251
|
Rascovan N, Duraisamy R, Desnues C. Metagenomics and the Human Virome in Asymptomatic Individuals. Annu Rev Microbiol 2017; 70:125-41. [PMID: 27607550 DOI: 10.1146/annurev-micro-102215-095431] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
High-throughput sequencing technologies have revolutionized how we think about viruses. Investigators can now go beyond pathogenic viruses and have access to the thousands of viruses that inhabit our bodies without causing clinical symptoms. By studying their interactions with each other, with other microbes, and with host genetics and immune systems, we can learn how they affect health and disease. This article reviews current knowledge of the composition and diversity of the human virome in physiologically healthy individuals. It focuses on recent results from metagenomics studies and discusses the contribution of bacteriophages and eukaryotic viruses to human health.
Collapse
Affiliation(s)
- Nicolás Rascovan
- Faculté de Médecine, Aix Marseille Université, 13385 Marseille, France.,URMITE, UM63, CNRS 7278, IRD 198, INSERM 1095, 13385 Marseille, France;
| | - Raja Duraisamy
- Faculté de Médecine, Aix Marseille Université, 13385 Marseille, France.,URMITE, UM63, CNRS 7278, IRD 198, INSERM 1095, 13385 Marseille, France;
| | - Christelle Desnues
- Faculté de Médecine, Aix Marseille Université, 13385 Marseille, France.,URMITE, UM63, CNRS 7278, IRD 198, INSERM 1095, 13385 Marseille, France;
| |
Collapse
|
252
|
The Human Virome: Implications for Clinical Practice in Transplantation Medicine. J Clin Microbiol 2017; 55:2884-2893. [PMID: 28724557 DOI: 10.1128/jcm.00489-17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Advances in DNA sequencing technology have provided an unprecedented opportunity to study the human virome. Transplant recipients and other immunocompromised hosts are at particular risk for developing virus-related pathology; thus, the impact of the virome on health and disease may be even more relevant in this population. Here, we discuss technical considerations in studying the human virome, the current literature on the virome in transplant recipients, and near-future applications of sequence-based findings that can further our understanding of viruses in transplantation medicine.
Collapse
|
253
|
Rutvisuttinunt W, Klungthong C, Thaisomboonsuk B, Chinnawirotpisan P, Ajariyakhajorn C, Manasatienkij W, Phonpakobsin T, Lon C, Saunders D, Wangchuk S, Shrestha SK, Velasco JMS, Alera MTP, Simasathien S, Buddhari D, Jarman RG, Macareo LR, Yoon IK, Fernandez S. Retrospective use of next-generation sequencing reveals the presence of Enteroviruses in acute influenza-like illness respiratory samples collected in South/South-East Asia during 2010-2013. J Clin Virol 2017; 94:91-99. [PMID: 28779659 PMCID: PMC7106496 DOI: 10.1016/j.jcv.2017.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 06/29/2017] [Accepted: 07/08/2017] [Indexed: 01/15/2023]
Abstract
Next-generation Sequencing (NGS) was adopted in routine respiratory pathogen surveillance from South/South East (S/SE) Asia during 2010–2013. From 12,865 respiratory collections from ILI patients, 324 CPE-positive from 4,478 viral isolations were negative by standard assays. The CPE-positive samples were pooled, screened using NGS and validated the presence of the pathogens identified from NGS. Herpes simplex virus type 1, parainfluenza, adenovirus, coronavirus, human metapneumovirus, mumps virus and enterovirus genus were detected. NGS on pooled samples can be applied to surveillance work, identifying medically important viruses which may have missed by conventional methods.
Background Emerging and re-emerging respiratory pathogens represent an increasing threat to public health. Etiological determination during outbreaks generally relies on clinical information, occasionally accompanied by traditional laboratory molecular or serological testing. Often, this limited testing leads to inconclusive findings. The Armed Forces Research Institute of Medical Sciences (AFRIMS) collected 12,865 nasopharyngeal specimens from acute influenza-like illness (ILI) patients in five countries in South/South East Asia during 2010–2013. Three hundred and twenty-four samples which were found to be negative for influenza virus after screening with real-time RT-PCR and cell-based culture techniques demonstrated the potential for viral infection with evident cytopathic effect (CPE) in several cell lines. Objective To assess whether whole genome next-generation sequencing (WG-NGS) together with conventional molecular assays can be used to reveal the etiology of influenza negative, but CPE positive specimens. Study design The supernatant of these CPE positive cell cultures were grouped in 32 pools containing 2–26 supernatants per pool. Three WG-NGS runs were performed on these supernatant pools. Sequence reads were used to identify positive pools containing viral pathogens. Individual samples in the positive pools were confirmed by qRT-PCR, RT-PCR, PCR and Sanger sequencing from the CPE culture and original clinical specimens. Results WG-NGS was an effective way to expand pathogen identification in surveillance studies. This enabled the identification of a viral agent in 71.3% (231/324) of unidentified surveillance samples, including common respiratory pathogens (100/324; 30.9%): enterovirus (16/100; 16.0%), coxsackievirus (31/100; 31.0%), echovirus (22/100; 22.0%), human rhinovirus (3/100; 3%), enterovirus genus (2/100; 2.0%), influenza A (9/100; 9.0%), influenza B, (5/100; 5.0%), human parainfluenza (4/100; 4.0%), human adenovirus (3/100; 3.0%), human coronavirus (1/100; 1.0%), human metapneumovirus (2/100; 2.0%), and mumps virus (2/100; 2.0%), in addition to the non-respiratory pathogen herpes simplex virus type 1 (HSV-1) (172/324; 53.1%) and HSV-1 co-infection with respiratory viruses (41/324; 12.7%).
Collapse
Affiliation(s)
- Wiriya Rutvisuttinunt
- Department of Virology, Armed Forces Research Institute of Medical Sciences, 315/6, Rajavithi Road, Rajathewi, Bangkok, Thailand; Walter Reed/AFRIMS Research Unit Nepal, Kathmandu, Nepal.
| | - Chonticha Klungthong
- Department of Virology, Armed Forces Research Institute of Medical Sciences, 315/6, Rajavithi Road, Rajathewi, Bangkok, Thailand
| | - Butsaya Thaisomboonsuk
- Department of Virology, Armed Forces Research Institute of Medical Sciences, 315/6, Rajavithi Road, Rajathewi, Bangkok, Thailand
| | - Piyawan Chinnawirotpisan
- Department of Virology, Armed Forces Research Institute of Medical Sciences, 315/6, Rajavithi Road, Rajathewi, Bangkok, Thailand
| | - Chuanpis Ajariyakhajorn
- Department of Virology, Armed Forces Research Institute of Medical Sciences, 315/6, Rajavithi Road, Rajathewi, Bangkok, Thailand
| | - Wudtichai Manasatienkij
- Department of Virology, Armed Forces Research Institute of Medical Sciences, 315/6, Rajavithi Road, Rajathewi, Bangkok, Thailand
| | - Thipwipha Phonpakobsin
- Department of Virology, Armed Forces Research Institute of Medical Sciences, 315/6, Rajavithi Road, Rajathewi, Bangkok, Thailand
| | - Chanthap Lon
- Department of Immunology and Medicine, Armed Forces Research Institute of Medical Sciences, 315/6, Rajavithi Road, Rajathewi, Bangkok, Thailand
| | - David Saunders
- Department of Immunology and Medicine, Armed Forces Research Institute of Medical Sciences, 315/6, Rajavithi Road, Rajathewi, Bangkok, Thailand
| | - Sonam Wangchuk
- Royal Centre for Disease Control, Department of Public Health, Ministry of Health, Thimphu, Bhutan
| | - Sanjaya K Shrestha
- Walter Reed/AFRIMS Research Unit Nepal, Kathmandu, Nepal; Center for International Health, University of Bergen, Norway
| | - John Mark S Velasco
- Department of Virology, Armed Forces Research Institute of Medical Sciences, 315/6, Rajavithi Road, Rajathewi, Bangkok, Thailand
| | - Maria Theresa P Alera
- Department of Virology, Armed Forces Research Institute of Medical Sciences, 315/6, Rajavithi Road, Rajathewi, Bangkok, Thailand
| | | | - Darunee Buddhari
- Department of Virology, Armed Forces Research Institute of Medical Sciences, 315/6, Rajavithi Road, Rajathewi, Bangkok, Thailand
| | - Richard G Jarman
- Department of Virology, Armed Forces Research Institute of Medical Sciences, 315/6, Rajavithi Road, Rajathewi, Bangkok, Thailand
| | - Louis R Macareo
- Department of Virology, Armed Forces Research Institute of Medical Sciences, 315/6, Rajavithi Road, Rajathewi, Bangkok, Thailand
| | - In-Kyu Yoon
- Department of Virology, Armed Forces Research Institute of Medical Sciences, 315/6, Rajavithi Road, Rajathewi, Bangkok, Thailand
| | - Stefan Fernandez
- Department of Virology, Armed Forces Research Institute of Medical Sciences, 315/6, Rajavithi Road, Rajathewi, Bangkok, Thailand.
| |
Collapse
|
254
|
Albert E, Solano C, Pascual T, Torres I, Macera L, Focosi D, Maggi F, Giménez E, Amat P, Navarro D. Dynamics of Torque Teno virus plasma DNAemia in allogeneic stem cell transplant recipients. J Clin Virol 2017; 94:22-28. [PMID: 28710997 DOI: 10.1016/j.jcv.2017.07.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 05/05/2017] [Accepted: 07/03/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND Torque Teno virus (TTV) plasma DNA load directly correlate with the level of immunosuppresion in different clinical settings. It is uncertain whether this may be the case in allogeneic hematopoietic stem cell transplant recipients (allo-HSCT). OBJECTIVES We characterized the dynamics of TTV DNAemia in patients undergoing T-cell replete allo-SCT. STUDY DESIGN Retrospective single-center observational study including 72 allo-HSCT patients. Plasma TTV DNA loads were quantified before initiating the conditioning regimen and at different time-points after transplant by real-time PCR. White blood cells (WBC) and absolute lymphocyte counts (ALC) were measured by flow cytometry. RESULTS A dramatic drop in plasma TTV DNA load was observed shortly after conditioning. The TTV DNA load increased steadily after engraftment reaching its peak at day +90 after transplant. The increase in TTV DNA load paralleled that of ALC, and was of greater magnitude in patients who developed severe (grades II-IV) acute graft vs. host disease. CONCLUSION Repopulation of lymphocytes early after allo-HSCT correlates with an increase of plasma TTV DNA load. Prospective studies are nevertheless needed to determine whether the kinetics of TTV DNAemia may allow inference of the degree of overall immunocompetence in these patients.
Collapse
Affiliation(s)
- Eliseo Albert
- Microbiology Service, Hospital Clínico Universitario, Institute for Research INCLIVA, Valencia, Spain
| | - Carlos Solano
- Hematology Service, Hospital Clínico Universitario, Institute for Research INCLIVA, Valencia, Spain; Department of Medicine, School of Medicine, University of Valencia, Valencia, Spain
| | - Tania Pascual
- Microbiology Service, Hospital Clínico Universitario, Institute for Research INCLIVA, Valencia, Spain
| | - Ignacio Torres
- Microbiology Service, Hospital Clínico Universitario, Institute for Research INCLIVA, Valencia, Spain
| | - Lisa Macera
- Virology Unit, Pisa University Hospital, Pisa, Italy
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | | | - Estela Giménez
- Microbiology Service, Hospital Clínico Universitario, Institute for Research INCLIVA, Valencia, Spain
| | - Paula Amat
- Hematology Service, Hospital Clínico Universitario, Institute for Research INCLIVA, Valencia, Spain
| | - David Navarro
- Microbiology Service, Hospital Clínico Universitario, Institute for Research INCLIVA, Valencia, Spain; Department of Microbiology, School of Medicine, University of Valencia, Valencia, Spain.
| |
Collapse
|
255
|
Kandathil AJ, Breitwieser FP, Sachithanandham J, Robinson M, Mehta SH, Timp W, Salzberg SL, Thomas DL, Balagopal A. Presence of Human Hepegivirus-1 in a Cohort of People Who Inject Drugs. Ann Intern Med 2017; 167:1-7. [PMID: 28586923 PMCID: PMC5721525 DOI: 10.7326/m17-0085] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Next-generation metagenomic sequencing (NGMS) has opened new frontiers in microbial discovery but has been clinically characterized in only a few settings. OBJECTIVE To explore the plasma virome of persons who inject drugs and to characterize the sensitivity and accuracy of NGMS compared with quantitative clinical standards. DESIGN Longitudinal and cross-sectional studies. SETTING A clinical trial (ClinicalTrials.gov: NCT01285050) and a well-characterized cohort study of persons who have injected drugs. PARTICIPANTS Persons co-infected with hepatitis C virus (HCV) and HIV. MEASUREMENTS Viral nucleic acid in plasma by NGMS and quantitative polymerase chain reaction (PCR). RESULTS Next-generation metagenomic sequencing generated a total of 600 million reads, which included the expected HIV and HCV RNA sequences. HIV and HCV reads were consistently identified only when samples contained more than 10 000 copies/mL or IU/mL, respectively, as determined by quantitative PCR. A novel RNA virus, human hepegivirus-1 (HHpgV-1), was also detected by NGMS in 4 samples from 2 persons in the clinical trial. Through use of a quantitative PCR assay for HHpgV-1, infection was also detected in 17 (10.9%) of 156 members of a cohort of persons who injected drugs. In these persons, HHpgV-1 viremia persisted for a median of at least 4538 days and was associated with detection of other bloodborne viruses, such as HCV RNA and SEN virus D. LIMITATION The medical importance of HHpgV-1 infection is unknown. CONCLUSION Although NGMS is insensitive for detection of viruses with relatively low plasma nucleic acid concentrations, it may have broad potential for discovery of new viral infections of possible medical importance, such as HHpgV-1. PRIMARY FUNDING SOURCE National Institutes of Health.
Collapse
Affiliation(s)
- Abraham J Kandathil
- From Johns Hopkins University and Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Florian P Breitwieser
- From Johns Hopkins University and Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | | | - Matthew Robinson
- From Johns Hopkins University and Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Shruti H Mehta
- From Johns Hopkins University and Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Winston Timp
- From Johns Hopkins University and Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Steven L Salzberg
- From Johns Hopkins University and Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - David L Thomas
- From Johns Hopkins University and Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Ashwin Balagopal
- From Johns Hopkins University and Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| |
Collapse
|
256
|
Torque Teno Virus Load-Inverse Association With Antibody-Mediated Rejection After Kidney Transplantation. Transplantation 2017; 101:360-367. [PMID: 27525643 DOI: 10.1097/tp.0000000000001455] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Antibody-mediated rejection (AMR) represents one of the cardinal causes of late allograft loss after kidney transplantation, and there is great need for noninvasive tools improving early diagnosis of this rejection type. One promising strategy might be the quantification of peripheral blood DNA levels of the highly prevalent and apathogenic Torque Teno virus (TTV), which might mirror the overall level of immunosuppression and thus help determine the risk of alloimmune response. METHODS To assess the association between TTV load in the peripheral blood and AMR, 715 kidney transplant recipients (median, 6.3 years posttransplantation) were subjected to a systematical cross-sectional AMR screening and, in parallel, TTV quantification. RESULTS Eighty-six of these recipients had donor-specific antibodies and underwent protocol biopsy, AMR-positive patients (n = 46) showed only 25% of the TTV levels measured in patients without AMR (P = 0.003). In a generalized linear model, higher TTV levels were associated with a decreased risk for AMR after adjustment for potential confounders (risk ratio 0.94 per TTV log level; 95% confidence interval 0.90-0.99; P = 0.02). CONCLUSIONS Future studies will have to clarify whether longitudinal assessment of TTV load might predict AMR risk and help guide the type and intensity of immunosuppression to prevent antibody-mediated graft injury.
Collapse
|
257
|
Wekerle T, Segev D, Lechler R, Oberbauer R. Strategies for long-term preservation of kidney graft function. Lancet 2017; 389:2152-2162. [PMID: 28561006 DOI: 10.1016/s0140-6736(17)31283-7] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 03/13/2017] [Accepted: 03/16/2017] [Indexed: 12/21/2022]
Abstract
Kidney transplantation has become a routine procedure in the treatment of patients with kidney failure, and requires collaboration of experts from different disciplines, such as nephrology, surgery, immunology, pathology, infectious disease medicine, cardiology, and oncology. Grafts can be obtained from deceased or living donors, with different logistical requirements and implications for long-term graft patency. 1-year graft survival rates are greater than 95% in many centres but improvement of long-term function remains a challenge. New developments in molecular immunology and computational biology have increased precision of donor and recipient matching of HLA and non-HLA compatibility. Individual omics-wide molecular diagnostics, extracorporeal therapies, and drug developments allow for precise individual decision making and treatment. Tolerance induction by mixed chimerism without toxic conditioning and with a low risk of graft versus host disease is a visionary but realistic goal. Some of these innovations are already used in modern transplant centres and will allow advancement in long-term allograft preservation.
Collapse
Affiliation(s)
- Thomas Wekerle
- Department of Surgery, Section of Transplantation Immunology, Medical University of Vienna, Vienna, Austria
| | - Dorry Segev
- Department of Surgery, Johns Hopkins University, Baltimore, MD, USA
| | - Robert Lechler
- MRC Centre for Transplantation, King's College London, London, UK
| | - Rainer Oberbauer
- Department of Nephrology and Dialysis, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
258
|
Agbor-Enoh S, Tunc I, De Vlaminck I, Fideli U, Davis A, Cuttin K, Bhatti K, Marishta A, Solomon MA, Jackson A, Graninger G, Harper B, Luikart H, Wylie J, Wang X, Berry G, Marboe C, Khush K, Zhu J, Valantine H. Applying rigor and reproducibility standards to assay donor-derived cell-free DNA as a non-invasive method for detection of acute rejection and graft injury after heart transplantation. J Heart Lung Transplant 2017. [PMID: 28624139 DOI: 10.1016/j.healun.2017.05.026] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Use of new genomic techniques in clinical settings requires that such methods are rigorous and reproducible. Previous studies have shown that quantitation of donor-derived cell-free DNA (%ddcfDNA) by unbiased shotgun sequencing is a sensitive, non-invasive marker of acute rejection after heart transplantation. The primary goal of this study was to assess the reproducibility of %ddcfDNA measurements across technical replicates, manual vs automated platforms, and rejection phenotypes in distinct patient cohorts. METHODS After developing and validating the %ddcfDNA assay, we subjected the method to a rigorous test of its reproducibility. We measured %ddcfDNA in technical replicates performed by 2 independent laboratories and verified the reproducibility of %ddcfDNA patterns of 2 rejection phenotypes: acute cellular rejection and antibody-mediated rejection in distinct patient cohorts. RESULTS We observed strong concordance of technical-replicate %ddcfDNA measurements across 2 independent laboratories (slope = 1.02, R2 > 0.99, p < 10-6), as well as across manual and automated platforms (slope = 0.80, R2 = 0.92, p < 0.001). The %ddcfDNA measurements in distinct heart transplant cohorts had similar baselines and error rates. The %ddcfDNA temporal patterns associated with rejection phenotypes were similar in both patient cohorts; however, the quantity of ddcfDNA was significantly higher in samples with severe vs mild histologic rejection grade (2.73% vs 0.14%, respectively; p < 0.001). CONCLUSIONS The %ddcfDNA assay is precise and reproducible across laboratories and in samples from 2 distinct types of heart transplant rejection. These findings pave the way for larger studies to assess the clinical utility of %ddcfDNA as a marker of acute rejection after heart transplantation.
Collapse
Affiliation(s)
- Sean Agbor-Enoh
- Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland; Pulmonary and Critical Care Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland; Laboratory of Transplantation Genomics, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Ilker Tunc
- Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Iwijn De Vlaminck
- Department of Bioengineering, Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Ulgen Fideli
- Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland; Laboratory of Transplantation Genomics, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Andrew Davis
- Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland; Laboratory of Transplantation Genomics, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Karen Cuttin
- Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland; Laboratory of Transplantation Genomics, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Kenneth Bhatti
- Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland; Laboratory of Transplantation Genomics, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Argit Marishta
- Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland; Laboratory of Transplantation Genomics, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Michael A Solomon
- Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland; Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Annette Jackson
- Pulmonary and Critical Care Medicine, The Johns Hopkins School of Medicine, Baltimore, Maryland; Laboratory of Transplantation Genomics, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Grace Graninger
- Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Bonnie Harper
- Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Helen Luikart
- Department of Medicine, Stanford University School of Medicine, Palo Alto, California
| | - Jennifer Wylie
- Department of Medicine, Stanford University School of Medicine, Palo Alto, California
| | - Xujing Wang
- Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Gerald Berry
- Department of Medicine, Stanford University School of Medicine, Palo Alto, California
| | - Charles Marboe
- Department of Medicine, New York Presbyterian University Hospital of Cornell and Columbia, New York, New York
| | - Kiran Khush
- Department of Medicine, New York Presbyterian University Hospital of Cornell and Columbia, New York, New York
| | - Jun Zhu
- Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland
| | - Hannah Valantine
- Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, Maryland; Laboratory of Transplantation Genomics, National Heart, Lung, and Blood Institute, Bethesda, Maryland.
| |
Collapse
|
259
|
Precision monitoring of immunotherapies in solid organ and hematopoietic stem cell transplantation. Adv Drug Deliv Rev 2017. [PMID: 28625828 DOI: 10.1016/j.addr.2017.06.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Pharmacological immunotherapies are a key component of post-transplant therapy in solid-organ and hematopoietic stem cell transplantation. In current clinical practice, immunotherapies largely follow a one-size fits all approach, leaving a large portion of transplant recipients either over- or under-immunosuppressed, and consequently at risk of infections or immune-mediated complications. Our goal here is to review recent and rapid advances in precision and genomic medicine approaches to monitoring of post-transplant immunotherapies. We will discuss recent advances in precision measurements of pharmacological immunosuppression, measurements of the plasma and gut microbiome, strategies to monitor for allograft injury and post-transplant malignancies via circulating cell-free DNA, and comprehensive measurements of the B and T cell immune cell repertoire.
Collapse
|
260
|
Riella LV, Bagley J, Iacomini J, Alegre ML. Impact of environmental factors on alloimmunity and transplant fate. J Clin Invest 2017; 127:2482-2491. [PMID: 28481225 DOI: 10.1172/jci90596] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Although gene-environment interactions have been investigated for many years to understand people's susceptibility to autoimmune diseases or cancer, a role for environmental factors in modulating alloimmune responses and transplant outcomes is only now beginning to emerge. New data suggest that diet, hyperlipidemia, pollutants, commensal microbes, and pathogenic infections can all affect T cell activation, differentiation, and the kinetics of graft rejection. These observations reveal opportunities for novel therapeutic interventions to improve graft outcomes as well as for noninvasive biomarker discovery to predict or diagnose graft deterioration before it becomes irreversible. In this Review, we will focus on the impact of these environmental factors on immune function and, when known, on alloimmune function, as well as on transplant fate.
Collapse
Affiliation(s)
- Leonardo V Riella
- Schuster Family Transplantation Research Center, Renal Division, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Jessamyn Bagley
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Sackler School of Biomedical Sciences Programs in Immunology and Genetics, Boston, Massachusetts, USA
| | - John Iacomini
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, Sackler School of Biomedical Sciences Programs in Immunology and Genetics, Boston, Massachusetts, USA
| | | |
Collapse
|
261
|
Abbas AA, Diamond J, Chehoud C, Chang B, Kotzin J, Young J, Imai I, Haas A, Cantu E, Lederer D, Meyer K, Milewski R, Olthoff K, Shaked A, Christie J, Bushman F, Collman R. The Perioperative Lung Transplant Virome: Torque Teno Viruses Are Elevated in Donor Lungs and Show Divergent Dynamics in Primary Graft Dysfunction. Am J Transplant 2017; 17:1313-1324. [PMID: 27731934 PMCID: PMC5389935 DOI: 10.1111/ajt.14076] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 09/12/2016] [Accepted: 09/26/2016] [Indexed: 01/25/2023]
Abstract
Primary graft dysfunction (PGD) is a principal cause of early morbidity and mortality after lung transplantation, but its pathogenic mechanisms are not fully clarified. To date, studies using standard clinical assays have not linked microbial factors to PGD. We previously used comprehensive metagenomic methods to characterize viruses in lung allografts >1 mo after transplant and found that levels of Anellovirus, mainly torque teno viruses (TTVs), were significantly higher than in nontransplanted healthy controls. We used quantitative polymerase chain reaction to analyze TTV and shotgun metagenomics to characterize full viral communities in acellular bronchoalveolar lavage from donor organs and postreperfusion allografts in PGD and non-PGD lung transplant recipient pairs. Unexpectedly, TTV DNA levels were elevated 100-fold in donor lungs compared with healthy adults (p = 0.0026). Although absolute TTV levels did not differ by PGD status, PGD cases showed a smaller increase in TTV levels from before to after transplant than did control recipients (p = 0.041). Metagenomic sequencing revealed mainly TTV and bacteriophages of respiratory tract bacteria, but no viral taxa distinguished PGD cases from controls. These findings suggest that conditions associated with brain death promote TTV replication and that greater immune activation or tissue injury associated with PGD may restrict TTV abundance in the lung.
Collapse
Affiliation(s)
- A. A. Abbas
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - J.M. Diamond
- Pulmonary, Allergy and Critical Care Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA,Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - C. Chehoud
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - B. Chang
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - J.J. Kotzin
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - J.C. Young
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - I. Imai
- Pulmonary, Allergy and Critical Care Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - A.R. Haas
- Pulmonary, Allergy and Critical Care Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - E. Cantu
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - D.J. Lederer
- Departments of Medicine and Epidemiology, College of Physicians and Surgeons, Columbia University, New York, NY
| | - K. Meyer
- School of Medicine and Public Health, University of Wisconsin, Madison, WI
| | - R.K. Milewski
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - K.M. Olthoff
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - A. Shaked
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - J.D. Christie
- Pulmonary, Allergy and Critical Care Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA,Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - F.D. Bushman
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA,Corresponding authors: Frederic Bushman: , Ronald Collman:
| | - R.G. Collman
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA,Pulmonary, Allergy and Critical Care Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA,Corresponding authors: Frederic Bushman: , Ronald Collman:
| |
Collapse
|
262
|
Sartor RB, Wu GD. Roles for Intestinal Bacteria, Viruses, and Fungi in Pathogenesis of Inflammatory Bowel Diseases and Therapeutic Approaches. Gastroenterology 2017; 152:327-339.e4. [PMID: 27769810 PMCID: PMC5511756 DOI: 10.1053/j.gastro.2016.10.012] [Citation(s) in RCA: 535] [Impact Index Per Article: 76.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 10/13/2016] [Accepted: 10/14/2016] [Indexed: 02/08/2023]
Abstract
Intestinal microbiota are involved in the pathogenesis of Crohn's disease, ulcerative colitis, and pouchitis. We review the mechanisms by which these gut bacteria, fungi, and viruses mediate mucosal homeostasis via their composite genes (metagenome) and metabolic products (metabolome). We explain how alterations to their profiles and functions under conditions of dysbiosis contribute to inflammation and effector immune responses that mediate inflammatory bowel diseases (IBD) in humans and enterocolitis in mice. It could be possible to engineer the intestinal environment by modifying the microbiota community structure or function to treat patients with IBD-either with individual agents, via dietary management, or as adjuncts to immunosuppressive drugs. We summarize the latest information on therapeutic use of fecal microbial transplantation and propose improved strategies to selectively normalize the dysbiotic microbiome in personalized approaches to treatment.
Collapse
Affiliation(s)
- R Balfour Sartor
- Departments of Medicine, Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| | - Gary D Wu
- Division of Gastroenterology, Perelman School of Medicine, the University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
263
|
Janowski AB, Krishnamurthy SR, Lim ES, Zhao G, Brenchley JM, Barouch DH, Thakwalakwa C, Manary MJ, Holtz LR, Wang D. Statoviruses, A novel taxon of RNA viruses present in the gastrointestinal tracts of diverse mammals. Virology 2017; 504:36-44. [PMID: 28152382 PMCID: PMC5515247 DOI: 10.1016/j.virol.2017.01.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/13/2017] [Accepted: 01/17/2017] [Indexed: 01/21/2023]
Abstract
Next-generation sequencing has expanded our understanding of the viral populations that constitute the mammalian virome. We describe a novel taxon of viruses named Statoviruses, for Stool associated Tombus-like viruses, present in multiple metagenomic datasets. These viruses define a novel clade that is phylogenetically related to the RNA virus families Tombusviridae and Flaviviridae. Five distinct statovirus types were identified in human, macaque, mouse, and cow gastrointestinal tract samples. The prototype genome, statovirus A, was frequently identified in macaque stool samples from multiple geographically distinct cohorts. Another genome, statovirus C1, was discovered in a stool sample from a human child with fever, cough, and rash. Further experimental data will clarify whether these viruses are infectious to mammals or if they originate from another source present in the mammalian gastrointestinal tract.
Collapse
Affiliation(s)
- Andrew B Janowski
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
| | - Siddharth R Krishnamurthy
- Department of Molecular Microbiology and Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Efrem S Lim
- Department of Molecular Microbiology and Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Guoyan Zhao
- Department of Molecular Microbiology and Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Jason M Brenchley
- Lab of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Dan H Barouch
- Center for Virology and Vaccine Research Beth Israel Deaconess Medical Center, Boston, MA, USA; Ragon Institute of MGH, MIT, and Harvard, Boston, MA, USA
| | - Chrissie Thakwalakwa
- Department of Community Health, College of Medicine, University of Malawi, Blantyre 3, Malawi
| | - Mark J Manary
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
| | - Lori R Holtz
- Department of Pediatrics, Washington University School of Medicine, St Louis, MO, USA
| | - David Wang
- Department of Molecular Microbiology and Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
264
|
Godkin A, Smith KA. Chronic infections with viruses or parasites: breaking bad to make good. Immunology 2017; 150:389-396. [PMID: 28009488 PMCID: PMC5343343 DOI: 10.1111/imm.12703] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 12/02/2016] [Accepted: 12/16/2016] [Indexed: 12/19/2022] Open
Abstract
Eukaryotic forms of life have been continually invaded by microbes and larger multicellular parasites, such as helminths. Over a billion years ago bacterial endosymbionts permanently colonized eukaryotic cells leading to recognized organelles with a distinct genetic lineage, such as mitochondria and chloroplasts. Colonization of our skin and mucosal surfaces with bacterial commensals is now known to be important for host health. However, the contribution of chronic virus and parasitic infections to immune homeostasis is being increasingly questioned. Persistent infection does not necessarily equate to exhibiting a chronic illness: healthy hosts (e.g. humans) have chronic viral and parasitic infections with no evidence of disease. Indeed, there are now examples of complex interactions between these microbes and hosts that seem to confer an advantage to the host at a particular time, suggesting that the relationship has progressed along an axis from parasitic to commensal to one of a mutualistic symbiosis. This concept is explored using examples from viruses and parasites, considering how the relationships may be not only detrimental but also beneficial to the human host.
Collapse
Affiliation(s)
- Andrew Godkin
- Division of Infection and Immunity, Cardiff University, Cardiff, Glamorgan, UK
| | - Katherine A Smith
- Division of Infection and Immunity, Cardiff University, Cardiff, Glamorgan, UK
| |
Collapse
|
265
|
VirusSeeker, a computational pipeline for virus discovery and virome composition analysis. Virology 2017; 503:21-30. [PMID: 28110145 DOI: 10.1016/j.virol.2017.01.005] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/07/2017] [Accepted: 01/10/2017] [Indexed: 01/21/2023]
Abstract
The advent of Next Generation Sequencing (NGS) has vastly increased our ability to discover novel viruses and to systematically define the spectrum of viruses present in a given specimen. Such studies have led to the discovery of novel viral pathogens as well as broader associations of the virome with diverse diseases including inflammatory bowel disease, severe acute malnutrition and HIV/AIDS. Critical to the success of these efforts are robust bioinformatic pipelines for rapid classification of microbial sequences. Existing computational tools are typically focused on either eukaryotic virus discovery or virome composition analysis but not both. Here we present VirusSeeker, a BLAST-based NGS data analysis pipeline designed for both purposes. VirusSeeker has been successfully applied in several previously published virome studies. Here we demonstrate the functionality of VirusSeeker in both novel virus discovery and virome composition analysis.
Collapse
|
266
|
Li DK, Chen H, Ferber J, Odouli R. Infection and antibiotic use in infancy and risk of childhood obesity: a longitudinal birth cohort study. Lancet Diabetes Endocrinol 2017; 5:18-25. [PMID: 27815091 DOI: 10.1016/s2213-8587(16)30281-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/13/2016] [Accepted: 09/14/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND Data from previous studies have suggested a possible association between antibiotic use in infancy and risk of childhood obesity, with implications for health-care delivery and obesity prevention strategies. However, whether the observed association was due to antibiotic use or underlying infection, or both, is unclear. We aimed to disentangle the effect of antibiotic use in infancy from that of underlying infection on the risk of childhood obesity. METHODS In this longitudinal birth cohort study, we included infants in the Kaiser Permanente Northern California population born between Jan 1, 1997, and March 31, 2013. We used electronic medical records to ascertain data for antibiotic use, infection diagnosis, and anthropometric measurements (and thus BMI and obesity status) from birth up to age 18 years. We used standard mixed-effects logistic regression for repeated measurements to analyse multiple BMI measurements per child (median five measurements) and to obtain odds ratios (ORs) and 95% CIs for obesity risk. We also did a substudy in 547 same-sex twin pairs with discordant exposure status to substantiate our findings. FINDINGS 260 556 individuals were included in our analysis. After controlling for maternal age, race or ethnic origin, pre-pregnancy BMI, preterm delivery, low birthweight, maternal antibiotic use, and infection during pregnancy, infection without antibiotic use in infancy was associated with an increased risk of childhood obesity compared with controls without infection (OR 1·25, 95% CI 1·20-1·29). A clear dose-response relation was seen between infection episodes and risk of childhood obesity (ptrend <0·0001). By contrast, compared with infants with untreated infection, antibiotic use during infancy was not associated with risk of childhood obesity (1·01, 0·98-1·04). Neither broad-spectrum nor narrow-spectrum antibiotics were associated with risk of childhood obesity. These findings were supported by the results of the twin set analysis. INTERPRETATION Infection, but not antibiotic use, during infancy is associated with risk of childhood obesity. This finding will need to be replicated in future studies. Although our results do not rule out a potential effect of antibiotics on microbiome composition and the use of antibiotics should always be judicious, they suggest that treatment of common infections with antibiotics in infancy is unlikely to be a main contributor to childhood obesity. FUNDING Kaiser Permanente Center for Effectiveness & Safety Research.
Collapse
Affiliation(s)
- De-Kun Li
- Division of Research, Kaiser Foundation Research Institute, Kaiser Permanente Northern California, Oakland, CA, USA.
| | - Hong Chen
- Division of Research, Kaiser Foundation Research Institute, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Jeannette Ferber
- Division of Research, Kaiser Foundation Research Institute, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Roxana Odouli
- Division of Research, Kaiser Foundation Research Institute, Kaiser Permanente Northern California, Oakland, CA, USA
| |
Collapse
|
267
|
Mitchell AB, Oliver BGG, Glanville AR. Translational Aspects of the Human Respiratory Virome. Am J Respir Crit Care Med 2016; 194:1458-1464. [DOI: 10.1164/rccm.201606-1278ci] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
268
|
|
269
|
Abstract
The human immune system is highly variable between individuals but relatively stable over time within a given person. Recent conceptual and technological advances have enabled systems immunology analyses, which reveal the composition of immune cells and proteins in populations of healthy individuals. The range of variation and some specific influences that shape an individual's immune system is now becoming clearer. Human immune systems vary as a consequence of heritable and non-heritable influences, but symbiotic and pathogenic microbes and other non-heritable influences explain most of this variation. Understanding when and how such influences shape the human immune system is key for defining metrics of immunological health and understanding the risk of immune-mediated and infectious diseases.
Collapse
Affiliation(s)
- Petter Brodin
- Science for Life Laboratory, Department of Medicine, Solna, Karolinska Institutet, Stockholm 17165, Sweden.,Department of Neonatology, Karolinska University Hospital, Stockholm 14186, Sweden
| | - Mark M Davis
- Department of Microbiology and Immunology, Stanford University School of Medicine.,Institute of Immunity, Transplantation and Infection, Stanford University School of Medicine.,Howard Hughes Medical Institute, Stanford University School of Medicine, California 94304, USA
| |
Collapse
|
270
|
Taur Y. Intestinal microbiome changes and stem cell transplantation: Lessons learned. Virulence 2016; 7:930-938. [PMID: 27805463 PMCID: PMC5160401 DOI: 10.1080/21505594.2016.1250982] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 10/14/2016] [Accepted: 10/17/2016] [Indexed: 02/08/2023] Open
Abstract
Studies of the microbiome in the setting of haematopoietic stem cell transplantation (SCT) have shown evidence that intestinal microbes appear to play a particularly important role in determining the outcome of treatment, impacting complications such as infection or graft-versus-host disease. Past studies may vary in terms of the level at which the microbiome is examined, leading to different but overlapping systems of taxonomy or nomenclature, which may be difficult for non-specialists to understand. This article will review the current body of work examining the clinical impact of the microbiome on SCT, and will provide a basic framework for the bacterial phylogenetic structure upon which the results of these studies rest. With this framework it can be shown that recurring patterns do emerge in prior studies identifying the microbes that confer benefit in this population.
Collapse
Affiliation(s)
- Ying Taur
- Medicine, Infectious Diseases Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
271
|
Georgel P. Innate immune receptors in solid organ transplantation. Hum Immunol 2016; 77:1071-1075. [DOI: 10.1016/j.humimm.2016.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 03/18/2016] [Accepted: 04/04/2016] [Indexed: 12/13/2022]
|
272
|
A Snapshot of Antibody-Mediated Rejection Goes Viral. Transplantation 2016; 101:227. [PMID: 27779575 DOI: 10.1097/tp.0000000000001540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
273
|
Ngoi CN, Siqueira J, Li L, Deng X, Mugo P, Graham SM, Price MA, Sanders EJ, Delwart E. The plasma virome of febrile adult Kenyans shows frequent parvovirus B19 infections and a novel arbovirus (Kadipiro virus). J Gen Virol 2016; 97:3359-3367. [PMID: 27902331 DOI: 10.1099/jgv.0.000644] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Viral nucleic acids present in the plasma of 498 Kenyan adults with unexplained fever were characterized by metagenomics analysis of 51 sample pools. The highest to lowest fraction of plasma pools was positive for parvovirus B19 (75 %), pegivirus C (GBV-C) (67 %), alpha anellovirus (59 %), gamma anellovirus (55 %), beta anellovirus (41 %), dengue virus genotype 2 (DENV-2) (16 %), human immunodeficiency virus type 1 (6 %), human herpesvirus 6 (6 %), HBV (4 %), rotavirus (4 %), hepatitis B virus (4 %), rhinovirus C (2 %), Merkel cell polyomavirus (MCPyV; 2 %) and Kadipiro virus (2 %). Ranking by overall percentage of viral reads yielded similar results. Characterization of viral nucleic acids in the plasma of a febrile East African population showed a high frequency of parvovirus B19 and DENV infections and detected a reovirus (Kadipiro virus) previously reported only in Asian Culex mosquitoes, providing a baseline to compare with future virome studies to detect emerging viruses in this region.
Collapse
Affiliation(s)
- Carolyne N Ngoi
- Centre for Geographic Medicine Research - Coast, Kenya Medical Research Institute, Kilifi, Kenya
- Blood Systems Research Institute, San Francisco, CA, USA
| | - Juliana Siqueira
- Blood Systems Research Institute, San Francisco, CA, USA
- Programa de Oncovirologia, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Linlin Li
- Blood Systems Research Institute, San Francisco, CA, USA
| | - Xutao Deng
- Blood Systems Research Institute, San Francisco, CA, USA
| | - Peter Mugo
- Centre for Geographic Medicine Research - Coast, Kenya Medical Research Institute, Kilifi, Kenya
| | - Susan M Graham
- Centre for Geographic Medicine Research - Coast, Kenya Medical Research Institute, Kilifi, Kenya
- University of Washington, Seattle, WA, USA
| | - Matt A Price
- International AIDS Vaccine Initiative, New York, NY, USA
- Department of Epidemiology and Biostatistics, University of California at San Francisco, CA, USA
| | - Eduard J Sanders
- Centre for Geographic Medicine Research - Coast, Kenya Medical Research Institute, Kilifi, Kenya
- Nuffield Department of Medicine, University of Oxford, Headington, UK
| | - Eric Delwart
- Department of Laboratory Medicine, University of California at San Francisco, CA, USA
- Blood Systems Research Institute, San Francisco, CA, USA
| |
Collapse
|
274
|
Görzer I, Jaksch P, Strassl R, Klepetko W, Puchhammer-Stöckl E. Association between plasma Torque teno virus level and chronic lung allograft dysfunction after lung transplantation. J Heart Lung Transplant 2016; 36:366-368. [PMID: 27876413 DOI: 10.1016/j.healun.2016.10.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 10/13/2016] [Accepted: 10/16/2016] [Indexed: 01/03/2023] Open
Affiliation(s)
- Irene Görzer
- Department of Virology, Medical University of Vienna, Vienna, Austria
| | - Peter Jaksch
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | - Robert Strassl
- Division of Clinical Virology, Medical University of Vienna, Vienna, Austria
| | - Walter Klepetko
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | | |
Collapse
|
275
|
Olivieri F, Albertini MC, Orciani M, Ceka A, Cricca M, Procopio AD, Bonafè M. DNA damage response (DDR) and senescence: shuttled inflamma-miRNAs on the stage of inflamm-aging. Oncotarget 2016; 6:35509-21. [PMID: 26431329 PMCID: PMC4742121 DOI: 10.18632/oncotarget.5899] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 09/17/2015] [Indexed: 12/31/2022] Open
Abstract
A major issue in aging research is how cellular phenomena affect aging at the systemic level. Emerging evidence suggests that DNA damage response (DDR) signaling is a key mechanism linking DNA damage accumulation, cell senescence, and organism aging. DDR activation in senescent cells promotes acquisition of a proinflammatory secretory phenotype (SASP), which in turn elicits DDR and SASP activation in neighboring cells, thereby creating a proinflammatory environment extending at the local and eventually the systemic level. DDR activation is triggered by genomic lesions as well as emerging bacterial and viral metagenomes. Therefore, the buildup of cells with an activated DDR probably fuels inflamm-aging and predisposes to the development of the major age-related diseases (ARDs). Micro (mi)-RNAs - non-coding RNAs involved in gene expression modulation - are released locally and systemically by a variety of shuttles (exosomes, lipoproteins, proteins) that likely affect the efficiency of their biological effects. Here we suggest that some miRNAs, previously found to be associated with inflammation and senescence - miR-146, miR-155, and miR-21 - play a central role in the interplay among DDR, cell senescence and inflamm-aging. The identification of the functions of shuttled senescence-associated miRNAs is expected to shed light on the aging process and on how to delay ARD development.
Collapse
Affiliation(s)
- Fabiola Olivieri
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Ancona, Italy.,Center of Clinical Pathology and Innovative Therapy, Italian National Research Center on Aging, INRCA-IRCCS, Ancona, Italy
| | - Maria Cristina Albertini
- Department of Biomolecular Sciences, Biochemistry and Molecular Biology, Università degli Studi di Urbino "Carlo Bo", Urbino, Italy
| | - Monia Orciani
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Ancona, Italy
| | - Artan Ceka
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Ancona, Italy
| | - Monica Cricca
- Department of Experimental, Diagnostic and Specialty Medicine, DIMES, University of Bologna, Bologna, Italy
| | - Antonio Domenico Procopio
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Ancona, Italy.,Center of Clinical Pathology and Innovative Therapy, Italian National Research Center on Aging, INRCA-IRCCS, Ancona, Italy
| | - Massimiliano Bonafè
- Department of Experimental, Diagnostic and Specialty Medicine, DIMES, University of Bologna, Bologna, Italy
| |
Collapse
|
276
|
Gordon PMK, Khan A, Sajid U, Chang N, Suresh V, Dimnik L, Lamont RE, Parboosingh JS, Martin SR, Pon RT, Weatherhead J, Wegener S, Isaac D, Greenway SC. An Algorithm Measuring Donor Cell-Free DNA in Plasma of Cellular and Solid Organ Transplant Recipients That Does Not Require Donor or Recipient Genotyping. Front Cardiovasc Med 2016; 3:33. [PMID: 27713880 PMCID: PMC5031701 DOI: 10.3389/fcvm.2016.00033] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 09/09/2016] [Indexed: 11/22/2022] Open
Abstract
Cell-free DNA (cfDNA) has significant potential in the diagnosis and monitoring of clinical conditions. However, accurately and easily distinguishing the relative proportion of DNA molecules in a mixture derived from two different sources (i.e., donor and recipient tissues after transplantation) is challenging. In human cellular transplantation, there is currently no useable method to detect in vivo engraftment, and blood-based non-invasive tests for allograft rejection in solid organ transplantation are either non-specific or absent. Elevated levels of donor cfDNA have been shown to correlate with solid organ rejection, but complex methodology limits implementation of this promising biomarker. We describe a cost-effective method to quantify donor cfDNA in recipient plasma using a panel of high-frequency single nucleotide polymorphisms, next-generation (semiconductor) sequencing, and a novel mixture model algorithm. In vitro, our method accurately and rapidly determined donor:recipient DNA admixture. For in vivo testing, donor cfDNA was serially quantified in an infant with a urea cycle disorder after receiving six daily infusions of donor liver cells. Donor cfDNA isolated from 1 to 2 ml of recipient plasma was detected as late as 24 weeks after infusion suggesting engraftment. The percentage of circulating donor cfDNA was also assessed in pediatric and adult heart transplant recipients undergoing routine endomyocardial biopsy with levels observed to be stable over time and generally measuring <1% in cases without moderate or severe cellular rejection. Unlike existing non-invasive methods used to define the proportion of donor cfDNA in solid organ transplant patients, our assay does not require sex mismatch, donor genotyping, or whole-genome sequencing and potentially has broad application to detect cellular engraftment or allograft injury after transplantation.
Collapse
Affiliation(s)
- Paul M K Gordon
- Alberta Children's Hospital Research Institute, University of Calgary , Calgary, AB , Canada
| | - Aneal Khan
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada; Department of Paediatrics, University of Calgary, Calgary, AB, Canada; Department of Medical Genetics, University of Calgary, Calgary, AB, Canada
| | - Umair Sajid
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada; Department of Paediatrics, University of Calgary, Calgary, AB, Canada
| | - Nicholas Chang
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada; Department of Paediatrics, University of Calgary, Calgary, AB, Canada
| | - Varun Suresh
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada; Department of Paediatrics, University of Calgary, Calgary, AB, Canada
| | - Leo Dimnik
- Molecular Diagnostic Laboratory, Genetic Laboratory Services-South, Alberta Health Services , Calgary, AB , Canada
| | - Ryan E Lamont
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada; Department of Medical Genetics, University of Calgary, Calgary, AB, Canada; Molecular Diagnostic Laboratory, Genetic Laboratory Services-South, Alberta Health Services, Calgary, AB, Canada
| | - Jillian S Parboosingh
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada; Department of Medical Genetics, University of Calgary, Calgary, AB, Canada; Molecular Diagnostic Laboratory, Genetic Laboratory Services-South, Alberta Health Services, Calgary, AB, Canada
| | - Steven R Martin
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada; Department of Paediatrics, University of Calgary, Calgary, AB, Canada
| | - Richard T Pon
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada; Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Jene Weatherhead
- Alberta Children's Hospital Research Institute, University of Calgary , Calgary, AB , Canada
| | - Shelly Wegener
- Alberta Children's Hospital Research Institute, University of Calgary , Calgary, AB , Canada
| | - Debra Isaac
- Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, University of Calgary , Calgary, AB , Canada
| | - Steven C Greenway
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada; Department of Paediatrics, University of Calgary, Calgary, AB, Canada; Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada; Department of Cardiac Sciences, Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
277
|
Rani A, Ranjan R, McGee HS, Metwally A, Hajjiri Z, Brennan DC, Finn PW, Perkins DL. A diverse virome in kidney transplant patients contains multiple viral subtypes with distinct polymorphisms. Sci Rep 2016; 6:33327. [PMID: 27633952 PMCID: PMC5025891 DOI: 10.1038/srep33327] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 08/19/2016] [Indexed: 01/08/2023] Open
Abstract
Recent studies have established that the human urine contains a complex microbiome, including a virome about which little is known. Following immunosuppression in kidney transplant patients, BK polyomavirus (BKV) has been shown to induce nephropathy (BKVN), decreasing graft survival. In this study we investigated the urine virome profile of BKV+ and BKV- kidney transplant recipients. Virus-like particles were stained to confirm the presence of VLP in the urine samples. Metagenomic DNA was purified, and the virome profile was analyzed using metagenomic shotgun sequencing. While the BK virus was predominant in the BKV+ group, it was also found in the BKV- group patients. Additional viruses were also detected in all patients, notably including JC virus (JCV) and Torque teno virus (TTV) and interestingly, we detected multiple subtypes of the BKV, JCV and TTV. Analysis of the BKV subtypes showed that nucleotide polymorphisms were detected in the VP1, VP2 and Large T Antigen proteins, suggesting potential functional effects for enhanced pathogenicity. Our results demonstrate a complex urinary virome in kidney transplant patients with multiple viruses with several distinct subtypes warranting further analysis of virus subtypes in immunosuppressed hosts.
Collapse
Affiliation(s)
- Asha Rani
- Department of Medicine, University of Illinois, Chicago, IL 60612, USA
| | - Ravi Ranjan
- Department of Medicine, University of Illinois, Chicago, IL 60612, USA
| | - Halvor S. McGee
- Department of Medicine, University of Illinois, Chicago, IL 60612, USA
| | - Ahmed Metwally
- Department of Medicine, University of Illinois, Chicago, IL 60612, USA
- Department of Bioengineering, University of Illinois, Chicago, IL 60612, USA
| | - Zahraa Hajjiri
- Department of Medicine, University of Illinois, Chicago, IL 60612, USA
| | - Daniel C. Brennan
- Division of Renal Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Patricia W. Finn
- Department of Medicine, University of Illinois, Chicago, IL 60612, USA
| | - David L. Perkins
- Department of Medicine, University of Illinois, Chicago, IL 60612, USA
- Department of Bioengineering, University of Illinois, Chicago, IL 60612, USA
- Department of Surgery, University of Illinois, Chicago, IL 60612, USA
| |
Collapse
|
278
|
Gentile G, Micozzi A. Speculations on the clinical significance of asymptomatic viral infections. Clin Microbiol Infect 2016; 22:585-8. [PMID: 27450587 DOI: 10.1016/j.cmi.2016.07.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 07/01/2016] [Accepted: 07/05/2016] [Indexed: 12/12/2022]
Abstract
A detailed understanding of asymptomatic chronic viral infections is critical to analyse their pathogenesis, assess the severity and burden of disease and, where required, optimize public health control measures. Recent studies on herpesviruses showed that the host-virus interactions are modulated by co-infections, emphasizing the relevance of co-infections in determining the clinical expression (from asymptomatic to symptomatic infections) and the severity of herpesvirus-associated diseases (either neoplastic or infectious diseases). To demonstrate causality between viruses (virome) and diseases, Koch's postulates should be adapted adding new knowledge on host-microbe relationship and microbial interactions. In the present review we aim to provide an update on asymptomatic chronic infections and criteria for causality and on the virological, immunological and host-virus interactions in asymptomatic chronic infections in human hosts, focusing on herpetic infections.
Collapse
Affiliation(s)
- G Gentile
- Department of Cellular Biotechnologies and Haematology, Sapienza University, Rome, Italy.
| | - A Micozzi
- Department of Cellular Biotechnologies and Haematology, Sapienza University, Rome, Italy
| |
Collapse
|
279
|
Abril MK, Barnett AS, Wegermann K, Fountain E, Strand A, Heyman BM, Blough BA, Swaminathan AC, Sharma-Kuinkel B, Ruffin F, Alexander BD, McCall CM, Costa SF, Arcasoy MO, Hong DK, Blauwkamp TA, Kertesz M, Fowler VG, Kraft BD. Diagnosis of Capnocytophaga canimorsus Sepsis by Whole-Genome Next-Generation Sequencing. Open Forum Infect Dis 2016; 3:ofw144. [PMID: 27704003 PMCID: PMC5047422 DOI: 10.1093/ofid/ofw144] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/19/2016] [Indexed: 01/21/2023] Open
Abstract
We report the case of a 60-year-old man with septic shock due to Capnocytophaga canimorsus that was diagnosed in 24 hours by a novel whole-genome next-generation sequencing assay. This technology shows great promise in identifying fastidious pathogens, and, if validated, it has profound implications for infectious disease diagnosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Chad M McCall
- Department of Pathology , Duke University Medical Center , Durham, North Carolina
| | | | | | | | | | | | | | | |
Collapse
|
280
|
Zou S, Caler L, Colombini-Hatch S, Glynn S, Srinivas P. Research on the human virome: where are we and what is next. MICROBIOME 2016; 4:32. [PMID: 27341799 PMCID: PMC4919837 DOI: 10.1186/s40168-016-0177-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 06/10/2016] [Indexed: 06/06/2023]
Abstract
The National Heart, Lung, and Blood Institute (NHLBI) of the National Institutes of Health convened a Working Group on the Microbiome in Cardiovascular, Pulmonary and Hematologic Health and Diseases from June 25, 2014, to June 26, 2014. The Working Group's central goal was to define what major microbiome research areas warranted additional study in the context of heart, lung, and blood (HLB) diseases. The Working Group identified studies of the human virome a key priority.
Collapse
Affiliation(s)
- Shimian Zou
- National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), 6701, Rockledge Drive, Room 9144, Bethesda, MD 20892-7950 USA
| | - Lis Caler
- National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), 6701, Rockledge Drive, Room 9144, Bethesda, MD 20892-7950 USA
| | - Sandra Colombini-Hatch
- National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), 6701, Rockledge Drive, Room 9144, Bethesda, MD 20892-7950 USA
| | - Simone Glynn
- National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), 6701, Rockledge Drive, Room 9144, Bethesda, MD 20892-7950 USA
| | - Pothur Srinivas
- National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), 6701, Rockledge Drive, Room 9144, Bethesda, MD 20892-7950 USA
| |
Collapse
|
281
|
Abstract
Increasing evidence indicates that microbes have a large influence on immune function. Previous studies have linked pathogenic microorganisms with decreased allograft tolerance and subsequent rejection. In this issue of the JCI, Lei and colleagues demonstrate that commensal organisms also influence the host response to allograft transplantation. Using murine skin and cardiac transplant models, the authors demonstrate that allograft rejection is accelerated in mice with a normal microbiome compared with germ-free animals and antibiotic-treated mice. The increased graft rejection observed in conventional animals was due to enhanced T cell priming and was mediated through type I IFN. Together, these results suggest that altering a patient's microbial community prior to transplant could improve allograft acceptance.
Collapse
|
282
|
Burnham P, Kim MS, Agbor-Enoh S, Luikart H, Valantine HA, Khush KK, De Vlaminck I. Single-stranded DNA library preparation uncovers the origin and diversity of ultrashort cell-free DNA in plasma. Sci Rep 2016; 6:27859. [PMID: 27297799 PMCID: PMC4906518 DOI: 10.1038/srep27859] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 05/26/2016] [Indexed: 12/18/2022] Open
Abstract
Circulating cell-free DNA (cfDNA) is emerging as a powerful monitoring tool in cancer, pregnancy and organ transplantation. Nucleosomal DNA, the predominant form of plasma cfDNA, can be adapted for sequencing via ligation of double-stranded DNA (dsDNA) adapters. dsDNA library preparations, however, are insensitive to ultrashort, degraded cfDNA. Drawing inspiration from advances in paleogenomics, we have applied a single-stranded DNA (ssDNA) library preparation method to sequencing of cfDNA in the plasma of lung transplant recipients (40 samples, six patients). We found that ssDNA library preparation yields a greater portion of sub-100 bp nuclear genomic cfDNA (p 10−5, Mann-Whitney U Test), and an increased relative abundance of mitochondrial (10.7x, p 10−5) and microbial cfDNA (71.3x, p10−5). The higher yield of microbial sequences from this method increases the sensitivity of cfDNA-based monitoring for infections following transplantation. We detail the fragmentation pattern of mitochondrial, nuclear genomic and microbial cfDNA over a broad fragment length range. We report the observation of donor-specific mitochondrial cfDNA in the circulation of lung transplant recipients. A ssDNA library preparation method provides a more informative window into understudied forms of cfDNA, including mitochondrial and microbial derived cfDNA and short nuclear genomic cfDNA, while retaining information provided by standard dsDNA library preparation methods.
Collapse
Affiliation(s)
- Philip Burnham
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Min Seong Kim
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | | | - Helen Luikart
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford CA 94305, USA
| | | | - Kiran K Khush
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford CA 94305, USA
| | - Iwijn De Vlaminck
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
283
|
Accounting for reciprocal host–microbiome interactions in experimental science. Nature 2016; 534:191-9. [DOI: 10.1038/nature18285] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 04/26/2016] [Indexed: 12/13/2022]
|
284
|
Abstract
Survival after lung transplantation is limited in large part due to the high incidence of chronic rejection, known as chronic lung allograft dysfunction (CLAD). Pulmonary infections are a frequent complication in lung transplant recipients, due both to immunosuppressive medications and constant exposure of the lung allograft to the external environment via the airways. Infection is a recognized risk factor for the development of CLAD, and both acute infection and chronic lung allograft colonization with microorganisms increase the risk for CLAD. Acute infection by community acquired respiratory viruses, and the bacteria Pseudomonas aeruginosa and Staphylococcus aureus are increasingly recognized as important risk factors for CLAD. Colonization by the fungus Aspergillus may also augment the risk of CLAD. Fostering this transition from healthy lung to CLAD in each of these infectious episodes is the persistence of an inflammatory lung allograft environment.
Collapse
Affiliation(s)
- Aric L Gregson
- Division of Infectious Diseases, Department of Medicine, University of California, Box 957119, Warren Hall 14-154, Los Angeles, CA, 90995-7119, USA.
| |
Collapse
|
285
|
Thomasy SM, Maggs DJ. A review of antiviral drugs and other compounds with activity against feline herpesvirus type 1. Vet Ophthalmol 2016; 19 Suppl 1:119-30. [PMID: 27091747 DOI: 10.1111/vop.12375] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Feline herpesvirus type 1 (FHV-1) is a common and important cause of ocular surface disease, dermatitis, respiratory disease, and potentially intraocular disease in cats. Many antiviral drugs developed for the treatment of humans infected with herpesviruses have been used to treat cats infected with FHV-1. Translational use of drugs in this manner ideally requires methodical investigation of their in vitro efficacy against FHV-1 followed by pharmacokinetic and safety trials in normal cats. Subsequently, placebo-controlled efficacy studies in experimentally inoculated animals should be performed followed, finally, by carefully designed and monitored clinical trials in client-owned animals. This review is intended to provide a concise overview of the available literature regarding the efficacy of antiviral drugs and other compounds with proven or putative activity against FHV-1, as well as a discussion of their safety in cats.
Collapse
Affiliation(s)
- Sara M Thomasy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California-Davis, Davis, CA, 95616, USA
| | - David J Maggs
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California-Davis, Davis, CA, 95616, USA
| |
Collapse
|
286
|
Focosi D, Antonelli G, Pistello M, Maggi F. Torquetenovirus: the human virome from bench to bedside. Clin Microbiol Infect 2016; 22:589-93. [PMID: 27093875 DOI: 10.1016/j.cmi.2016.04.007] [Citation(s) in RCA: 153] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 04/04/2016] [Accepted: 04/10/2016] [Indexed: 12/15/2022]
Abstract
Torquetenovirus (TTV) is the most abundant component of human virome. Virologists have long ignored this orphan and highly divergent virus, in part because TTV cannot be cultured and because it lacks serology reagents and animal models. Nevertheless, it is almost endemic worldwide and is insensitive to current antiviral drugs, so its monitoring is useful in various conditions. To date, TTV as a marker has proved useful in at least two circumstances: to identify anthropogenic pollution and to assess functional immune competence in immunosuppressed individuals. This review summarizes recent findings about TTV and discusses the main hurdles in translating them into clinical diagnostics.
Collapse
Affiliation(s)
- D Focosi
- North-Western Tuscany Blood Bank, Italy
| | - G Antonelli
- Laboratory of Virology, Department of Molecular Medicine, and Pasteur Institute-Cenci Bolognetti Foundation, Sapienza University of Rome, Rome, Italy
| | - M Pistello
- Virology Unit and Retrovirus Centre, Pisa University Hospital, Italy; Retrovirus Center and Virology Section, Department of Translational Research, University of Pisa, Pisa, Italy
| | - F Maggi
- Virology Unit and Retrovirus Centre, Pisa University Hospital, Italy.
| |
Collapse
|
287
|
Krishnamurthy SR, Janowski AB, Zhao G, Barouch D, Wang D. Hyperexpansion of RNA Bacteriophage Diversity. PLoS Biol 2016; 14:e1002409. [PMID: 27010970 PMCID: PMC4807089 DOI: 10.1371/journal.pbio.1002409] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Accepted: 02/18/2016] [Indexed: 12/20/2022] Open
Abstract
Bacteriophage modulation of microbial populations impacts critical processes in ocean, soil, and animal ecosystems. However, the role of bacteriophages with RNA genomes (RNA bacteriophages) in these processes is poorly understood, in part because of the limited number of known RNA bacteriophage species. Here, we identify partial genome sequences of 122 RNA bacteriophage phylotypes that are highly divergent from each other and from previously described RNA bacteriophages. These novel RNA bacteriophage sequences were present in samples collected from a range of ecological niches worldwide, including invertebrates and extreme microbial sediment, demonstrating that they are more widely distributed than previously recognized. Genomic analyses of these novel bacteriophages yielded multiple novel genome organizations. Furthermore, one RNA bacteriophage was detected in the transcriptome of a pure culture of Streptomyces avermitilis, suggesting for the first time that the known tropism of RNA bacteriophages may include gram-positive bacteria. Finally, reverse transcription PCR (RT-PCR)-based screening for two specific RNA bacteriophages in stool samples from a longitudinal cohort of macaques suggested that they are generally acutely present rather than persistent. This study uses computational metagenomics and molecular experimentation to massively expand the known genomic and ecological diversity of RNA bacteriophages, identifying novel tropisms and genes. Bacteriophages (viruses that infect bacteria) can alter biological processes in numerous ecosystems. While there are numerous studies describing the role of bacteriophages with DNA genomes in these processes, the role of bacteriophages with RNA genomes (RNA bacteriophages) is poorly understood. This gap in knowledge is in part because of the limited diversity of known RNA bacteriophages. Here, we begin to address the question by identifying 122 novel RNA bacteriophage partial genome sequences present in metagenomic datasets that are highly divergent from each other and previously described RNA bacteriophages. Additionally, many of these sequences contained novel properties, including novel genes, segmentation, and host range, expanding the frontiers of RNA bacteriophage genomics, evolution, and tropism. These novel RNA bacteriophage sequences were globally distributed from numerous ecological niches, including animal-associated and environmental habitats. These findings will facilitate our understanding of the role of the RNA bacteriophage in microbial communities. Furthermore, there are likely many more unrecognized RNA bacteriophages that remain to be discovered.
Collapse
Affiliation(s)
- Siddharth R. Krishnamurthy
- Departments of Molecular Microbiology and Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Andrew B. Janowski
- Departments of Molecular Microbiology and Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Guoyan Zhao
- Departments of Molecular Microbiology and Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Dan Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America, and Ragon Institute of MGH, MIT, and Harvard, Boston, Massachusetts, United States of America
| | - David Wang
- Departments of Molecular Microbiology and Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
288
|
Infectious Diseases in Transplantation--Report of the 20th Nantes Actualités Transplantation Meeting. Transplantation 2016; 99:2444-7. [PMID: 26627674 DOI: 10.1097/tp.0000000000000997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The 20th Nantes Actualités Transplantation (NAT) meeting was held on June 11, 2015, and June 12, 2015. This year, the local organizing committee selected an update on infectious diseases in solid organ and hematopoietic stem cell transplantation. With an attendance of close to 170 clinicians, researchers, students, engineers, technicians, invited speakers, and guests from North and South America, Germany, Switzerland, Netherlands, and France, the meeting was well attended. Invited speakers' expertise covered basic as well as translational microbiology, immunology, transplantation, and intensive care medicine. This report identifies a number of advances presented during the meeting in the care and management of infectious diseases in transplantation and immunocompromised patients. New antiviral immune responses and their modulation by pathogens in addition to novel antimicrobial therapeutic strategies, cell therapies, and genomic analysis were discussed.
Collapse
|
289
|
Monaco CL, Gootenberg DB, Zhao G, Handley SA, Ghebremichael MS, Lim ES, Lankowski A, Baldridge MT, Wilen CB, Flagg M, Norman JM, Keller BC, Luévano JM, Wang D, Boum Y, Martin JN, Hunt PW, Bangsberg DR, Siedner MJ, Kwon DS, Virgin HW. Altered Virome and Bacterial Microbiome in Human Immunodeficiency Virus-Associated Acquired Immunodeficiency Syndrome. Cell Host Microbe 2016; 19:311-22. [PMID: 26962942 PMCID: PMC4821831 DOI: 10.1016/j.chom.2016.02.011] [Citation(s) in RCA: 287] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 12/31/2015] [Accepted: 02/19/2016] [Indexed: 02/07/2023]
Abstract
Human immunodeficiency virus (HIV) infection is associated with increased intestinal translocation of microbial products and enteropathy as well as alterations in gut bacterial communities. However, whether the enteric virome contributes to this infection and resulting immunodeficiency remains unknown. We characterized the enteric virome and bacterial microbiome in a cohort of Ugandan patients, including HIV-uninfected or HIV-infected subjects and those either treated with anti-retroviral therapy (ART) or untreated. Low peripheral CD4 T cell counts were associated with an expansion of enteric adenovirus sequences and this increase was independent of ART treatment. Additionally, the enteric bacterial microbiome of patients with lower CD4 T counts exhibited reduced phylogenetic diversity and richness with specific bacteria showing differential abundance, including increases in Enterobacteriaceae, which have been associated with inflammation. Thus, immunodeficiency in progressive HIV infection is associated with alterations in the enteric virome and bacterial microbiome, which may contribute to AIDS-associated enteropathy and disease progression.
Collapse
Affiliation(s)
- Cynthia L Monaco
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Guoyan Zhao
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Scott A Handley
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | | | - Efrem S Lim
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Alex Lankowski
- Division of Infectious Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Megan T Baldridge
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Craig B Wilen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Meaghan Flagg
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | | | - Brian C Keller
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - David Wang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Yap Boum
- Médecins Sans Frontières Epicentre, 1956 Mbarara, Uganda
| | - Jeffrey N Martin
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA 94143, USA
| | - Peter W Hunt
- Department of Medicine, University of California San Francisco, San Francisco, CA 94110, USA
| | - David R Bangsberg
- Division of Infectious Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Harvard School of Public Health, Boston, MA 02114, USA
| | - Mark J Siedner
- Division of Infectious Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Douglas S Kwon
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA; Division of Infectious Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | - Herbert W Virgin
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
290
|
Vignolini T, Macera L, Antonelli G, Pistello M, Maggi F, Giannecchini S. Investigation on torquetenovirus (TTV) microRNA transcriptome in vivo. Virus Res 2016; 217:18-22. [PMID: 26959653 DOI: 10.1016/j.virusres.2016.03.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 03/02/2016] [Accepted: 03/03/2016] [Indexed: 01/03/2023]
Abstract
Torquetenovirus (TTV) is a widespread anellovirus that establishes persistent infections in human showing an increased viremia in immunosuppressed patients. TTV possesses microRNA (miRNA)-coding sequences that might be involved in viral immune evasion. Here, the presence of TTV DNA and miRNAs expression was investigated in plasma samples of 77 diseased (20 infected with human immunodeficiency virus (HIV), 18 infected with hepatitis B (HBV) virus, 18 infected with hepatitis C (HCV) virus, 21 solid organ transplanted) patients, and 25 healthy controls. TTV prevalence was significantly different in healthy controls (60%, 15/25) versus diseased patients (80%, 62/77), showing the highest TTV loads in transplant recipients. Genetic TTV analysis showed the highest prevalence of group 1, followed by groups 3, 4 and 5, and a lack of isolates of group 2. The expression of at least one TTV miRNAs of group 1, 3 and 5 was found in exosomes of plasma of the great majority of individuals (96%, 98/102 subjects) showing the higher prevalence of miRNAs of TTV group 3 (90%, 92/102), followed by miRNAs of group 1 (66%, 67/102), and miRNA of group 5 (49%, 50/102). TTV miRNAs expression and TTV viremia were not always directly correlated, and significant differences appeared in production of some TTV miRNAs between healthy controls and diseased patients. The reported TTV miRNAs status in exosomes encourages further investigation to understand their potential role in the expansion of anelloviruses upon immunosuppression.
Collapse
Affiliation(s)
- Tiziano Vignolini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Lisa Macera
- Virology Unit, Pisa University Hospital, Pisa, Italy
| | - Guido Antonelli
- Pasteur Institute-Cenci Bolognetti Foundation, Department of Molecular Medicine, Laboratory of Virology, Sapienza University of Rome, Italy
| | | | | | - Simone Giannecchini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| |
Collapse
|
291
|
The Human Virome in Health and Disease. Mol Microbiol 2016. [DOI: 10.1128/9781555819071.ch14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
292
|
King KR, Grazette LP, Paltoo DN, McDevitt JT, Sia SK, Barrett PM, Apple FS, Gurbel PA, Weissleder R, Leeds H, Iturriaga EJ, Rao AK, Adhikari B, Desvigne-Nickens P, Galis ZS, Libby P. Point-of-Care Technologies for Precision Cardiovascular Care and Clinical Research: National Heart, Lung, and Blood Institute Working Group. JACC Basic Transl Sci 2016; 1:73-86. [PMID: 26977455 PMCID: PMC4787294 DOI: 10.1016/j.jacbts.2016.01.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 01/20/2016] [Indexed: 12/26/2022]
Abstract
Point-of-care technologies (POC or POCT) are enabling innovative cardiovascular diagnostics that promise to improve patient care across diverse clinical settings. The National Heart, Lung, and Blood Institute convened a working group to discuss POCT in cardiovascular medicine. The multidisciplinary working group, which included clinicians, scientists, engineers, device manufacturers, regulatory officials, and program staff, reviewed the state of the POCT field; discussed opportunities for POCT to improve cardiovascular care, realize the promise of precision medicine, and advance the clinical research enterprise; and identified barriers facing translation and integration of POCT with existing clinical systems. A POCT development roadmap emerged to guide multidisciplinary teams of biomarker scientists, technologists, health care providers, and clinical trialists as they: 1) formulate needs assessments; 2) define device design specifications; 3) develop component technologies and integrated systems; 4) perform iterative pilot testing; and 5) conduct rigorous prospective clinical testing to ensure that POCT solutions have substantial effects on cardiovascular care.
Collapse
Affiliation(s)
- Kevin R. King
- Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Luanda P. Grazette
- Division of Cardiovascular Medicine, University of Southern California, Los Angeles, California
| | - Dina N. Paltoo
- Office of Science Policy, Office of the Director, National Institutes of Health, Bethesda, Maryland
| | - John T. McDevitt
- Departments of Bioengineering and Chemistry, Rice University, Houston, Texas
| | - Samuel K. Sia
- Department of Biomedical Engineering, Columbia University, New York, New York
| | | | - Fred S. Apple
- Hennepin County Medical Center and University of Minnesota, Department of Laboratory Medicine and Pathology, Minneapolis, Minnesota
| | - Paul A. Gurbel
- Inova Center for Thrombosis Research and Drug Development, Inova Heart and Vascular Institute, Falls Church, Virginia
| | - Ralph Weissleder
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Hilary Leeds
- Office of Science Policy, Office of the Director, National Institutes of Health, Bethesda, Maryland
| | - Erin J. Iturriaga
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Anupama K. Rao
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Bishow Adhikari
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | | | - Zorina S. Galis
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Peter Libby
- Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| |
Collapse
|
293
|
Barbian HJ, Li Y, Ramirez M, Klase Z, Lipende I, Mjungu D, Moeller AH, Wilson ML, Pusey AE, Lonsdorf EV, Bushman FD, Hahn BH. Destabilization of the gut microbiome marks the end-stage of simian immunodeficiency virus infection in wild chimpanzees. Am J Primatol 2015; 80. [PMID: 26676710 DOI: 10.1002/ajp.22515] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 10/20/2015] [Accepted: 12/01/2015] [Indexed: 12/17/2022]
Abstract
Enteric dysbiosis is a characteristic feature of progressive human immunodeficiency virus type 1 (HIV-1) infection but has not been observed in simian immunodeficiency virus (SIVmac)-infected macaques, including in animals with end-stage disease. This has raised questions concerning the mechanisms underlying the HIV-1 associated enteropathy, with factors other than virus infection, such as lifestyle and antibiotic use, implicated as playing possible causal roles. Simian immunodeficiency virus of chimpanzees (SIVcpz) is also associated with increased mortality in wild-living communities, and like HIV-1 and SIVmac, can cause CD4+ T cell depletion and immunodeficiency in infected individuals. Given the central role of the intestinal microbiome in mammalian health, we asked whether gut microbial constituents could be identified that are indicative of SIVcpz status and/or disease progression. Here, we characterized the gut microbiome of SIVcpz-infected and -uninfected chimpanzees in Gombe National Park, Tanzania. Subjecting a small number of fecal samples (N = 9) to metagenomic (shotgun) sequencing, we found bacteria of the family Prevotellaceae to be enriched in SIVcpz-infected chimpanzees. However, 16S rRNA gene sequencing of a larger number of samples (N = 123) failed to show significant differences in both the composition and diversity (alpha and beta) of gut bacterial communities between infected (N = 24) and uninfected (N = 26) chimpanzees. Similarly, chimpanzee stool-associated circular virus (Chi-SCV) and chimpanzee adenovirus (ChAdV) identified by metagenomic sequencing were neither more prevalent nor more abundant in SIVcpz-infected individuals. However, fecal samples collected from SIVcpz-infected chimpanzees within 5 months before their AIDS-related death exhibited significant compositional changes in their gut bacteriome. These data indicate that SIVcpz-infected chimpanzees retain a stable gut microbiome throughout much of their natural infection course, with a significant destabilization of bacterial (but not viral) communities observed only in individuals with known immunodeficiency within the last several months before their death. Am. J. Primatol. 80:e22515, 2018. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Hannah J Barbian
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Yingying Li
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Miguel Ramirez
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Zachary Klase
- Department of Biological Sciences, University of the Sciences, Philadelphia, Pennsylvania
| | | | - Deus Mjungu
- Gombe Stream Research Center, Kigoma, Tanzania
| | - Andrew H Moeller
- Department of Integrative Biology, University of California, Berkeley, California.,Miller Institute for Basic Research, University of California, Berkeley, California
| | - Michael L Wilson
- Department of Anthropology, University of Minnesota, Minneapolis, Minnesota.,Department of Ecology, Evolution and Behavior, University of Minnesota, St. Paul, Minnesota
| | - Anne E Pusey
- Department of Evolutionary Anthropology, Duke University, Durham, North Carolina
| | - Elizabeth V Lonsdorf
- Department of Psychology, Franklin and Marshall College, Lancaster, Pennsylvania
| | - Frederic D Bushman
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Beatrice H Hahn
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
294
|
Masouridi-Levrat S, Pradier A, Simonetta F, Kaiser L, Chalandon Y, Roosnek E. Torque teno virus in patients undergoing allogeneic hematopoietic stem cell transplantation for hematological malignancies. Bone Marrow Transplant 2015; 51:440-2. [DOI: 10.1038/bmt.2015.262] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
295
|
Abstract
Non-targeted identification of microbes is now possible directly in biological samples, based on whole-genome-NGS (WG-NGS) techniques that allow deep sequencing of nucleic acids, data mining and sorting out of sequences of pathogens without any a priori hypothesis. WG-NGS was first only used as a research tool due to its cost, complexity and lack of standardization. Recent improvements in sample preparation and bioinformatics pipelines and decrease in cost now allow actionable diagnostics in patients. The potency and limits of WG-NGS and possible future indications are discussed here. WG-NGS will likely soon become a standard procedure in microbiological diagnosis.
Collapse
Affiliation(s)
- Marc Lecuit
- Institut Pasteur, Inserm Unit 1117, Biology of Infection Unit, Paris, France
| | - Marc Eloit
- b Biology of Infection Unit , Institut Pasteur, Inserm U1117, Pathogen Discovery Laboratory , Paris , France.,c Pathoquest , Paris , France
| |
Collapse
|
296
|
Pérez-Brocal V, García-López R, Nos P, Beltrán B, Moret I, Moya A. Metagenomic Analysis of Crohn's Disease Patients Identifies Changes in the Virome and Microbiome Related to Disease Status and Therapy, and Detects Potential Interactions and Biomarkers. Inflamm Bowel Dis 2015; 21:2515-32. [PMID: 26313691 DOI: 10.1097/mib.0000000000000549] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND The aim of this study was to survey the bacterial and viral communities in different types of samples from patients with Crohn's disease (CD) at different stages of the disease to relate their distribution with the origin and progression of this disorder. METHODS A total of 42 fecal samples and 15 biopsies from 20 patients with CD and 20 healthy control individuals were collected for bacterial 16S rRNA gene profiling and DNA/RNA virome metagenomic analysis through 454 pyrosequencing. Their composition, abundance, and diversity were analyzed, and comparisons of disease status, patient status, and sample origin were used to determine statistical differences between the groups. RESULTS Bacterial composition and relative abundance in new-onset patients with CD differed markedly from control individuals. Individual variability and sample origin had a stronger impact on viral communities than the disease, contrary to what was observed for bacterial populations although increased numbers of overrepresented viruses were observed in feces from patients with CD. Correlation-based networks were constructed to show potential relations between bacteria and between those and viruses. CONCLUSIONS The bacterial community reflects the disease status of individuals more accurately than their viral counterparts. However, numerous viral biomarkers specifically associated with CD disease were identified. Because viruses can modulate bacterial communities, the correlation networks between both communities constitute a step forward in unraveling their interactions under normal and CD disease conditions.
Collapse
Affiliation(s)
- Vicente Pérez-Brocal
- *Genomics and Health Area, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO)-Salud Pública, Valencia, Spain; †Institut Cavanilles de Biodiversitat i Biologia Evolutiva, Universitat de València, Paterna, Spain; ‡CIBER en Epidemiología y Salud Pública (CIBERESP), Madrid, Spain; §Servicio de Medicina Digestiva, Hospital Universitari i Politècnic La Fe, Valencia, Spain; ‖CIBER en Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain; and ¶Servicio de Medicina Digestiva, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | | | | | | | | | | |
Collapse
|
297
|
Faria MMP, Conly JM, Surette MG. The development and application of a molecular community profiling strategy to identify polymicrobial bacterial DNA in the whole blood of septic patients. BMC Microbiol 2015; 15:215. [PMID: 26474751 PMCID: PMC4609058 DOI: 10.1186/s12866-015-0557-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 10/08/2015] [Indexed: 02/08/2023] Open
Abstract
Background The application of molecular based diagnostics in sepsis has had limited success to date. Molecular community profiling methods have indicated that polymicrobial infections are more common than suggested by standard clinical culture. A molecular profiling approach was developed to investigate the propensity for polymicrobial infections in patients predicted to have bacterial sepsis. Results Disruption of blood cells with saponin and hypotonic shock enabled the recovery of microbial cells with no significant changes in microbial growth when compared to CFU/ml values immediately prior to the addition of saponin. DNA extraction included a cell-wall digestion step with both lysozyme and mutanolysin, which increased the recovery of terminal restriction fragments by 2.4 fold from diverse organisms. Efficiencies of recovery and limits of detection using Illumina sequencing of the 16S rRNA V3 region were determined for both viable cells and DNA using mock bacterial communities inoculated into whole blood. Bacteria from pre-defined communities could be recovered following lysis and removal of host cells with > 97 % recovery of total DNA present. Applying the molecular profiling methodology to three septic patients in the intensive care unit revealed microbial DNA from blood had consistent alignment with cultured organisms from the primary infection site providing evidence for a bloodstream infection in the absence of a clinical lab positive blood culture result in two of the three cases. In addition, the molecular profiling indicated greater diversity was present in the primary infection sample when compared to clinical diagnostic culture. Conclusions A method for analyzing bacterial DNA from whole blood was developed in order to characterize the bacterial DNA profile of sepsis infections. Preliminary results indicated that sepsis infections were polymicrobial in nature with the bacterial DNA recovered suggesting a more complex etiology when compared to blood culture data.
Collapse
Affiliation(s)
- M M P Faria
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, T2N 4 N1, Canada. .,Department of Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, T2N 4 N1, Canada. .,Farncombe Family Digestive Health Research Institute, Departments of Medicine and Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, 1280 Main Street, HSC 3 N 8 F, Hamilton, ON, L8S 4 K1, Canada.
| | - J M Conly
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, T2N 4 N1, Canada. .,Department of Medicine, University of Calgary, Calgary, AB, T2N 4 N1, Canada. .,Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, T2N 4 N1, Canada. .,Department of Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, T2N 4 N1, Canada.
| | - M G Surette
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, T2N 4 N1, Canada. .,Department of Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, T2N 4 N1, Canada. .,Farncombe Family Digestive Health Research Institute, Departments of Medicine and Biochemistry and Biomedical Sciences, Faculty of Health Sciences, McMaster University, 1280 Main Street, HSC 3 N 8 F, Hamilton, ON, L8S 4 K1, Canada. .,Department of Medicine, McMaster University, Hamilton, ON, L8S 4 K1, Canada. .,Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, L8S 4 K1, Canada.
| |
Collapse
|
298
|
Lewis JD, Chen EZ, Baldassano RN, Otley AR, Griffiths AM, Lee D, Bittinger K, Bailey A, Friedman ES, Hoffmann C, Albenberg L, Sinha R, Compher C, Gilroy E, Nessel L, Grant A, Chehoud C, Li H, Wu GD, Bushman FD. Inflammation, Antibiotics, and Diet as Environmental Stressors of the Gut Microbiome in Pediatric Crohn's Disease. Cell Host Microbe 2015; 18:489-500. [PMID: 26468751 PMCID: PMC4633303 DOI: 10.1016/j.chom.2015.09.008] [Citation(s) in RCA: 540] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 07/31/2015] [Accepted: 09/22/2015] [Indexed: 12/13/2022]
Abstract
Abnormal composition of intestinal bacteria--"dysbiosis"-is characteristic of Crohn's disease. Disease treatments include dietary changes and immunosuppressive anti-TNFα antibodies as well as ancillary antibiotic therapy, but their effects on microbiota composition are undetermined. Using shotgun metagenomic sequencing, we analyzed fecal samples from a prospective cohort of pediatric Crohn's disease patients starting therapy with enteral nutrition or anti-TNFα antibodies and reveal the full complement and dynamics of bacteria, fungi, archaea, and viruses during treatment. Bacterial community membership was associated independently with intestinal inflammation, antibiotic use, and therapy. Antibiotic exposure was associated with increased dysbiosis, whereas dysbiosis decreased with reduced intestinal inflammation. Fungal proportions increased with disease and antibiotic use. Dietary therapy had independent and rapid effects on microbiota composition distinct from other stressor-induced changes and effectively reduced inflammation. These findings reveal that dysbiosis results from independent effects of inflammation, diet, and antibiotics and shed light on Crohn disease treatments.
Collapse
Affiliation(s)
- James D Lewis
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Gastroenterology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eric Z Chen
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert N Baldassano
- Department of Pediatric Gastroenterology Hepatology and Nutrition, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Anthony R Otley
- Department of Pediatrics, IWK Health Centre, Halifax, NS B3K 6R8, Canada
| | - Anne M Griffiths
- Division of Gastroenterology Hepatology and Nutrition, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Dale Lee
- Department of Pediatric Gastroenterology Hepatology and Nutrition, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Division of Pediatric Gastroenterology Hepatology and Nutrition, Seattle Children's Hospital, University of Washington, Seattle, WA 98105, USA
| | - Kyle Bittinger
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aubrey Bailey
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elliot S Friedman
- Division of Gastroenterology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Christian Hoffmann
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lindsey Albenberg
- Department of Pediatric Gastroenterology Hepatology and Nutrition, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Rohini Sinha
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Charlene Compher
- School of Nursing, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Erin Gilroy
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lisa Nessel
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amy Grant
- Department of Pediatrics, IWK Health Centre, Halifax, NS B3K 6R8, Canada
| | - Christel Chehoud
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hongzhe Li
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gary D Wu
- Division of Gastroenterology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Frederic D Bushman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
299
|
Noninvasive monitoring of infection and rejection after lung transplantation. Proc Natl Acad Sci U S A 2015; 112:13336-41. [PMID: 26460048 DOI: 10.1073/pnas.1517494112] [Citation(s) in RCA: 221] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The survival rate following lung transplantation is among the lowest of all solid-organ transplants, and current diagnostic tests often fail to distinguish between infection and rejection, the two primary posttransplant clinical complications. We describe a diagnostic assay that simultaneously monitors for rejection and infection in lung transplant recipients by sequencing of cell-free DNA (cfDNA) in plasma. We determined that the levels of donor-derived cfDNA directly correlate with the results of invasive tests of rejection (area under the curve 0.9). We also analyzed the nonhuman cfDNA as a hypothesis-free approach to test for infections. Cytomegalovirus is most frequently assayed clinically, and the levels of CMV-derived sequences in cfDNA are consistent with clinical results. We furthermore show that hypothesis-free monitoring for pathogens using cfDNA reveals undiagnosed cases of infection, and that certain infectious pathogens such as human herpesvirus (HHV) 6, HHV-7, and adenovirus, which are not often tested clinically, occur with high frequency in this cohort.
Collapse
|
300
|
Li L, Deng X, Da Costa AC, Bruhn R, Deeks SG, Delwart E. Virome analysis of antiretroviral-treated HIV patients shows no correlation between T-cell activation and anelloviruses levels. J Clin Virol 2015; 72:106-13. [PMID: 26479202 DOI: 10.1016/j.jcv.2015.09.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/01/2015] [Accepted: 09/16/2015] [Indexed: 01/29/2023]
Abstract
BACKGROUND Abnormally high levels of T-cell activation can persist in HIV-infected subjects despite effective anti-retroviral therapy (ART) and has been associated with negative health outcomes. The nature of the antigenic drivers or other causes of this residual T-cell activation remain uncertain. Anelloviruses are universally acquired soon after birth, resulting in persistent viremia, and considered part of the commensal human virome. Reduced immunocompetence results in increased anellovirus levels. OBJECTIVES To test whether increased levels of anelloviruses or other viruses in plasma are associated with higher levels of persistent T-cell activation during ART. STUDY DESIGN Two amplification methods combined with next generation sequencing were used to detect all viruses and estimate relative anellovirus levels in plasma from 19 adults on effective ART who exhibited a wide range of T-cell activation levels. RESULTS Nucleic acids from HBV and HCV were detected in one patient each while pegivirus A (GBV-C) was found in three patients. Anellovirus DNA was detected in all patients with some individuals carrying up to eight different genotypes. Specific anellovirus genotypes or higher level of co-infections were not detected in subjects with higher levels of T-cell activation. No association was detected between relative plasma anellovirus DNA levels and the percentage of activated CD4 or CD8 T cells. CONCLUSIONS Human anelloviruses were detected in all HIV suppressed subjects, exhibited a wide range of viremia levels, and were genetically highly diverse. The level of persistent T-cell activation was not correlated with the level of viremia or genotypes present indicating that anellovirus antigens are unlikely to be a dominant source of antigens driving chronic T-cell activation.
Collapse
Affiliation(s)
- Linlin Li
- Blood Systems Research Institute, San Francisco, CA, USA; Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Xutao Deng
- Blood Systems Research Institute, San Francisco, CA, USA; Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Antonio Charlys Da Costa
- Blood Systems Research Institute, San Francisco, CA, USA; Institute of Tropical Medicine, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Roberta Bruhn
- Blood Systems Research Institute, San Francisco, CA, USA
| | - Steven G Deeks
- Positive Health Program, San Francisco General Hospital, San Francisco, CA, USA
| | - Eric Delwart
- Blood Systems Research Institute, San Francisco, CA, USA; Department of Laboratory Medicine, University of California, San Francisco, CA, USA.
| |
Collapse
|