detected in the spiral ganglion neurons and the endolymph area, but not in the perilymph area. The number of neurons decreased, and their ultrastructures changed. Moreover, with age, the number of neurons dramatically decreased, and the ultrastructural lesions of neurons became much more severe. Conclusions The results suggest that the direct injection of MCMV into the placenta may efficiently cause fetal infection and disturb the intrauterine development of the fetus, and placental inoculation itself has no obvious adverse effects on offspring. The reduction in the number of spiral ganglion neurons and the ultrastructural lesions of the neurons may be the major cause of congenital CMV infection-induced progressive SNHL.
Collapse
Affiliation(s)
- Chen Juanjuan
- Department of Obstetrics and Gynecology, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kanai K, Yamada S, Yamamoto Y, Fukui Y, Kurane I, Inoue N. Re-evaluation of the genome sequence of guinea pig cytomegalovirus. J Gen Virol 2011; 92:1005-1020. [PMID: 21270288 DOI: 10.1099/vir.0.027789-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Congenital infection by human cytomegalovirus (HCMV) is a major cause of birth defects and developmental abnormalities. Since guinea pig cytomegalovirus (GPCMV) crosses the placenta and causes infection in utero, GPCMV models are useful for studies of the mechanisms of transplacental transmission. During our characterization of a genomic locus required for GPCMV dissemination in animals, we found that the nucleotide sequence in and around the nearby immediate-early genes in our lineage of GPCMV strain 22122 [designated GPCMV (ATCC-P5)] showed clear differences from that reported previously for the same strain [designated GPCMV (UMN)] passaged extensively in vitro. Since in vitro passaging of HCMV is known to result in genetic alterations, especially in the UL128-UL131A locus, and loss of growth ability in particular cell types, in this study we determined the complete genome sequence of GPCMV (ATCC-P5), which grows efficiently in animals. A total of 359 differences were identified between the genome sequences of GPCMV (UMN) and GPCMV (ATCC-P5), and these resulted in structural differences in 29 protein-encoding regions. In addition, some genes predicted from our analysis but not from GPCMV (UMN) are well conserved among cytomegaloviruses. An additional 18 passages of GPCMV (ATCC-P5) in vitro generated no further marked alterations in these genes or in the locus corresponding to the HCMV UL128-UL131A. Our analyses indicate that the published sequence of GPCMV (UMN) contains a substantial number of sequencing errors and, possibly, some mutations resulting from a long history of passaging in vitro. Our re-evaluation of the genetic content of GPCMV will provide a solid foundation for future studies.
Collapse
Affiliation(s)
- Kyosuke Kanai
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Souichi Yamada
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yumiko Yamamoto
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yoshiko Fukui
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Ichiro Kurane
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Naoki Inoue
- Department of Virology I, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
36
|
In vivo imaging assay for the convenient evaluation of antiviral compounds against cytomegalovirus in mice. Antiviral Res 2010; 88:45-52. [DOI: 10.1016/j.antiviral.2010.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2009] [Revised: 06/29/2010] [Accepted: 07/06/2010] [Indexed: 11/22/2022]
|
37
|
Noyola DE, Jiménez-Capdeville ME, Demmler-Harrison GJ. Central nervous system disorders in infants with congenital cytomegalovirus infection. Neurol Res 2010; 32:278-84. [PMID: 20406606 DOI: 10.1179/016164110x12645013284293] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
BACKGROUND Congenital cytomegalovirus is the most common cause of congenital infection in developed countries and a major etiology for neurological disability in children. In many countries, there is low awareness of the importance of this virus as a cause of neurological disorders. METHODS We reviewed current knowledge regarding neurological disorders associated to congenital cytomegalovirus infection and analysed the epidemiology of this infection in Latin American countries. RESULTS The incidence of congenital cytomegalovirus infection reported from Latin American countries ranges from 0 to 6.8% in different settings. Congenital cytomegalovirus infection is a common cause of hearing deficits and mental impairment in children. However, the impact of this infection as a cause of neurological disorders in Latin American countries remains poorly documented. DISCUSSION Actions are needed to increase knowledge regarding the frequency and impact of congenital cytomegalovirus infection on Latin American children, as well as to increase awareness of the general population and the medical community regarding the need to identify infants infected in utero by this virus and to carefully evaluate their neurological development throughout childhood.
Collapse
Affiliation(s)
- Daniel E Noyola
- Department of Microbiology, Facultad de Medicina, Universidad Autónoma de San Luis Potosí, San Luis Potosí, Mexico.
| | | | | |
Collapse
|
38
|
Schleiss MR, McVoy MA. Guinea Pig Cytomegalovirus (GPCMV): A Model for the Study of the Prevention and Treatment of Maternal-Fetal Transmission. Future Virol 2010; 5:207-217. [PMID: 23308078 DOI: 10.2217/fvl.10.8] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A major public health challenge today is the problem of congenital cytomegalovirus (CMV) transmission. Maternal-fetal CMV infections are common, occurring in 0.5-2% of pregnancies, and these infections often lead to long-term injury of the newborn infant. In spite of the well-recognized burden that these infections place on society, there are as yet no clearly established interventions available to prevent transmission of CMV. In order to study potential interventions, such as vaccines or antiviral therapies, an animal model of congenital CMV transmission is required. The best small animal model of CMV transmission is the guinea pig cytomegalovirus (GPCMV) model. This article summarizes the GPCMV model, putting it into the larger context of how studies in this system have relevance to human health. An emphasis is placed on how the vertical transmission of GPCMV recapitulates the pathogenesis of congenital CMV in infants, making this a uniquely well-suited model for the study of potential CMV vaccines.
Collapse
Affiliation(s)
- Mark R Schleiss
- Center for Infectious Diseases and Microbiology Translational Research, University of Minnesota Medical School, 2001 6 Street SE, Minneapolis, MN 55455,
| | | |
Collapse
|
39
|
Smit AL, Stokroos RJ, Litjens SGH, Kremer B, Kramer BW. Potential role for lipopolysaccharide in congenital sensorineural hearing loss. J Med Microbiol 2010; 59:377-383. [PMID: 20093374 DOI: 10.1099/jmm.0.015792-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Congenital sensorineural hearing loss (SNHL) is common. In the Western world, the incidence is 1-3 per 1000 live births. The aetiology encompasses genetic and non-genetic factors accounting for 55 % and 45 % of cases, respectively. Reports that describe the contribution of intrauterine infection to the occurrence of congenital SNHL are limited, and comparative analysis of the different pathogens is lacking. Lipopolysaccharide (LPS), a product of bacteriolysis, has been demonstrated to be associated with inner ear damage in experimental studies. To elucidate the potential role of this toxin in congenital SNHL and to identify the pathogenesis and transmission routes, we reviewed the literature. We speculate that different routes of exposure to LPS in utero may result in congenital inner ear damage.
Collapse
Affiliation(s)
- A L Smit
- Department of Otorhinolaryngology/Head and NeckSurgery, Maastricht University Medical Centre, PO Box 5800, Maastricht, TheNetherlands
| | - R J Stokroos
- Department of Otorhinolaryngology/Head and NeckSurgery, Maastricht University Medical Centre, PO Box 5800, Maastricht, TheNetherlands
| | - S G H Litjens
- Faculty of Health, Medicine and Life Sciences, Universityof Maastricht, PO Box 616, Maastricht, The Netherlands
| | - B Kremer
- School of Oncology and Developmental Biology, Universityof Maastricht, PO Box 5800, Maastricht, The Netherlands.,Department of Otorhinolaryngology/Head and NeckSurgery, Maastricht University Medical Centre, PO Box 5800, Maastricht, TheNetherlands
| | - B W Kramer
- Department of Pediatrics, Maastricht University MedicalCentre, PO Box 5800, Maastricht, The Netherlands.,School of Oncology and Developmental Biology, Universityof Maastricht, PO Box 5800, Maastricht, The Netherlands
| |
Collapse
|
40
|
Yamada S, Nozawa N, Katano H, Fukui Y, Tsuda M, Tsutsui Y, Kurane I, Inoue N. Characterization of the guinea pig cytomegalovirus genome locus that encodes homologs of human cytomegalovirus major immediate-early genes, UL128, and UL130. Virology 2009; 391:99-106. [PMID: 19559454 DOI: 10.1016/j.virol.2009.05.034] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2009] [Revised: 05/21/2009] [Accepted: 05/22/2009] [Indexed: 11/30/2022]
Abstract
We reported previously that the guinea pig cytomegalovirus (CMV) stock purchased from the American Type Culture Collection contained two types of strains, one containing and the other lacking a 1.6 kb locus, and that the 1.6 kb locus was required for efficient viral growth in animals but not in cell culture. In this study, we characterized the genetic contents of the locus, and found that i) the 1.6 kb locus encodes homologs of human CMV UL128 and UL130, GP129 and GP131, respectively, ii) these genes are expressed with late gene kinetics, iii) GP131 protein (pGP131) localized to cell surface only in the presence of glycoproteins H and L, and iv) pGP131 is a virion component. Therefore, it is plausible that pGP131 forms a complex with glycoproteins H and L and becomes a virion component as does UL130 protein (pUL130). Since pUL130 is one of the glycoproteins essential for infection of endothelial and epithelial cells in human and primates, functional and immunological analyses of this GPCMV homolog of pUL130 may help to illuminate the in vivo role of pUL130.
Collapse
Affiliation(s)
- Souichi Yamada
- Department of Virology I, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Lee JH, Park K, Kang TC, Choung YH. Three-dimensional anatomy of the temporal bone in normal mice. Anat Histol Embryol 2009; 38:311-5. [PMID: 19519734 DOI: 10.1111/j.1439-0264.2009.00946.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
This study was performed to determine the three-dimensional (3D) structure of the murine temporal bone and to provide a survey atlas of the temporal bone structures in mice. The temporal bones of adult BALB/c mice were examined and 3D high-resolution reconstructions of the temporal bone were obtained using a micro-CT system. Using the system described here, the bony labyrinth and membranous labyrinth could be investigated in a non-destructive manner. The turning rate of the cochlea was two (human rate: two and a half). The shapes of the superior and posterior semicircular canals were more flexed than those in humans. The malleus manubrium was directed anteriorly and was thin and fan-shaped like a Persian sword. The size of the incus relative to the malleus was smaller than that in the human ossicles. The 3D reconstruction of murine temporal bone described in this study provides anatomical information that will be useful in future studies using mouse model.
Collapse
Affiliation(s)
- J H Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Hallym University College of Medicine, Chuncheon, Kangwon, Korea.
| | | | | | | |
Collapse
|
42
|
Cheeran MCJ, Lokensgard JR, Schleiss MR. Neuropathogenesis of congenital cytomegalovirus infection: disease mechanisms and prospects for intervention. Clin Microbiol Rev 2009; 22:99-126, Table of Contents. [PMID: 19136436 PMCID: PMC2620634 DOI: 10.1128/cmr.00023-08] [Citation(s) in RCA: 348] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Congenital cytomegalovirus (CMV) infection is the leading infectious cause of mental retardation and hearing loss in the developed world. In recent years, there has been an improved understanding of the epidemiology, pathogenesis, and long-term disabilities associated with CMV infection. In this review, current concepts regarding the pathogenesis of neurological injury caused by CMV infections acquired by the developing fetus are summarized. The pathogenesis of CMV-induced disabilities is considered in the context of the epidemiology of CMV infection in pregnant women and newborn infants, and the clinical manifestations of brain injury are reviewed. The prospects for intervention, including antiviral therapies and vaccines, are summarized. Priorities for future research are suggested to improve the understanding of this common and disabling illness of infancy.
Collapse
Affiliation(s)
- Maxim C-J Cheeran
- Departments of Medicine, Center for Infectious Diseases and Microbiology Translational Research, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| | | | | |
Collapse
|
43
|
Cristobal R, Oghalai JS. Hearing loss in children with very low birth weight: current review of epidemiology and pathophysiology. Arch Dis Child Fetal Neonatal Ed 2008; 93:F462-8. [PMID: 18941031 PMCID: PMC3597102 DOI: 10.1136/adc.2007.124214] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
An association between birth weight <1500 g (very low birth weight (VLBW)) and hearing loss has been long recognised. As universal hearing screening programmes have become widely implemented and the survival rate of VLBW babies in modern intensive care units has increased, we have gained a substantially better understanding of the nature of this problem. However, many gaps in our knowledge base exist. This review describes recent data on hearing loss in the VLBW population and explains the current level of understanding about the physiological basis underlying the auditory deficits in these patients. Although VLBW alone may not have a severe impact on hearing, it is commonly associated with multiple other risk factors that can alter hearing in a synergistic fashion. Therefore, the risk of hearing loss is substantially higher than in the general newborn population. Also, it is important to perform a more comprehensive audiometric evaluation than standard otoacoustic emission screening for infants who are in the neonatal intensive care unit in order not to miss hearing loss due to retrocochlear pathology. Furthermore, children with VLBW are also at increased risk of experiencing progressive or delayed-onset hearing loss, and thus should continue to have serial hearing evaluations after discharge from the neonatal intensive care unit.
Collapse
Affiliation(s)
- R Cristobal
- Bobby R Alford Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, Texas, USA
| | - J S Oghalai
- Bobby R Alford Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, Texas, USA,The Hearing Center at Texas Children’s Hospital, Houston, Texas, USA,Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA,Department of Bioengineering, Rice University, Houston, Texas, USA
| |
Collapse
|
44
|
Nozawa N, Yamamoto Y, Fukui Y, Katano H, Tsutsui Y, Sato Y, Yamada S, Inami Y, Nakamura K, Yokoi M, Kurane I, Inoue N. Identification of a 1.6 kb genome locus of guinea pig cytomegalovirus required for efficient viral growth in animals but not in cell culture. Virology 2008; 379:45-54. [PMID: 18656220 DOI: 10.1016/j.virol.2008.06.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2008] [Revised: 03/10/2008] [Accepted: 06/10/2008] [Indexed: 11/17/2022]
Abstract
Guinea pig cytomegalovirus (GPCMV) provides a useful model for studies of congenital CMV infection. During characterization of the GPCMV genome sequence, we identified two types of strains in a virus stock purchased from ATCC. One of them, GPCMV/del, lacks a 1.6 kb locus that positionally corresponds to murine CMV (MCMV) M129-M133. Growth of GPCMV/del in cell culture was marginally better than that of the other strain, GPCMV/full, which harbors the 1.6 kb locus. However, in animals infected intraperitoneally with virus stocks containing both strains, GPCMV/full disseminated more efficiently than GPCMV/del, including 200-fold greater viral load in salivary glands. Viral DNA, transcripts of the immediate-early 2 gene homolog, and viral antigens were more abundant in animals infected with GPCMV/full than in those infected with GPCMV/del. Although the observed phenomena have some similarity with the growth properties of MCMV strains defective in mck-1/mck-2(M129/131) and those defective in sgg(M132), no M129-M132 homologs were found in the 1.6 kb locus. Since one of the ORFs in the locus has a weak sequence similarity with HCMV UL130, which relates to cell tropism, further studies will be required to learn the mechanism for efficient GPCMV growth in animal.
Collapse
Affiliation(s)
- Naoki Nozawa
- Department of Virology I, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Induction of cytomegalovirus-infected labyrinthitis in newborn mice by lipopolysaccharide: a model for hearing loss in congenital CMV infection. J Transl Med 2008; 88:722-30. [PMID: 18475257 DOI: 10.1038/labinvest.2008.39] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Congenital cytomegalovirus (CMV) infection is the most common infectious cause of sensorineural hearing loss in children. Here, we established an experimental model of hearing loss after systemic infection with murine CMV (MCMV) in newborn mice. Although almost no viral infection was observed in the inner ears and brains by intraperitoneal (i.p.) infection with MCMV in newborn mice, infection in these regions was induced in combination with intracerebral (i.c.) injection of bacterial lipopolysaccharide (LPS). The susceptibility of the inner ears was higher than that of the brains in terms of viral titer per unit weight. In the labyrinths, the viral infection was associated with the mesenchymal vessels and accompanied by inflammatory cells induced by LPS, causing hematogenous targets of infection in the labyrinths. Viral infection also spread in the perilymph regions such as the scala tympani and scala vestibuli, probably from infected brains via meningogenic and cochlear nerve routes. Viral infection was not observed in the scala media in the endolymph, including the Corti organ. However, viral infection was observed in the spiral limbus, including the stria vascularis. These results suggest that hearing loss caused by labyrinthitis after congenital CMV infection may be enhanced by inflammation caused by systemic bacterial infection in the neonatal period.
Collapse
|
46
|
Establishment of a cell-based assay for screening of compounds inhibiting very early events in the cytomegalovirus replication cycle and characterization of a compound identified using the assay. Antimicrob Agents Chemother 2008; 52:2420-7. [PMID: 18458124 DOI: 10.1128/aac.00134-08] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To simplify the detection of infectious human cytomegalovirus (HCMV), we generated a cell line that produced luciferase in a dose-dependent manner upon HCMV infection. Using this cell line, we identified anti-HCMV compounds from a diverse library of 9,600 compounds. One of them, 1-(3,5-dichloro-4-pyridyl)piperidine-4-carboxamide (DPPC), was effective against HCMV (Towne strain) infection of human lung fibroblast cells at a 50% effective concentration of 2.5 microM. DPPC also inhibited the growth of clinical HCMV isolates and guinea pig and mouse cytomegaloviruses. Experiments using various time frames for treatment of the cells with DPPC demonstrated that DPPC was effective during the first 24 h after HCMV infection. DPPC treatment decreased not only viral DNA replication but also IE1 and IE2 expression at mRNA and protein levels in the HCMV-infected cells. However, DPPC did not inhibit the attachment of HCMV particles to the cell surface. DPPC is a unique compound that targets the very early phase of cytomegalovirus infection, probably by disrupting a pathway that is important after viral entry but before immediate-early gene expression.
Collapse
|
47
|
Woolf NK, Jaquish DV, Koehrn FJ. Transplacental murine cytomegalovirus infection in the brain of SCID mice. Virol J 2007; 4:26. [PMID: 17349048 PMCID: PMC1838414 DOI: 10.1186/1743-422x-4-26] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2006] [Accepted: 03/09/2007] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Congenital cytomegalovirus (CMV) infection is the most common congenital viral infection in humans and the major nonhereditary cause of central nervous system (CNS) developmental disorders. Previous attempts to develop a murine CMV (MCMV) model of natural congenital human CMV (HCMV) infection have failed because MCMV does not cross the placenta in immunocompetent mice. RESULTS In marked contrast with immunocompetent mice, C.B-17 SCID (severe combined immunodeficient) mice were found to be highly susceptible to natural MCMV transplacental transmission and congenital infection. Timed-pregnant SCID mice were intraperitoneally (IP) injected with MCMV at embryonic (E) stages E0-E7, and vertical MCMV transmission was evaluated using nested polymerase chain reaction (nPCR), in situ hybridization (ISH) and immunohistochemical (IHC) assays. SCID mouse dams IP injected at E0 with 102 PFU of MCMV died or resorbed their fetuses by E18. Viable fetuses collected at E18 from SCID mice IP injected with 102-104 PFU of MCMV at E7 did not demonstrate vertical MCMV transmission. Notably, transplacental MCMV transmission was confirmed in E18 fetuses from SCID mice IP injected with 103 PFU of MCMV at stages E3-E5. The maximum rate of transplacental MCMV transmission (53%) at E18 occurred when SCID mouse dams were IP injected with 103 PFU of MCMV at E4. Congenital infection was confirmed by IHC immunostaining of MCMV antigens in 26% of the MCMV nPCR positive E18 fetuses. Transplacental MCMV transmission was associated with intrauterine growth retardation and microcephaly. Additionally, E18 fetuses with MCMV nPCR positive brains had cerebral interleukin-1alpha (IL-1alpha) expression significantly upregulated and cerebral IL-1 receptor II (IL-1RII) transcription significantly downregulated. However, MCMV-induced changes in cerebral cytokine expression were not associated with any histological signs of MCMV infection or inflammation in the brain. CONCLUSION Severe T- and B-cell immunodeficiencies in SCID mice significantly enhance the rate of natural MCMV transplacental transmission and congenital infection. During gestation MCMV exhibits a tissue tropism for the developing brain, and vertical MCMV transmission is correlated with fetal growth retardation and abnormal cerebral proinflammatory cytokine expression. These data confirm that natural vertical MCMV infection in SCID mice constitutes a useful new experimental rodent model of congenital HCMV infection.
Collapse
Affiliation(s)
- Nigel K Woolf
- Departments of Surgery/Anatomy, University of California Medical School at San Diego Life, 9500 Gilman Drive, La Jolla, CA 92093-0604, USA
- Department of Surgery, Veterans Affairs Research Service, VA San Diego Healthcare System, 3350 La Jolla Village Drive, La Jolla, CA 92161, USA
| | - Dawn V Jaquish
- Departments of Surgery/Anatomy, University of California Medical School at San Diego Life, 9500 Gilman Drive, La Jolla, CA 92093-0604, USA
- Department of Surgery, Veterans Affairs Research Service, VA San Diego Healthcare System, 3350 La Jolla Village Drive, La Jolla, CA 92161, USA
| | - Fred J Koehrn
- Departments of Surgery/Anatomy, University of California Medical School at San Diego Life, 9500 Gilman Drive, La Jolla, CA 92093-0604, USA
- Department of Surgery, Veterans Affairs Research Service, VA San Diego Healthcare System, 3350 La Jolla Village Drive, La Jolla, CA 92161, USA
| |
Collapse
|