1
|
Vijayan K. K. V, De Paris K. Nonhuman primate models of pediatric viral diseases. Front Cell Infect Microbiol 2024; 14:1493885. [PMID: 39691699 PMCID: PMC11649651 DOI: 10.3389/fcimb.2024.1493885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/08/2024] [Indexed: 12/19/2024] Open
Abstract
Infectious diseases are the leading cause of death in infants and children under 5 years of age. In utero exposure to viruses can lead to spontaneous abortion, preterm birth, congenital abnormalities or other developmental defects, often resulting in lifelong health sequalae. The underlying biological mechanisms are difficult to study in humans due to ethical concerns and limited sample access. Nonhuman primates (NHP) are closely related to humans, and pregnancy and immune ontogeny in infants are very similar to humans. Therefore, NHP are a highly relevant model for understanding fetal and postnatal virus-host interactions and to define immune mechanisms associated with increased morbidity and mortality in infants. We will discuss NHP models of viruses causing congenital infections, respiratory diseases in early life, and HIV. Cytomegalovirus (CMV) remains the most common cause of congenital defects worldwide. Measles is a vaccine-preventable disease, yet measles cases are resurging. Zika is an example of an emerging arbovirus with devastating consequences for the developing fetus and the surviving infant. Among the respiratory viruses, we will discuss influenza and Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). We will finish with HIV as an example of a lifelong infection without a cure or vaccine. The review will highlight (i) the impact of viral infections on fetal and infant immune development, (ii) how differences in infant and adult immune responses to infection alter disease outcome, and emphasize the invaluable contribution of pediatric NHP infection models to the design of effective treatment and prevention strategies, including vaccines, for human infants.
Collapse
Affiliation(s)
- Vidya Vijayan K. K.
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, United States
| | - Kristina De Paris
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, United States
- Center for AIDS Research, University of North Carolina, Chapel Hill, NC, United States
- Children’s Research Institute, University of North Carolina, Chapel Hill, NC, United States
| |
Collapse
|
2
|
Mudd JC. Quantitative and Qualitative Distinctions between HIV-1 and SIV Reservoirs: Implications for HIV-1 Cure-Related Studies. Viruses 2024; 16:514. [PMID: 38675857 PMCID: PMC11054464 DOI: 10.3390/v16040514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/07/2024] [Accepted: 03/16/2024] [Indexed: 04/28/2024] Open
Abstract
The persistence of the latent viral reservoir is the main hurdle to curing HIV-1 infection. SIV infection of non-human primates (NHPs), namely Indian-origin rhesus macaques, is the most relevant and widely used animal model to evaluate therapies that seek to eradicate HIV-1. The utility of a model ultimately rests on how accurately it can recapitulate human disease, and while reservoirs in the NHP model behave quantitatively very similar to those of long-term suppressed persons with HIV-1 (PWH) in the most salient aspects, recent studies have uncovered key nuances at the clonotypic level that differentiate the two in qualitative terms. In this review, we will highlight differences relating to proviral intactness, clonotypic structure, and decay rate during ART between HIV-1 and SIV reservoirs and discuss the relevance of these distinctions in the interpretation of HIV-1 cure strategies. While these, to some degree, may reflect a unique biology of the virus or host, distinctions among the proviral landscape in SIV are likely to be shaped significantly by the condensed timeframe of NHP studies. ART is generally initiated earlier in the disease course, and animals are virologically suppressed for shorter periods before receiving interventions. Because these are experimental variables dictated by the investigator, we offer guidance on study design for cure-related studies performed in the NHP model. Finally, we highlight the case of GS-9620 (Vesatolimod), an antiviral TLR7 agonist tested in multiple independent pre-clinical studies in which virological outcomes may have been influenced by study-related variables.
Collapse
Affiliation(s)
- Joseph C. Mudd
- Tulane National Primate Research Center, Covington, LA 70433, USA;
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
3
|
Kaur A, Vaccari M. Exploring HIV Vaccine Progress in the Pre-Clinical and Clinical Setting: From History to Future Prospects. Viruses 2024; 16:368. [PMID: 38543734 PMCID: PMC10974975 DOI: 10.3390/v16030368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/08/2024] [Accepted: 02/21/2024] [Indexed: 04/01/2024] Open
Abstract
The human immunodeficiency virus (HIV) continues to pose a significant global health challenge, with millions of people affected and new cases emerging each year. While various treatment and prevention methods exist, including antiretroviral therapy and non-vaccine approaches, developing an effective vaccine remains the most crucial and cost-effective solution to combating the HIV epidemic. Despite significant advancements in HIV research, the HIV vaccine field has faced numerous challenges, and only one clinical trial has demonstrated a modest level of efficacy. This review delves into the history of HIV vaccines and the current efforts in HIV prevention, emphasizing pre-clinical vaccine development using the non-human primate model (NHP) of HIV infection. NHP models offer valuable insights into potential preventive strategies for combating HIV, and they play a vital role in informing and guiding the development of novel vaccine candidates before they can proceed to human clinical trials.
Collapse
Affiliation(s)
- Amitinder Kaur
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA;
- School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Monica Vaccari
- Division of Immunology, Tulane National Primate Research Center, Covington, LA 70433, USA;
- School of Medicine, Tulane University, New Orleans, LA 70112, USA
| |
Collapse
|
4
|
Kirkland JM, Patel I, Ardeshna MS, Kopec AM. Microglial synaptic pruning in the nucleus accumbens during adolescence sex-specifically influences splenic immune outcomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.03.539317. [PMID: 37205376 PMCID: PMC10187280 DOI: 10.1101/2023.05.03.539317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Strong social support promotes a variety of positive health outcomes in humans and rodent models, while social isolation in rodents shortens lifespan, perceived social isolation (i.e. loneliness) can increase mortality by up to 50% in humans. How social relationships lead to these drastic health effects is unclear, but may involve modulation of the peripheral immune system. The reward circuitry of the brain and social behaviors undergo a critical period of development during adolescence. We published that microglia-mediated synaptic pruning occurs in the nucleus accumbens (NAc) reward region during adolescence to mediate social development in male and female rats. We hypothesized that if reward circuitry activity and social relationships directly impact the peripheral immune system, then natural developmental changes in the reward circuitry and social behaviors during adolescence should also directly impact the peripheral immune system. To test this, we inhibited microglial pruning in the NAc during adolescence, and then collected spleen tissue for mass spectrometry proteomic analysis and ELISA validation. We found that the global proteomic consequences of inhibiting microglial pruning in the NAc were similar between the sexes, but target-specific examination suggests that NAc pruning impacts Th1 cell-related immune markers in the spleen in males, but not females, and broad neurochemical systems in the spleen in females, but not males.
Collapse
Affiliation(s)
- J. M. Kirkland
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College
| | - Ishan Patel
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College
| | - Monali S. Ardeshna
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College
| | - Ashley M. Kopec
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College
| |
Collapse
|
5
|
Milligan EC, Olstad K, Williams CA, Mallory M, Cano P, Cross KA, Munt JE, Garrido C, Lindesmith L, Watanabe J, Usachenko JL, Hopkins L, Immareddy R, Shaan Lakshmanappa Y, Elizaldi SR, Roh JW, Sammak RL, Pollard RE, Yee JL, Herbek S, Scobey T, Miehlke D, Fouda G, Ferrari G, Gao H, Shen X, Kozlowski PA, Montefiori D, Hudgens MG, Edwards DK, Carfi A, Corbett KS, Graham BS, Fox CB, Tomai M, Iyer SS, Baric R, Reader R, Dittmer DP, Van Rompay KKA, Permar SR, De Paris K. Infant rhesus macaques immunized against SARS-CoV-2 are protected against heterologous virus challenge 1 year later. Sci Transl Med 2023; 15:eadd6383. [PMID: 36454813 PMCID: PMC9765459 DOI: 10.1126/scitranslmed.add6383] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
The U.S. Food and Drug Administration only gave emergency use authorization of the BNT162b2 and mRNA-1273 SARS-CoV-2 vaccines for infants 6 months and older in June 2022. Yet questions regarding the durability of vaccine efficacy, especially against emerging variants, in this age group remain. We demonstrated previously that a two-dose regimen of stabilized prefusion Washington SARS-CoV-2 S-2P spike (S) protein encoded by mRNA encapsulated in lipid nanoparticles (mRNA-LNP) or purified S-2P mixed with 3M-052, a synthetic Toll-like receptor (TLR) 7/8 agonist, in a squalene emulsion (Protein+3M-052-SE) was safe and immunogenic in infant rhesus macaques. Here, we demonstrate that broadly neutralizing and spike-binding antibodies against variants of concern (VOCs), as well as T cell responses, persisted for 12 months. At 1 year, corresponding to human toddler age, we challenged vaccinated rhesus macaques and age-matched nonvaccinated controls intranasally and intratracheally with a high dose of heterologous SARS-CoV-2 B.1.617.2 (Delta). Seven of eight control rhesus macaques exhibited severe interstitial pneumonia and high virus replication in the upper and lower respiratory tract. In contrast, vaccinated rhesus macaques had faster viral clearance with mild to no pneumonia. Neutralizing and binding antibody responses to the B.1.617.2 variant at the day of challenge correlated with lung pathology and reduced virus replication. Overall, the Protein+3M-052-SE vaccine provided superior protection to the mRNA-LNP vaccine, emphasizing opportunities for optimization of current vaccine platforms. The observed efficacy of both vaccines 1 year after vaccination supports the implementation of an early-life SARS-CoV-2 vaccine.
Collapse
Affiliation(s)
- Emma C Milligan
- Department of Microbiology and Immunology, Children's Research Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Katherine Olstad
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Caitlin A Williams
- Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Michael Mallory
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Patricio Cano
- Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kaitlyn A Cross
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jennifer E Munt
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Carolina Garrido
- Center for Immunology and Infectious Diseases, University of California at Davis, Davis, CA 95616, USA
| | - Lisa Lindesmith
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jennifer Watanabe
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Jodie L Usachenko
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Lincoln Hopkins
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Ramya Immareddy
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | | | - Sonny R Elizaldi
- Center for Immunology and Infectious Diseases, University of California at Davis, Davis, CA 95616, USA.,Graduate Group in Immunology, University of California at Davis, Davis, CA 95616, USA
| | - Jamin W Roh
- Center for Immunology and Infectious Diseases, University of California at Davis, Davis, CA 95616, USA.,Graduate Group in Immunology, University of California at Davis, Davis, CA 95616, USA
| | - Rebecca L Sammak
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Rachel E Pollard
- School of Veterinary Medicine, University of California at Davis, Davis, CA 95616, USA
| | - JoAnn L Yee
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Savannah Herbek
- Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Trevor Scobey
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Dieter Miehlke
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA.,Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Genevieve Fouda
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA.,Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Guido Ferrari
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA.,Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Hongmei Gao
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Xiaoying Shen
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Pamela A Kozlowski
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - David Montefiori
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Michael G Hudgens
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | - Kizzmekia S Corbett
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Christopher B Fox
- Access to Advanced Health Institute, Seattle, WA 98102, USA.,Department of Global Health, University of Washington, Seattle, WA 98105, USA
| | - Mark Tomai
- 3M Corporate Research Materials Laboratory, Saint Paul, MN 55144, USA
| | - Smita S Iyer
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA.,Center for Immunology and Infectious Diseases, University of California at Davis, Davis, CA 95616, USA
| | - Ralph Baric
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Rachel Reader
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Dirk P Dittmer
- Department of Microbiology and Immunology, Children's Research Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Koen K A Van Rompay
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA.,Department of Pathology, Microbiology and Immunology, University of California at Davis, Davis, CA 95616, USA
| | - Sallie R Permar
- Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Kristina De Paris
- Department of Microbiology and Immunology, Children's Research Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
6
|
Capitanio JP, Del Rosso LA, Spinner A. Variation in infant rhesus monkeys' (Macaca mulatta) neutrophil-to-lymphocyte ratio is associated with environmental conditions, emotionality, and cortisol concentrations, and predicts disease-related outcomes. Brain Behav Immun 2023; 109:105-116. [PMID: 36681357 DOI: 10.1016/j.bbi.2023.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
The neutrophil-to-lymphocyte ratio (NLR) is a predictor of morbidity for a variety of medical conditions, but little is known about how variation in NLR arises. We examined variation in this measure in a sample of 4577 infant rhesus monkeys (54.8 % female), who participated in the BioBehavioral Assessment program at the California National Primate Research Center at 3-4 months of age. Lower values for NLR were seen for animals reared indoors, for animals that were raised to be free of specific pathogens, and for males. In addition lower NLR was associated with higher stress values of cortisol and with greater emotionality in response to an acute stressor. Finally, lower NLR in infancy was associated with greater risk for developing airways hyperresponsiveness (a hallmark of asthma); with display of diarrhea up to 3.97 years later; and with greater viral load when infected with the simian immunodeficiency virus at a mean of 6.1 years of age. Infant NLR was a better predictor of viral load than was a contemporaneously obtained measure of NLR. Infant and adult values of NLR were only modestly correlated; one reason may be that the infant measure was obtained during stressful conditions and the adult measure was obtained under baseline conditions. We propose that NLR is an integrated outcome measure reflecting organization and interaction of stress-response and immune systems. As such, assessment of NLR under conditions of stress may be a particularly useful marker of individual differences in morbidity, especially for conditions in which stress plays an important role, as in asthma, diarrhea/colitis, and AIDS.
Collapse
Affiliation(s)
- John P Capitanio
- Department of Psychology, University of California, Davis, CA, USA; California National Primate Research Center, University of California, Davis, CA, USA.
| | - Laura A Del Rosso
- California National Primate Research Center, University of California, Davis, CA, USA
| | - Abigail Spinner
- California National Primate Research Center, University of California, Davis, CA, USA
| |
Collapse
|
7
|
Yee JL, Strelow LI, White JA, Rosenthal AN, Barry PA. Horizontal transmission of endemic viruses among rhesus macaques (Macaca mulatta): Implications for human cytomegalovirus vaccine/challenge design. J Med Primatol 2023; 52:53-63. [PMID: 36151734 PMCID: PMC9825633 DOI: 10.1111/jmp.12621] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Rhesus macaques are natural hosts to multiple viruses including rhesus cytomegalovirus (RhCMV), rhesus rhadinovirus (RRV), and Simian Foamy Virus (SFV). While viral infections are ubiquitous, viral transmissions to uninfected animals are incompletely defined. Management procedures of macaque colonies include cohorts that are Specific Pathogen Free (SPF). Greater understanding of viral transmission would augment SPF protocols. Moreover, vaccine/challenge studies of human viruses would be enhanced by leveraging transmission of macaque viruses to recapitulate expected challenges of human vaccine trials. MATERIALS AND METHODS This study characterizes viral transmissions to uninfected animals following inadvertent introduction of RhCMV/RRV/SFV-infected adults to a cohort of uninfected juveniles. Following co-housing with virus-positive adults, juveniles were serially evaluated for viral infection. RESULTS Horizontal viral transmission was rapid and absolute, reaching 100% penetrance between 19 and 78 weeks. CONCLUSIONS This study provides insights into viral natural histories with implications for colony management and modeling vaccine-mediated immune protection studies.
Collapse
Affiliation(s)
- JoAnn L Yee
- California National Primate Research Center, Davis, California, USA
- University of California, Davis, Davis, California, USA
| | - Lisa I Strelow
- California National Primate Research Center, Davis, California, USA
- University of California, Davis, Davis, California, USA
- Center for Immunology and Infectious Diseases, Davis, California, USA
| | - Jessica A White
- California National Primate Research Center, Davis, California, USA
- University of California, Davis, Davis, California, USA
| | - Ann N Rosenthal
- California National Primate Research Center, Davis, California, USA
- University of California, Davis, Davis, California, USA
| | - Peter A Barry
- California National Primate Research Center, Davis, California, USA
- University of California, Davis, Davis, California, USA
- Center for Immunology and Infectious Diseases, Davis, California, USA
| |
Collapse
|
8
|
Smiley Evans T, Lowenstine LJ, Ssebide B, Barry PA, Kinani JF, Nizeyimana F, Noheli JB, Okello R, Mudakikwa A, Cranfield MR, Mazet JAK, Johnson CK, Gilardi KV. Simian homologues of human herpesviruses and implications for novel viral introduction to free-living mountain gorillas. Am J Primatol 2023; 85:e23439. [PMID: 36263518 PMCID: PMC11017921 DOI: 10.1002/ajp.23439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/17/2022] [Accepted: 09/05/2022] [Indexed: 01/05/2023]
Abstract
The endangered mountain gorilla (Gorilla beringei beringei) in Rwanda, Uganda, and the Democratic Republic of Congo is frequently in contact with humans through tourism, research activities, and illegal entry of people into protected gorilla habitat. Herpesviruses, which are ubiquitous in primates, have the potential to be shared in any setting where humans and gorillas share habitat. Based on serological findings and clinical observations of orofacial ulcerated lesions resembling herpetic lesions, an alpha-herpesvirus resembling human herpes simplex virus type 1 (HSV-1) has long been suspected to be present in human-habituated mountain gorillas in the wild. While the etiology of orofacial lesions in the wild has not been confirmed, HSV-1 has been suspected in captively-housed mountain gorillas and confirmed in a co-housed confiscated Grauer's gorilla (Gorilla beringei graueri). To better characterize herpesviruses infecting mountain gorillas and to determine the presence/absence of HSV-1 in the free-living population, we conducted a population-wide survey to test for the presence of orally shed herpesviruses. DNA was extracted from discarded chewed plants collected from 294 individuals from 26 groups, and samples were screened by polymerase chain reaction using pan-herpesvirus and HSV-1-specific assays. We found no evidence that human herpesviruses had infected free-ranging mountain gorillas. However, we found gorilla-specific homologs to human herpesviruses, including cytomegaloviruses (GbbCMV-1 and 2), a lymphocryptovirus (GbbLCV-1), and a new rhadinovirus (GbbRHV-1) with similar characteristics (i.e., timing of primary infection, shedding in multiple age groups, and potential modes of transmission) to their human counterparts, human cytomegalovirus, Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus, respectively.
Collapse
Affiliation(s)
- Tierra Smiley Evans
- Gorilla Doctors, Karen C. Drayer Wildlife Health Center, One Health Institute, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | - Linda J Lowenstine
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | - Benard Ssebide
- Gorilla Doctors, Karen C. Drayer Wildlife Health Center, One Health Institute, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | - Peter A Barry
- Department of Pathology and Laboratory Medicine, Center for Immunology and Infectious Diseases, California National Primate Research Center, University of California Davis, Davis, California, USA
| | - Jean Felix Kinani
- One Health Approach for Conservation (OHAC), Gorilla Health, Kigali, Rwanda
| | - Fred Nizeyimana
- Gorilla Doctors, Karen C. Drayer Wildlife Health Center, One Health Institute, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | - Jean Bosco Noheli
- Gorilla Doctors, Karen C. Drayer Wildlife Health Center, One Health Institute, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | - Ricky Okello
- Gorilla Doctors, Karen C. Drayer Wildlife Health Center, One Health Institute, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | | | - Michael R Cranfield
- Gorilla Doctors, Karen C. Drayer Wildlife Health Center, One Health Institute, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | - Jonna A K Mazet
- Gorilla Doctors, Karen C. Drayer Wildlife Health Center, One Health Institute, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | - Christine K Johnson
- Epicenter for Disease Dynamics, One Health Institute, University of California Davis, Davis, California, USA
| | - Kirsten V Gilardi
- Gorilla Doctors, Karen C. Drayer Wildlife Health Center, One Health Institute, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| |
Collapse
|
9
|
Chin N, Narayan NR, Méndez-Lagares G, Ardeshir A, Chang WLW, Deere JD, Fontaine JH, Chen C, Kieu HT, Lu W, Barry PA, Sparger EE, Hartigan-O'Connor DJ. Cytomegalovirus infection disrupts the influence of short-chain fatty acid producers on Treg/Th17 balance. MICROBIOME 2022; 10:168. [PMID: 36210471 PMCID: PMC9549678 DOI: 10.1186/s40168-022-01355-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 08/15/2022] [Indexed: 06/01/2023]
Abstract
BACKGROUND Both the gut microbiota and chronic viral infections have profound effects on host immunity, but interactions between these influences have been only superficially explored. Cytomegalovirus (CMV), for example, infects approximately 80% of people globally and drives significant changes in immune cells. Similarly, certain gut-resident bacteria affect T-cell development in mice and nonhuman primates. It is unknown if changes imposed by CMV on the intestinal microbiome contribute to immunologic effects of the infection. RESULTS We show that rhesus cytomegalovirus (RhCMV) infection is associated with specific differences in gut microbiota composition, including decreased abundance of Firmicutes, and that the extent of microbial change was associated with immunologic changes including the proliferation, differentiation, and cytokine production of CD8+ T cells. Furthermore, RhCMV infection disrupted the relationship between short-chain fatty acid producers and Treg/Th17 balance observed in seronegative animals, showing that some immunologic effects of CMV are due to disruption of previously existing host-microbe relationships. CONCLUSIONS Gut microbes have an important influence on health and disease. Diet is known to shape the microbiota, but the influence of concomitant chronic viral infections is unclear. We found that CMV influences gut microbiota composition to an extent that is correlated with immunologic changes in the host. Additionally, pre-existing correlations between immunophenotypes and gut microbes can be subverted by CMV infection. Immunologic effects of CMV infection on the host may therefore be mediated by two different mechanisms involving gut microbiota. Video Abstract.
Collapse
Affiliation(s)
- Ning Chin
- California National Primate Research Center, University of California, Davis, Davis, USA
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, USA
| | - Nicole R Narayan
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, USA
| | - Gema Méndez-Lagares
- California National Primate Research Center, University of California, Davis, Davis, USA
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, USA
| | - Amir Ardeshir
- California National Primate Research Center, University of California, Davis, Davis, USA
| | - W L William Chang
- California National Primate Research Center, University of California, Davis, Davis, USA
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, USA
| | - Jesse D Deere
- California National Primate Research Center, University of California, Davis, Davis, USA
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, USA
| | - Justin H Fontaine
- California National Primate Research Center, University of California, Davis, Davis, USA
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, USA
| | - Connie Chen
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, USA
| | - Hung T Kieu
- California National Primate Research Center, University of California, Davis, Davis, USA
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, USA
| | - Wenze Lu
- California National Primate Research Center, University of California, Davis, Davis, USA
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, USA
| | - Peter A Barry
- Center for Immunology and Infectious Diseases, University of California, Davis, Davis, USA
| | - Ellen E Sparger
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, Davis, USA
| | - Dennis J Hartigan-O'Connor
- California National Primate Research Center, University of California, Davis, Davis, USA.
- Department of Medical Microbiology and Immunology, School of Medicine, University of California, Davis, Davis, USA.
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, USA.
| |
Collapse
|
10
|
Capitanio JP, Sommet N, Del Rosso L. The relationship of maternal rank, 5-HTTLPR genotype, and MAOA-LPR genotype to temperament in infant rhesus monkeys (Macaca mulatta). Am J Primatol 2022; 84:e23374. [PMID: 35322905 PMCID: PMC10461592 DOI: 10.1002/ajp.23374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/17/2022] [Accepted: 02/27/2022] [Indexed: 11/11/2022]
Abstract
Temperament is a construct whose manifestations are quantifiable from an early age, and whose origins have been proposed as "biological." Our goal was to determine whether maternal rank and infant genotype are associated with five measures of temperament in 3- to 4-month old rhesus monkeys (Macaca mulatta), all of whom were born and reared by their mothers in large, outdoor, half-acre cages. Maternal rank was defined as the proportion of animals outranked by each female, and the two genes of interest to us were monoamine oxidase and serotonin transporter, both of which are polymorphic in their promoter regions (MAOA-LPR and 5-HTTLPR, respectively), with one allele of each gene considered a "plasticity" allele, conferring increased sensitivity to environmental events. Our large sample size (n = 2014-3140) enabled us to examine the effects of individual genotypes rather than combining genotypes as is often done. Rank was positively associated with Confident temperament, but only for animals with the 5-repeat allele for MAOA-LPR. Rank had no other effect on temperament. In contrast, genotype had many different effects, with 5-HTTLPR associated with behavioral inhibition, and MAOA-LPR associated with ratings-based measures of temperament. We also examined the joint effect of the two genotypes and found some evidence for a dose-response: animals with the plasticity alleles for both genes were more likely to be behaviorally inhibited. Our results suggest phenotypic differences between animals possessing alleles for MAOA-LPR that show functional equivalence based on in vitro tests, and our data for 5-HTTLPR revealed differences between short/short homozygotes and long/short heterozygotes, strongly suggesting that combining genotypes for statistical analysis should be avoided if possible. Our analysis also provides evidence of sex differences in temperament, and, to our knowledge, the only evidence of differences in temperament based on specific pathogen-free status. We suggest several directions for future research.
Collapse
Affiliation(s)
- John P. Capitanio
- Neuroscience and Behavior Unit, California National Primate Research CenterUniversity of CaliforniaDavisCaliforniaUSA
- Department of PsychologyUniversity of CaliforniaDavisCaliforniaUSA
| | - Nicolas Sommet
- LIVES Center, Faculty of Social and Political SciencesUniversity of LausanneLausanneSwitzerland
| | - Laura Del Rosso
- Neuroscience and Behavior Unit, California National Primate Research CenterUniversity of CaliforniaDavisCaliforniaUSA
| |
Collapse
|
11
|
Abstract
Nonhuman primates are critically important animal models in which to study complex human diseases, understand biological functions, and address the safety of new diagnostics and therapies proposed for human use. They have genetic, physiologic, immunologic, and developmental similarities when compared to humans and therefore provide important preclinical models of human health and disease. This review highlights select research areas that demonstrate the importance of nonhuman primates in translational research. These include pregnancy and developmental disorders, infectious diseases, gene therapy, somatic cell genome editing, and applications of in vivo imaging. The power of the immune system and our increasing understanding of the role it plays in acute and chronic illnesses are being leveraged to produce new treatments for a range of medical conditions. Given the importance of the human immune system in health and disease, detailed study of the immune system of nonhuman primates is essential to advance preclinical translational research. The need for nonhuman primates continues to remain a high priority, which has been acutely evident during the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) global pandemic. Nonhuman primates will continue to address key questions and provide predictive models to identify the safety and efficiency of new diagnostics and therapies for human use across the lifespan.
Collapse
Affiliation(s)
- Alice F Tarantal
- Departments of Pediatrics and Cell Biology and Human Anatomy, University of California, Davis, California, USA;
- California National Primate Research Center, University of California, Davis, California, USA
| | - Stephen C Noctor
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, California, USA;
| | - Dennis J Hartigan-O'Connor
- California National Primate Research Center, University of California, Davis, California, USA
- Medical Microbiology and Immunology, School of Medicine, University of California, Davis, California, USA;
| |
Collapse
|
12
|
Pathogenesis of wild-type-like rhesus cytomegalovirus strains following oral exposure of immune-competent rhesus macaques. J Virol 2021; 96:e0165321. [PMID: 34788083 DOI: 10.1128/jvi.01653-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Rhesus cytomegalovirus (RhCMV) infection of rhesus macaques (Macaca mulatta) is a valuable nonhuman primate model of human CMV (HCMV) persistence and pathogenesis. In vivo studies predominantly use tissue culture-adapted variants of RhCMV that contain multiple genetic mutations compared to wild-type (WT) RhCMV. In many studies, animals have been inoculated by non-natural routes (e.g., subcutaneous, intravenous) that do not recapitulate disease progression via the normative route of mucosal exposure. Accordingly, the natural history of RhCMV would be more accurately reproduced by infecting macaques with strains of RhCMV that reflect the WT genome using natural routes of mucosal transmission. Herein, we tested two WT-like RhCMV strains, UCD52 and UCD59, and demonstrated that systemic infection and frequent, high-titer viral shedding in bodily fluids occurred following oral inoculation. RhCMV disseminated to a broad range of tissues, including the central nervous system and reproductive organs. Commonly infected tissues included the thymus, spleen, lymph nodes, kidneys, bladder, and salivary glands. Histological examination revealed prominent nodular hyperplasia in spleens and variable levels of lymphoid lymphofollicular hyperplasia in lymph nodes. One of six inoculated animals had limited viral dissemination and shedding, with commensurately weak antibody responses to RhCMV antigens. These data suggest that long-term RhCMV infection parameters might be restricted by local innate factors and/or de novo host immune responses in a minority of primary infections. Together, we have established an oral RhCMV infection model that mimics natural HCMV infection. The virological and immunological parameters characterized in this study will greatly inform HCMV vaccine designs for human immunization. IMPORTANCE Human cytomegalovirus (HCMV) is globally ubiquitous with high seroprevalence rates in all communities. HCMV infections can occur vertically following mother-to-fetus transmission across the placenta and horizontally following shedding of virus in bodily fluids in HCMV infected hosts and subsequent exposure of susceptible individuals to virus-laden fluids. Intrauterine HCMV has long been recognized as an infectious threat to fetal growth and development. Since vertical HCMV infections occur following horizontal HCMV transmission to the pregnant mother, the nonhuman primate model of HCMV pathogenesis was used to characterize the virological and immunological parameters of infection following primary mucosal exposures to rhesus cytomegalovirus.
Collapse
|
13
|
Sarfas C, White AD, Sibley L, Morrison AL, Gullick J, Lawrence S, Dennis MJ, Marsh PD, Fletcher HA, Sharpe SA. Characterization of the Infant Immune System and the Influence and Immunogenicity of BCG Vaccination in Infant and Adult Rhesus Macaques. Front Immunol 2021; 12:754589. [PMID: 34707617 PMCID: PMC8542880 DOI: 10.3389/fimmu.2021.754589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/27/2021] [Indexed: 11/13/2022] Open
Abstract
In many countries where tuberculosis (TB) is endemic, the Bacillus Calmette–Guérin (BCG) vaccine is given as close to birth as possible to protect infants and children from severe forms of TB. However, BCG has variable efficacy and is not as effective against adult pulmonary TB. At present, most animal models used to study novel TB vaccine candidates rely on the use of adult animals. Human studies show that the infant immune system is different to that of an adult. Understanding how the phenotypic profile and functional ability of the immature host immune system compares to that of a mature adult, together with the subsequent BCG immune response, is critical to ensuring that new TB vaccines are tested in the most appropriate models. BCG-specific immune responses were detected in macaques vaccinated within a week of birth from six weeks after immunization indicating that neonatal macaques are able to generate a functional cellular response to the vaccine. However, the responses measured were significantly lower than those typically observed following BCG vaccination in adult rhesus macaques and infant profiles were skewed towards the activation and attraction of macrophages and monocytes and the synthesis in addition to release of pro-inflammatory cytokines such as IL-1, IL-6 and TNF-α. The frequency of specific immune cell populations changed significantly through the first three years of life as the infants developed into young adult macaques. Notably, the CD4:CD8 ratio significantly declined as the macaques aged due to a significant decrease in the proportion of CD4+ T-cells relative to a significant increase in CD8+ T-cells. Also, the frequency of both CD4+ and CD8+ T-cells expressing the memory marker CD95, and memory subset populations including effector memory, central memory and stem cell memory, increased significantly as animals matured. Infant macaques, vaccinated with BCG within a week of birth, possessed a significantly higher frequency of CD14+ classical monocytes and granulocytes which remained different throughout the first three years of life compared to unvaccinated age matched animals. These findings, along with the increase in monokines following vaccination in infants, may provide an insight into the mechanism by which vaccination with BCG is able to provide non-specific immunity against non-mycobacterial organisms.
Collapse
Affiliation(s)
- Charlotte Sarfas
- National Infection Service, UK Health Security Agency, Salisbury, United Kingdom
| | - Andrew D White
- National Infection Service, UK Health Security Agency, Salisbury, United Kingdom
| | - Laura Sibley
- National Infection Service, UK Health Security Agency, Salisbury, United Kingdom
| | - Alexandra L Morrison
- National Infection Service, UK Health Security Agency, Salisbury, United Kingdom
| | - Jennie Gullick
- National Infection Service, UK Health Security Agency, Salisbury, United Kingdom
| | - Steve Lawrence
- National Infection Service, UK Health Security Agency, Salisbury, United Kingdom
| | - Mike J Dennis
- National Infection Service, UK Health Security Agency, Salisbury, United Kingdom
| | - Philip D Marsh
- National Infection Service, UK Health Security Agency, Salisbury, United Kingdom
| | - Helen A Fletcher
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Sally A Sharpe
- National Infection Service, UK Health Security Agency, Salisbury, United Kingdom
| |
Collapse
|
14
|
Méndez-Lagares G, Chin N, Chang WW, Lee J, Rosás-Umbert M, Kieu HT, Merriam D, Lu W, Kim S, Adamson L, Brander C, Luciw PA, Barry PA, Hartigan-O’Connor DJ. Cytomegalovirus mediates expansion of IL-15-responsive innate-memory cells with SIV killing function. J Clin Invest 2021; 131:148542. [PMID: 34153005 PMCID: PMC8321572 DOI: 10.1172/jci148542] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 06/03/2021] [Indexed: 12/13/2022] Open
Abstract
Interindividual immune variability is driven predominantly by environmental factors, including exposure to chronic infectious agents such as cytomegalovirus (CMV). We investigated the effects of rhesus CMV (RhCMV) on composition and function of the immune system in young macaques. Within months of infection, RhCMV was associated with impressive changes in antigen presenting cells, T cells, and NK cells-and marked expansion of innate-memory CD8+ T cells. These cells express high levels of NKG2A/C and the IL-2 and IL-15 receptor beta chain, CD122. IL-15 was sufficient to drive differentiation of the cells in vitro and in vivo. Expanded NKG2A/C+CD122+CD8+ T cells in RhCMV-infected macaques, but not their NKG2-negative counterparts, were endowed with cytotoxicity against class I-deficient K562 targets and prompt IFN-γ production in response to stimulation with IL-12 and IL-18. Because RhCMV clone 68-1 forms the viral backbone of RhCMV-vectored SIV vaccines, we also investigated immune changes following administration of RhCMV 68-1-vectored SIV vaccines. These vaccines led to impressive expansion of NKG2A/C+CD8+ T cells with capacity to inhibit SIV replication ex vivo. Thus, CMV infection and CMV-vectored vaccination drive expansion of functional innate-like CD8 cells via host IL-15 production, suggesting that innate-memory expansion could be achieved by other vaccine platforms expressing IL-15.
Collapse
Affiliation(s)
- Gema Méndez-Lagares
- California National Primate Research Center
- Department of Medical Microbiology and Immunology
| | - Ning Chin
- California National Primate Research Center
- Department of Medical Microbiology and Immunology
| | - W.L. William Chang
- California National Primate Research Center
- Department of Medical Microbiology and Immunology
| | - Jaewon Lee
- Graduate Group in Immunology, and
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
| | | | - Hung T. Kieu
- California National Primate Research Center
- Department of Medical Microbiology and Immunology
| | - David Merriam
- California National Primate Research Center
- Department of Medical Microbiology and Immunology
| | - Wenze Lu
- California National Primate Research Center
- Department of Medical Microbiology and Immunology
| | - Sungjin Kim
- Department of Medical Microbiology and Immunology
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
| | - Lourdes Adamson
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
| | - Christian Brander
- IrsiCaixa - AIDS Research Institute, Badalona, Barcelona, Spain
- Universitat de Vic-Universitat Central de Catalunya (UVic-UCC), Vic, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Paul A. Luciw
- California National Primate Research Center
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
- Department of Pathology and Laboratory Medicine, University of California, Davis, California, USA
| | - Peter A. Barry
- California National Primate Research Center
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
- Department of Pathology and Laboratory Medicine, University of California, Davis, California, USA
| | - Dennis J. Hartigan-O’Connor
- California National Primate Research Center
- Department of Medical Microbiology and Immunology
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, California, USA
| |
Collapse
|
15
|
He Z, Fahlberg MD, Takahashi N, Slisarenko N, Rout N, Didier ES, Kuroda MJ. Declining neutrophil production despite increasing G-CSF levels is associated with chronic inflammation in elderly rhesus macaques. J Leukoc Biol 2021; 109:1033-1043. [PMID: 33974319 DOI: 10.1002/jlb.1hi1120-779r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/27/2021] [Accepted: 02/10/2021] [Indexed: 01/07/2023] Open
Abstract
Aging is characterized by a loss of bone marrow hematopoietic tissue, systemic chronic inflammation, and higher susceptibility to infectious and noninfectious diseases. We previously reported the tightly regulated kinetics and massive daily production of neutrophils during homeostasis in adult rhesus macaques aged 3 to 19 yr (equivalent to approximately 10 to 70 yr of age in humans). In the current study, we observed an earlier release of recently dividing neutrophils from bone marrow and greater in-group variability of neutrophil kinetics based on in vivo BrdU labeling in a group of older rhesus macaques of 20-26 yr of age. Comparing neutrophil numbers and circulating cytokine levels in rhesus macaques spanning 2 to 26 yr of age, we found a negative correlation between age and blood neutrophil counts and a positive correlation between age and plasma G-CSF levels. Hierarchic clustering analysis also identified strong associations between G-CSF with the proinflammatory cytokines, IL-1β and MIP-1α. Furthermore, neutrophils from older macaques expressed less myeloperoxidase and comprised higher frequencies of polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) compared to the young adult macaques. In summary, we observed an earlier release from bone marrow and a reduced production of neutrophils despite the increased levels of plasma G-CSF, especially in the elderly rhesus macaques. This lower neutrophil production capacity associated with increased production of proinflammatory cytokines as well as an earlier release of less mature neutrophils and PMN-MDSCs may contribute to the chronic inflammation and greater susceptibility to infectious and noninfectious diseases during aging.
Collapse
Affiliation(s)
- Ziyuan He
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, USA.,Division of Immunology, Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Marissa D Fahlberg
- Division of Immunology, Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Naofumi Takahashi
- Division of Immunology, Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA.,Joint Research Center for Human Retrovirus Infections, Kumamoto University, Kumamoto, Japan
| | - Nadia Slisarenko
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Namita Rout
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Elizabeth S Didier
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA.,Center for Immunology and Infectious Diseases and the California National Primate Research Center, University of California Davis, Davis, California, USA
| | - Marcelo J Kuroda
- Division of Immunology, Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA.,Center for Immunology and Infectious Diseases and the California National Primate Research Center, University of California Davis, Davis, California, USA
| |
Collapse
|
16
|
Rioux M, McNeil M, Francis ME, Dawe N, Foley M, Langley JM, Kelvin AA. The Power of First Impressions: Can Influenza Imprinting during Infancy Inform Vaccine Design? Vaccines (Basel) 2020; 8:E546. [PMID: 32961707 PMCID: PMC7563765 DOI: 10.3390/vaccines8030546] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 12/15/2022] Open
Abstract
Influenza virus infection causes severe respiratory illness in people worldwide, disproportionately affecting infants. The immature respiratory tract coupled with the developing immune system, and lack of previous exposure to the virus is thought to synergistically play a role in the increased disease severity in younger age groups. No influenza vaccines are available for those under six months, although maternal influenza immunization is recommended. In children aged six months to two years, vaccine immunogenicity is dampened compared to older children and adults. Unlike older children and adults, the infant immune system has fewer antigen-presenting cells and soluble immune factors. Paradoxically, we know that a person's first infection with the influenza virus during infancy or childhood leads to the establishment of life-long immunity toward that particular virus strain. This is called influenza imprinting. We contend that by understanding the influenza imprinting event in the context of the infant immune system, we will be able to design more effective influenza vaccines for both infants and adults. Working through the lens of imprinting, using infant influenza animal models such as mice and ferrets which have proven useful for infant immunity studies, we will gain a better understanding of imprinting and its implications regarding vaccine design. This review examines literature regarding infant immune and respiratory development, current vaccine strategies, and highlights the importance of research into the imprinting event in infant animal models to develop more effective and protective vaccines for all including young children.
Collapse
Affiliation(s)
- Melissa Rioux
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.); (M.M.); (M.E.F.); (N.D.); (M.F.)
| | - Mara McNeil
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.); (M.M.); (M.E.F.); (N.D.); (M.F.)
| | - Magen E. Francis
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.); (M.M.); (M.E.F.); (N.D.); (M.F.)
- Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), Saskatoon, SK S7N 5E3, Canada
| | - Nicholas Dawe
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.); (M.M.); (M.E.F.); (N.D.); (M.F.)
| | - Mary Foley
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.); (M.M.); (M.E.F.); (N.D.); (M.F.)
| | - Joanne M. Langley
- Department of Pediatrics, Division of Infectious Disease, Faculty of Medicine, Dalhousie University, Halifax, NS B3K 6R8, Canada;
- The Canadian Center for Vaccinology (IWK Health Centre, Dalhousie University and the Nova Scotia Health Authority), Halifax, NS B3K 6R8, Canada
- Department of Community Health and Epidemiology, Faculty of Medicine, Dalhousie University, Halifax, NS B3K 6R8, Canada
| | - Alyson A. Kelvin
- Department of Microbiology and Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; (M.R.); (M.M.); (M.E.F.); (N.D.); (M.F.)
- Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), Saskatoon, SK S7N 5E3, Canada
- Department of Pediatrics, Division of Infectious Disease, Faculty of Medicine, Dalhousie University, Halifax, NS B3K 6R8, Canada;
- The Canadian Center for Vaccinology (IWK Health Centre, Dalhousie University and the Nova Scotia Health Authority), Halifax, NS B3K 6R8, Canada
| |
Collapse
|
17
|
Abstract
: The use of cytomegalovirus (CMV) as a vaccine vector to express antigens against multiple infectious diseases, including simian immunodeficiency virus, Ebola virus, plasmodium, and mycobacterium tuberculosis, in rhesus macaques has generated extraordinary levels of protective immunity against subsequent pathogenic challenge. Moreover, the mechanisms of immune protection have altered paradigms about viral vector-mediated immunity against ectopically expressed vaccine antigens. Further optimization of CMV-vectored vaccines, particularly as this approach moves to human clinical trials will be augmented by a more complete understanding of how CMV engenders mechanisms of immune protection. This review summarizes the particulars of the specific CMV vaccine vector that has been used to date (rhesus CMV strain 68-1) in relation to CMV natural history.
Collapse
|
18
|
Magden ER, Nehete BP, Chitta S, Williams LE, Simmons JH, Abee CR, Nehete PN. Comparative Analysis of Cellular Immune Responses in Conventional and SPF Olive Baboons ( Papio anubis). Comp Med 2020; 70:160-169. [PMID: 32014083 DOI: 10.30802/aalas-cm-19-000035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Olive baboons (P. anubis) have provided a useful model of human diseases and conditions, including cardiac, respiratory, and infectious diseases; diabetes; and involving genetics, immunology, aging, and xenotransplantation. The development of a immunologically defined SPF baboons has advanced research further, especially for studies involving the immune system and immunosuppression. In this study, we compare normal immunologic changes of PBMC subsets, and their function in age-matched conventional and SPF baboons. Our results revealed that both groups have comparable numbers of different lymphocyte subsets, but phenotypic differences in central and effector memory T-cell subsets are more pronounced in CD4+ T cells. Despite equal proportions of CD3+ T cells among the conventional and SPF baboons, PBMC from the conventional group showed greater proliferative responses to phytohemagglutinin and pokeweed mitogen and higher numbers of IFNγ-producing cells after stimulation with concanavalin A or pokeweed mitogen, whereas plasma levels of the inflammatory cytokine TNFα were significantly higher in SPF baboons. Exposure of PBMC from conventional baboons to various Toll-like (TLR) ligands, including TLR3, TLR4, and TLR8, yielded increased numbers of IFNγ producing cells, whereas PBMC from SPF baboons stimulated with TLR5 or TLR6 ligand had more IFNγ-producing cells. These findings suggest that although lymphocyte subsets share many phenotypic and functional similarities in conventional and SPF baboons, specific differences in the immune function of lymphocytes could differentially influence the quality and quantity of their innate and adaptive immune responses. These differences should be considered in interpreting experimental outcomes, specifically in studies measuring immunologic endpoints.
Collapse
Affiliation(s)
- Elizabeth R Magden
- The University of Texas MD Anderson Cancer Center Bastrop, Department of Comparative Medicine, Houston, Texas
| | - Bharti P Nehete
- The University of Texas MD Anderson Cancer Center Bastrop, Department of Comparative Medicine, Houston, Texas;,
| | - Sriram Chitta
- The University of Texas MD Anderson Cancer Center Bastrop, Department of Comparative Medicine, Houston, Texas
| | - Lawrence E Williams
- The University of Texas MD Anderson Cancer Center Bastrop, Department of Comparative Medicine, Houston, Texas
| | - Joe H Simmons
- The University of Texas MD Anderson Cancer Center Bastrop, Department of Comparative Medicine, Houston, Texas
| | - Christian R Abee
- The University of Texas MD Anderson Cancer Center Bastrop, Department of Comparative Medicine, Houston, Texas
| | - Pramod N Nehete
- The University of Texas MD Anderson Cancer Center Bastrop, Department of Comparative Medicine, Houston, Texas; The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| |
Collapse
|
19
|
Vera Cruz D, Nelson CS, Tran D, Barry PA, Kaur A, Koelle K, Permar SR. Intrahost cytomegalovirus population genetics following antibody pretreatment in a monkey model of congenital transmission. PLoS Pathog 2020; 16:e1007968. [PMID: 32059027 PMCID: PMC7046290 DOI: 10.1371/journal.ppat.1007968] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 02/27/2020] [Accepted: 12/02/2019] [Indexed: 12/12/2022] Open
Abstract
Human cytomegalovirus (HCMV) infection is the leading non-genetic cause of congenital birth defects worldwide. While several studies have addressed the genetic composition of viral populations in newborns diagnosed with HCMV, little is known regarding mother-to-child viral transmission dynamics and how therapeutic interventions may impact within-host viral populations. Here, we investigate how preexisting CMV-specific antibodies shape the maternal viral population and intrauterine virus transmission. Specifically, we characterize the genetic composition of CMV populations in a monkey model of congenital CMV infection to examine the effects of passively-infused hyperimmune globulin (HIG) on viral population genetics in both maternal and fetal compartments. In this study, 11 seronegative, pregnant monkeys were challenged with rhesus CMV (RhCMV), including a group pretreated with a standard potency HIG preparation (n = 3), a group pretreated with a high-neutralizing potency HIG preparation (n = 3), and an untreated control group (n = 5). Targeted amplicon deep sequencing of RhCMV glycoprotein B and L genes revealed that one of the three strains present in the viral inoculum (UCD52) dominated maternal and fetal viral populations. We identified minor haplotypes of this strain and characterized their dynamics. Many of the identified haplotypes were consistently detected at multiple timepoints within sampled maternal tissues, as well as across tissue compartments, indicating haplotype persistence over time and transmission between maternal compartments. However, haplotype numbers and diversity levels were not appreciably different between control, standard-potency, and high-potency pretreatment groups. We found that while the presence of maternal antibodies reduced viral load and congenital infection, it had no apparent impact on intrahost viral genetic diversity at the investigated loci. Interestingly, some minor haplotypes present in fetal and maternal-fetal interface tissues were also identified as minor haplotypes in corresponding maternal tissues, providing evidence for a loose RhCMV mother-to-fetus transmission bottleneck even in the presence of preexisting antibodies.
Collapse
Affiliation(s)
- Diana Vera Cruz
- Computational Biology and Bioinformatics program / Duke Center for Genomic and Computational Biology, Duke University, Durham, North Carolina, United States of America
| | - Cody S. Nelson
- Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Dollnovan Tran
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, United States of America
| | - Peter A. Barry
- Center for Comparative Medicine, Department of Pathology and Laboratory Medicine, University of California, Davis, California, United States of America
| | - Amitinder Kaur
- Tulane National Primate Research Center, Tulane University, Covington, Louisiana, United States of America
| | - Katia Koelle
- Department of Biology, Emory University, Atlanta, Georgia, United States of America
| | - Sallie R. Permar
- Human Vaccine Institute, Duke University School of Medicine, Durham, North Carolina, United States of America
| |
Collapse
|
20
|
Zheng L, Li H, Fu L, Liu S, Yan Q, Leng SX. Blocking cellular N-glycosylation suppresses human cytomegalovirus entry in human fibroblasts. Microb Pathog 2020; 138:103776. [PMID: 31600539 DOI: 10.1016/j.micpath.2019.103776] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/02/2019] [Accepted: 10/04/2019] [Indexed: 12/21/2022]
Abstract
N-glycosylation plays an important role in the pathogenesis of viral infections. However, the role of host cell N-glycosylation in human cytomegalovirus (hCMV) infection remains to be elucidated. In this study, we found that blocking or removal of cellular N-glycosylation by tunicamycin, peptide-N-glycosidase F (PNGase F) treatment, or N-acetylglucosaminyltransferase I (MGAT1) knockdown resulted in suppression of hCMV infection in human fibroblasts. This suppression was reversed following N-glycosylation restoration. Immunofluorescence and flow cytometry analysis showed that blockade of cellular N-glycosylation interfered with hCMV entry rather than binding. Removal of N-glycosylation on epidermal growth factor (EGFR) and integrin β3, two proposed hCMV receptors, blocked their interaction with hCMV glycoproteins B and H. It also suppressed activation of these receptors and downstream integrin β3/Src signaling. Taken together, these results suggest that N-glycosylation of host cell glycoproteins including two proposed hCMV receptors is critical for hCMV entry rather than attachment. They provide novel insights into the biological process important for the early stage of hCMV infection with potential therapeutic implications.
Collapse
Affiliation(s)
- Luping Zheng
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Laboratory of Glycobiology and Glycoengineering, Dalian, Liaoning Province, China
| | - Huifen Li
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Li Fu
- Institute of Dalian Fusheng Natural Medicine, Development District, Dalian, Liaoning Province, China
| | - Sally Liu
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Qiu Yan
- Department of Biochemistry and Molecular Biology, Dalian Medical University, Liaoning Provincial Core Laboratory of Glycobiology and Glycoengineering, Dalian, Liaoning Province, China.
| | - Sean X Leng
- Division of Geriatric Medicine and Gerontology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
21
|
Merino KM, Slisarenko N, Taylor JM, Falkenstein KP, Gilbert MH, Bohm RP, Blanchard JL, Ardeshir A, Didier ES, Kim WK, Kuroda MJ. Clinical and Immunological Metrics During Pediatric Rhesus Macaque Development. Front Pediatr 2020; 8:388. [PMID: 32766187 PMCID: PMC7378395 DOI: 10.3389/fped.2020.00388] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 06/08/2020] [Indexed: 12/18/2022] Open
Abstract
Background: Clinical measurements commonly used to evaluate overall health of laboratory animals including complete blood count, serum chemistry, weight, and immunophenotyping, differ with respect to age, development, and environment. This report provides comprehensive clinical and immunological reference ranges for pediatric rhesus macaques over the first year of life. Methods: We collected and analyzed blood samples from 151 healthy rhesus macaques, aged 0-55 weeks, and compared mother-reared infants to two categories of nursery-reared infants; those on an active research protocol and those under derivation for the expanded specific-pathogen-free breeding colony. Hematology was performed on EDTA-anticoagulated blood using a Sysmex XT2000i, and serum clinical chemistry was performed using the Beckman AU480 chemistry analyzer. Immunophenotyping of whole blood was performed with immunofluorescence staining and subsequent flow cytometric analysis on a BD LSRFortessa. Plasma cytokine analysis was performed using a Millipore multiplex Luminex assay. Results: For hematological and chemistry measurements, pediatric reference ranges deviate largely from adults. Comparison of mother-reared and nursery-reared animals revealed that large differences depend on rearing conditions and diet. Significant differences found between two nursery-reared cohorts (research and colony animals) indicate large influences of experimental factors and anesthetic events on these parameters. Immune cells and cytokine responses presented with distinct patterns for infants depending on age, birth location, and rearing conditions. Conclusions: Our results illustrate how the immune system changed over time and that there was variability among pediatric age groups. Reference ranges of results reported here will support interpretations for how infection and treatment may skew common immune correlates used for assessment of pathology or protection in research studies as well as help veterinarians in the clinical care of infant non-human primates. We highlighted the importance of using age-specific reference comparisons for pediatric studies and reiterated the utility of rhesus macaques as a model for human studies. Given the rapid transformation that occurs in multiple tissue compartments after birth and cumulative exposures to antigens as individuals grow, a better understanding of immunological development and how this relates to timing of infection or vaccination will support optimal experimental designs for developing vaccines and treatment interventions.
Collapse
Affiliation(s)
- Kristen M Merino
- Division of Immunology, Tulane National Primate Research Center, Covington, LA, United States.,Walter Reed Army Institute of Research, National Academy of Sciences, Engineering and Medicine Fellow, Silver Spring, MD, United States
| | - Nadia Slisarenko
- Division of Immunology, Tulane National Primate Research Center, Covington, LA, United States
| | - Joshua M Taylor
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, United States
| | - Kathrine P Falkenstein
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, United States
| | - Margaret H Gilbert
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, United States
| | - Rudolf P Bohm
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, United States
| | - James L Blanchard
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, LA, United States
| | - Amir Ardeshir
- California National Primate Research Center, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Elizabeth S Didier
- Center for Immunology and Infectious Diseases, California National Primate Research Center, University of California, Davis, Davis, CA, United States.,Division of Microbiology, Tulane National Primate Research Center, Covington, LA, United States
| | - Woong-Ki Kim
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Marcelo J Kuroda
- Division of Immunology, Tulane National Primate Research Center, Covington, LA, United States.,Division of Microbiology, Tulane National Primate Research Center, Covington, LA, United States
| |
Collapse
|
22
|
Rubino G, Bulati M, Aiello A, Aprile S, Gambino CM, Gervasi F, Caruso C, Accardi G. Sicilian centenarian offspring are more resistant to immune ageing. Aging Clin Exp Res 2019; 31:125-133. [PMID: 29594822 DOI: 10.1007/s40520-018-0936-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 03/21/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Immunosenescence constitutes a major indirect cause of morbidity and mortality in the elderly. Previous analysis of immune signatures in a cohort of centenarian offspring showed an intermediate immunophenotype between age-matched and younger controls. AIMS To confirm and extend the previous studies performing further phenotypical analysis in centenarian offspring and controls. METHODS Analysis of Treg cells, γδ T cells, mucosal-associated invariant T cells, and senescent immune T cells was performed in centenarian offspring and controls. RESULTS We report significant differences between elderly and centenarian offspring in most of the studied subsets, showing that centenarian offspring subsets present an intermediate phenotyping between elderly and younger people. CONCLUSION The whole present data confirm and extend the previous results showing that centenarian offspring retain more youthful immunological parameters and that the exhaustion of the immune system is less evident than in elderly without centenarian parents, though further investigations are warranted.
Collapse
|
23
|
Subclinical Cytomegalovirus Infection Is Associated with Altered Host Immunity, Gut Microbiota, and Vaccine Responses. J Virol 2018; 92:JVI.00167-18. [PMID: 29669841 DOI: 10.1128/jvi.00167-18] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/14/2018] [Indexed: 12/15/2022] Open
Abstract
Subclinical viral infections (SVI), including cytomegalovirus (CMV), are highly prevalent in humans, resulting in lifelong persistence. However, the impact of SVI on the interplay between the host immunity and gut microbiota in the context of environmental exposures is not well defined. We utilized the preclinical nonhuman primate (NHP) model consisting of SVI-free (specific-pathogen-free [SPF]) rhesus macaques and compared them to the animals with SVI (non-SPF) acquired through natural exposure and investigated the impact of SVI on immune cell distribution and function, as well as on gut microbiota. These changes were examined in animals housed in the outdoor environment compared to the controlled indoor environment. We report that SVI are associated with altered immune cell subsets and gut microbiota composition in animals housed in the outdoor environment. Non-SPF animals harbored a higher proportion of potential butyrate-producing Firmicutes and higher numbers of lymphocytes, effector T cells, and cytokine-producing T cells. Surprisingly, these differences diminished following their transfer to the controlled indoor environment, suggesting that non-SPFs had increased responsiveness to environmental exposures. An experimental infection of indoor SPF animals with CMV resulted in an increased abundance of butyrate-producing bacteria, validating that CMV enhanced colonization of butyrate-producing commensals. Finally, non-SPF animals displayed lower antibody responses to influenza vaccination compared to SPF animals. Our data show that subclinical CMV infection heightens host immunity and gut microbiota changes in response to environmental exposures. This may contribute to the heterogeneity in host immune response to vaccines and environmental stimuli at the population level.IMPORTANCE Humans harbor several latent viruses that modulate host immunity and commensal microbiota, thus introducing heterogeneity in their responses to pathogens, vaccines, and environmental exposures. Most of our understanding of the effect of CMV on the immune system is based on studies of children acquiring CMV or of immunocompromised humans with acute or reactivated CMV infection or in ageing individuals. The experimental mouse models are genetically inbred and are completely adapted to the indoor laboratory environment. In contrast, nonhuman primates are genetically outbred and are raised in the outdoor environment. Our study is the first to report the impact of long-term subclinical CMV infection on host immunity and gut microbiota, which is evident only in the outdoor environment but not in the indoor environment. The significance of this study is in highlighting the impact of SVI on enhancing host immune susceptibility to environmental exposures and immune heterogeneity.
Collapse
|
24
|
Sparger EE, Murphy BG, Kamal FM, Arzi B, Naydan D, Skouritakis CT, Cox DP, Skorupski K. Investigation of immune cell markers in feline oral squamous cell carcinoma. Vet Immunol Immunopathol 2018; 202:52-62. [PMID: 30078599 DOI: 10.1016/j.vetimm.2018.06.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 05/04/2018] [Accepted: 06/13/2018] [Indexed: 01/22/2023]
Abstract
Squamous cell carcinoma is the most common oral cancer in the cat and presents as a locally aggressive lesion for which an effective therapeutic protocol remains elusive. Feline oral squamous cell carcinoma (OSCC) shares many clinical characteristics with human head and neck squamous cell carcinoma (HNSCC). Accordingly, present studies were conducted to determine similarities for immune markers shared by feline OSCC and human HNSCC. Biopsies harvested from a feline patient cohort-1 (n = 12) were analyzed for lymphoid cell infiltrates by immunohistochemistry (IHC). Results revealed unique patterns of T cell infiltration involving both neoplastic epithelium and stroma that were detected in most patient tumor biopsies (92%) examined by IHC staining for CD3. Intratumoral B cell infiltrates were detected within tumor stroma only, based on IHC staining for CD79a and CD20 for all patients within the same cohort-1. Infiltration of tumors by a regulatory CD4 T cell subset (Tregs) defined by expression of the forkhead transcription factor FoxP3, was also detected in biopsies from 57% of patients and involved infiltration of neoplastic epithelium and stroma. Patient biopsies were also examined for expression of immunomodulator cyclooxygenase (COX)-2 and revealed positive but weak staining of neoplastic epithelium in a significant proportion of cases (75%). Interestingly, COX-2 expression was detected in both neoplastic epithelium and stroma. Blood collected from a second cohort of feline OSCC patients (n = 9) revealed an increased frequency of circulating CD4+FoxP3+ T cells when compared to healthy adult controls (n = 7) (P = 0.045), although frequencies of CD4+CD25+FoxP3+ T cells were comparable between patients and healthy pet cat controls. Lastly, biopsies from feline OSCC patients were characterized for histologic subtype using a classification scheme previously described for human HNSCC. This analysis revealed the conventional subtype as the predominant variant (75%) with conventional subtypes split evenly between well differentiated and moderately differentiated carcinomas. Two cases were classified as papillary and one case as basaloid subtypes. Correlations between subtype, immune marker scores or circulating Treg frequencies and clinical characteristics or outcome were not detected, most likely due to small patient numbers within patient cohorts. However, findings from these studies provide a preliminary step in the characterization of immune and histologic markers that will be critical to defining prognostic immune markers for feline OSCC and potential targets for testing of immunotherapeutics also relevant to human HNSCC in future studies.
Collapse
Affiliation(s)
- Ellen E Sparger
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, CA, USA.
| | - Brian G Murphy
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Farina Mustaffa Kamal
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Boaz Arzi
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Diane Naydan
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Chrisoula T Skouritakis
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Darren P Cox
- Department of Orofacial Sciences, University of California, San Francisco, CA, USA
| | - Katherine Skorupski
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA, USA
| |
Collapse
|
25
|
Amedee AM, Phillips B, Jensen K, Robichaux S, Lacour N, Burke M, Piatak M, Lifson JD, Kozlowski PA, Van Rompay KK, De Paris K. Early Sites of Virus Replication After Oral SIV mac251 Infection of Infant Macaques: Implications for Pathogenesis. AIDS Res Hum Retroviruses 2018; 34:286-299. [PMID: 29237287 DOI: 10.1089/aid.2017.0169] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Despite optimization of preventative measures for vertical HIV-1 transmission, daily, roughly 400 infants become HIV infected, most of them through breastfeeding. Viral entry has been presumed to occur in the gastrointestinal tract; however, the exact entry site(s) have not been defined. Therefore, we quantified simian immunodeficiency virus (SIV) RNA and DNA in oral, intestinal, and systemic tissues of 15 infant macaques within 48-96 h after oral SIVmac251 exposure. SIV DNA was detected as early as 48 h, whereas SIV RNA was typically detected at later time points (72-96 h). Transmitted founder viruses were identical or very similar to a single genotype in the SIVmac251 challenge stock. SIV RNA and DNA were most frequently found in lymph nodes (LNs) draining the oral cavity and in the ileum. Using in situ hybridization, SIV-infected cells in LNs were exclusively represented by CD3+ T cells. SIV RNA and DNA were also detected in the lungs of 20% of the animals, and 60% of the animals had detectable SIV DNA in the cerebrum. The early detection of viral RNA or DNA in lung and brain tissues emphasizes the need for early treatment of pediatric HIV infection to prevent damage not only to the immune system but also to the respiratory tract and central nervous system.
Collapse
Affiliation(s)
- Angela M. Amedee
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Bonnie Phillips
- Department of Microbiology and Immunology and Center for AIDS Research, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Kara Jensen
- Department of Microbiology and Immunology and Center for AIDS Research, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Spencer Robichaux
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Nedra Lacour
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Mark Burke
- Howard University, Washington, District of Columbia
| | - Michael Piatak
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Pamela A. Kozlowski
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Koen K.A. Van Rompay
- California National Primate Research Center, University of California, Davis, Davis, California
| | - Kristina De Paris
- Department of Microbiology and Immunology and Center for AIDS Research, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
26
|
Modeling aging in HIV infection in nonhuman primates to address an emerging challenge of the post-ART era. Curr Opin Virol 2017; 25:66-75. [PMID: 28803049 DOI: 10.1016/j.coviro.2017.07.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/17/2017] [Accepted: 07/18/2017] [Indexed: 12/22/2022]
Abstract
The advent of antiretroviral therapy (ART) has dramatically improved both quality and length of life for subjects infected with human immunodeficiency virus (HIV), delaying or preventing progression to acquired immunodeficiency syndrome (AIDS). However, the virus induces aging-related changes to the immune system which confound treatment. Additionally, the normal physiologic events that occur during aging lead to deficiencies in immunity which not only exacerbate HIV pathogenesis but also trigger a variety of comorbidities. Here, the synergistic linkage between aging and HIV infection is examined in regard to the immunological and pathological mechanisms that drive both senescence and disease progression. The use of NHPs to investigate potential therapeutic strategies to control the deleterious consequences of aging with HIV infection is also reviewed.
Collapse
|
27
|
Bulati M, Caruso C, Colonna-Romano G. From lymphopoiesis to plasma cells differentiation, the age-related modifications of B cell compartment are influenced by "inflamm-ageing". Ageing Res Rev 2017; 36:125-136. [PMID: 28396185 DOI: 10.1016/j.arr.2017.04.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/04/2017] [Accepted: 04/05/2017] [Indexed: 12/26/2022]
Abstract
Ageing is a complex process characterized by a general decline in physiological functions with increasing morbidity and mortality. The most important aspect of ageing is the chronic inflammatory status, named "inflamm-ageing", strictly associated with the deterioration of the immune function, termed "immunosenescence". Both are causes of increased susceptibility of elderly to infectious diseases, cancer, dementia, cardiovascular diseases and autoimmunity, and of a decreased response to vaccination. It has been widely demonstrated that ageing has a strong impact on the remodelling of the B cell branch of immune system. The first evident effect is the significant decrease in circulating B cells, primarily due to the reduction of new B cell coming from bone marrow (BM) progenitors, as inflammation directly impacts on B lymphopoiesis. Besides, in aged individuals, there is a shift from naïve to memory immunoglobulins production, accompanied by the impaired ability to produce high affinity protective antibodies against newly encountered antigens. This is accompanied by the increase of expanded clones of B cells, which correlates with poor health status. Age-related modifications also occur in naïve/memory B cells subsets. Indeed, in the elderly, there is a reduction of naïve B cells, accompanied by the expansion of memory B cells that show a senescence-associated phenotype. Finally, elderly show the impaired ability of memory B cells to differentiate into plasma cells. It can be concluded that inflammation is the leading cause of the age-related impairment of B cell compartment, which play certainly a key role in the development of age-related diseases. This makes study of B cells in the aged an important tool for monitoring immunosenescence, chronic inflammatory disorders and the effectiveness of vaccines or pharmacological therapies.
Collapse
|
28
|
Abstract
Human cytomegalovirus (HCMV) encodes numerous proteins and microRNAs that function to evade the immune response and allow the virus to replicate and disseminate in the face of a competent innate and acquired immune system. The establishment of a latent infection by CMV, which if completely quiescent at the level of viral gene expression would represent an ultimate in immune evasion strategies, is not sufficient for lifelong persistence and dissemination of the virus. CMV needs to reactivate and replicate in a lytic cycle of infection in order to disseminate further, which occurs in the face of a fully primed secondary immune response. Without reactivation, latency itself would be redundant for the virus. It is also becoming clear that latency is not a totally quiescent state, but is characterized by limited viral gene expression. Therefore, the virus also needs immune evasion strategies during latency. An effective immune response to CMV is required or viral replication will cause morbidity and ultimately mortality in the host. There is clearly a complex balance between virus immune evasion and host immune recognition over a lifetime. This poses the important question of whether long-term evasion or manipulation of the immune response driven by CMV is detrimental to health. In this meeting report, three groups used the murine model of CMV (MCMV) to examine if the contribution of the virus to immune senescence is set by the (i) initial viral inoculum, (ii) inflation of T cell responses, (iii) or the balance between functionally distinct effector CD4+ T cells. The work of other groups studying the CMV response in humans is discussed. Their work asks whether the ability to make immune responses to new antigens is compromised by (i) age and HCMV carriage, (ii) long-term exposure to HCMV giving rise to an overall immunosuppressive environment and increased levels of latent virus, or (iii) adapted virus mutants (used as potential vaccines) that have the capacity to elicit conventional and unconventional T cell responses.
Collapse
|
29
|
Chang WLW, Gonzalez DF, Kieu HT, Castillo LD, Messaoudi I, Shen X, Tomaras GD, Shacklett BL, Barry PA, Sparger EE. Changes in Circulating B Cell Subsets Associated with Aging and Acute SIV Infection in Rhesus Macaques. PLoS One 2017; 12:e0170154. [PMID: 28095513 PMCID: PMC5240950 DOI: 10.1371/journal.pone.0170154] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 12/29/2016] [Indexed: 12/21/2022] Open
Abstract
Aging and certain viral infections can negatively impact humoral responses in humans. To further develop the nonhuman primate (NHP) model for investigating B cell dynamics in human aging and infectious disease, a flow cytometric panel was developed to characterize circulating rhesus B cell subsets. Significant differences between human and macaque B cells included the proportions of cells within IgD+ and switched memory populations and a prominent CD21-CD27+ unswitched memory population detected only in macaques. We then utilized the expanded panel to analyze B cell alterations associated with aging and acute simian immunodeficiency virus (SIV) infection in the NHP model. In the aging study, distinct patterns of B cell subset frequencies were observed for macaques aged one to five years compared to those between ages 5 and 30 years. In the SIV infection study, B cell frequencies and absolute number were dramatically reduced following acute infection, but recovered within four weeks of infection. Thereafter, the frequencies of activated memory B cells progressively increased; these were significantly correlated with the magnitude of SIV-specific IgG responses, and coincided with impaired maturation of anti-SIV antibody avidity, as previously reported for HIV-1 infection. These observations further validate the NHP model for investigation of mechanisms responsible for B cells alterations associated with immunosenescence and infectious disease.
Collapse
Affiliation(s)
- W. L. William Chang
- Center for Comparative Medicine, University of California Davis, Davis, California, United States of America
- * E-mail:
| | - Denise F. Gonzalez
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Hung T. Kieu
- Center for Comparative Medicine, University of California Davis, Davis, California, United States of America
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Luis D. Castillo
- Center for Comparative Medicine, University of California Davis, Davis, California, United States of America
| | - Ilhem Messaoudi
- Department of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, California, United States of America
| | - Xiaoying Shen
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Georgia D. Tomaras
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Barbara L. Shacklett
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, California, United States of America
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of California Davis, Davis, California, United States of America
| | - Peter A. Barry
- Center for Comparative Medicine, University of California Davis, Davis, California, United States of America
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Davis, Davis, California, United States of America
- California National Primate Research Center, University of California Davis, Davis, California, United States of America
| | - Ellen E. Sparger
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| |
Collapse
|
30
|
Jensen K, Dela Pena-Ponce MG, Piatak M, Shoemaker R, Oswald K, Jacobs WR, Fennelly G, Lucero C, Mollan KR, Hudgens MG, Amedee A, Kozlowski PA, Estes JD, Lifson JD, Van Rompay KKA, Larsen M, De Paris K. Balancing Trained Immunity with Persistent Immune Activation and the Risk of Simian Immunodeficiency Virus Infection in Infant Macaques Vaccinated with Attenuated Mycobacterium tuberculosis or Mycobacterium bovis BCG Vaccine. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:e00360-16. [PMID: 27655885 PMCID: PMC5216431 DOI: 10.1128/cvi.00360-16] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 09/12/2016] [Indexed: 12/14/2022]
Abstract
Our goal is to develop a pediatric combination vaccine to protect the vulnerable infant population against human immunodeficiency virus type 1 (HIV-1) and tuberculosis (TB) infections. The vaccine consists of an auxotroph Mycobacterium tuberculosis strain that coexpresses HIV antigens. Utilizing an infant rhesus macaque model, we have previously shown that this attenuated M. tuberculosis (AMtb)-simian immunodeficiency virus (SIV) vaccine is immunogenic, and although the vaccine did not prevent oral SIV infection, a subset of vaccinated animals was able to partially control virus replication. However, unexpectedly, vaccinated infants required fewer SIV exposures to become infected compared to naive controls. Considering that the current TB vaccine, Mycobacterium bovis bacillus Calmette-Guérin (BCG), can induce potent innate immune responses and confer pathogen-unspecific trained immunity, we hypothesized that an imbalance between enhanced myeloid cell function and immune activation might have influenced the outcome of oral SIV challenge in AMtb-SIV-vaccinated infants. To address this question, we used archived samples from unchallenged animals from our previous AMtb-SIV vaccine studies and vaccinated additional infant macaques with BCG or AMtb only. Our results show that vaccinated infants, regardless of vaccine strain or regimen, had enhanced myeloid cell responses. However, CD4+ T cells were concurrently activated, and the persistence of these activated target cells in oral and/or gastrointestinal tissues may have facilitated oral SIV infection. Immune activation was more pronounced in BCG-vaccinated infant macaques than in AMtb-vaccinated infant macaques, indicating a role for vaccine attenuation. These findings underline the importance of understanding the interplay of vaccine-induced immunity and immune activation and its effect on HIV acquisition risk and outcome in infants.
Collapse
Affiliation(s)
- Kara Jensen
- Department of Microbiology and Immunology and Center for AIDS Research, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Myra Grace Dela Pena-Ponce
- Department of Microbiology and Immunology and Center for AIDS Research, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Michael Piatak
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Rebecca Shoemaker
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Kelli Oswald
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | | | - Glenn Fennelly
- Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Carissa Lucero
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Katie R Mollan
- Lineberger Cancer Center and Center for AIDS Research, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Michael G Hudgens
- Gillings School of Global Public Health and Center for AIDS Research, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Angela Amedee
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Pamela A Kozlowski
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Jacob D Estes
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Jeffrey D Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Koen K A Van Rompay
- California National Primate Research Center, University of California, Davis, Davis, California, USA
| | - Michelle Larsen
- Albert Einstein College of Medicine, New York, New York, USA
| | - Kristina De Paris
- Department of Microbiology and Immunology and Center for AIDS Research, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
31
|
Peverengo L, Prochetto E, Rodeles L, Valenzuela I, Marcipar IS, Bottasso O, Vicco MH. Antibody profiles induced by Trypanosoma cruzi in chagasic patients with previous or current exposure to mycobacteria. Pathog Dis 2016; 74:ftw109. [PMID: 27815312 DOI: 10.1093/femspd/ftw109] [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] [Revised: 07/31/2016] [Accepted: 11/02/2016] [Indexed: 11/12/2022] Open
Abstract
Since the immune response mounted by the host to a particular microorganism might be influenced by the acquired immunological experience due to previous contact with other microorganisms, we performed a cross-sectional study to explore the pattern of Trypanosoma cruzi infection-related antibodies in T. cruzi-seropositive individuals presenting concomitant tuberculosis, or the antecedent of BCG vaccination. Sampled individuals were grouped as follows: patients with Chagas disease, not vaccinated with BCG, who further developed pulmonary tuberculosis; individuals with Chagas disease, BCG-vaccinated; and subjects with Chagas disease, presenting neither BCG scar nor tuberculosis disease. Non-vaccinated individuals or without tuberculosis, presented the highest values of anti-PH (P < 0.001), anti-FRA (P < 0.001), anti-p2β (P = 0.0023) and anti-B13 (P < 0.001) antibodies. The present findings constitute the first demonstration of the potential influence of concomitant tuberculosis on Chagas disease.
Collapse
Affiliation(s)
- Luz Peverengo
- Laboratorio de Tecnología Inmunológica, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Estefanía Prochetto
- Laboratorio de Tecnología Inmunológica, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Luz Rodeles
- Laboratorio de Tecnología Inmunológica, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina.,Facultad de Ciencias Médicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Ignacio Valenzuela
- Facultad de Ciencias Médicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Iván Sergio Marcipar
- Laboratorio de Tecnología Inmunológica, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Oscar Bottasso
- Instituto de Inmunología Clínica y Experimental de Rosario, UNR-CONICET, Santa Fe, Argentina
| | - Miguel Hernán Vicco
- Laboratorio de Tecnología Inmunológica, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, Santa Fe, Argentina .,Facultad de Ciencias Médicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| |
Collapse
|
32
|
Exploitation of Interleukin-10 (IL-10) Signaling Pathways: Alternate Roles of Viral and Cellular IL-10 in Rhesus Cytomegalovirus Infection. J Virol 2016; 90:9920-9930. [PMID: 27558431 DOI: 10.1128/jvi.00635-16] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 08/18/2016] [Indexed: 12/16/2022] Open
Abstract
There is accumulating evidence that the viral interleukin-10 (vIL-10) ortholog of both human and rhesus cytomegalovirus (HCMV and RhCMV, respectively) suppresses the functionality of cell types that are critical to contain virus dissemination and help shape long-term immunity during the earliest virus-host interactions. In particular, exposure of macrophages, peripheral blood mononuclear cells, monocyte-derived dendritic cells, and plasmacytoid dendritic cells to vIL-10 suppresses multiple effector functions including, notably, those that link innate and adaptive immune responses. Further, vaccination of RhCMV-uninfected rhesus macaques with nonfunctional forms of RhCMV vIL-10 greatly restricted parameters of RhCMV infection following RhCMV challenge of the vaccinees. Vaccinees exhibited significantly reduced shedding of RhCMV in saliva and urine following RhCMV challenge compared to shedding in unvaccinated controls. Based on the evidence that vIL-10 is critical during acute infection, the role of vIL-10 during persistent infection was analyzed in rhesus macaques infected long term with RhCMV to determine whether postinfection vaccination against vIL-10 could change the virus-host balance. RhCMV-seropositive macaques, which shed RhCMV in saliva, were vaccinated with nonfunctional RhCMV vIL-10, and shedding levels of RhCMV in saliva were evaluated. Following robust increases in vIL-10-binding and vIL-10-neutralizing antibodies, shedding levels of RhCMV modestly declined, consistent with the interpretation that vIL-10 may play a functional role during persistent infection. However, a more significant association was observed between the levels of cellular IL-10 secreted in peripheral blood mononuclear cells exposed to RhCMV antigens and shedding of RhCMV in saliva. This result implies that RhCMV persistence is associated with the induction of cellular IL-10 receptor-mediated signaling pathways. IMPORTANCE Human health is adversely impacted by viruses that establish lifelong infections that are often accompanied with increased morbidity and mortality (e.g., infections with HIV, hepatitis C virus, or human cytomegalovirus). A longstanding but unfulfilled goal has been to develop postinfection vaccine strategies that could "reboot" the immune system of an infected individual in ways that would enable the infected host to develop immune responses that clear reservoirs of persistent virus infection, effectively curing the host of infection. This concept was evaluated in rhesus macaques infected long term with rhesus cytomegalovirus by repeatedly immunizing infected animals with nonfunctional versions of the rhesus cytomegalovirus-encoded viral interleukin-10 immune-modulating protein. Following vaccine-mediated boosting of antibody titers to viral interleukin-10, there was modest evidence for increased immunological control of the virus following vaccination. More significantly, data were also obtained that indicated that rhesus cytomegalovirus is able to persist due to upregulation of the cellular interleukin-10 signaling pathway.
Collapse
|