151
|
Inaba Y, Motobayashi M, Nishioka M, Kaneko T, Yamauchi S, Kawasaki Y, Shiba N, Nishio SY, Moteki H, Miyagawa M, Takumi Y, Usami SI, Koike K. Correlation Between White Matter Lesions and Intelligence Quotient in Patients With Congenital Cytomegalovirus Infection. Pediatr Neurol 2016; 55:52-7. [PMID: 26778145 DOI: 10.1016/j.pediatrneurol.2015.11.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 11/26/2015] [Accepted: 11/27/2015] [Indexed: 11/29/2022]
Abstract
BACKGROUND It is well known that congenital cytomegalovirus infection exhibits white matter and other types of lesions in magnetic resonance imaging (MRI), but little is known on the clinical significance of white matter lesions because they are also present in asymptomatic congenital cytomegalovirus infection. We investigated for relationships among white matter lesions, intelligence quotient, and other neurodevelopmental features. METHODS Nine children (five boys and four girls; mean age: 87.4 months, range: 63-127 months) with sensorineural hearing loss (five bilateral and four unilateral) had been diagnosed as having congenital cytomegalovirus infection by positive polymerase chain reaction findings of dried umbilical cords. They were evaluated for the presence of autistic features, tested using Wechsler Intelligence Scale for Children-Fourth Edition for intelligence quotient, and underwent brain MRI to measure white matter lesion localization and volume. RESULTS At the time of MRI examination (mean age: 69.4 months, range: 19-92 months), white matter lesions were detected in eight of nine patients. Five subjects were diagnosed as having autism spectrum disorders. We observed increased white matter lesion volume was associated with lower intelligence quotient scores (R(2) = 0.533, P = 0.026) but not with autism spectrum disorders. CONCLUSIONS In individuals with congenital cytomegalovirus, an increased white matter lesion volume is associated with lower intelligence quotient scores but not with an increased likelihood of autistic behavior.
Collapse
Affiliation(s)
- Yuji Inaba
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan.
| | - Mitsuo Motobayashi
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan
| | - Makoto Nishioka
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan
| | - Tomoki Kaneko
- Department of Radiology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Shoko Yamauchi
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yoichiro Kawasaki
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan
| | - Naoko Shiba
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan
| | - Shin-ya Nishio
- Department of Otolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hideaki Moteki
- Department of Otolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Maiko Miyagawa
- Department of Otolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yutaka Takumi
- Department of Otolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Shin-ichi Usami
- Department of Otolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Kenichi Koike
- Department of Pediatrics, Shinshu University School of Medicine, Matsumoto, Japan
| |
Collapse
|
152
|
Bergevin A, Zick CD, McVicar SB, Park AH. Cost-benefit analysis of targeted hearing directed early testing for congenital cytomegalovirus infection. Int J Pediatr Otorhinolaryngol 2015; 79:2090-3. [PMID: 26432541 DOI: 10.1016/j.ijporl.2015.09.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Revised: 09/12/2015] [Accepted: 09/14/2015] [Indexed: 10/23/2022]
Abstract
OBJECTIVES In this study, we estimate an ex ante cost-benefit analysis of a Utah law directed at improving early cytomegalovirus (CMV) detection. STUDY DESIGN We use a differential cost of treatment analysis for publicly insured CMV-infected infants detected by a statewide hearing-directed CMV screening program. METHODS Utah government administrative data and multi-hospital accounting data are used to estimate and compare costs and benefits for the Utah infant population. RESULTS If antiviral treatment succeeds in mitigating hearing loss for one infant per year, the public savings will offset the public costs incurred by screening and treatment. If antiviral treatment is not successful, the program represents a net cost, but may still have non-monetary benefits such as accelerated achievement of diagnostic milestones. CONCLUSIONS The CMV education and treatment program costs are modest and show potential for significant cost savings.
Collapse
Affiliation(s)
- Anna Bergevin
- Center for Public Policy & Administration, University of Utah, Salt Lake City, UT, United States
| | - Cathleen D Zick
- Department of Family & Consumer Studies, University of Utah, Salt Lake City, UT, United States.
| | | | - Albert H Park
- Division of Otolaryngology-Head and Neck Surgery, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
153
|
Zhang W, Yao Y, Chen J, Wang M. Unconserved C terminal of human cytomegalovirus tegument protein pUL76 elicits nuclear aggresome formation and induces DNA damage in transfected cells. J Biomed Sci 2015; 22:95. [PMID: 26494186 PMCID: PMC4618751 DOI: 10.1186/s12929-015-0205-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 10/10/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The HCMV UL76 gene is a member of UL24 family in herpes virus and encodes a highly conserved herpes virus protein. Inherited from common ancestor, members of Herpes_UL24 family encode proteins with a conserved N terminal and varied in C terminal region. To define which region (conserved N terminal or unconserved C terminal) of UL76 was responsible for its ability to induce DNA damage and aggresome formation, the wild-type UL76 gene and two deletion mutants were transfected to cells and analyzed by immunofluorescent staining, Western blotting and comet assay. RESULTS We report that the EGFP-fusion proteins present as globular aggresomes and colocalize with γ-H2AX in cells transfected with either pEGFP-UL76 or pEGFP-UL76C. The relative expression level of γ-H2AX and percentage of cells with comet tails were elevated in pEGFP-UL76 or pEGFP-UL76C transfection groups compared to the control. CONCLUSION Our findings suggest that the unconserved C terminal (not the conserved N terminal) of pUL76 was sufficient to induce DNA damage and aggresome formation in transfected cells.
Collapse
Affiliation(s)
- Wenchang Zhang
- Department of Microbiology, Anhui Medical University, 81st Meishan Road, Hefei, 230032, Anhui, China.
| | - Yao Yao
- Department of Microbiology, Anhui Medical University, 81st Meishan Road, Hefei, 230032, Anhui, China.
| | - Jingxian Chen
- Department of Pathology & Cell Biology, Columbia University, New York, 10032, USA.
| | - Mingli Wang
- Department of Microbiology, Anhui Medical University, 81st Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
154
|
Abstract
Hearing loss is the most common congenital sensory impairment. According to National Health and Nutrition Examination Survey data from 2001 to 2008, 20.3% of subjects aged greater than or equal to 12 had unilateral or bilateral hearing loss. The World Health Organization notes that, worldwide, there are 360 million people with disabling hearing loss, with 50% preventable. Although many hearing losses are acquired, many others are manifestations of preexisting conditions. The purpose of a pediatric hearing evaluation is to identify the degree and type of hearing loss and etiology and to outline a comprehensive strategy that supports language and social development and communication.
Collapse
Affiliation(s)
- Margaret A Kenna
- Otolaryngology and Communication Enhancement, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, BCH3129, Boston, MA 02115, USA.
| |
Collapse
|
155
|
Engman ML, Sundin M, Miniscalco C, Westerlund J, Lewensohn-Fuchs I, Gillberg C, Fernell E. Prenatal acquired cytomegalovirus infection should be considered in children with autism. Acta Paediatr 2015; 104:792-5. [PMID: 25900322 DOI: 10.1111/apa.13032] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2014] [Revised: 03/10/2015] [Accepted: 04/17/2015] [Indexed: 01/02/2023]
Abstract
AIM The aim of the study was to evaluate the prevalence of congenital cytomegalovirus infection (CMV) in a representative sample of children with autism spectrum disorder. METHODS In a representative group of 115 preschool children with autism spectrum disorder, of whom 33 also had intellectual disability, the dried blood spots from the newborn metabolic screening were analysed for CMV DNA using TaqMan polymerase chain reaction. RESULTS One of the 33 children with autism spectrum disorder and intellectual disability - 3% of that group - had congenital CMV infection. The corresponding prevalence in newborn infants in Sweden is 0.2%. None of the 82 children without intellectual disability had congenital CMV. CONCLUSION The finding lends some further support for congenital CMV being one of the many aetiologies underlying autism spectrum disorder with intellectual disability. The rate of 3% of congenital CMV in children with autism spectrum disorder with intellectual disability has implications for the medical work-up. The finding of congenital CMV also indicates the need for repeated hearing assessments in the child. There is a need for similar studies with much larger samples.
Collapse
Affiliation(s)
- Mona-Lisa Engman
- Division of Pediatrics; Department of Clinical Science; Intervention and Technology; Karolinska Institutet; Stockholm Sweden
- Division of Pediatrics, Emergency and General Pediatrics Section; Astrid Lindgren Children's Hospital; Karolinska University Hospital; Stockholm Sweden
| | - Mikael Sundin
- Division of Pediatrics; Department of Clinical Science; Intervention and Technology; Karolinska Institutet; Stockholm Sweden
- Division of Pediatrics, Hematology, Immunology and SCT Section; Astrid Lindgren Children's Hospital; Karolinska University Hospital; Stockholm Sweden
| | - Carmela Miniscalco
- Gillberg Neuropsychiatry Centre; Department of Neuroscience and Physiology; Gothenburg University; Gothenburg Sweden
| | - Joakim Westerlund
- Gillberg Neuropsychiatry Centre; Department of Neuroscience and Physiology; Gothenburg University; Gothenburg Sweden
- Department of Psychology; Stockholm University; Stockholm Sweden
| | - Ilona Lewensohn-Fuchs
- Department of Clinical Microbiology; Karolinska University Laboratory; Karolinska University Hospital; Stockholm Sweden
| | - Christopher Gillberg
- Gillberg Neuropsychiatry Centre; Department of Neuroscience and Physiology; Gothenburg University; Gothenburg Sweden
| | - Elisabeth Fernell
- Gillberg Neuropsychiatry Centre; Department of Neuroscience and Physiology; Gothenburg University; Gothenburg Sweden
| |
Collapse
|
156
|
Çelikel E, Tezer H, Kanik-Yuksek S, Gülhan B, Ozkaya-Parlakay A, Yaralı N. Evaluation of 98 immunocompetent children with cytomegalovirus infection: importance of neurodevelopmental follow-up. Eur J Pediatr 2015; 174:1101-7. [PMID: 25762027 DOI: 10.1007/s00431-015-2513-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 02/17/2015] [Accepted: 03/02/2015] [Indexed: 12/22/2022]
Abstract
UNLABELLED This study aims to analyze and evaluate the clinic and demographic features of immunocompetent children that have been diagnosed with cytomegalovirus (CMV) infection. The data of children diagnosed with CMV infection between January 2005 and December 2010 and their follow-ups for 2 years were retrospectively evaluated. Ninety-eight patients were included, and the median age at admission was 5.6 months (5 days-36 months). 54.1% was male. The diagnosis of CMV infection was performed by measurement of serum anti-CMV specific Ig M and IgG titers and PCR method in blood and/or urine. In 3.06% of the patients, congenital infection was detected, whereas possible congenital infection was observed in 36.7% of the patients. Furthermore, 44 patients (44.8%) were detected to have perinatal infection while postnatal infection was spotted in 15.3% of the patients. The common presenting manifestations were prolonged jaundice, diarrhea, vomiting, abdominal distension, skin eruption, and seizure. And the most common physical examination findings were hepatosplenomegaly, microcephaly, jaundice, and petechia. The mainstream laboratory results were elevated transaminases (50%), anemia (30.6%), leukocytosis (27.5%), and thrombocytopenia (18.3%). There were intracranial calcification in 5.1% and eye findings in 5.1%. On follow-up of patients, complete improvement (59.1%), neuromotor developmental delay (11.2%), epilepsy (10.2%), hearing loss (3.06 %), hemolytic anemia (2.04%), and growth retardation (1.02%) were detected. CONCLUSION CMV infection is a significant disease both in congenital and perinatal period. It must be considered that diagnosed patients need to be monitored for a long time with special attention to their neurodevelopmental follow-ups.
Collapse
Affiliation(s)
- Elif Çelikel
- Department of Pediatrics, Ankara Hematology Oncology Children's Training and Research Hospital, Ankara, Turkey,
| | | | | | | | | | | |
Collapse
|
157
|
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: 1.1] [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
|
158
|
Child Care Provider Awareness and Prevention of Cytomegalovirus and Other Infectious Diseases. CHILD & YOUTH CARE FORUM 2015. [DOI: 10.1007/s10566-015-9325-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
159
|
Wu SE, Miller WE. The human cytomegalovirus lytic cycle is induced by 1,25-dihydroxyvitamin D3 in peripheral blood monocytes and in the THP-1 monocytic cell line. Virology 2015; 483:83-95. [PMID: 25965798 DOI: 10.1016/j.virol.2015.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 01/12/2015] [Accepted: 04/02/2015] [Indexed: 12/11/2022]
Abstract
Human cytomegalovirus (HCMV) resides in a latent form in hematopoietic progenitors and undifferentiated cells within the myeloid lineage. Maturation and differentiation along the myeloid lineage triggers lytic replication. Here, we used peripheral blood monocytes and the monocytic cell line THP-1 to investigate the effects of 1,25-dihydroxyvitamin D3 on HCMV replication. Interestingly, 1,25-dihydroxyvitamin D3 induces lytic replication marked by upregulation of HCMV gene expression and production of infectious virus. Moreover, we demonstrate that the effects of 1,25-dihydroxyvitamin D3 correlate with maturation/differentiation of the monocytes and not by directly stimulating the MIEP. These results are somewhat surprising as 1,25-dihydroxyvitamin D3 typically boosts immunity to bacteria and viruses rather than driving the infectious life cycle as it does for HCMV. Defining the signaling pathways kindled by 1,25-dihydroxyvitamin D3 will lead to a better understanding of the underlying molecular mechanisms that determine the fate of HCMV once it infects cells in the myeloid lineage.
Collapse
Affiliation(s)
- Shu-En Wu
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267-0524, United States
| | - William E Miller
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267-0524, United States.
| |
Collapse
|
160
|
Harrison GJ. Current controversies in diagnosis, management, and prevention of congenital cytomegalovirus: updates for the pediatric practitioner. Pediatr Ann 2015; 44:e115-25. [PMID: 25996198 DOI: 10.3928/00904481-20150512-11] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Congenital cytomegalovirus (CMV) infection has been called "the elephant in our living room" because it is a major public health problem that for decades has been unrecognized and unaddressed. Congenital CMV infection is a common cause of sensorineural hearing loss, vision loss, neurodevelopment disabilities, liver disease, and growth failure. Diagnostic tests are now widely available to identify newborns with congenital CMV infection, congenitally infected newborns now can be easily assessed for evidence of organ involvement, and there are now antiviral treatments and other interventions available to improve the outcome in children with congenital CMV disease. A licensed vaccine to prevent CMV infection is not yet available; however, a "CMV knowledge vaccine," composed of "an ounce of CMV awareness and three simple precautions" and that is endorsed by the Centers for Disease Control and Prevention is available for pregnant women who wish to reduce their contact with potentially CMV-infected secretions and therefore reduce their risk of acquiring CMV during pregnancy. Medical experts in the field of congenital CMV have been called upon for a consensus statement for diagnosis and treatment, and nonprofit organizations of families affected by congenital CMV from around the world have formed a collaborative coalition to facilitate the spread of CMV knowledge and awareness.
Collapse
|
161
|
Toumpas CJ, Clark J, Harris A, Beswick R, Nourse CB. Congenital cytomegalovirus infection is a significant cause of moderate to profound sensorineural hearing loss in Queensland children. J Paediatr Child Health 2015; 51:541-544. [PMID: 25412949 DOI: 10.1111/jpc.12776] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/11/2014] [Indexed: 11/30/2022]
Abstract
AIM To investigate the proportion of children with moderate to profound hearing loss who have congenital cytomegalovirus (cCMV) infection. METHOD Retrospective analysis of CMV dried blood spot (DBS) polymerase chain reaction (PCR) in children with moderate to profound hearing impairment referred to tertiary referral centres in Queensland. Participants were under 18 years old with no readily identified cause of hearing impairment, between 2008 and 2011. The primary outcome measure was DBS CMV PCR. Other outcome measures for cases referred to the Childhood Hearing Clinic (CHC) at the Mater Children's Hospital were level of hearing impairment and the neonatal hearing screen result. RESULTS Of DBS CMV PCR testing for 106 children at the CHC for 2008 to 2011 inclusive, nine (8.5%) were positive (five with bilateral hearing impairment, four with unilateral hearing impairment). The prevalence of cCMV infection in children with moderate to profound hearing impairment was 8.4%, consistent with the statewide rate of 9.4% for 2008 to mid-2011. CONCLUSION cCMV is a significant cause of hearing impairment in Queensland children. Investigation for cCMV by retrospective DBS CMV PCR should be part of the routine investigation of all babies and young children with hearing impairment. However early diagnosis is preferable and could be achieved by routine early screening of all newborns with hearing impairment for CMV before 3 weeks of age. The healthy hearing screening programme is a routine part of neonatal care. Enhancing the integration of screening for cCMV may reduce the current delays in diagnosis and should be evaluated.
Collapse
Affiliation(s)
- Christopher J Toumpas
- Department of Paediatrics, Mater Children's Hospital, South Brisbane, Queensland, Australia.,Department of Paediatrics, Greenslopes Private Hospital, South Brisbane, Queensland, Australia.,School of Medicine, University of Queensland, South Brisbane, Queensland, Australia
| | - Julia Clark
- Department of Paediatrics, Lady Cilento Children's Hospital, South Brisbane, Queensland, Australia.,Department of Paediatrics, University of Queensland, South Brisbane, Queensland, Australia
| | - Alison Harris
- Department of Paediatrics, Lady Cilento Children's Hospital, South Brisbane, Queensland, Australia.,Department of Paediatrics, University of Queensland, South Brisbane, Queensland, Australia
| | - Rachael Beswick
- Health Services Support Agency, Brisbane, Queensland, Australia
| | - Clare B Nourse
- Department of Paediatrics, Lady Cilento Children's Hospital, South Brisbane, Queensland, Australia.,Department of Paediatrics, University of Queensland, South Brisbane, Queensland, Australia
| |
Collapse
|
162
|
Koontz D, Baecher K, Amin M, Nikolova S, Gallagher M, Dollard S. Evaluation of DNA extraction methods for the detection of Cytomegalovirus in dried blood spots. J Clin Virol 2015; 66:95-9. [PMID: 25866346 DOI: 10.1016/j.jcv.2015.03.015] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 02/24/2015] [Accepted: 03/14/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND Dried blood spots (DBS) are collected universally from newborns and may be valuable for the diagnosis of congenital Cytomegalovirus (CMV) infection. The reported analytical sensitivity for DBS testing compared to urine or saliva varies greatly across CMV studies. The purpose of this study was to directly compare the performance of various DNA extraction methods for identification of CMV in DBS including those used most often in CMV studies. STUDY DESIGN Whatman(®) Grade 903 filter paper cards were spotted with blood samples from 25 organ transplant recipients who had confirmed CMV viremia. Six DNA extraction methods were compared for relative yield of viral and cellular DNA: 2 manual solution-based methods (Gentra Puregene, thermal shock), 2 manual silica column-based methods (QIAamp DNA Mini, QIAamp DNA Investigator), and 2 automated methods (M48 MagAttract Mini, QIAcube Investigator). DBS extractions were performed in triplicate followed by real-time quantitative PCR (qPCR). RESULTS For extraction of both viral and cellular DNA, two methods (QIAamp DNA Investigator and thermal shock) consistently gave the highest yields, and two methods (M48 MagAttract Mini and QIAamp DNA Mini) consistently gave the lowest yields. There was an average 3-fold difference in DNA yield between the highest and lowest yield methods. CONCLUSION The choice of DNA extraction method is a major factor in the ability to detect low levels of CMV in DBS and can largely account for the wide range of DBS sensitivities reported in studies to date.
Collapse
Affiliation(s)
- D Koontz
- Newborn Screening and Molecular Biology Branch, Centers for Disease Control and Prevention, 4770 Buford Hwy. NE, Atlanta, GA 30341, USA.
| | - K Baecher
- Newborn Screening and Molecular Biology Branch, Centers for Disease Control and Prevention, 4770 Buford Hwy. NE, Atlanta, GA 30341, USA
| | - M Amin
- Measles, Mumps, Rubella, and Herpesvirus Laboratory Branch, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30329, USA
| | - S Nikolova
- Newborn Screening and Molecular Biology Branch, Centers for Disease Control and Prevention, 4770 Buford Hwy. NE, Atlanta, GA 30341, USA
| | - M Gallagher
- Newborn Screening and Molecular Biology Branch, Centers for Disease Control and Prevention, 4770 Buford Hwy. NE, Atlanta, GA 30341, USA
| | - S Dollard
- Measles, Mumps, Rubella, and Herpesvirus Laboratory Branch, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30329, USA
| |
Collapse
|
163
|
Kimberlin DW, Jester PM, Sánchez PJ, Ahmed A, Arav-Boger R, Michaels MG, Ashouri N, Englund JA, Estrada B, Jacobs RF, Romero JR, Sood SK, Whitworth MS, Abzug MJ, Caserta MT, Fowler S, Lujan-Zilbermann J, Storch GA, DeBiasi RL, Han JY, Palmer A, Weiner LB, Bocchini JA, Dennehy PH, Finn A, Griffiths PD, Luck S, Gutierrez K, Halasa N, Homans J, Shane AL, Sharland M, Simonsen K, Vanchiere JA, Woods CR, Sabo DL, Aban I, Kuo H, James SH, Prichard MN, Griffin J, Giles D, Acosta EP, Whitley RJ. Valganciclovir for symptomatic congenital cytomegalovirus disease. N Engl J Med 2015; 372:933-43. [PMID: 25738669 PMCID: PMC4401811 DOI: 10.1056/nejmoa1404599] [Citation(s) in RCA: 475] [Impact Index Per Article: 52.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND The treatment of symptomatic congenital cytomegalovirus (CMV) disease with intravenous ganciclovir for 6 weeks has been shown to improve audiologic outcomes at 6 months, but the benefits wane over time. METHODS We conducted a randomized, placebo-controlled trial of valganciclovir therapy in neonates with symptomatic congenital CMV disease, comparing 6 months of therapy with 6 weeks of therapy. The primary end point was the change in hearing in the better ear ("best-ear" hearing) from baseline to 6 months. Secondary end points included the change in hearing from baseline to follow-up at 12 and 24 months and neurodevelopmental outcomes, with each end point adjusted for central nervous system involvement at baseline. RESULTS A total of 96 neonates underwent randomization, of whom 86 had follow-up data at 6 months that could be evaluated. Best-ear hearing at 6 months was similar in the 6-month group and the 6-week group (2 and 3 participants, respectively, had improvement; 36 and 37 had no change; and 5 and 3 had worsening; P=0.41). Total-ear hearing (hearing in one or both ears that could be evaluated) was more likely to be improved or to remain normal at 12 months in the 6-month group than in the 6-week group (73% vs. 57%, P=0.01). The benefit in total-ear hearing was maintained at 24 months (77% vs. 64%, P=0.04). At 24 months, the 6-month group, as compared with the 6-week group, had better neurodevelopmental scores on the Bayley Scales of Infant and Toddler Development, third edition, on the language-composite component (P=0.004) and on the receptive-communication scale (P=0.003). Grade 3 or 4 neutropenia occurred in 19% of the participants during the first 6 weeks. During the next 4.5 months of the study, grade 3 or 4 neutropenia occurred in 21% of the participants in the 6-month group and in 27% of those in the 6-week group (P=0.64). CONCLUSIONS Treating symptomatic congenital CMV disease with valganciclovir for 6 months, as compared with 6 weeks, did not improve hearing in the short term but appeared to improve hearing and developmental outcomes modestly in the longer term. (Funded by the National Institute of Allergy and Infectious Diseases; ClinicalTrials.gov number, NCT00466817.).
Collapse
|
164
|
Yan XC, Wang JH, Wang B, Huang LL, Zhou LQ, Zhu B, Liang Y. Study of human cytomegalovirus replication in body fluids, placental infection, and miscarriage during the first trimester of pregnancy. J Med Virol 2015; 87:1046-53. [PMID: 25732959 DOI: 10.1002/jmv.24158] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2015] [Indexed: 11/08/2022]
Abstract
Intrauterine infection caused by human cytomegalovirus (HCMV) can lead to embryo, fetal, and neonatal damage. The prevalence of HCMV replication in body fluids (blood, urine, and cervicovaginal secretion) was investigated, and its effects on HCMV vertical transmission and miscarriages in early pregnant women were evaluated. HCMV DNA in body fluids was detected in 1,064 early pregnant women (624 normal pregnancies and 440 miscarriages). There were 101 cases who were HCMV DNA positive in cervicovaginal secretion and the rates were 10.9% (48/440 cases) and 8.5% (53/624 cases) in miscarriages and normal pregnancies, respectively (P > 0.05). A total of 101 cases (63 and 38 cases with and without HCMV DNA in cervicovaginal secretion, respectively) were given HCMV DNA detection in placental villi/deciduas. There were five cases (7.9%; two normal pregnancies and three miscarriages) with HCMV DNA in placental villi/deciduas among the 63 cases with HCMV DNA in cervicovaginal secretion, whereas none of the other 38 cases were detected HCMV DNA positive in their placental villi/deciduas. The percentage of HCMV DNA in placental villi/deciduas was higher in miscarriage group (9.1% [3/33]) than that in the normal pregnancy group (6.7% [2/30]), but there was no statistical significance (P > 0.05). Two cases with a higher HCMV loads in cervicovaginal secretion and placental villi/deciduas had miscarriages. These findings suggest that HCMV replication in cervicovaginal secretion can involve in placental HCMV infection, and high HCMV DNA loads in cervicovaginal secretion and placental villi/deciduas are associated with miscarriage.
Collapse
Affiliation(s)
- Xiao Chuan Yan
- Department of General Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | | | | | | | | | | | | |
Collapse
|
165
|
MicroRNA miR-21 attenuates human cytomegalovirus replication in neural cells by targeting Cdc25a. J Virol 2014; 89:1070-82. [PMID: 25378484 DOI: 10.1128/jvi.01740-14] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
UNLABELLED Congenital human cytomegalovirus (HCMV) infection is a leading cause of birth defects, primarily manifesting as neurological disorders. HCMV infection alters expression of cellular microRNAs (miRs) and induces cell cycle arrest, which in turn modifies the cellular environment to favor virus replication. Previous observations found that HCMV infection reduces miR-21 expression in neural progenitor/stem cells (NPCs). Here, we show that infection of NPCs and U-251MG cells represses miR-21 while increasing the levels of Cdc25a, a cell cycle regulator and known target of miR-21. These opposing responses to infection prompted an investigation of the relationship between miR-21, Cdc25a, and viral replication. Overexpression of miR-21 in NPCs and U-251MG cells inhibited viral gene expression, genome replication, and production of infectious progeny, while shRNA-knockdown of miR-21 in U-251MG cells increased viral gene expression. In contrast, overexpression of Cdc25a in U-251MG cells increased viral gene expression and production of infectious progeny and overcame the inhibitory effects of miR-21 overexpression. Three viral gene products-IE1, pp71, and UL26-were shown to inhibit miR-21 expression at the transcriptional level. These results suggest that Cdc25a promotes HCMV replication and elevation of Cdc25a levels after HCMV infection are due in part to HCMV-mediated repression of miR-21. Thus, miR-21 is an intrinsic antiviral factor that is modulated by HCMV infection. This suggests a role for miR-21 downregulation in the neuropathogenesis of HCMV infection of the developing CNS. IMPORTANCE Human cytomegalovirus (HCMV) is a ubiquitous pathogen and has very high prevalence among population, especially in China, and congenital HCMV infection is a major cause for birth defects. Elucidating virus-host interactions that govern HCMV replication in neuronal cells is critical to understanding the neuropathogenesis of birth defects resulting from congenital infection. In this study, we confirm that HCMV infection downregulates miR-21 but upregulates Cdc25a. Further determined the negative effects of cellular miRNA miR-21 on HCMV replication in neural progenitor/stem cells and U-251MG glioblastoma/astrocytoma cells. More importantly, our results provide the first evidence that miR-21 negatively regulates HCMV replication by targeting Cdc25a, a vital cell cycle regulator. We further found that viral gene products of IE1, pp71, and UL26 play roles in inhibiting miR-21 expression, which in turn causes increases in Cdc25a and benefits HCMV replication. Thus, miR-21 appears to be an intrinsic antiviral factor that represents a potential target for therapeutic intervention.
Collapse
|
166
|
Congenital and acquired cytomegalovirus infection and hearing evaluation in children. Otolaryngol Pol 2014; 68:303-7. [DOI: 10.1016/j.otpol.2014.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 04/08/2014] [Accepted: 04/24/2014] [Indexed: 11/18/2022]
|
167
|
Karltorp E, Löfkvist U, Lewensohn-Fuchs I, Lindström K, Eriksson Westblad M, Teär Fahnehjelm K, Verrecchia L, Engman ML. Impaired balance and neurodevelopmental disabilities among children with congenital cytomegalovirus infection. Acta Paediatr 2014; 103:1165-73. [PMID: 25039817 DOI: 10.1111/apa.12745] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 06/03/2014] [Accepted: 07/09/2014] [Indexed: 11/27/2022]
Abstract
AIM Although cytomegalovirus (CMV) is the most common congenital infection, existing research has not provided us with a full picture of how this can affect children in the future. The aim of this case-control study was to evaluate disabilities in a well-defined group of children with congenital cytomegalovirus (CMV) infection, who had been fitted with cochlear implants because of severe hearing impairment. METHODS A multidisciplinary team assessed 26 children with congenital CMV infection for balance difficulties, neurodevelopmental disabilities and language and visual impairment. We also included a control group of 13 children with severe hearing impairment due to connexin 26 mutations. RESULTS The majority of the children with congenital CMV infection (88%) displayed balance disturbances, including walking at a later age, but there were no cases in the control group. The CMV group also displayed frequent neurodevelopmental disabilities and feeding difficulties. CONCLUSION Congenital CMV infection affects the general development of the brain and gives rise to a complex pattern of difficulties. Identifying comorbid conditions is very important, as children with associated difficulties and disabilities need more support than children with just hearing impairment. Congenital CMV infection needs to be considered in children with hearing impairment and/or balance disturbance and/or neurodevelopmental disabilities.
Collapse
Affiliation(s)
- Eva Karltorp
- Department of Clintec; Karolinska Institutet; Stockholm Sweden
- Department of Otorhinolaryngology; Karolinska University Hospital; Stockholm Sweden
| | - Ulrika Löfkvist
- Department of Clintec; Karolinska Institutet; Stockholm Sweden
- Department of Speech and Language Pathology; Karolinska University Hospital; Stockholm Sweden
- HEAD Graduate School; Karolinska University Hospital; Stockholm Sweden
| | - Ilona Lewensohn-Fuchs
- Department of Clinical Microbiology; Karolinska Institutet; Stockholm Sweden
- Division of Clinical Microbiology; Karolinska University Hospital; Stockholm Sweden
| | - Katarina Lindström
- Department of Clintec; Karolinska Institutet; Stockholm Sweden
- Division of Pediatrics; Karolinska University Hospital; Stockholm Sweden
| | | | - Kristina Teär Fahnehjelm
- Department of Clinical Neuroscience; Karolinska Institutet; Stockholm Sweden
- Department of Pediatric Ophthalmology and Strabismus; St Erik Eye Hospital; Karolinska University Hospital; Stockholm Sweden
| | - Luca Verrecchia
- Department of Clintec; Karolinska Institutet; Stockholm Sweden
- Department of Audiology and Neurotology; Karolinska University Hospital; Stockholm Sweden
| | - Mona-Lisa Engman
- Department of Clintec; Karolinska Institutet; Stockholm Sweden
- Division of Pediatrics; Karolinska University Hospital; Stockholm Sweden
| |
Collapse
|
168
|
Goderis J, De Leenheer E, Smets K, Van Hoecke H, Keymeulen A, Dhooge I. Hearing loss and congenital CMV infection: a systematic review. Pediatrics 2014; 134:972-82. [PMID: 25349318 DOI: 10.1542/peds.2014-1173] [Citation(s) in RCA: 341] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Hearing loss caused by congenital cytomegalovirus (cCMV) infection was first observed in 1964. Today cCMV is the most common cause of nonhereditary sensorineural hearing loss in childhood. Our objective was to provide an overview of the prevalence of cCMV-related hearing loss, to better define the nature of cCMV-associated hearing loss, and to investigate the importance of cCMV infection in hearing-impaired children. METHODS Two reviewers independently used Medline and manual searches of references from eligible studies and review articles to select cohort studies on children with cCMV infection with audiological follow-up and extracted data on population characteristics and hearing outcomes. RESULTS Thirty-seven studies were included: 10 population-based natural history studies, 14 longitudinal cohort studies, and 13 retrospective studies. The prevalence of cCMV in developed countries is 0.58% (95% confidence interval, 0.41-0.79). Among these newborns 12.6% (95% confidence interval, 10.2-16.5) will experience hearing loss: 1 out of 3 symptomatic children and 1 out of 10 asymptomatic children. Among symptomatic children, the majority have bilateral loss; among asymptomatic children, unilateral loss predominates. In both groups the hearing loss is mainly severe to profound. Hearing loss can have a delayed onset, and it is unstable, with fluctuations and progression. Among hearing-impaired children, cCMV is the causative agent in 10% to 20%. Despite strict selection criteria, some heterogeneity was found between selected studies. CONCLUSIONS This systematic review underscores the importance of cCMV as a cause of sensorineural hearing loss in childhood.
Collapse
|
169
|
The human cytomegalovirus UL26 protein antagonizes NF-κB activation. J Virol 2014; 88:14289-300. [PMID: 25275128 DOI: 10.1128/jvi.02552-14] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
UNLABELLED Viral infection frequently triggers activation of host innate immune pathways that attempt to limit viral spread. The NF-κB pathway is a critical component that governs this response. We have found that the human cytomegalovirus (HCMV) U(L)26 protein antagonizes NF-κB activation. Upon infection, an HCMV strain lacking the U(L)26 gene (ΔU(L)26) induced the nuclear translocation of the NF-κB RelB subunit and activated expression and secretion of interleukin-6 (IL-6), an NF-κB target gene. The ΔU(L)26 mutant was also more sensitive to challenge with tumor necrosis factor alpha (TNF-α), a canonical NF-κB inducer. Further, expression of U(L)26 in the absence of other viral proteins blocked NF-κB activation induced by either TNF-α treatment or infection with Sendai virus (SeV). Our results indicate that U(L)26 expression is sufficient to block TNF-α-induced NF-κB nuclear translocation and IκB degradation. Last, U(L)26 blocks TNF-α-induced IκB-kinase (IKK) phosphorylation, a key step in NF-κB activation. Combined, our results indicate that U(L)26 is part of a viral program to antagonize innate immunity through modulation of NF-κB signaling. IMPORTANCE The NF-κB signaling pathway regulates innate immunity, an integral host process that limits viral pathogenesis. Viruses have evolved mechanisms to modulate NF-κB signaling to ensure their replication. HCMV is a major cause of birth defects and disease in immunosuppressed populations. HCMV is known to actively target the NF-κB pathway, which is important for HCMV infection. Our results indicate that the HCMV U(L)26 gene is a key modulator of NF-κB pathway activity. We find the U(L)26 gene is both necessary and sufficient to block NF-κB activation upon challenge with antiviral cytokines. Further, U(L)26 attenuates the phosphorylation and activation of a key NF-κB activating kinase complex, IKK. Our study provides new insight into how HCMV targets the NF-κB pathway. Given its importance to viral infection, the mechanisms through which viruses target the NF-κB pathway highlight areas of vulnerability that could be therapeutically targeted to attenuate viral replication.
Collapse
|
170
|
Smithers-Sheedy H, Raynes-Greenow C, Badawi N, McIntyre S, Jones CA. Congenital cytomegalovirus is associated with severe forms of cerebral palsy and female sex in a retrospective population-based study. Dev Med Child Neurol 2014; 56:846-52. [PMID: 24749557 DOI: 10.1111/dmcn.12467] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/22/2014] [Indexed: 11/30/2022]
Abstract
AIM Congenital cytomegalovirus (cCMV) infection can result in poor outcomes including cerebral palsy (CP). The aim of this study was to describe the incidence and comorbidities of CP reported to the Australian Cerebral Palsy Register (ACPR) as attributed to cCMV infection. METHOD This was a retrospective population-based study. Cases were drawn from Australian state CP registers with population level ascertainment, 1993 to 2003 (n=2265; 56.4% males, Gross Motor Function Classification System [GMFCS] ratings available for Victorian cases only: 70% GMFCS levels I to III and 30% GMFCS levels IV to V). Clinical data were extracted and cases with cCMV reported as a known cause were compared with cases where cCMV was not reported. RESULTS Children with cCMV (n=34; 12 males, 22 females; mean [SD] gestational age, 36.4 wk [4.4], range 24-41 wk) accounted for 1.5% of CP cases; 2.9 per 100,000 live births, (95% confidence intervals 1.9-3.9). When compared with CP cases where cCMV was not reported, proportionally, more CP cases with cCMV were born to younger mothers (p<0.001), were female (64% vs 43%, p=0.014), had spastic quadriplegia (73% vs 21%, p<0.001), required wheeled mobility i.e. GMFCS IV or V (78% vs 28%, p<0.001), had epilepsy (70% vs 30%, p<0.001), deafness (40% vs 2%, p<0.001), functional blindness (20% vs 5%, p<0.001), and severe communication impairment (71% vs 25%, p<0.001). INTERPRETATION cCMV is an important potentially preventable cause of CP and is associated with severe disability and female sex in cases reported to the ACPR. Future studies utilising prospective sample collection for cCMV testing are needed to confirm these findings.
Collapse
Affiliation(s)
- Hayley Smithers-Sheedy
- Discipline of Paediatrics & Child Health, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia; Centre for Perinatal Infection Research or Grace Centre for Newborn Care, The Children's Hospital at Westmead, Sydney, NSW, Australia; Cerebral Palsy Alliance Research Institute, University of Notre Dame Australia, Sydney, NSW, Australia; Marie Bashir Institute for Infectious Diseases and Biosecurity, The University of Sydney, Sydney, NSW, Australia
| | | | | | | | | | | |
Collapse
|
171
|
The M33 G protein-coupled receptor encoded by murine cytomegalovirus is dispensable for hematogenous dissemination but is required for growth within the salivary gland. J Virol 2014; 88:11811-24. [PMID: 25100846 DOI: 10.1128/jvi.01006-14] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a pathogen found worldwide and is a serious threat to immunocompromised individuals and developing fetuses. Due to the species specificity of cytomegaloviruses, murine cytomegalovirus (MCMV) has been used as a model for in vivo studies of HCMV pathogenesis. The MCMV genome, like the genomes of other beta- and gammaherpesviruses, encodes G protein-coupled receptors (GPCRs) that modulate host signaling pathways presumably to facilitate viral replication and dissemination. Among these viral receptors, the M33 GPCR carried by MCMV is an activator of CREB, NF-κB, and phospholipase C-β signaling pathways and has been implicated in aspects of pathogenesis in vivo, including persistence in the salivary glands of BALB/c mice. In this study, we used immunocompetent nonobese diabetic (NOD) and immunocompromised NOD-scid-gamma (NSG) mice to further investigate the salivary gland defect exhibited by M33 deficiency. Interestingly, we demonstrate that virus with an M33 deletion (ΔM33) can replicate in the salivary gland of immunocompromised animals, albeit with a 400-fold growth defect compared with the growth of wild-type virus. Moreover, we determined that M33 does not have a role in cell-associated hematogenous dissemination but is required for viral amplification once the virus reaches the salivary gland. We conclude that the reduced replicative capacity of the ΔM33 virus is due to a specific defect occurring within the localized environment of the salivary gland. Importantly, since the salivary gland represents a site essential for persistence and horizontal transmission, an understanding of the mechanisms of viral replication within this site could lead to the generation of novel therapeutics useful for the prevention of HCMV spread. Importance: Human cytomegalovirus infects the majority of the American people and can reside silently in infected individuals for the duration of their lives. Under a number of circumstances, the virus can reactivate, leading to a variety of diseases in both adults and developing babies, and therefore, identifying the function of viral proteins is essential to understand how the virus spreads and causes disease. We aim to utilize animal models to study the function of an important class of viral proteins termed G protein-coupled receptors with the ultimate goal of developing inhibitors to these proteins that could one day be used to prevent viral spread.
Collapse
|
172
|
Cohen BE, Durstenfeld A, Roehm PC. Viral causes of hearing loss: a review for hearing health professionals. Trends Hear 2014; 18:18/0/2331216514541361. [PMID: 25080364 PMCID: PMC4222184 DOI: 10.1177/2331216514541361] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
A number of viral infections can cause hearing loss. Hearing loss induced by these viruses can be congenital or acquired, unilateral or bilateral. Certain viral infections can directly damage inner ear structures, others can induce inflammatory responses which then cause this damage, and still others can increase susceptibility or bacterial or fungal infection, leading to hearing loss. Typically, virus-induced hearing loss is sensorineural, although conductive and mixed hearing losses can be seen following infection with certain viruses. Occasionally, recovery of hearing after these infections can occur spontaneously. Most importantly, some of these viral infections can be prevented or treated. For many of these viruses, guidelines for their treatment or prevention have recently been revised. In this review, we outline many of the viruses that cause hearing loss, their epidemiology, course, prevention, and treatment.
Collapse
Affiliation(s)
| | - Anne Durstenfeld
- Department of Surgery, Temple University School of Medicine, Philadelphia, PA, USA
| | - Pamela C Roehm
- Department of Otolaryngology-Head and Neck Surgery, Temple University School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
173
|
Hearing impairment in Estonia: an algorithm to investigate genetic causes in pediatric patients. Adv Med Sci 2014; 58:419-28. [PMID: 24222258 DOI: 10.2478/ams-2013-0001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
PURPOSE The present study was initiated to establish the etiological causes of early onset hearing loss (HL) among Estonian children between 2000-2009. METHODS The study group consisted of 233 probands who were first tested with an arrayed primer extension assay, which covers 199 mutations in 7 genes (GJB2, GJB6, GJB3, SLC26A4, SLC26A5 genes, and two mitochondrial genes - 12S rRNA, tRNASer(UCN)). From probands whose etiology of HL remained unknown, DNA analysis of congenital cytomegalovirus (CMV) infection and G-banded karyotype and/or chromosomal microarray analysis (CMA) were performed. RESULTS In 110 (47%) cases, the etiology of HL was genetic and in 5 (2%) congenital CMV infection was diagnosed. We found mutations with clinical significance in GJB2 (100 children, 43%) and in 2 mitochondrial genes (2 patients, 1%). A single mutation in SLC26A4 gene was detected in 5 probands (2.2%) and was considered diagnostic. In 4 probands a heterozygous IVS2-2A>G change in the SLC26A5 gene was found. We did not find any instances of homozygosity for this splice variant in the probands. CMA identified in 4 probands chromosomal regions with the loss of one allele. In 2 of them we were able to conclude that the found abnormalities are definitely pathogenic (12q13.3-q14.2 and 17q22-23.2 microdeletion), but the pathogenity of 2 other findings (3p26.2 and 1p33 microdeletion) remained unknown. CONCLUSION This practical diagnostic algorithm confirmed the etiology of early onset HL for 115 Estonian patients (49%). This algorithm may be generalized to other populations for clinical application.
Collapse
|
174
|
Chisholm KM, Aziz N, McDowell M, Guo FP, Srinivas N, Benitz WE, Norton ME, Gutierrez K, Folkins AK, Pinsky BA. Evaluation of serial urine viral cultures for the diagnosis of cytomegalovirus infection in neonates and infants. Pediatr Dev Pathol 2014; 17:176-80. [PMID: 24617645 DOI: 10.2350/14-01-1432-oa.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Cytomegalovirus (CMV) is the most common cause of congenital infection worldwide. Urine viral culture is the standard for CMV diagnosis in neonates and infants. The objectives of this study were to compare the performance of serial paired rapid shell vial cultures (SVC) and routine viral cultures (RVC), and to determine the optimal number of cultures needed to detect positive cases. From 2001 to 2011, all paired CMV SVC and RVC performed on neonates and infants less than 100 days of age were recorded. Testing episodes were defined as sets of cultures performed within 7 days of one another. A total of 1264 neonates and infants underwent 1478 testing episodes; 68 (5.4%) had at least one episode with a positive CMV culture. In episodes where CMV was detected before day 21 of life, the first specimen was positive in 100% (16/16) of cases. When testing occurred after 21 days of life, the first specimen was positive in 82.7% (43/52) of cases, requiring three cultures to reach 100% detection. The SVC was more prone to assay failure than RVC. Overall, when RVC was compared to SVC, there was 86.0% positive agreement and 99.9% negative agreement. In conclusion, three serial urine samples are necessary for detection of CMV in specimens collected between day of life 22 and 99, while one sample may be sufficient on or before day of life 21. Though SVC was more sensitive than RVC, the risk of SVC failure supports the use of multimodality testing to optimize detection.
Collapse
Affiliation(s)
- Karen M Chisholm
- 1 Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
175
|
Bates M, Tembo J, Zumla A. Congenital cytomegalovirus infections in sub-Saharan Africa - a neglected and growing problem. Trop Med Int Health 2014; 19:996-8. [DOI: 10.1111/tmi.12317] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Matthew Bates
- University of Zambia and University College London Medical School Research and Training Programme; University Teaching Hospital; Lusaka Zambia
- Division of Infection and Immunity; Department of Infection; Center for Clinical Microbiology; University College London, and UCL Hospitals NHS Foundation Trust; London UK
| | - John Tembo
- University of Zambia and University College London Medical School Research and Training Programme; University Teaching Hospital; Lusaka Zambia
| | - Alimuddin Zumla
- University of Zambia and University College London Medical School Research and Training Programme; University Teaching Hospital; Lusaka Zambia
- Division of Infection and Immunity; Department of Infection; Center for Clinical Microbiology; University College London, and UCL Hospitals NHS Foundation Trust; London UK
| |
Collapse
|
176
|
Duan YL, Ye HQ, Zavala AG, Yang CQ, Miao LF, Fu BS, Seo KS, Davrinche C, Luo MH, Fortunato EA. Maintenance of large numbers of virus genomes in human cytomegalovirus-infected T98G glioblastoma cells. J Virol 2014; 88:3861-73. [PMID: 24453365 PMCID: PMC3993548 DOI: 10.1128/jvi.01166-13] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 01/14/2014] [Indexed: 01/28/2023] Open
Abstract
UNLABELLED After infection, human cytomegalovirus (HCMV) persists for life. Primary infections and reactivation of latent virus can both result in congenital infection, a leading cause of central nervous system birth defects. We previously reported long-term HCMV infection in the T98G glioblastoma cell line (1). HCMV infection has been further characterized in T98Gs, emphasizing the presence of HCMV DNA over an extended time frame. T98Gs were infected with either HCMV Towne or AD169-IE2-enhanced green fluorescent protein (eGFP) strains. Towne infections yielded mixed IE1 antigen-positive and -negative (Ag(+)/Ag(-)) populations. AD169-IE2-eGFP infections also yielded mixed populations, which were sorted to obtain an IE2(-) (Ag(-)) population. Viral gene expression over the course of infection was determined by immunofluorescent analysis (IFA) and reverse transcription-PCR (RT-PCR). The presence of HCMV genomes was determined by PCR, nested PCR (n-PCR), and fluorescence in situ hybridization (FISH). Compared to the HCMV latency model, THP-1, Towne-infected T98Gs expressed IE1 and latency-associated transcripts for longer periods, contained many more HCMV genomes during early passages, and carried genomes for a greatly extended period of passaging. Large numbers of HCMV genomes were also found in purified Ag(-) AD169-infected cells for the first several passages. Interestingly, latency transcripts were observed from very early times in the Towne-infected cells, even when IE1 was expressed at low levels. Although AD169-infected Ag(-) cells expressed no detectable levels of either IE1 or latency transcripts, they also maintained large numbers of genomes within the cell nuclei for several passages. These results identify HCMV-infected T98Gs as an attractive new model in the study of the long-term maintenance of virus genomes in the context of neural cell types. IMPORTANCE Our previous work showed that T98G glioblastoma cells were semipermissive to HCMV infection; virus trafficked to the nucleus, and yet only a proportion of cells stained positive for viral antigens, thus allowing continual subculturing and passaging. The cells eventually transitioned to a state where viral genomes were maintained without viral antigen expression or virion production. Here we report that during long-term T98G infection, large numbers of genomes were maintained within all of the cells' nuclei for the first several passages (through passage 4 [P4]), even in the presence of continual cellular division. Surprisingly, genomes were maintained, albeit at a lower level, through day 41. This is decidedly longer than in any other latency model system that has been described to date. We believe that this system offers a useful model to aid in unraveling the cellular components involved in viral genome maintenance (and presumably replication) in cells carrying long-term latent genomes in a neural context.
Collapse
Affiliation(s)
- Ying-Liang Duan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Han-Qing Ye
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Anamaria G. Zavala
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, USA
| | - Cui-Qing Yang
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Ling-Feng Miao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Bi-Shi Fu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Keun Seok Seo
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, USA
| | | | - Min-Hua Luo
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | | |
Collapse
|
177
|
American College of Medical Genetics and Genomics guideline for the clinical evaluation and etiologic diagnosis of hearing loss. Genet Med 2014; 16:347-55. [PMID: 24651602 DOI: 10.1038/gim.2014.2] [Citation(s) in RCA: 161] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 01/06/2014] [Indexed: 01/24/2023] Open
Abstract
Hearing loss is a common and complex condition that can occur at any age, can be inherited or acquired, and is associated with a remarkably wide array of etiologies. The diverse causes of hearing loss, combined with the highly variable and often overlapping presentations of different forms of hearing loss, challenge the ability of traditional clinical evaluations to arrive at an etiologic diagnosis for many deaf and hard-of-hearing individuals. However, identifying the etiology of a hearing loss may affect clinical management, improve prognostic accuracy, and refine genetic counseling and assessment of the likelihood of recurrence for relatives of deaf and hard-of-hearing individuals. Linguistic and cultural identities associated with being deaf or hard of hearing can complicate access to and the effectiveness of clinical care. These concerns can be minimized when genetic and other health-care services are provided in a linguistically and culturally sensitive manner. This guideline offers information about the frequency, causes, and presentations of hearing loss and suggests approaches to the clinical evaluation of deaf and hard-of-hearing individuals aimed at identifying an etiologic diagnosis and providing informative and effective patient education and genetic counseling.
Collapse
|
178
|
Systematic review of the birth prevalence of congenital cytomegalovirus infection in developing countries. Int J Infect Dis 2014; 22:44-8. [PMID: 24631522 DOI: 10.1016/j.ijid.2013.12.010] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 11/12/2013] [Accepted: 12/07/2013] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Congenital cytomegalovirus (CMV) infection is the leading infectious cause of congenital hearing loss and neurodevelopmental disability in developed countries. Information on congenital CMV infection in developing countries appears to be lacking. METHODS We conducted a systematic literature review to identify studies from developing countries with population-based samples of at least 300 infants that used laboratory methods established as reliable for the diagnosis of congenital CMV infection. RESULTS Most studies were excluded due to biased samples or inadequate diagnostic methods; consequently the search identified just 11 studies that were from Africa, Asia, and Latin America. The number of newborns tested ranged from 317 to 12 195. Maternal CMV seroprevalence ranged from 84% to 100%. CMV birth prevalence varied from 0.6% to 6.1%. CMV-associated impairments were not documented in most studies. CONCLUSIONS Birth prevalence ranges were higher than for Europe and North America, as expected based on the higher maternal CMV seroprevalence. With very limited data available on sequelae, the disease burden of congenital CMV in developing countries remains largely unknown at this time.
Collapse
|
179
|
Philips B, Maes LK, Keppler H, Dhooge I. Cochlear implants in children deafened by congenital cytomegalovirus and matched Connexin 26 peers. Int J Pediatr Otorhinolaryngol 2014; 78:410-5. [PMID: 24485973 DOI: 10.1016/j.ijporl.2013.11.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 11/07/2013] [Accepted: 11/09/2013] [Indexed: 11/15/2022]
Abstract
OBJECTIVE To compare the long-term speech perception and production outcomes after cochlear implantation (CI) in children deafened by congenital cytomegalovirus (cCMV) with a matched group of Cx26-CI children by controlling for chronological age and magnetic resonance imaging (MRI) findings. METHODS Retrospective review of 12 cCMV-CI children and matched Cx26-CI children for speech perception and speech production outcomes. RESULTS Two trends were seen in our data. First, cCMV-CI children with normal MRI scans perform equally or even slightly better on speech perception tests compared to their Cx26-CI peers during the first three years. The majority of cCMV-CI children with normal MRI scans (5 out of 7), suffered from a delayed-onset SNHL. Their mean age at first implantation (2y9m, range 15-82m) was higher compared to their matched Cx26 peers (9m, range 7-12m). Before being implanted, the majority of these delayed-onset hearing impaired children had benefited from a certain period of normal hearing (with or without amplification of a hearing aid). Possibly, this input might have led to an advantage the first three years after CI. Second, results between cCMV-CI children with and cCMV-CI children without MRI abnormalities and their matched Cx26-CI counterparts tentatively suggest that, over a 5-yr follow-up period, cCMV-CI children with abnormalities on MRI scans catch up for speech perception, but lag behind for speech production. CONCLUSION cCMV-CI children with normal MRI scans perform equally or even slightly better on speech perception tests compared to their Cx26-CI peers during the first three years, whereas results between cCMV-CI children with and cCMV-CI children without MRI abnormalities and their matched Cx26-CI counterparts tentatively suggest that, over a 5-yr follow-up period, cCMV-CI children with abnormal MRI scans catch up for speech perception, but lag behind for speech production. In future, the inclusion of MRI results may assist in improved counseling of parents with cCMV deafened children seeking CI.
Collapse
Affiliation(s)
- Birgit Philips
- Ghent University, Faculty of Medicine and Health Sciences, Belgium.
| | - Leen K Maes
- Ghent University, Faculty of Medicine and Health Sciences, Belgium
| | - Hannah Keppler
- Ghent University, Faculty of Medicine and Health Sciences, Belgium
| | - Ingeborg Dhooge
- Ghent University, Faculty of Medicine and Health Sciences, Belgium; Ghent University Hospital, ENT-Department, De Pintelaan 185, 1P1, 9000 Ghent, Belgium
| |
Collapse
|
180
|
N’Diaye DS, Yazdanpanah Y, Krivine A, Andrieu T, Rozenberg F, Picone O, Tsatsaris V, Goffinet F, Launay O. Predictive factors of cytomegalovirus seropositivity among pregnant women in Paris, France. PLoS One 2014; 9:e89857. [PMID: 24587077 PMCID: PMC3933677 DOI: 10.1371/journal.pone.0089857] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 01/27/2014] [Indexed: 11/18/2022] Open
Abstract
Background Cytomegalovirus (CMV) is the most frequent cause of congenital infection. The objective of this study was to evaluate predictive factors for CMV seronegativity in a cohort of pregnant women in Paris, France. Methods Pregnant women enrolled in a prospective cohort during the 2009 A/H1N1 pandemic were tested for CMV IgG antibodies. Variables collected were age, geographic origin, lifestyle, work characteristics, socioeconomic status, gravidity, parity and number of children at home. A multivariate logistic regression model was used to identify independent predictive factors for CMV seropositivity. Results Among the 826 women enrolled, 389 (47.1%) were primiparous, and 552 (67.1%) had Metropolitan France as a geographic origin. Out of these, 355 (i.e. 57.0%, 95% confidence interval (CI): [53.6%–60.4%]) were CMV seropositive: 43.7% (95% CI:[39.5%–47.9%]) in those whose geographic origin was Metropolitan France and 84.1% in those with other origins (95% CI:[79.2%–88.3%]). Determinants associated with CMV seropositivity in a multivariate logistic regression model were: (i) geographic origin (p<0.001(compared with Metropolitan France, geographic origins of Africa adjusted odds ratio (aOR) 21.2, 95% CI:[9.7–46.5], French overseas departments and territories and other origin, aOR 7.5, 95% CI:[3.9–14.6], and Europe or Asia, aOR 2.2, 95% CI: [1.3–3.7]); and (ii) gravidity (p = 0.019), (compared with gravidity = 1, if gravidity≥3, aOR = 1.5, 95% CI: [1.1–2.2]; if gravidity = 2, aOR = 1.0, 95% CI: [0.7–1.4]). Work characteristics and socioeconomic status were not independently associated with CMV seropositivity. Conclusions In this cohort of pregnant women, a geographic origin of Metropolitan France and a low gravidity were predictive factors for CMV low seropositivity. Such women are therefore the likely target population for prevention of CMV infection during pregnancy in France.
Collapse
Affiliation(s)
- Dieynaba S. N’Diaye
- Infection, Antimicrobiens, Modélisation, Evolution (IAME), Unité Mixte de Recherche (UMR) 1137, Institut National de la Santé et de la Recherche Médicale (INSERM), Université (Univ) Paris Diderot, Sorbonne Paris Cité, Paris, France
- Université Pierre et Marie Curie (UPMC) Univ Paris 06, École doctorale (ED)393, Paris, France
- * E-mail:
| | - Yazdan Yazdanpanah
- Infection, Antimicrobiens, Modélisation, Evolution (IAME), Unité Mixte de Recherche (UMR) 1137, Institut National de la Santé et de la Recherche Médicale (INSERM), Université (Univ) Paris Diderot, Sorbonne Paris Cité, Paris, France
- Assistance-Publique Hôpitaux de Paris (AP-HP), Groupe Hospitalier Bichat-Claude Bernard, Hôpitaux Universitaires Paris Nord Val de Seine (HUPNVS), Service des Maladies Infectieuses, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Anne Krivine
- AP-HP, Groupe Hospitalier Cochin–Broca–Hôtel Dieu, Département de Virologie, Paris, France
| | - Thibaut Andrieu
- INSERM Unité (U)953, Recherche Epidémiologique en Santé Périnatale et Santé des Femmes et des Enfants, Université Pierre-et-Marie-Curie, Paris, France
| | - Flore Rozenberg
- AP-HP, Groupe Hospitalier Cochin–Broca–Hôtel Dieu, Département de Virologie, Paris, France
- Université Paris Descartes, Paris Sorbonne Cité, Paris, France
| | | | - Vassilis Tsatsaris
- Université Paris Descartes, Paris Sorbonne Cité, Paris, France
- AP-HP, Hôpital Cochin, Maternité, Paris, France
| | - François Goffinet
- INSERM Unité (U)953, Recherche Epidémiologique en Santé Périnatale et Santé des Femmes et des Enfants, Université Pierre-et-Marie-Curie, Paris, France
- Université Paris Descartes, Paris Sorbonne Cité, Paris, France
- AP-HP, Hôpital Cochin, Maternité, Paris, France
- INSERM Centre d’Investigation Clinique (CIC) P0901, Paris, France
| | - Odile Launay
- Université Paris Descartes, Paris Sorbonne Cité, Paris, France
- INSERM, CIC BT505, Paris, France
- AP-HP, Hôpital Cochin, CIC de Vaccinologie Cochin-Pasteur, Paris, France
| |
Collapse
|
181
|
Mathers C, Spencer CM, Munger J. Distinct domains within the human cytomegalovirus U(L)26 protein are important for wildtype viral replication and virion stability. PLoS One 2014; 9:e88101. [PMID: 24505393 PMCID: PMC3914908 DOI: 10.1371/journal.pone.0088101] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 01/09/2014] [Indexed: 11/21/2022] Open
Abstract
The human cytomegalovirus (HCMV) UL26 gene encodes a virion protein that is important for high titer viral replication. To identify specific domains within the UL26 protein that contribute to viral infection, we created a panel of site-directed UL26 mutant viruses and assessed their impact on phenotypes attributed to UL26. We find that the C-terminal 38 amino acids of the UL26 protein are absolutely necessary for UL26 function. A stop-insertion mutant that produced a truncated UL26 protein lacking this region behaved identically to UL26-null viruses. This included reduced accumulation of IE1 protein at early time points, smaller plaque size, reduced virion stability, and growth with similarly attenuated kinetics. This C-terminal truncation decreased the amount of UL26 packaged into the virion resulting in reduced delivery of UL26 to newly infected cells. Further, this C-terminal truncated UL26 exhibited substantially reduced nuclear localization compared to wildtype UL26. Translation of UL26 mRNA is initiated from two separate in frame methionines that give rise to a long and a short isoform of UL26. We find that the N-terminal 34 amino acids, which are unique to the long isoform of UL26, are also important for the function of the UL26 protein. A viral mutant that produces only the short isoform of UL26 and lacks these N-terminal 34 amino acids exhibits delayed IE1 accumulation, and demonstrates intermediate defects in viral plaque size, virion stability and viral growth kinetics. Ablation of the short UL26 isoform in the presence of the long UL26 isoform did not impact any of the in vitro phenotypes tested. These experiments highlight important domains within the UL26 protein that contribute to HCMV infection.
Collapse
Affiliation(s)
- Chun Mathers
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Cody M. Spencer
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, New York, United States of America
| | - Joshua Munger
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, New York, United States of America
- * E-mail:
| |
Collapse
|
182
|
Moteki H, Suzuki M, Naito Y, Fujiwara K, Oguchi K, Nishio SY, Iwasaki S, Usami SI. Evaluation of cortical processing of language by use of positron emission tomography in hearing loss children with congenital cytomegalovirus infection. Int J Pediatr Otorhinolaryngol 2014; 78:285-9. [PMID: 24370468 DOI: 10.1016/j.ijporl.2013.11.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 11/22/2013] [Accepted: 11/24/2013] [Indexed: 10/26/2022]
Abstract
OBJECTIVE To predict cochlear implant efficacy and investigate the cortical processing of the visual component of language in profoundly deafened patients with asymptomatic congenital cytomegalovirus (CMV) infection. METHODS AND CASES The cortical activity of two children with CMV-related hearing loss was evaluated with fluorodeoxyglucose-positron emission tomography (FDG-PET) with a visual language task before cochlear implantation. Total development and auditory perception ability were assessed one year after implantation. RESULTS The two children with CMV-related hearing loss showed activation in the auditory association area where no activation was found in the controls, and exhibited nearly identical cortical activation patterns to those seen in patients with profound congenital hearing loss. In contrast, differences in total development in verbal ability and discrimination of sentences between the two cases were revealed one year after implantation. CONCLUSION These results might indicate that the differences of cortical activities according to hearing abilities could have been influenced by CMV infection that involves higher function of the brain directly and/or affects the cochlea peripherally. Additionally, if CMV infection might have affected only the cochlea, these cortical activation patterns were influenced secondary by the time course of hearing loss characterized by CMV infection, which had varied manifestations. Accurate diagnosis and cochlear implantation at the appropriate time are important for successful speech development, and each patient needs a personalized habilitation program based on their etiology and brain function.
Collapse
Affiliation(s)
- Hideaki Moteki
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Mika Suzuki
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Yasushi Naito
- Department of Otorhinolaryngology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Keizo Fujiwara
- Department of Otorhinolaryngology, Kobe City Medical Center General Hospital, Kobe, Japan
| | | | - Shin-ya Nishio
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Satoshi Iwasaki
- Department of Hearing Implant Sciences, Shinshu University School of Medicine, Matsumoto, Japan
| | - Shin-ichi Usami
- Department of Otorhinolaryngology, Shinshu University School of Medicine, Matsumoto, Japan.
| |
Collapse
|
183
|
Diagnosis of congenital CMV using PCR performed on formalin-fixed, paraffin-embedded placental tissue. Am J Surg Pathol 2013; 37:1413-20. [PMID: 23797721 DOI: 10.1097/pas.0b013e318290f171] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Congenital cytomegalovirus (CMV) infection may be asymptomatic until hearing loss manifests in childhood. Because diagnosis of congenital CMV requires viral detection within an infant's first 21 days of life, CMV polymerase chain reaction (PCR) on formalin-fixed, paraffin-embedded (FFPE) placental tissue provides a unique opportunity to identify congenital exposure in cases in which CMV is not initially suspected. To assess the utility of this approach, a database of all CMV cultures performed from July 2001 to March 2012 was used to identify infants in whom urine CMV cultures were obtained within 100 days of life. Corresponding placentas were then identified through the pathology database. The database was also queried to identify placentas in which CMV immunohistochemical analysis had been performed. CMV PCR was positive in FFPE placental tissue from 100% (5/5) of cases in which the first urine culture collected before the first 21 days of life was positive. Placentas from 20 infants with negative CMV urine cultures were CMV PCR negative. Interestingly, CMV was detected in 12.5% (1/8) of placentas in which the first CMV-positive urine culture was collected after the first 21 days of life. Furthermore, 4% (1/26) of placentas with chronic villitis by histology (no urine cultures available) were CMV PCR positive. In the 10 CMV PCR-positive placentas, including 3 cases of fetal demise, CMV immunohistochemistry was positive in just 6 cases. These results suggest that the confirmation of CMV exposure in utero by PCR of FFPE placental tissue provides a useful adjunct to histologic evaluation and may identify infants requiring close clinical follow-up.
Collapse
|
184
|
Mwaanza N, Chilukutu L, Tembo J, Kabwe M, Musonda K, Kapasa M, Chabala C, Sinyangwe S, Mwaba P, Zumla A, Bates M. High rates of congenital cytomegalovirus infection linked with maternal HIV infection among neonatal admissions at a large referral center in sub-Saharan Africa. Clin Infect Dis 2013; 58:728-35. [PMID: 24265360 DOI: 10.1093/cid/cit766] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Congenital cytomegalovirus (CMV) infection is the major infectious cause of birth defects and hearing loss globally. There is a growing recognition of the potential clinical impact of congenital CMV infections in high-seroprevalence settings. METHODS A cross-sectional study of neonatal admissions at a large referral center in sub-Saharan Africa to determine the prevalence of both symptomatic and asymptomatic congenital CMV infection was performed. Real-time polymerase chain reaction was used to screen DNA-extracted sera, urine, and saliva, and an enzyme-linked immunosorbent assay was used to screen serum samples for anti-CMV immunoglobulin M. Multivariate binary logistic regression was used to identify risk factors associated with increased odds of congenital CMV infection. RESULTS Congenital CMV was detected in 3.8% (15/395) of neonates. Among these infants, 6 of 15 (40%) presented with jaundice, 1 of whom also had petechiae. Congenital CMV infection was detected in 9 of 79 (11.4%; 95% confidence interval [CI], 6.1%-20.3%) neonates born to human immunodeficiency virus (HIV)-infected mothers, and both maternal HIV (odds ratio [OR], 6.661 [95% CI, 2.126-20.876], P = .001) and jaundice (OR, 5.701 [95% CI, 1.776-18.306], P = .003) were independently linked with significantly increased odds of congenital CMV infection. CONCLUSIONS Congenital and early infant CMV infections may have important consequences for child health in sub-Saharan Africa and other high HIV and CMV seroprevalence populations globally.
Collapse
Affiliation(s)
- Nyaxewo Mwaanza
- University of Zambia and University College London Medical School Research and Training Programme
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
185
|
Murine cytomegalovirus protein pM92 is a conserved regulator of viral late gene expression. J Virol 2013; 88:131-42. [PMID: 24131717 DOI: 10.1128/jvi.02684-13] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In this study, we report that murine cytomegalovirus (MCMV) protein pM92 regulates viral late gene expression during virus infection. Previously, we have shown that MCMV protein pM79 and its human cytomegalovirus (HCMV) homologue pUL79 are required for late viral gene transcription. Identification of additional factors involved is critical to dissecting the mechanism of this regulation. We show here that pM92 accumulated abundantly at late times of infection in a DNA synthesis-dependent manner and localized to nuclear viral replication compartments. To investigate the role of pM92, we constructed a recombinant virus SMin92, in which pM92 expression was disrupted by an insertional/frameshift mutation. During infection, SMin92 accumulated representative viral immediate-early gene products, early gene products, and viral DNA sufficiently but had severe reduction in the accumulation of late gene products and was thus unable to produce infectious progeny. Coimmunoprecipitation and mass spectrometry analysis revealed an interaction between pM92 and pM79, as well as between their HCMV homologues pUL92 and pUL79. Importantly, we showed that the growth defect of pUL92-deficient HCMV could be rescued in trans by pM92. This study indicates that pM92 is an additional viral regulator of late gene expression, that these regulators (represented by pM92 and pM79) may need to complex with each other for their activity, and that pM92 and pUL92 share a conserved function in CMV infection. pM92 represents a potential new target for therapeutic intervention in CMV disease, and a gateway into studying a largely uncharted viral process that is critical to the viral life cycle.
Collapse
|
186
|
Gabrielli L, Bonasoni MP, Santini D, Piccirilli G, Chiereghin A, Guerra B, Landini MP, Capretti MG, Lanari M, Lazzarotto T. Human fetal inner ear involvement in congenital cytomegalovirus infection. Acta Neuropathol Commun 2013; 1:63. [PMID: 24252374 PMCID: PMC3893406 DOI: 10.1186/2051-5960-1-63] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 09/23/2013] [Indexed: 01/12/2023] Open
Abstract
Background Congenital cytomegalovirus (CMV) infection is a leading cause of sensorineural hearing loss (SNHL). The mechanisms of pathogenesis of CMV-related SNHL are still unclear. The aim is to study congenital CMV-related damage in the fetal inner ear, in order to better understand the underlying pathophysiology behind CMV-SNHL. Results We studied inner ears and brains of 20 human fetuses, all at 21 week gestational age, with a high viral load in the amniotic fluid, with and without ultrasound (US) brain abnormalities. We evaluated histological brain damage, inner ear infection, local inflammatory response and tissue viral load. Immunohistochemistry revealed that CMV was positive in 14/20 brains (70%) and in the inner ears of 9/20 fetuses (45%). In the cases with inner ear infection, the marginal cell layer of the stria vascularis was always infected, followed by infection in the Reissner’s membrane. The highest tissue viral load was observed in the inner ear with infected Organ of Corti. Vestibular labyrinth showed CMV infection of sensory cells in the utricle and in the crista ampullaris. US cerebral anomalies were detected in 6 cases, and in all those cases, the inner ear was always involved. In the other 14 cases with normal brain scan, histological brain damage was present in 8 fetuses and 3 of them presented inner ear infection. Conclusions CMV-infection of the marginal cell layer of the stria vascularis may alter potassium and ion circulation, dissipating the endocochlear potential with consequent SNHL. Although abnormal cerebral US is highly predictive of brain and inner ear damage, normal US findings cannot exclude them either.
Collapse
|
187
|
Kadambari S, Luck S, Davis A, Williams EJ, Berrington J, Griffiths PD, Sharland M. Clinically targeted screening for congenital CMV - potential for integration into the National Hearing Screening Programme. Acta Paediatr 2013; 102:928-33. [PMID: 23927434 DOI: 10.1111/apa.12335] [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: 09/10/2012] [Revised: 03/09/2013] [Accepted: 06/25/2013] [Indexed: 12/01/2022]
Abstract
UNLABELLED Screening for a condition should only be undertaken if certain strict criteria are met. Congenital CMV (cCMV) is a leading cause of sensorineuronal hearing loss (SNHL) and meets many of these criteria, but is not currently screened for in the UK. Ganciclovir reduces CMV-induced progressive SNHL if treatment is begun in the first month of life. The Newborn Hearing Screening Programme (NHSP) has been shown to identify SNHL at the earliest possible age. The potential of integrating screening for cCMV into the NHSP is discussed to consolidate the link between screening, early diagnosis and management. CONCLUSION The early diagnosis and treatment of cCMV may prevent a small proportion of late SNHL. In the absence of any screening programme, we provide evidence that clinically targeted screening through the NHSP is a potential option in the UK, enhancing the diagnostic pathway and enabling appropriate early treatment to reduce long-term morbidity.
Collapse
Affiliation(s)
- S Kadambari
- Paediatric Infectious Diseases Research Group; Jenner Wing; St George's University of London; London; UK
| | - S Luck
- Centre for Virology; University College London Medical School and Royal Free Hospital; London; UK
| | - A Davis
- Public Health England; London; UK
| | - EJ Williams
- Newcastle Neonatal Service; Royal Victoria Infirmary; Newcastle-upon-Tyne; UK
| | - J Berrington
- Newcastle Neonatal Service; Royal Victoria Infirmary; Newcastle-upon-Tyne; UK
| | - PD Griffiths
- Centre for Virology; University College London Medical School and Royal Free Hospital; London; UK
| | - M Sharland
- Paediatric Infectious Diseases Research Group; Jenner Wing; St George's University of London; London; UK
| |
Collapse
|
188
|
Dorn M, Lidzba K, Bevot A, Goelz R, Hauser TK, Wilke M. Long-term neurobiological consequences of early postnatal hCMV-infection in former preterms: a functional MRI study. Hum Brain Mapp 2013; 35:2594-606. [PMID: 24027137 DOI: 10.1002/hbm.22352] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 05/17/2013] [Accepted: 05/31/2013] [Indexed: 11/07/2022] Open
Abstract
Early postnatal infection with human cytomegalovirus (hCMV) may contribute to an adverse cognitive outcome in early preterm-born children (PT). We here set out to explore whether long-term neurobiological consequences of such an infection are detectable using fMRI in children and adolescents who were born very preterm and who either did (PThCMV+ ) or did not (PT(hCMV-)) suffer from an early postnatal hCMV-infection, when compared with typically developing healthy control (HC) subjects. Overall, data from 71 children and adolescents could be included, 34 PT (of which 15 were PT(hCMV+) and 19 were PT(hCMV-)) and 37 HC. Using a recently established "dual use" fMRI task, we investigated language and visuospatial functions. There were significant activation differences in the left hippocampus (PT > HC and PT(hCMV+) > HC), and in the right anterior cingulate cortex (PT(hCMV-) > PT(hCMV+)) when performing the language task. Surprisingly, only a small region in the occipital cortex showed a significant activation difference (HC > PT(HCMV-)) when performing the visuospatial task. Targeted analyses revealed differences in gray matter volume, but not density, in several brain regions. Our results suggest that long-term neurobiological consequences of an early postnatal hCMV infection are detectable even in older children and adolescents formerly born very preterm, compatible with a higher effort when performing a cognitive task. This suggests that measures to prevent such an infection are warranted. Furthermore, an interrelation of brain structure and function was detected that may constitute a severe confound when using fMRI to compare structurally differing groups.
Collapse
Affiliation(s)
- Maik Dorn
- Department of Pediatric Neurology and Developmental Medicine, Children's Hospital, University Hospital, Eberhard Karls University, Tübingen, Germany; Experimental Pediatric Neuroimaging, Children's Hospital, University Hospital, Eberhard Karls University, Tübingen, Germany; Department of Neuroradiology, University Hospital, Eberhard Karls University, Tübingen, Germany
| | | | | | | | | | | |
Collapse
|
189
|
Does congenital cytomegalovirus infection lead to hearing loss by inducing mutation of the GJB2 gene? Pediatr Res 2013; 74:121-6. [PMID: 23665763 DOI: 10.1038/pr.2013.77] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 02/11/2013] [Indexed: 11/08/2022]
Abstract
BACKGROUND Congenital cytomegalovirus (CMV) infection and mutation of the gap junction β-2 (GJB2) gene are important causes of sensorineural hearing loss (SNHL). This study aims to determine if congenital CMV infection leads to deafness by inducing GJB2 mutation. METHODS GJB2 gene sequencing and auditory brainstem response testing were performed in 159 neonates (63 with and 96 without CMV infection) from August 2008 to August 2011. For neonates with GJB2 mutation, their parents were further screened for GJB2 sequence. RESULTS The incidence of SNHL was 12.7% in CMV-infected but 0% in uninfected children aged 1-1.5 y (P = 0.000). Similar mutation rates of the GJB2 gene were observed in neonates with or without CMV infection (34.9 vs. 32.3%, respectively, P = 0.734). No significant difference in the mutation rate of GJB2 was found among neonates with CMV infection and SNHL, those with CMV infection and normal hearing, and uninfected newborns with normal hearing (P = 0.438). Mutations 79G>A, 109G>A, 341A>G, and 608T>C were found in neonates with and without CMV infection. All of the above mutations were also found in both or one of the corresponding parents. CONCLUSION Congenital CMV infections may cause deafness in neonates, but this might be independent of GJB2 gene mutation.
Collapse
|
190
|
Later passages of neural progenitor cells from neonatal brain are more permissive for human cytomegalovirus infection. J Virol 2013; 87:10968-79. [PMID: 23903847 DOI: 10.1128/jvi.01120-13] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Congenital human cytomegalovirus (HCMV) infection is the most frequent infectious cause of birth defects, primarily neurological disorders. Neural progenitor/stem cells (NPCs) are the major cell type in the subventricular zone and are susceptible to HCMV infection. In culture, the differentiation status of NPCs may change with passage, which in turn may alter susceptibility to virus infection. Previously, only early-passage (i.e., prior to passage 9) NPCs were studied and shown to be permissive to HCMV infection. In this study, NPC cultures derived at different gestational ages were evaluated after short (passages 3 to 6) and extended (passages 11 to 20) in vitro passages for biological and virological parameters (i.e., cell morphology, expression of NPC markers and HCMV receptors, viral entry efficiency, viral gene expression, virus-induced cytopathic effect, and release of infectious progeny). These parameters were not significantly influenced by the gestational age of the source tissues. However, extended-passage cultures showed evidence of initiation of differentiation, increased viral entry, and more efficient production of infectious progeny. These results confirm that NPCs are fully permissive for HCMV infection and that extended-passage NPCs initiate differentiation and are more permissive for HCMV infection. Later-passage NPCs being differentiated and more permissive for HCMV infection suggest that HCMV infection in fetal brain may cause more neural cell loss and give rise to severe neurological disabilities with advancing brain development.
Collapse
|
191
|
Abstract
Toxoplasma gondii, rubella, cytomegalovirus and herpes simplex virus have in common that they can cause congenital (TORCH) infection, leading to fetal and neonatal morbidity and mortality. During the last decades, TORCH screening, which is generally considered to be single serum testing, has been increasingly used inappropriately and questions have been raised concerning the indications and cost-effectiveness of TORCH testing. The problems of TORCH screening lie in requesting the screening for the wrong indications, wrong interpretation of the single serum results and in case there is a good indication for diagnosis of congenital infection, sending in the wrong materials. This review provides an overview of the pathogenesis, epidemiology and clinical consequences of congenital TORCH infections and discusses the indications for, and interpretation of, TORCH screens.
Collapse
Affiliation(s)
- Eveline P de Jong
- Department of Paediatrics, Juliana Children’s Hospital, HAGA Hospital, The Hague, The Netherlands
| | | | | | | |
Collapse
|
192
|
Abstract
Cytomegalovirus is the commonest congenital viral infection in the developed world, with an overall prevalence of approximately 0.6%. Approximately 10% of congenitally infected infants have signs and symptoms of disease at birth, and these symptomatic infants have a substantial risk of subsequent neurologic sequelae. These include sensorineural hearing loss, mental retardation, microcephaly, development delay, seizure disorders, and cerebral palsy. Antiviral therapy for children with symptomatic congenital cytomegalovirus infection is effective at reducing the risk of long-term disabilities and should be offered to families with affected newborns. An effective preconceptual vaccine against CMV could protect against long-term neurologic sequelae and other disabilities.
Collapse
|
193
|
Martines F, Martines E, Mucia M, Sciacca V, Salvago P. Prelingual sensorineural hearing loss and infants at risk: Western Sicily report. Int J Pediatr Otorhinolaryngol 2013; 77:513-8. [PMID: 23332734 DOI: 10.1016/j.ijporl.2012.12.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 12/05/2012] [Accepted: 12/18/2012] [Indexed: 10/27/2022]
Abstract
OBJECTIVE To evaluate independent etiologic factor associated with sensorineural hearing loss (SNHL) in newborn at risk; to study the role of their interaction especially in NICU infants who present often multiple risk factors for SNHL. METHODS The main risk factors for SNHL reported by JCIH 2007 were evaluated on 508 infant at risk ranging from 4 to 20 weeks of life, transferred to the Audiology Department of Palermo from the main births centers of Western Sicily. After a global audiological assessment, performed with TEOAE, tympanometry and ABR, the prevalence and the effect of risk factors was statistically studied through univariate and multivariate analysis on the total population (normal and deaf subjects). RESULTS Fifty-one infants (10.03%) were diagnosed with SNHL (45 bilateral and 6 monolateral) with a mean hearing threshold of 87.39 ± 28.25 dB HL; from logistic regression analysis family history of hearing impairment (HI) and TORCH infections resulted independent significant risk factors (P<0.00001 and P=0.024 respectively). High SNHL percentages were evidenced also in NICU babies, due to the various pathologies and risk factors presented by these infants, and among newborns who suffered from hyperbilirubinemia requiring exchange transfusion (11.97% and 9.52% respectively). Craniofacial abnormalities (CFA) and syndromes associated to HI showed an important relationship (P<0.00001) with conductive hearing loss (CHL). Multiple regression analysis of the variation in SNHL among NICU infants evidenced an increased risk for SNHL of 21.24% and of 19.33% respectively in preterm infants and in case of hyperbilirubinemia if respiratory distress is concomitant with these risk factors. It was also observed an higher risk of SNHL (99.66%) in case of coexistence of prematurity and hyperbilirubinemia. Finally among infants with very low birth weight (VLBW) it was evidenced a statistically difference between the mean weight of SNHL infants respect to NHL newborns (P=0.048). CONCLUSION The high SNHL prevalence (10.03%) in our cohort underlines how infants at risk are more susceptible to suffer from SNHL; in particular NICU newborns have a 33% greater chance of developing SNHL, because of the presence of multiple risk factors (or=1.33) and their interaction. As the number of coexisting risk factors increases, the prevalence of SNHL also increases (r(2)=0.93).
Collapse
Affiliation(s)
- Francesco Martines
- Università degli Studi di Palermo, Dipartimento di Biomedicina Sperimentale e Neuroscienze Cliniche, BioNeC, Sezione di Otorinolaringoiatria, Via del Vespro 129, 90127 Palermo, Italy.
| | | | | | | | | |
Collapse
|
194
|
Prevalence and risk factors for sensorineural hearing loss: Western Sicily overview. Eur Arch Otorhinolaryngol 2013; 270:3049-56. [DOI: 10.1007/s00405-013-2379-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 01/22/2013] [Indexed: 12/31/2022]
|
195
|
Nuñez-Ramos R, Becerril J, Blázquez D, Rojo P, de Vergas J, Folgueira D. Diagnóstico precoz de la infección congénita por citomegalovirus: oportunidades perdidas. Enferm Infecc Microbiol Clin 2013; 31:93-6. [DOI: 10.1016/j.eimc.2012.06.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 06/25/2012] [Accepted: 06/26/2012] [Indexed: 11/25/2022]
|
196
|
Schuster K, Henneke P, Huzly D, Speckmann C. Zytomegalievirusinfektionen im ersten Lebensjahr. Monatsschr Kinderheilkd 2013. [DOI: 10.1007/s00112-012-2807-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
197
|
Ward JA, Sidell DR, Nassar M, Reece AL, Choo DI. Safety of cidofovir by intratympanic delivery technique. Antivir Ther 2013; 19:97-105. [DOI: 10.3851/imp2693] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2013] [Indexed: 10/26/2022]
|
198
|
Manicklal S, Emery VC, Lazzarotto T, Boppana SB, Gupta RK. The "silent" global burden of congenital cytomegalovirus. Clin Microbiol Rev 2013; 26:86-102. [PMID: 23297260 PMCID: PMC3553672 DOI: 10.1128/cmr.00062-12] [Citation(s) in RCA: 680] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Human cytomegalovirus (CMV) is a leading cause of congenital infections worldwide. In the developed world, following the virtual elimination of circulating rubella, it is the commonest nongenetic cause of childhood hearing loss and an important cause of neurodevelopmental delay. The seroprevalence of CMV in adults and the incidence of congenital CMV infection are highest in developing countries (1 to 5% of births) and are most likely driven by nonprimary maternal infections. However, reliable estimates of prevalence and outcome from developing countries are not available. This is largely due to the dogma that maternal preexisting seroimmunity virtually eliminates the risk for sequelae. However, recent data demonstrating similar rates of sequelae, especially hearing loss, following primary and nonprimary maternal infection have underscored the importance of congenital CMV infection in resource-poor settings. Although a significant proportion of congenital CMV infections are attributable to maternal primary infection in well-resourced settings, the absence of specific interventions for seronegative mothers and uncertainty about fetal prognosis have discouraged routine maternal antibody screening. Despite these challenges, encouraging results from prototype vaccines have been reported, and the first randomized phase III trials of prenatal interventions and prolonged postnatal antiviral therapy are under way. Successful implementation of strategies to prevent or reduce the burden of congenital CMV infection will require heightened global awareness among clinicians and the general population. In this review, we highlight the global epidemiology of congenital CMV and the implications of growing knowledge in areas of prevention, diagnosis, prognosis, and management for both low (50 to 70%)- and high (>70%)-seroprevalence settings.
Collapse
Affiliation(s)
- Sheetal Manicklal
- Division of Medical Virology, Department of Clinical Laboratory Sciences, National Health Laboratory Service, Groote Schuur Hospital/University of Cape Town, Cape Town, South Africa
| | - Vincent C. Emery
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Tiziana Lazzarotto
- Operative Unit of Microbiology, St. Orsola Malpighi General Hospital/University of Bologna, Bologna, Italy
| | - Suresh B. Boppana
- Pediatrics and Microbiology, University of Alabama School of Medicine, Birmingham, Alabama, USA
| | - Ravindra K. Gupta
- Division of Infection and Immunity, University College London, London, United Kingdom
| |
Collapse
|
199
|
Melnick M, Jaskoll T. An in vitro mouse model of congenital cytomegalovirus-induced pathogenesis of the inner ear cochlea. ACTA ACUST UNITED AC 2012; 97:69-78. [PMID: 23281115 DOI: 10.1002/bdra.23105] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Revised: 10/22/2012] [Accepted: 11/06/2012] [Indexed: 11/10/2022]
Abstract
Congenital human cytomegalovirus (CMV) infection is the leading nongenetic etiology of sensorineural hearing loss (SNHL) at birth and prelingual SNHL not expressed at birth. The paucity of temporal bone autopsy specimens from infants with congenital CMV infection has hindered the critical correlation of histopathology with pathogenesis. Here, we present an in vitro embryonic mouse model of CMV-infected cochleas that mimics the human sites of viral infection and associated pathology. There is a striking dysplasia/hyperplasia in mouse CMV-infected cochlear epithelium and mesenchyme, including organ of Corti hair and supporting cells and stria vascularis. This is concomitant with significant dysregulation of p19, p21, p27, and Pcna gene expression, as well as proliferating cell nuclear antigen (PCNA) protein expression. Other pathologies similar to those arising from known deafness gene mutations include downregulation of KCNQ1 protein expression in the stria vascularis, as well as hypoplastic and dysmorphic melanocytes. Thus, this model provides a relevant and reliable platform within which the detailed cell and molecular biology of CMV-induced deafness may be studied.
Collapse
Affiliation(s)
- Michael Melnick
- Laboratory for Developmental Genetics, University of Southern California, Los Angeles, CA 90089, USA.
| | | |
Collapse
|
200
|
Cordier A, Guitton S, Vauloup-Fellous C, Grangeot-Keros L, Benachi A, Picone O. Awareness and knowledge of congenital cytomegalovirus infection among health care providers in France. J Clin Virol 2012; 55:158-63. [DOI: 10.1016/j.jcv.2012.06.022] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 05/22/2012] [Accepted: 06/29/2012] [Indexed: 10/28/2022]
|