1
|
Balamayooran G, Tooze JA, Gardin JF, Long MC, Caudell DL, Cline JM, Kock ND, Paitsel M, Moore S, Jorgensen MJ. Age and sex associated organ weight differences in vervets/African green monkeys (Chlorocebus aethiops sabaeus). J Med Primatol 2024; 53:e12721. [PMID: 39048121 PMCID: PMC11378953 DOI: 10.1111/jmp.12721] [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: 04/24/2024] [Revised: 06/03/2024] [Accepted: 06/17/2024] [Indexed: 07/27/2024]
Abstract
AbstractBackgroundAfrican green monkeys (AGMs, also known as vervets, Cholorocebus aethiops sabaeus) have been used in a variety of biomedical research studies. The aim of this study was to generate a reference for normal organ weights and percentage organ weights in AGMs of different age categories and sex.MethodsThe organ weights were compiled from 479 AGMs (285 females and 194 males) from 2004 to 2021. Age and sex differences of absolute and relative organ weights were analyzed using analysis of variance.ResultsThe findings demonstrate that males had higher body and organ weights than age‐matched females, but relative organ weights did not differ between males and females. At maturity, adrenal gland, brain, kidney, liver, thymus, and thyroid gland weights as a percentage of body weight declined, but relative weights of prostate gland, testes, and uterus were higher.ConclusionThese data should be beneficial to biomedical researchers and pathologists working with AGMs.
Collapse
Affiliation(s)
- Gayathriy Balamayooran
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Janet A Tooze
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Jean F Gardin
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Margaret C Long
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - David L Caudell
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - J Mark Cline
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Nancy D Kock
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Monica Paitsel
- Animal Resources Program, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Stacy Moore
- Animal Resources Program, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Matthew J Jorgensen
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
2
|
Moyo-Gwete T, Ayres F, Mzindle NB, Makhado Z, Manamela NP, Richardson SI, Kitchin D, van Graan S, van Heerden J, Parbhoo N, Chege GK, Moore PL. Evaluating the antibody response elicited by diverse HIV envelope immunogens in the African green monkey (Vervet) model. Sci Rep 2024; 14:13311. [PMID: 38858452 PMCID: PMC11164991 DOI: 10.1038/s41598-024-63703-7] [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: 06/20/2023] [Accepted: 05/31/2024] [Indexed: 06/12/2024] Open
Abstract
African Green (Vervet) monkeys have been extensively studied to understand the pathogenesis of infectious diseases. Using vervet monkeys as pre-clinical models may be an attractive option for low-resourced areas as they are found abundantly and their maintenance is more cost-effective than bigger primates such as rhesus macaques. We assessed the feasibility of using vervet monkeys as animal models to examine the immunogenicity of HIV envelope trimer immunogens in pre-clinical testing. Three groups of vervet monkeys were subcutaneously immunized with either the BG505 SOSIP.664 trimer, a novel subtype C SOSIP.664 trimer, CAP255, or a combination of BG505, CAP255 and CAP256.SU SOSIP.664 trimers. All groups of vervet monkeys developed robust binding antibodies by the second immunization with the peak antibody response occurring after the third immunization. Similar to binding, antibody dependent cellular phagocytosis was also observed in all the monkeys. While all animals developed potent, heterologous Tier 1 neutralizing antibody responses, autologous neutralization was limited with only half of the animals in each group developing responses to their vaccine-matched pseudovirus. These data suggest that the vervet monkey model may yield distinct antibody responses compared to other models. Further study is required to further determine the utility of this model in HIV immunization studies.
Collapse
Affiliation(s)
- Thandeka Moyo-Gwete
- SA MRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
- Centre for HIV and STIs, National Institute or Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa.
| | - Frances Ayres
- SA MRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Centre for HIV and STIs, National Institute or Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa
| | - Nonkululeko B Mzindle
- SA MRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Centre for HIV and STIs, National Institute or Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa
| | - Zanele Makhado
- SA MRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Centre for HIV and STIs, National Institute or Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa
| | - Nelia P Manamela
- SA MRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Centre for HIV and STIs, National Institute or Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa
| | - Simone I Richardson
- SA MRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Centre for HIV and STIs, National Institute or Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa
| | - Dale Kitchin
- SA MRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Centre for HIV and STIs, National Institute or Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa
| | - Strauss van Graan
- SA MRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Centre for HIV and STIs, National Institute or Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa
| | - Joritha van Heerden
- Primate Unit and Delft Animal Centre, Centre and Platform Office, South African Medical Research Council, Cape Town, South Africa
| | - Nishal Parbhoo
- Department of Life and Consumer Sciences, College of Agriculture and Environmental Sciences, University of South Africa, Johannesburg, South Africa
| | - Gerald K Chege
- Primate Unit and Delft Animal Centre, Centre and Platform Office, South African Medical Research Council, Cape Town, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Penny L Moore
- SA MRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Centre for HIV and STIs, National Institute or Communicable Diseases (NICD) of the National Health Laboratory Service (NHLS), Johannesburg, South Africa
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| |
Collapse
|
3
|
Tang W, Liang P. The identification of retro-DNAs in primate genomes as DNA transposons mobilizing via retrotransposition. F1000Res 2024; 12:255. [PMID: 38915770 PMCID: PMC11195612 DOI: 10.12688/f1000research.130043.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/23/2024] [Indexed: 06/26/2024] Open
Abstract
Background Mobile elements (MEs) constitute a major portion of the genome in primates and other higher eukaryotes, and they play important role in genome evolution and gene function. MEs can be divided into two fundamentally different classes: DNA transposons which transpose in the genome in a "cut-and-paste" style, and retrotransposons which propagate in a "copy-and-paste" fashion via a process involving transcription and reverse-transcription. In primate genomes, DNA transposons are mostly dead, while many retrotransposons are still highly active. We report here the identification of a unique group of MEs, which we call "retro-DNAs", for their combined characteristics of these two fundamentally different ME classes. Methods A comparative computational genomic approach was used to analyze the reference genome sequences of 10 primate species consisting of five apes, four monkeys, and marmoset. Results From our analysis, we identified a total of 1,750 retro-DNAs, representing 748 unique insertion events in the genomes of ten primate species including human. These retro-DNAs contain sequences of DNA transposons but lack the terminal inverted repeats (TIRs), the hallmark of DNA transposons. Instead, they show characteristics of retrotransposons, such as polyA tails, longer target-site duplications (TSDs), and the "TT/AAAA" insertion site motif, suggesting the use of the L1-based target- primed reverse transcription (TPRT) mechanism. At least 40% of these retro-DNAs locate into genic regions, presenting potentials for impacting gene function. More interestingly, some retro-DNAs, as well as their parent sites, show certain levels of expression, suggesting that they have the potential to create more retro-DNA copies in the present primate genomes. Conclusions Although small in number, the identification of these retro-DNAs reveals a new mean for propagating DNA transposons in primate genomes without active canonical DNA transposon activity. Our data also suggest that the TPRT machinery may transpose a wider variety of DNA sequences in the genomes.
Collapse
Affiliation(s)
- Wangxiangfu Tang
- Department of Biological Sciences, Brock University, St. Catharines, Ontario, L2S 3A1, Canada
| | - Ping Liang
- Department of Biological Sciences, Brock University, St. Catharines, Ontario, L2S 3A1, Canada
- Centre of Biotechnology, Brock University, St. Catharines, Ontario, L2S 3A1, Canada
| |
Collapse
|
4
|
Pletcher JS, Zimmer JL, Liu CC, Beierschmitt A, Lewin AC. Ocular examination findings and selected ophthalmic diagnostic tests in African green monkeys (Chlorocebus aethiops sabaeus). Vet Ophthalmol 2024; 27:158-169. [PMID: 37442802 DOI: 10.1111/vop.13132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 06/26/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023]
Abstract
OBJECTIVE To document ocular lesions and establish ophthalmic diagnostic test reference values in a colony of African green monkeys (Chlorocebus aethiops sabaeus). ANIMALS STUDIED Fifty one geriatric (GAGM, 19-30 years old), 10 adult (AAGM, 5-9 years old) and 10 juvenile (JAGM, <2 years old) African green monkeys housed in a single Caribbean research colony. PROCEDURES Ocular biomicroscopy, indirect fundoscopy, Schirmer tear test (STT), rebound tonometry (TonoVet®) and corneal fluorescein staining were performed. Mixed ANCOVA tests were performed to compare STT and IOP between groups. RESULTS Common ocular lesions in GAGM included vitreal degeneration (27/51, 51/102 eyes) and cataracts (21/51, 32/102 eyes). Vitreal degeneration was also common in AAGM (8/10, 16/20 eyes) and infrequent in JAGM (3/10, 6/20 eyes). Cataracts were not present in any JAGM or AAGM. All eyes in all three groups had perilimbal corneal pigmentation and faint lace-like anterior corneal stromal opacification. Median (range) STT values were 16.0 (18) mm/min in GAGM. Mean (SD) STT values were 14.2 (4.6) mm/min in AAGM, and 8.9 (3.4) mm/min in JAGM. Median (range) IOP values were 16.5 (27) mmHg in GAGM. Mean (SD) IOP values were 18.0 (2.8) mmHg in AAGM, and 14.1 (2.2) mmHg in JAGM. JAGM had significantly lower STT and IOP values compared to AAGM (p = .0449, .0057, respectively) and GAGM (p = .0002, .0130, respectively). CONCLUSIONS Spontaneous ocular lesions were common in geriatric monkeys in this research colony. IOP and STT values were lower in juvenile African green monkeys relative to adult or geriatric animals.
Collapse
Affiliation(s)
- Jacklin S Pletcher
- Matthew J. Ryan Veterinary Hospital, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jennifer L Zimmer
- Oakland Veterinary Referral Services, Bloomfield Township, Michigan, USA
| | - Chin-Chi Liu
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Amy Beierschmitt
- Behavioural Science Foundation, Estridge Estate, Saint Kitts and Nevis
| | - Andrew C Lewin
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| |
Collapse
|
5
|
Jocher J, Edenhofer FC, Müller S, Janssen P, Briem E, Geuder J, Enard W. Generation and characterization of two Vervet monkey induced pluripotent stem cell lines derived from fibroblasts. Stem Cell Res 2024; 75:103315. [PMID: 38277709 DOI: 10.1016/j.scr.2024.103315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 12/21/2023] [Accepted: 01/16/2024] [Indexed: 01/28/2024] Open
Abstract
Cross-species comparisons using pluripotent stem cells from primates are crucial to better understand human biology, disease, and evolution. The Vervet monkey (Chlorocebus aethiops sabaeus) serves as an important primate model for such studies, and therefore we reprogrammed skin fibroblasts derived from a male and a female individual, resulting in two induced pluripotent stem cell lines (iPSCs). These iPSCs display the characteristic ESC-like colony morphology, express key pluripotency markers, and possess the ability to differentiate into cells representing all three germ layers. Importantly, both generated cell lines can be maintained in feeder-free culture conditions using commercially available medium.
Collapse
Affiliation(s)
- Jessica Jocher
- Anthropology & Human Genomics, Faculty of Biology, Ludwig-Maximilians-Universität München, Großhaderner Straße 2, 82152 Martinsried, Germany
| | - Fiona C Edenhofer
- Anthropology & Human Genomics, Faculty of Biology, Ludwig-Maximilians-Universität München, Großhaderner Straße 2, 82152 Martinsried, Germany
| | - Stefan Müller
- Institute of Human Genetics, Munich University Hospital, Ludwig-Maximilians-Universität München, 80336 Munich, Germany
| | - Philipp Janssen
- Anthropology & Human Genomics, Faculty of Biology, Ludwig-Maximilians-Universität München, Großhaderner Straße 2, 82152 Martinsried, Germany
| | - Eva Briem
- Anthropology & Human Genomics, Faculty of Biology, Ludwig-Maximilians-Universität München, Großhaderner Straße 2, 82152 Martinsried, Germany
| | - Johanna Geuder
- Anthropology & Human Genomics, Faculty of Biology, Ludwig-Maximilians-Universität München, Großhaderner Straße 2, 82152 Martinsried, Germany
| | - Wolfgang Enard
- Anthropology & Human Genomics, Faculty of Biology, Ludwig-Maximilians-Universität München, Großhaderner Straße 2, 82152 Martinsried, Germany.
| |
Collapse
|
6
|
Hungwe FTT, Laycock KM, Ntereke TD, Mabaka R, Paganotti GM. A historical perspective on arboviruses of public health interest in Southern Africa. Pathog Glob Health 2024; 118:131-159. [PMID: 38082563 PMCID: PMC11141323 DOI: 10.1080/20477724.2023.2290375] [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] [Indexed: 05/31/2024] Open
Abstract
Arboviruses are an existing and expanding threat globally, with the potential for causing devastating health and socioeconomic impacts. Mitigating this threat necessitates a One Health approach that integrates vector surveillance, rapid disease detection, and innovative prevention and control measures. In Southern Africa, limited data on the epidemiology of arboviruses, their vectors, and their hosts prevent an effective response. We reviewed the current knowledge on arboviruses in Southern Africa and identified opportunities for further research. A literature search was conducted to identify studies published on arboviruses in 10 tropical and temperate countries of the Southern African Development Community (SADC) from 1900 onward. We identified 280 studies, half (51.1%) originating from South Africa, that described 31 arboviral species, their vectors, and their clinical effects on hosts reported in the region. Arboviral research flourished in the SADC in the mid-20th century but then declined, before reemerging in the last two decades. Recent research consists largely of case reports describing outbreaks. Historical vector surveillance and serosurveys from the mid-20th century suggest that arboviruses are plentiful across Southern Africa, but large gaps remain in the current understanding of arboviral distribution, transmission dynamics, and public health impact.
Collapse
Affiliation(s)
- Faith T. T. Hungwe
- School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
- Department of Molecular Medicine, Karolinska Institute, Stockholm, Sweden
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Katherine M. Laycock
- The Ryan White Center for Pediatric Infectious Disease and Global Health, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Rorisang Mabaka
- School of Allied Health Sciences, Faculty of Health Sciences, University of Botswana, Gaborone, Botswana
| | - Giacomo M. Paganotti
- Botswana-University of Pennsylvania Partnership, Gaborone, Botswana
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biomedical Sciences, University of Botswana, Gaborone, Botswana
| |
Collapse
|
7
|
Solingapuram Sai KK, Erichsen JM, Gollapelli KK, Krizan I, Miller M, Bansode A, Jorgensen MJ, Register T, Cazzola C, Gandhi R, Suman J, Craft S. First Biodistribution Study of [68Ga]Ga-NOTA-Insulin Following Intranasal Administration in Adult Vervet Monkeys. J Alzheimers Dis 2024; 101:309-320. [PMID: 39213084 DOI: 10.3233/jad-240484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Background Intranasal insulin (INI) is being explored as a treatment for Alzheimer's disease (AD). Improved memory, functional ability, and cerebrospinal fluid (CSF) AD biomarker profiles have been observed following INI administration. However, the method of intranasal delivery may significantly affect outcomes. Objective To show reliable delivery of insulin to the brain using the Aptar Cartridge Pump System (CPS) intranasal delivery system. Methods To visualize INI biodistribution, we developed a novel PET radiotracer, Gallium 68-radiolabeled (NOTA-conjugated) insulin, [68Ga]Ga-NOTA-insulin. We used the Aptar CPS to administer [68Ga]Ga-NOTA-insulin to anesthetized healthy adult vervet monkeys and measured brain regional activity and whole-body dosimetry following PET/CT scans. Results We observed brain penetration of [68Ga]Ga-NOTA-insulin following intranasal administration with the Aptar CPS. Radioactive uptake was seen in multiple regions, including the amygdala, putamen, hypothalamus, hippocampus, and choroid plexus. A safety profile and whole-body dosimetry were also established in a second cohort of vervets. Safety was confirmed: vitals remained stable, blood glucose levels were unchanged, and no organ was exposed to more than 2.5 mSv of radioactivity. Extrapolations from vervet organ distribution allowed for estimation of the [68Ga]Ga-NOTA-insulin absorbed dose in humans, and the maximum dose of [68Ga]Ga-NOTA-insulin that can be safely administered to humans was determined to be 185 MBq. Conclusions The use of [68Ga]Ga-NOTA-insulin as a PET radiotracer is safe and effective for observing brain uptake in vervet monkeys. Further, the Aptar CPS successfully targets [68Ga]Ga-NOTA-insulin to the brain. The data will be essential in guiding future studies of intranasal [68Ga]Ga-NOTA-insulin administration in humans.
Collapse
Affiliation(s)
| | - Jennifer M Erichsen
- Department of Internal Medicine, Division of Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | - Ivan Krizan
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Mack Miller
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Avinash Bansode
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Mathew J Jorgensen
- Department of Pathology, Division of Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Thomas Register
- Department of Pathology, Division of Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | | | | | - Suzanne Craft
- Department of Internal Medicine, Division of Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
8
|
Bhoopal B, Gollapelli KK, Damuka N, Miller M, Krizan I, Bansode A, Register T, Frye BM, Kim J, Mintz A, Orr M, Craft S, Whitlow C, Lockhart SN, Shively CA, Solingapuram Sai KK. Preliminary PET Imaging of Microtubule-Based PET Radioligand [ 11C]MPC-6827 in a Nonhuman Primate Model of Alzheimer's Disease. ACS Chem Neurosci 2023; 14:3745-3751. [PMID: 37724996 PMCID: PMC10966409 DOI: 10.1021/acschemneuro.3c00527] [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] [Indexed: 09/21/2023] Open
Abstract
The microtubule (MT) instability observed in Alzheimer's disease (AD) is commonly attributed to hyperphosphorylation of the MT-associated protein, tau. In vivo PET imaging offers an opportunity to gain critical information about MT changes with the onset and development of AD and related dementia. We developed the first brain-penetrant MT PET ligand, [11C]MPC-6827, and evaluated its in vivo imaging utility in vervet monkeys. Consistent with our previous in vitro cell uptake and in vivo rodent imaging experiments, [11C]MPC-6827 uptake increased with MT destabilization. Radioactive uptake was inversely related to (cerebrospinal fluid) CSF Aβ42 levels and directly related to age in a nonhuman primate (NHP) model of AD. Additionally, in vitro autoradiography studies also corroborated PET imaging results. Here, we report the preliminary results of PET imaging with [11C]MPC-6827 in four female vervet monkeys with high or low CSF Aβ42 levels, which have been shown to correlate with the Aβ plaque burden, similar to humans.
Collapse
Affiliation(s)
- Bhuvanachandra Bhoopal
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Krishna Kumar Gollapelli
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Naresh Damuka
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Mack Miller
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Ivan Krizan
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Avinash Bansode
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Thomas Register
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Brett M Frye
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Jeongchul Kim
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Akiva Mintz
- Department of Radiology, Columbia University School of Medicine, New York, New York 10032, United States
| | - Miranda Orr
- Department of Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Suzanne Craft
- Department of Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Christopher Whitlow
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Samuel N Lockhart
- Department of Gerontology, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Carol A Shively
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina 27157, United States
| | | |
Collapse
|
9
|
Mrożek K, Marchewka J, Leszczyński B. A morphological study and the variability in the number of infraorbital foramina in the African green monkey (Grivet) (Chlorocebus aethiops) using microcomputed tomography. J Morphol 2023; 284:e21607. [PMID: 37458084 DOI: 10.1002/jmor.21607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/18/2023] [Accepted: 05/28/2023] [Indexed: 07/18/2023]
Abstract
Knowledge of the nonhuman primate morphology and anatomy related to craniofacial mechanoreception is essential for a fundamental understanding of the incidents that have occurred during the evolution of craniofacial features. The present study focuses on the variability in the number of infraorbital foramina and associated anatomical structures such as the infraorbital canal (IOC) and the infraorbital groove (IOG), as they are considered to play an important role in the behavioral ecology of these animals. A total of 19 skulls of Chlorocebus aethiops were analyzed. The number of infraorbital foramina was assessed macroscopically using a magnifying glass and a small diameter probe. Three dimensional (3D) projections and morphometric analysis of the infraorbital foramina, IOCs, and IOGs were performed using microcomputed tomography (micro-CT) for two skulls that represent one of the most common morphological types. Regardless of sex and body side, the most common morphological type observed in the studied species is the presence of three infraorbital foramina. The IOC takes a funnel or pinched shape. 3D projections were made to assess the course of the infraorbital vascular and nerve bundles in selected individuals. The results indicate a high morphological diversity within the species, although there appears to be a consistent distribution pattern of infraorbital neurovascular bundles in species of the Cercopithecidae family. The use of X-ray micro-CT allowed 3D visualization of the maxillary region to determine the variability of the infraorbital foramina and to track the division of the infraorbital neurovascular bundle in the case of the most common macroscopic expression of the number of the infraorbital foramen in C. aethiops, as well as the morphometric of the IOCs and IOGs which are related to mechanoreception of the primate's snout.
Collapse
Affiliation(s)
- Kamil Mrożek
- Nature Education Center, Jagiellonian University, Krakow, Poland
- Laboratory of Anthropology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Justyna Marchewka
- Department of Human Biology, Institute of Biological Sciences, Cardinal Stefan Wyszynski University, Warsaw, Poland
| | - Bartosz Leszczyński
- Department of Medical Physics, Marian Smoluchowski Institute of Physics, Jagiellonian University, Krakow, Poland
| |
Collapse
|
10
|
Georgiadis M, Menzel M, Reuter JA, Born DE, Kovacevich SR, Alvarez D, Taghavi HM, Schroeter A, Rudin M, Gao Z, Guizar-Sicairos M, Weiss TM, Axer M, Rajkovic I, Zeineh MM. Imaging crossing fibers in mouse, pig, monkey, and human brain using small-angle X-ray scattering. Acta Biomater 2023; 164:317-331. [PMID: 37098400 PMCID: PMC10811447 DOI: 10.1016/j.actbio.2023.04.029] [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: 11/28/2022] [Revised: 04/15/2023] [Accepted: 04/18/2023] [Indexed: 04/27/2023]
Abstract
Myelinated axons (nerve fibers) efficiently transmit signals throughout the brain via action potentials. Multiple methods that are sensitive to axon orientations, from microscopy to magnetic resonance imaging, aim to reconstruct the brain's structural connectome. As billions of nerve fibers traverse the brain with various possible geometries at each point, resolving fiber crossings is necessary to generate accurate structural connectivity maps. However, doing so with specificity is a challenging task because signals originating from oriented fibers can be influenced by brain (micro)structures unrelated to myelinated axons. X-ray scattering can specifically probe myelinated axons due to the periodicity of the myelin sheath, which yields distinct peaks in the scattering pattern. Here, we show that small-angle X-ray scattering (SAXS) can be used to detect myelinated, axon-specific fiber crossings. We first demonstrate the capability using strips of human corpus callosum to create artificial double- and triple-crossing fiber geometries, and we then apply the method in mouse, pig, vervet monkey, and human brains. We compare results to polarized light imaging (3D-PLI), tracer experiments, and to outputs from diffusion MRI that sometimes fails to detect crossings. Given its specificity, capability of 3-dimensional sampling and high resolution, SAXS could serve as a ground truth for validating fiber orientations derived using diffusion MRI as well as microscopy-based methods. STATEMENT OF SIGNIFICANCE: To study how the nerve fibers in our brain are interconnected, scientists need to visualize their trajectories, which often cross one another. Here, we show the unique capacity of small-angle X-ray scattering (SAXS) to study these fiber crossings without use of labeling, taking advantage of SAXS's specificity to myelin - the insulating sheath that is wrapped around nerve fibers. We use SAXS to detect double and triple crossing fibers and unveil intricate crossings in mouse, pig, vervet monkey, and human brains. This non-destructive method can uncover complex fiber trajectories and validate other less specific imaging methods (e.g., MRI or microscopy), towards accurate mapping of neuronal connectivity in the animal and human brain.
Collapse
Affiliation(s)
- Marios Georgiadis
- Department of Radiology, Stanford School of Medicine, Stanford, CA, USA; Institute for Biomedical Engineering, ETH Zurich and University of Zurich, Zurich, Switzerland.
| | - Miriam Menzel
- Institute of Neuroscience and Medicine (INM-1), Forschungszentrum Jülich GmbH, Jülich 52425, Germany; Department of Imaging Physics, Delft University of Technology, Delft, the Netherlands
| | - Jan A Reuter
- Institute of Neuroscience and Medicine (INM-1), Forschungszentrum Jülich GmbH, Jülich 52425, Germany
| | - Donald E Born
- Department of Pathology, Stanford School of Medicine, Stanford, CA, USA
| | | | - Dario Alvarez
- Department of Radiology, Stanford School of Medicine, Stanford, CA, USA
| | | | - Aileen Schroeter
- Institute for Biomedical Engineering, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Markus Rudin
- Institute for Biomedical Engineering, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Zirui Gao
- Institute for Biomedical Engineering, ETH Zurich and University of Zurich, Zurich, Switzerland
| | | | - Thomas M Weiss
- SLAC National Accelerator Laboratory, Stanford Synchrotron Radiation Lightsource, USA
| | - Markus Axer
- Institute of Neuroscience and Medicine (INM-1), Forschungszentrum Jülich GmbH, Jülich 52425, Germany
| | - Ivan Rajkovic
- SLAC National Accelerator Laboratory, Stanford Synchrotron Radiation Lightsource, USA
| | - Michael M Zeineh
- Department of Radiology, Stanford School of Medicine, Stanford, CA, USA
| |
Collapse
|
11
|
Meléndez GC, Kavanagh K, Gharraee N, Lacy JL, Goslen KH, Block M, Whitfield J, Widiapradja A, Levick SP. Replacement substance P reduces cardiac fibrosis in monkeys with type 2 diabetes. Biomed Pharmacother 2023; 160:114365. [PMID: 36758315 DOI: 10.1016/j.biopha.2023.114365] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM)-associated cardiac fibrosis contributes to heart failure. We previously showed that diabetic mice with cardiomyopathy, including cardiac fibrosis, exhibit low levels of the neuropeptide substance P; exogenous replacement of substance P reversed cardiac fibrosis, independent of body weight, blood glucose and blood pressure. We sought to elucidate the effectiveness and safety of replacement substance P to ameliorate or reverse cardiac fibrosis in type 2 diabetic monkeys. METHODS Four female T2DM African Green monkeys receive substance P (0.5 mg/Kg/day S.Q. injection) for 8 weeks. We obtained cardiac magnetic resonance imaging and blood samples to assess left ventricular function and fibrosis by T1 map-derived extracellular volume as well as circulating procollagen type I C-terminal propeptide. Hematological parameters for toxicities were also assessed in these monkeys and compared with three female T2DM monkeys receiving saline S.Q. as a safety comparison group. RESULTS Diabetic monkeys receiving replacement substance P exhibited a ∼20% decrease in extracellular volume (p = 0.01), concomitant with ∼25% decrease procollagen type I C-terminal propeptide levels (p = 0.008). Left ventricular ejection fraction was unchanged with substance P (p = 0.42); however, circumferential strain was improved (p < 0.01). Complete blood counts, glycosylated hemoglobin A1c, lipids, liver and pancreatic enzymes, and inflammation markers were unchanged (p > 0.05). CONCLUSIONS Replacement substance P reversed cardiac fibrosis in a large preclinical model of type 2 diabetes, independent of glycemic control. No hematological or organ-related toxicity was associated with replacement substance P. These results strongly support a potential application for replacement substance P as safe therapy for diabetic cardiac fibrosis.
Collapse
Affiliation(s)
- Giselle C Meléndez
- Internal Medicine, Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA; Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA.
| | - Kylie Kavanagh
- Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA; College of Health and Medicine, The University of Tasmania, Hobart, TAS, Australia
| | - Nazli Gharraee
- Internal Medicine, Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jessica L Lacy
- Internal Medicine, Section on Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Kevin H Goslen
- Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Masha Block
- Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jordyn Whitfield
- Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Alexander Widiapradja
- Kolling Institute, Royal North Shore Hospital, St Leonards, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia; Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| | - Scott P Levick
- Kolling Institute, Royal North Shore Hospital, St Leonards, NSW, Australia; Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia; Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
12
|
Vallender EJ, Hotchkiss CE, Lewis AD, Rogers J, Stern JA, Peterson SM, Ferguson B, Sayers K. Nonhuman primate genetic models for the study of rare diseases. Orphanet J Rare Dis 2023; 18:20. [PMID: 36721163 PMCID: PMC9887761 DOI: 10.1186/s13023-023-02619-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 01/15/2023] [Indexed: 02/01/2023] Open
Abstract
Pre-clinical research and development relies heavily upon translationally valid models of disease. A major difficulty in understanding the biology of, and developing treatments for, rare disease is the lack of animal models. It is important that these models not only recapitulate the presentation of the disease in humans, but also that they share functionally equivalent underlying genetic causes. Nonhuman primates share physiological, anatomical, and behavioral similarities with humans resulting from close evolutionary relationships and high genetic homology. As the post-genomic era develops and next generation sequencing allows for the resequencing and screening of large populations of research animals, naturally occurring genetic variation in nonhuman primates with clinically relevant phenotypes is regularly emerging. Here we review nonhuman primate models of multiple rare genetic diseases with a focus on the similarities and differences in manifestation and etiologies across species. We discuss how these models are being developed and how they can offer new tools and opportunities for researchers interested in exploring novel therapeutics for these and other genetic diseases. Modeling human genetic diseases in translationally relevant nonhuman primates presents new prospects for development of therapeutics and a better understanding of rare diseases. The post-genomic era offers the opportunity for the discovery and further development of more models like those discussed here.
Collapse
Affiliation(s)
- Eric J. Vallender
- University of Mississippi Medical Center, Jackson, MS USA
- Tulane National Primate Research Center, Covington, LA USA
| | - Charlotte E. Hotchkiss
- University of Washington, Seattle, WA USA
- Washington National Primate Research Center, Seattle, WA USA
| | - Anne D. Lewis
- Oregon Health and Sciences University, Beaverton, OR USA
- Oregon National Primate Research Center, Beaverton, OR USA
| | - Jeffrey Rogers
- Baylor College of Medicine, Houston, TX USA
- Wisconsin National Primate Research Center, Madison, WI USA
| | - Joshua A. Stern
- University of California-Davis, Davis, CA USA
- California National Primate Research Center, Davis, CA USA
| | - Samuel M. Peterson
- Oregon Health and Sciences University, Beaverton, OR USA
- Oregon National Primate Research Center, Beaverton, OR USA
| | - Betsy Ferguson
- Oregon Health and Sciences University, Beaverton, OR USA
- Oregon National Primate Research Center, Beaverton, OR USA
| | - Ken Sayers
- Texas Biomedical Research Institute, San Antonio, TX USA
- Southwest National Primate Research Center, San Antonio, TX USA
| |
Collapse
|
13
|
Jasinska AJ, Apetrei C, Pandrea I. Walk on the wild side: SIV infection in African non-human primate hosts-from the field to the laboratory. Front Immunol 2023; 13:1060985. [PMID: 36713371 PMCID: PMC9878298 DOI: 10.3389/fimmu.2022.1060985] [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: 10/04/2022] [Accepted: 12/15/2022] [Indexed: 01/15/2023] Open
Abstract
HIV emerged following cross-species transmissions of simian immunodeficiency viruses (SIVs) that naturally infect non-human primates (NHPs) from Africa. While HIV replication and CD4+ T-cell depletion lead to increased gut permeability, microbial translocation, chronic immune activation, and systemic inflammation, the natural hosts of SIVs generally avoid these deleterious consequences when infected with their species-specific SIVs and do not progress to AIDS despite persistent lifelong high viremia due to long-term coevolution with their SIV pathogens. The benign course of natural SIV infection in the natural hosts is in stark contrast to the experimental SIV infection of Asian macaques, which progresses to simian AIDS. The mechanisms of non-pathogenic SIV infections are studied mainly in African green monkeys, sooty mangabeys, and mandrills, while progressing SIV infection is experimentally modeled in macaques: rhesus macaques, pigtailed macaques, and cynomolgus macaques. Here, we focus on the distinctive features of SIV infection in natural hosts, particularly (1): the superior healing properties of the intestinal mucosa, which enable them to maintain the integrity of the gut barrier and prevent microbial translocation, thus avoiding excessive/pathologic immune activation and inflammation usually perpetrated by the leaking of the microbial products into the circulation; (2) the gut microbiome, the disruption of which is an important factor in some inflammatory diseases, yet not completely understood in the course of lentiviral infection; (3) cell population shifts resulting in target cell restriction (downregulation of CD4 or CCR5 surface molecules that bind to SIV), control of viral replication in the lymph nodes (expansion of natural killer cells), and anti-inflammatory effects in the gut (NKG2a/c+ CD8+ T cells); and (4) the genes and biological pathways that can shape genetic adaptations to viral pathogens and are associated with the non-pathogenic outcome of the natural SIV infection. Deciphering the protective mechanisms against SIV disease progression to immunodeficiency, which have been established through long-term coevolution between the natural hosts and their species-specific SIVs, may prompt the development of novel therapeutic interventions, such as drugs that can control gut inflammation, enhance gut healing capacities, or modulate the gut microbiome. These developments can go beyond HIV infection and open up large avenues for correcting gut damage, which is common in many diseases.
Collapse
Affiliation(s)
- Anna J. Jasinska
- Division of Infectious Diseases, Department of Medicine (DOM), School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Cristian Apetrei
- Division of Infectious Diseases, Department of Medicine (DOM), School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Infectious Diseases and Immunology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Ivona Pandrea
- Department of Infectious Diseases and Immunology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
14
|
Kessler SE, Tsangaras K, Rasoloharijaona S, Radespiel U, Greenwood AD. Long-term host-pathogen evolution of endogenous beta- and gammaretroviruses in mouse lemurs with little evidence of recent retroviral introgression. Virus Evol 2022; 9:veac117. [PMID: 36632481 PMCID: PMC9825726 DOI: 10.1093/ve/veac117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 11/03/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
Madagascar's flora and fauna have evolved in relative isolation since the island split from the African and Indian continents. When the last common ancestors of lemurs left Africa between 40 and 70 million years ago, they carried a subset of the viral diversity of the mainland population within them, which continued to evolve throughout the lemur radiation. Relative to other primate radiations, we know very little about the past or present viral diversity of lemurs, particularly mouse lemurs. Using high-throughput sequencing, we identified two gammaretroviruses and three betaretroviruses in the genomes of four species of wild mouse lemurs. The two gammaretroviruses and two betaretroviruses have not previously been described. One betaretrovirus was previously identified. All identified viruses are present in both Lorisiformes and Lemuriformes but absent from haplorrhine primates. The estimated ages of these viruses are consistent with the estimated divergence dates of the host lineages, suggesting they colonized the lemur genome after the Haplorrhine-Strepsirrhine split, but before the Lorisiformes-Lemuriformes split and before the colonization of Madagascar. The viral phylogenies connect multiple lineages of retroviruses from non-lemur and non-Madagascar-native species, suggesting substantial cross-species transmission occurred deep in the primate clade prior to its geographic dispersal. These phylogenies provide novel insights into known retroviral clades. They suggest that the origin of gammaretroviruses in rodents or bats may be premature and that the Jaagsiekte sheep virus clade may be older and more broadly distributed among mammals than previously thought.
Collapse
Affiliation(s)
| | - Kyriakos Tsangaras
- Department of Wildlife Diseases, Leibniz Institute for Zoo and Wildlife Research (IZW), Alfred-Kowalke-Straße 17, Berlin 10315, Germany,Department of Life and Health Sciences, University of Nicosia, 46 Makedonitissas Avenue, CY-2417, P.O. Box 24005, Nicosia, CY-1700, Cyprus
| | - Solofonirina Rasoloharijaona
- Faculty of Science, Technology and Environment, University of Mahajanga, 5 Georges V Street - Building KAKAL Mahajanga Be - Po. Box 652 , Mahajanga 401, Madagascar
| | - Ute Radespiel
- Institute of Zoology, University of Veterinary Medicine Hannover, Foundation, Buenteweg 17, Hannover 30559, Germany
| | | |
Collapse
|
15
|
Balamayooran G, Atkins HM, Andrews RN, Michalson KT, Hutchison AR, LeGrande AC, Wilson QN, Gee MK, Aycock ST, Jorgensen MJ, Young RW, Kock ND, Caudell DL. Epizootic Yersinia enterocolitica in captive African green monkeys ( Chlorocebus aethiops sabaeus). Front Vet Sci 2022; 9:922961. [PMID: 36504866 PMCID: PMC9727084 DOI: 10.3389/fvets.2022.922961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 11/04/2022] [Indexed: 11/24/2022] Open
Abstract
Yersinia enterocolitica is a Gram-negative bacterium that typical results in enterocolitis in humans and poses significant worldwide risks to public health. An outbreak of yersiniosis in the Vervet/African green monkey colony at the WFSM during the winter of 2015-2016 accounted for widespread systemic infection with high morbidity and mortality. Most of the cases had extensive necrosis with suppuration and large colonies of bacilli in the large bowel and associated lymph nodes; however, the small intestine, stomach, and other organs were also regularly affected. Positive cultures of Yersinia enterocolitica were recovered from affected tissues in 20 of the 23 cases. Carrier animals in the colony were suspected as the source of the infection because many clinically normal animals were culture-positive during and after the outbreak. In this study, we describe the gross and histology findings and immune cell profiles in different organs of affected animals. We found increased numbers of myeloid-derived phagocytes and CD11C-positive antigen-presenting cells and fewer adaptive T and B lymphocytes, suggesting an immunocompromised state in these animals. The pathogen-mediated microenvironment may have contributed to the immunosuppression and rapid spread of the infection in the vervets. Further studies in vervets could provide a better understanding of Yersinia-mediated pathogenesis and immunosuppression, which could be fundamental to understanding chronic and systemic inflammatory diseases in humans.
Collapse
|
16
|
Gagnon CM, Svardal H, Jasinska AJ, Danzy Cramer J, Freimer NB, Paul Grobler J, Turner TR, Schmitt CA. Evidence of selection in the uncoupling protein 1 gene region suggests local adaptation to solar irradiance in savannah monkeys ( Chlorocebus spp.). Proc Biol Sci 2022; 289:20221254. [PMID: 36100027 PMCID: PMC9470266 DOI: 10.1098/rspb.2022.1254] [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: 06/29/2022] [Accepted: 08/23/2022] [Indexed: 11/12/2022] Open
Abstract
In the last 300 thousand years, the genus Chlorocebus expanded from equatorial Africa into the southernmost latitudes of the continent, where colder climate was a probable driver of natural selection. We investigated population-level genetic variation in the mitochondrial uncoupling protein 1 (UCP1) gene region-implicated in non-shivering thermogenesis (NST)-in 73 wild savannah monkeys from three taxa representing this southern expansion (Chlorocebus pygerythrus hilgerti, Chlorocebus cynosuros and Chlorocebus pygerythrus pygerythrus) ranging from Kenya to South Africa. We found 17 single nucleotide polymorphisms with extended haplotype homozygosity consistent with positive selective sweeps, 10 of which show no significant linkage disequilibrium with each other. Phylogenetic generalized least-squares modelling with ecological covariates suggest that most derived allele frequencies are significantly associated with solar irradiance and winter precipitation, rather than overall low temperatures. This selection and association with irradiance is demonstrated by a relatively isolated population in the southern coastal belt of South Africa. We suggest that sunbathing behaviours common to savannah monkeys, in combination with the strength of solar irradiance, may mediate adaptations to thermal stress via NST among savannah monkeys. The variants we discovered all lie in non-coding regions, some with previously documented regulatory functions, calling for further validation and research.
Collapse
Affiliation(s)
| | - Hannes Svardal
- Department of Biology, University of Antwerp, Antwerp, Belgium
- Naturalis Biodiversity Center, Leiden, The Netherlands
| | - Anna J. Jasinska
- Center for Neurobehavioral Genetics, University of California, Los Angeles, CA 90095, USA
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
- Division of Infectious Diseases, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | | | - Nelson B. Freimer
- Center for Neurobehavioral Genetics, University of California, Los Angeles, CA 90095, USA
| | - J. Paul Grobler
- Department of Genetics, University of the Free State, Bloemfontein, Free State 9301, South Africa
| | - Trudy R. Turner
- Department of Genetics, University of the Free State, Bloemfontein, Free State 9301, South Africa
- Department of Anthropology, University of Wisconsin, Milwaukee, Milwaukee, WI, 53201, USA
| | - Christopher A. Schmitt
- Department of Anthropology, Boston University, Boston, MA 02215, USA
- Department of Biology, Boston University, Boston, MA 02215, USA
| |
Collapse
|
17
|
Campos JHC, Alves GV, Maricato JT, Braconi CT, Antoneli FM, Janini LMR, Briones MRS. The epitranscriptome of Vero cells infected with SARS-CoV-2 assessed by direct RNA sequencing reveals m6A pattern changes and DRACH motif biases in viral and cellular RNAs. Front Cell Infect Microbiol 2022; 12:906578. [PMID: 36051243 PMCID: PMC9425070 DOI: 10.3389/fcimb.2022.906578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 07/25/2022] [Indexed: 11/18/2022] Open
Abstract
The epitranscriptomics of the SARS-CoV-2 infected cell reveals its response to viral replication. Among various types of RNA nucleotide modifications, the m6A is the most common and is involved in several crucial processes of RNA intracellular location, maturation, half-life and translatability. This epitranscriptome contains a mixture of viral RNAs and cellular transcripts. In a previous study we presented the analysis of the SARS-CoV-2 RNA m6A methylation based on direct RNA sequencing and characterized DRACH motif mutations in different viral lineages. Here we present the analysis of the m6A transcript methylation of Vero cells (derived from African Green Monkeys) and Calu-3 cells (human) upon infection by SARS-CoV-2 using direct RNA sequencing data. Analysis of these data by nonparametric statistics and two computational methods (m6anet and EpiNano) show that m6A levels are higher in RNAs of infected cells. Functional enrichment analysis reveals increased m6A methylation of transcripts involved in translation, peptide and amine metabolism. This analysis allowed the identification of differentially methylated transcripts and m6A unique sites in the infected cell transcripts. Results here presented indicate that the cell response to viral infection not only changes the levels of mRNAs, as previously shown, but also its epitranscriptional pattern. Also, transcriptome-wide analysis shows strong nucleotide biases in DRACH motifs of cellular transcripts, both in Vero and Calu-3 cells, which use the signature GGACU whereas in viral RNAs the signature is GAACU. We hypothesize that the differences of DRACH motif biases, might force the convergent evolution of the viral genome resulting in better adaptation to target sequence preferences of writer, reader and eraser enzymes. To our knowledge, this is the first report on m6A epitranscriptome of the SARS-CoV-2 infected Vero cells by direct RNA sequencing, which is the sensu stricto RNA-seq.
Collapse
Affiliation(s)
- João H. C. Campos
- Center for Medical Bioinformatics, Escola Paulista de Medicina, UNIFESP, São Paulo, Brazil
| | - Gustavo V. Alves
- Center for Medical Bioinformatics, Escola Paulista de Medicina, UNIFESP, São Paulo, Brazil
| | - Juliana T. Maricato
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, UNIFESP, São Paulo, Brazil
| | - Carla T. Braconi
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, UNIFESP, São Paulo, Brazil
| | - Fernando M. Antoneli
- Center for Medical Bioinformatics, Escola Paulista de Medicina, UNIFESP, São Paulo, Brazil
| | - Luiz Mario R. Janini
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, UNIFESP, São Paulo, Brazil
| | - Marcelo R. S. Briones
- Center for Medical Bioinformatics, Escola Paulista de Medicina, UNIFESP, São Paulo, Brazil
| |
Collapse
|
18
|
Wakeman DR, Weed MR, Perez SE, Cline EN, Viola KL, Wilcox KC, Moddrelle DS, Nisbett EZ, Kurian AM, Bell AF, Pike R, Jacobson PB, Klein WL, Mufson EJ, Lawrence MS, Elsworth JD. Intrathecal amyloid-beta oligomer administration increases tau phosphorylation in the medial temporal lobe in the African green monkey: A nonhuman primate model of Alzheimer's disease. Neuropathol Appl Neurobiol 2022; 48:e12800. [PMID: 35156715 PMCID: PMC10902791 DOI: 10.1111/nan.12800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/31/2022] [Accepted: 02/05/2022] [Indexed: 11/26/2022]
Abstract
AIMS An obstacle to developing new treatment strategies for Alzheimer's disease (AD) has been the inadequate translation of findings in current AD transgenic rodent models to the prediction of clinical outcomes. By contrast, nonhuman primates (NHPs) share a close neurobiology with humans in virtually all aspects relevant to developing a translational AD model. The present investigation used African green monkeys (AGMs) to refine an inducible NHP model of AD based on the administration of amyloid-beta oligomers (AβOs), a key upstream initiator of AD pathology. METHODS AβOs or vehicle were repeatedly delivered over 4 weeks to age-matched young adult AGMs by intracerebroventricular (ICV) or intrathecal (IT) injections. Induction of AD-like pathology was assessed in subregions of the medial temporal lobe (MTL) by quantitative immunohistochemistry (IHC) using the AT8 antibody to detect hyperphosphorylated tau. Hippocampal volume was measured by magnetic resonance imaging (MRI) scans prior to, and after, intrathecal injections. RESULTS IT administration of AβOs in young adult AGMs revealed an elevation of tau phosphorylation in the MTL cortical memory circuit compared with controls. The largest increases were detected in the entorhinal cortex that persisted for at least 12 weeks after dosing. MRI scans showed a reduction in hippocampal volume following AβO injections. CONCLUSIONS Repeated IT delivery of AβOs in young adult AGMs led to an accelerated AD-like neuropathology in MTL, similar to human AD, supporting the value of this translational model to de-risk the clinical trial of diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
| | | | - Sylvia E Perez
- Neurobiology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Erika N Cline
- Neurobiology, Northwestern University, Evanston, Illinois, USA
| | - Kirsten L Viola
- Neurobiology, Northwestern University, Evanston, Illinois, USA
| | - Kyle C Wilcox
- Neurobiology, Northwestern University, Evanston, Illinois, USA
| | - David S Moddrelle
- Virscio Inc., St. Kitts Biomedical Research Foundation, St. Kitts, West Indies
| | - Ernell Z Nisbett
- Virscio Inc., St. Kitts Biomedical Research Foundation, St. Kitts, West Indies
| | | | - Amanda F Bell
- Virscio Inc., St. Kitts Biomedical Research Foundation, St. Kitts, West Indies
| | - Ricaldo Pike
- Virscio Inc., St. Kitts Biomedical Research Foundation, St. Kitts, West Indies
| | | | - William L Klein
- Neurobiology, Northwestern University, Evanston, Illinois, USA
| | - Elliott J Mufson
- Neurobiology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | | | | |
Collapse
|
19
|
Bjornson-Hooper ZB, Fragiadakis GK, Spitzer MH, Chen H, Madhireddy D, Hu K, Lundsten K, McIlwain DR, Nolan GP. A Comprehensive Atlas of Immunological Differences Between Humans, Mice, and Non-Human Primates. Front Immunol 2022; 13:867015. [PMID: 35359965 PMCID: PMC8962947 DOI: 10.3389/fimmu.2022.867015] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/16/2022] [Indexed: 01/01/2023] Open
Abstract
Animal models are an integral part of the drug development and evaluation process. However, they are unsurprisingly imperfect reflections of humans, and the extent and nature of many immunological differences are unknown. With the rise of targeted and biological therapeutics, it is increasingly important that we understand the molecular differences in the immunological behavior of humans and model organisms. However, very few antibodies are raised against non-human primate antigens, and databases of cross-reactivity between species are incomplete. Thus, we screened 332 antibodies in five immune cell populations in blood from humans and four non-human primate species generating a comprehensive cross-reactivity catalog that includes cell type-specificity. We used this catalog to create large mass cytometry universal cross-species phenotyping and signaling panels for humans, along with three of the model organisms most similar to humans: rhesus and cynomolgus macaques and African green monkeys; and one of the mammalian models most widely used in drug development: C57BL/6 mice. As a proof-of-principle, we measured immune cell signaling responses across all five species to an array of 15 stimuli using mass cytometry. We found numerous instances of different cellular phenotypes and immune signaling events occurring within and between species, and detailed three examples (double-positive T cell frequency and signaling; granulocyte response to Bacillus anthracis antigen; and B cell subsets). We also explore the correlation of herpes simian B virus serostatus on the immune profile. Antibody panels and the full dataset generated are available online as a resource to enable future studies comparing immune responses across species during the evaluation of therapeutics.
Collapse
Affiliation(s)
| | - Gabriela K. Fragiadakis
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
- Department of Medicine, Division of Rheumatology, University of California San Francisco, San Francisco, CA, United States
- Bakar ImmunoX Initiative, University of California San Francisco, San Francisco, CA, United States
- University of California, San Francisco (UCSF) Data Science CoLab and University of California, San Francisco (UCSF) Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Matthew H. Spitzer
- Immunology Program, Stanford University, Stanford, CA, United States
- Departments of Otolaryngology – Head and Neck Surgery and Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, United States
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, United States
- Chan Zuckerberg Biohub, San Francisco, CA, United States
| | - Han Chen
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| | - Deepthi Madhireddy
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| | - Kevin Hu
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| | - Kelly Lundsten
- BioLegend Inc, Advanced Cytometry, San Diego, CA, United States
| | - David R. McIlwain
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| | - Garry P. Nolan
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| |
Collapse
|
20
|
Frye BM, Craft S, Latimer CS, Keene CD, Montine TJ, Register TC, Orr ME, Kavanagh K, Macauley SL, Shively CA. Aging-related Alzheimer's disease-like neuropathology and functional decline in captive vervet monkeys (Chlorocebus aethiops sabaeus). Am J Primatol 2021; 83:e23260. [PMID: 33818801 PMCID: PMC8626867 DOI: 10.1002/ajp.23260] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/05/2021] [Accepted: 03/21/2021] [Indexed: 12/17/2022]
Abstract
Age-related neurodegeneration characteristic of late-onset Alzheimer's disease (LOAD) begins in middle age, well before symptoms. Translational models to identify modifiable risk factors are needed to understand etiology and identify therapeutic targets. Here, we outline the evidence supporting the vervet monkey (Chlorocebus aethiops sabaeus) as a model of aging-related AD-like neuropathology and associated phenotypes including cognitive function, physical function, glucose handling, intestinal physiology, and CSF, blood, and neuroimaging biomarkers. This review provides the most comprehensive multisystem description of aging in vervets to date. This review synthesizes a large body of evidence that suggests that aging vervets exhibit a coordinated suite of traits consistent with early AD and provide a powerful, naturally occurring model for LOAD. Notably, relationships are identified between AD-like neuropathology and modifiable risk factors. Gaps in knowledge and key limitations are provided to shape future studies to illuminate mechanisms underlying divergent neurocognitive aging trajectories and to develop interventions that increase resilience to aging-associated chronic disease, particularly, LOAD.
Collapse
Affiliation(s)
- Brett M. Frye
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine
| | - Suzanne Craft
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine
- Wake Forest Alzheimer’s Disease Research Center
- J. Paul Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest School of Medicine
| | - Caitlin S. Latimer
- Department of Laboratory Medicine and Pathology, University of Washington-Seattle
| | - C. Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington-Seattle
| | | | - Thomas C. Register
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine
- Wake Forest Alzheimer’s Disease Research Center
- J. Paul Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest School of Medicine
| | - Miranda E. Orr
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine
| | - Kylie Kavanagh
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine
| | - Shannon L. Macauley
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine
- Wake Forest Alzheimer’s Disease Research Center
| | - Carol A. Shively
- Department of Pathology/Comparative Medicine, Wake Forest School of Medicine
- Wake Forest Alzheimer’s Disease Research Center
| |
Collapse
|
21
|
Epigenetic clock and methylation studies in vervet monkeys. GeroScience 2021; 44:699-717. [PMID: 34591235 PMCID: PMC9135907 DOI: 10.1007/s11357-021-00466-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 09/20/2021] [Indexed: 11/26/2022] Open
Abstract
DNA methylation-based biomarkers of aging have been developed for many mammals but not yet for the vervet monkey (Chlorocebus sabaeus), which is a valuable non-human primate model for biomedical studies. We generated novel DNA methylation data from vervet cerebral cortex, blood, and liver using highly conserved mammalian CpGs represented on a custom array (HorvathMammalMethylChip40). We present six DNA methylation-based estimators of age: vervet multi-tissue epigenetic clock and tissue-specific clocks for brain cortex, blood, and liver. In addition, we developed two dual species clocks (human-vervet clocks) for measuring chronological age and relative age, respectively. Relative age was defined as ratio of chronological age to maximum lifespan to address the species differences in maximum lifespan. The high accuracy of the human-vervet clocks demonstrates that epigenetic aging processes are evolutionary conserved in primates. When applying these vervet clocks to tissue samples from another primate species, rhesus macaque, we observed high age correlations but strong offsets. We characterized CpGs that correlate significantly with age in the vervet. CpG probes that gain methylation with age across tissues were located near the targets of Polycomb proteins SUZ12 and EED and genes possessing the trimethylated H3K27 mark in their promoters. The epigenetic clocks are expected to be useful for anti-aging studies in vervets.
Collapse
|
22
|
Schmitt CA, Garrett EC. De-Scent with Modification: More Evidence and Caution Needed to Assess Whether the Loss of a Pheromone Signaling Protein Permitted the Evolution of Same-Sex Sexual Behavior in Primates. ARCHIVES OF SEXUAL BEHAVIOR 2021; 50:2301-2307. [PMID: 31741251 DOI: 10.1007/s10508-019-01583-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 10/30/2019] [Accepted: 11/02/2019] [Indexed: 06/10/2023]
Affiliation(s)
- Christopher A Schmitt
- Department of Anthropology, Boston University, 232 Bay State Road, Boston, MA, 02215, USA.
- Department of Biology, Boston University, Boston, MA, USA.
| | - Eva C Garrett
- Department of Anthropology, Boston University, 232 Bay State Road, Boston, MA, 02215, USA
| |
Collapse
|
23
|
Li M, Brokaw A, Furuta AM, Coler B, Obregon-Perko V, Chahroudi A, Wang HY, Permar SR, Hotchkiss CE, Golos TG, Rajagopal L, Adams Waldorf KM. Non-human Primate Models to Investigate Mechanisms of Infection-Associated Fetal and Pediatric Injury, Teratogenesis and Stillbirth. Front Genet 2021; 12:680342. [PMID: 34290739 PMCID: PMC8287178 DOI: 10.3389/fgene.2021.680342] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 05/25/2021] [Indexed: 12/25/2022] Open
Abstract
A wide array of pathogens has the potential to injure the fetus and induce teratogenesis, the process by which mutations in fetal somatic cells lead to congenital malformations. Rubella virus was the first infectious disease to be linked to congenital malformations due to an infection in pregnancy, which can include congenital cataracts, microcephaly, hearing impairment and congenital heart disease. Currently, human cytomegalovirus (HCMV) is the leading infectious cause of congenital malformations globally, affecting 1 in every 200 infants. However, our knowledge of teratogenic viruses and pathogens is far from complete. New emerging infectious diseases may induce teratogenesis, similar to Zika virus (ZIKV) that caused a global pandemic in 2016-2017; thousands of neonates were born with congenital microcephaly due to ZIKV exposure in utero, which also included a spectrum of injuries to the brain, eyes and spinal cord. In addition to congenital anomalies, permanent injury to fetal and neonatal organs, preterm birth, stillbirth and spontaneous abortion are known consequences of a broader group of infectious diseases including group B streptococcus (GBS), Listeria monocytogenes, Influenza A virus (IAV), and Human Immunodeficiency Virus (HIV). Animal models are crucial for determining the mechanism of how these various infectious diseases induce teratogenesis or organ injury, as well as testing novel therapeutics for fetal or neonatal protection. Other mammalian models differ in many respects from human pregnancy including placentation, labor physiology, reproductive tract anatomy, timeline of fetal development and reproductive toxicology. In contrast, non-human primates (NHP) most closely resemble human pregnancy and exhibit key similarities that make them ideal for research to discover the mechanisms of injury and for testing vaccines and therapeutics to prevent teratogenesis, fetal and neonatal injury and adverse pregnancy outcomes (e.g., stillbirth or spontaneous abortion). In this review, we emphasize key contributions of the NHP model pre-clinical research for ZIKV, HCMV, HIV, IAV, L. monocytogenes, Ureaplasma species, and GBS. This work represents the foundation for development and testing of preventative and therapeutic strategies to inhibit infectious injury of human fetuses and neonates.
Collapse
Affiliation(s)
- Miranda Li
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States
- Department of Biological Sciences, Columbia University, New York, NY, United States
| | - Alyssa Brokaw
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Anna M. Furuta
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Brahm Coler
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, United States
| | - Veronica Obregon-Perko
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
| | - Ann Chahroudi
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
- Center for Childhood Infections and Vaccines of Children’s Healthcare of Atlanta and Emory University, Atlanta, GA, United States
| | - Hsuan-Yuan Wang
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Sallie R. Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, United States
| | - Charlotte E. Hotchkiss
- Washington National Primate Research Center, University of Washington, Seattle, WA, United States
| | - Thaddeus G. Golos
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, United States
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, United States
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Lakshmi Rajagopal
- Department of Global Health, University of Washington, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Kristina M. Adams Waldorf
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
- Department of Obstetrics and Gynecology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
24
|
Frye BM, Valure PM, Craft S, Baxter MG, Scott C, Wise-Walden S, Bissinger DW, Register HM, Copeland C, Jorgensen MJ, Justice JN, Kritchevsky SB, Register TC, Shively CA. Temporal emergence of age-associated changes in cognitive and physical function in vervets (Chlorocebus aethiops sabaeus). GeroScience 2021; 43:1303-1315. [PMID: 33611720 PMCID: PMC8190425 DOI: 10.1007/s11357-021-00338-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 02/04/2021] [Indexed: 02/07/2023] Open
Abstract
Dual declines in gait speed and cognitive performance are associated with increased risk of developing dementia. Characterizing the patterns of such impairments therefore is paramount to distinguishing healthy from pathological aging. Nonhuman primates such as vervet/African green monkeys (Chlorocebus aethiops sabaeus) are important models of human neurocognitive aging, yet the trajectory of dual decline has not been characterized. We therefore (1) assessed whether cognitive and physical performance (i.e., gait speed) are lower in older aged animals; (2) explored the relationship between performance in a novel task of executive function (Wake Forest Maze Task-WFMT) and a well-established assessment of working memory (delayed response task-DR task); and (3) examined the association between baseline gait speed with executive function and working memory at 1-year follow-up. We found (1) physical and cognitive declines with age; (2) strong agreement between performance in the novel WFMT and DR task; and (3) that slow gait is associated with poor cognitive performance in both domains. Our results suggest that older aged vervets exhibit a coordinated suite of traits consistent with human aging and that slow gait may be a biomarker of cognitive decline. This integrative approach provides evidence that gait speed and cognitive function differ across the lifespan in female vervet monkeys, which advances them as a model that could be used to dissect relationships between trajectories of dual decline over time.
Collapse
Affiliation(s)
- Brett M Frye
- Wake Forest School of Medicine, Medical Center Blvd Winston-Salem, NC, 27157-1040, USA
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Department of Internal Medicine - Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, USA
| | - Payton M Valure
- Wake Forest School of Medicine, Medical Center Blvd Winston-Salem, NC, 27157-1040, USA
| | - Suzanne Craft
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Department of Internal Medicine - Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, USA
- Wake Forest Alzheimer's Disease Research Center, Winston-Salem, USA
| | - Mark G Baxter
- Nash Family Department of Neuroscience, IW, New York, USA
| | - Christie Scott
- Wake Forest School of Medicine, Medical Center Blvd Winston-Salem, NC, 27157-1040, USA
| | - Shanna Wise-Walden
- Wake Forest School of Medicine, Medical Center Blvd Winston-Salem, NC, 27157-1040, USA
| | - David W Bissinger
- Wake Forest School of Medicine, Medical Center Blvd Winston-Salem, NC, 27157-1040, USA
| | - Hannah M Register
- Wake Forest School of Medicine, Medical Center Blvd Winston-Salem, NC, 27157-1040, USA
| | - Carson Copeland
- Wake Forest School of Medicine, Medical Center Blvd Winston-Salem, NC, 27157-1040, USA
| | - Matthew J Jorgensen
- Wake Forest School of Medicine, Medical Center Blvd Winston-Salem, NC, 27157-1040, USA
| | - Jamie N Justice
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Department of Internal Medicine - Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, USA
| | - Stephen B Kritchevsky
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Department of Internal Medicine - Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, USA
| | - Thomas C Register
- Wake Forest School of Medicine, Medical Center Blvd Winston-Salem, NC, 27157-1040, USA
- Wake Forest Alzheimer's Disease Research Center, Winston-Salem, USA
| | - Carol A Shively
- Wake Forest School of Medicine, Medical Center Blvd Winston-Salem, NC, 27157-1040, USA.
- Wake Forest Alzheimer's Disease Research Center, Winston-Salem, USA.
| |
Collapse
|
25
|
Williams DM, Almanza SM, Sifuentes-Romero I, Detwiler KM. The history, taxonomy, and geographic origins of an introduced African monkey in the southeastern United States. Primates 2021; 62:617-627. [PMID: 33963936 DOI: 10.1007/s10329-021-00890-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 01/23/2021] [Indexed: 11/24/2022]
Abstract
The origins and taxonomy of the introduced vervet monkey population in Dania Beach, Florida has been unconfirmed due to a lack of documentation and genetic research. Our goal was to determine the introduction history, species identification, and geographic origins of the monkeys. Through interviews, historical archives, and popular media, we traced the monkeys to an escape from the Dania Chimpanzee Farm in 1948. The facility imported primates from Africa for medical research purposes. Historical archives suggest the monkeys were caught in Sierra Leone. We tested the hypothesis of West African origins using three genetic markers: one mitochondrial DNA gene (cytochrome b) and two fragments from the Y-chromosome, the sex-determining gene and the zinc-finger gene. We ran Bayesian and maximum-likelihood analyses to reconstruct phylogenetic trees. Results from all loci confirmed the species identification is Chlorocebus sabaeus. We found no variation among the sampled individuals and found the cytochrome b haplotype to be a complete match to a C. sabaeus sample from Senegal. Phylogenetic analyses showed strong support for the Dania Beach mitochondrial and Y-chromosome lineages to group within a monophyletic C. sabaeus clade endemic to West Africa. Our study provides critical baseline information to the scientific community about a little-known population of Chlorocebus monkeys that have adapted to a novel environment in the southeastern United States.
Collapse
Affiliation(s)
- Deborah M Williams
- Biological Sciences Department, Florida Atlantic University, Boca Raton, USA.
| | - Sandra M Almanza
- Anthropology Department, Florida Atlantic University, Boca Raton, USA
| | | | - Kate M Detwiler
- Biological Sciences Department, Florida Atlantic University, Boca Raton, USA
- Anthropology Department, Florida Atlantic University, Boca Raton, USA
| |
Collapse
|
26
|
Abstract
Despite its important biological role, the evolution of recombination rates remains relatively poorly characterized. This owes, in part, to the lack of high-quality genomic resources to address this question across diverse species. Humans and our closest evolutionary relatives, anthropoid apes, have remained a major focus of large-scale sequencing efforts, and thus recombination rate variation has been comparatively well studied in this group-with earlier work revealing a conservation at the broad- but not the fine-scale. However, in order to better understand the nature of this variation, and the time scales on which substantial modifications occur, it is necessary to take a broader phylogenetic perspective. I here present the first fine-scale genetic map for vervet monkeys based on whole-genome population genetic data from ten individuals and perform a series of comparative analyses with the great apes. The results reveal a number of striking features. First, owing to strong positive correlations with diversity and weak negative correlations with divergence, analyses suggest a dominant role for purifying and background selection in shaping patterns of variation in this species. Second, results support a generally reduced broad-scale recombination rate compared with the great apes, as well as a narrower fraction of the genome in which the majority of recombination events are observed to occur. Taken together, this data set highlights the great necessity of future research to identify genomic features and quantify evolutionary processes that are driving these rate changes across primates.
Collapse
Affiliation(s)
- Susanne P Pfeifer
- Center for Evolution and Medicine, School of Life Sciences, Arizona State University, Tempe, AZ
| |
Collapse
|
27
|
Pichon F, Shen Y, Busato F, P Jochems S, Jacquelin B, Grand RL, Deleuze JF, Müller-Trutwin M, Tost J. Analysis and annotation of DNA methylation in two nonhuman primate species using the Infinium Human Methylation 450K and EPIC BeadChips. Epigenomics 2021; 13:169-186. [PMID: 33471557 DOI: 10.2217/epi-2020-0200] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aim: Nonhuman primates are essential for research on many human diseases. The Infinium Human Methylation450/EPIC BeadChips are popular tools for the study of the methylation state across the human genome at affordable cost. Methods: We performed a precise evaluation and re-annotation of the BeadChip probes for the analysis of genome-wide DNA methylation patterns in rhesus macaques and African green monkeys through in silico analyses combined with functional validation by pyrosequencing. Results: Up to 165,847 of the 450K and 261,545 probes of the EPIC BeadChip can be reliably used. The annotation files are provided in a format compatible with a variety of standard bioinformatic pipelines. Conclusion: Our study will facilitate high-throughput DNA methylation analyses in Macaca mulatta and Chlorocebus sabaeus.
Collapse
Affiliation(s)
- Fabien Pichon
- Laboratory for Epigenetics & Environment, Centre National de Recherche en Génomique Humaine, CEA-Institut de Biologie François Jacob, Evry, France
| | - Yimin Shen
- Laboratory for Epigenetics & Environment, Centre National de Recherche en Génomique Humaine, CEA-Institut de Biologie François Jacob, Evry, France.,Laboratory for Bioinformatics, Fondation Jean Dausset - Centre d'Etude du Polymorphisme Humain, 75010 Paris, France
| | - Florence Busato
- Laboratory for Epigenetics & Environment, Centre National de Recherche en Génomique Humaine, CEA-Institut de Biologie François Jacob, Evry, France
| | - Simon P Jochems
- Institut Pasteur, HIV Inflammation & Persistence Unit, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France.,Leiden University Medical Center, Leiden, The Netherlands
| | | | - Roger Le Grand
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses, France
| | - Jean-Francois Deleuze
- Laboratory for Epigenetics & Environment, Centre National de Recherche en Génomique Humaine, CEA-Institut de Biologie François Jacob, Evry, France.,Laboratory for Bioinformatics, Fondation Jean Dausset - Centre d'Etude du Polymorphisme Humain, 75010 Paris, France
| | | | - Jörg Tost
- Laboratory for Epigenetics & Environment, Centre National de Recherche en Génomique Humaine, CEA-Institut de Biologie François Jacob, Evry, France
| |
Collapse
|
28
|
Lee JR, Choe SH, Kim YH, Cho HM, Park HR, Lee HE, Jin YB, Kim JS, Jeong KJ, Park SJ, Huh JW. Longitudinal profiling of the blood transcriptome in an African green monkey aging model. Aging (Albany NY) 2020; 13:846-864. [PMID: 33290253 PMCID: PMC7834999 DOI: 10.18632/aging.202190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/22/2020] [Indexed: 12/14/2022]
Abstract
African green monkeys (AGMs, Chlorocebus aethiops) are Old World monkeys which are used as experimental models in biomedical research. Recent technological advances in next generation sequencing are useful for unraveling the genetic mechanisms underlying senescence, aging, and age-related disease. To elucidate the normal aging mechanisms in older age, the blood transcriptomes of nine healthy, aged AGMs (15‒23 years old), were analyzed over two years. We identified 910‒1399 accumulated differentially expressed genes (DEGs) in each individual, which increased with age. Aging-related DEGs were sorted across the three time points. A major proportion of the aging-related DEGs belonged to gene ontology (GO) categories involved in translation and rRNA metabolic processes. Next, we sorted common aging-related DEGs across three time points over two years. Common aging-related DEGs belonged to GO categories involved in translation, cellular component biogenesis, rRNA metabolic processes, cellular component organization, biogenesis, and RNA metabolic processes. Furthermore, we identified 29 candidate aging genes that were upregulated across the time series analysis. These candidate aging genes were linked to protein synthesis. This study describes a changing gene expression pattern in AGMs during aging using longitudinal transcriptome sequencing. The candidate aging genes identified here may be potential targets for the treatment of aging.
Collapse
Affiliation(s)
- Ja-Rang Lee
- Primate Resource Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56216, Republic of Korea
| | - Se-Hee Choe
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Young-Hyun Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Hyeon-Mu Cho
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Hye-Ri Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Hee-Eun Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Yeung Bae Jin
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Ji-Su Kim
- Primate Resource Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56216, Republic of Korea
| | - Kang Jin Jeong
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Sang-Je Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Jae-Won Huh
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea.,Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| |
Collapse
|
29
|
Jasinska AJ, Dong TS, Lagishetty V, Katzka W, Jacobs JP, Schmitt CA, Cramer JD, Ma D, Coetzer WG, Grobler JP, Turner TR, Freimer N, Pandrea I, Apetrei C. Shifts in microbial diversity, composition, and functionality in the gut and genital microbiome during a natural SIV infection in vervet monkeys. MICROBIOME 2020; 8:154. [PMID: 33158452 PMCID: PMC7648414 DOI: 10.1186/s40168-020-00928-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/06/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND The microbiota plays an important role in HIV pathogenesis in humans. Microbiota can impact health through several pathways such as increasing inflammation in the gut, metabolites of bacterial origin, and microbial translocation from the gut to the periphery which contributes to systemic chronic inflammation and immune activation and the development of AIDS. Unlike HIV-infected humans, SIV-infected vervet monkeys do not experience gut dysfunction, microbial translocation, and chronic immune activation and do not progress to immunodeficiency. Here, we provide the first reported characterization of the microbial ecosystems of the gut and genital tract in a natural nonprogressing host of SIV, wild vervet monkeys from South Africa. RESULTS We characterized fecal, rectal, vaginal, and penile microbiomes in vervets from populations heavily infected with SIV from diverse locations across South Africa. Geographic site, age, and sex affected the vervet microbiome across different body sites. Fecal and vaginal microbiome showed marked stratification with three enterotypes in fecal samples and two vagitypes, which were predicted functionally distinct within each body site. External bioclimatic factors, biome type, and environmental temperature influenced microbiomes locally associated with vaginal and rectal mucosa. Several fecal microbial taxa were linked to plasma levels of immune molecules, for example, MIG was positively correlated with Lactobacillus and Escherichia/Shigella and Helicobacter, and IL-10 was negatively associated with Erysipelotrichaceae, Anaerostipes, Prevotella, and Anaerovibrio, and positively correlated with Bacteroidetes and Succinivibrio. During the chronic phase of infection, we observed a significant increase in gut microbial diversity, alterations in community composition (including a decrease in Proteobacteria/Succinivibrio in the gut) and functionality (including a decrease in genes involved in bacterial invasion of epithelial cells in the gut), and partial reversibility of acute infection-related shifts in microbial abundance observed in the fecal microbiome. As part of our study, we also developed an accurate predictor of SIV infection using fecal samples. CONCLUSIONS The vervets infected with SIV and humans infected with HIV differ in microbial responses to infection. These responses to SIV infection may aid in preventing microbial translocation and subsequent disease progression in vervets, and may represent host microbiome adaptations to the virus. Video Abstract.
Collapse
Affiliation(s)
- Anna J Jasinska
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Molecular Genetics, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland.
- Eye on Primates, Los Angeles, CA, USA.
| | - Tien S Dong
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Venu Lagishetty
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - William Katzka
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jonathan P Jacobs
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- UCLA Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | - Jennifer Danzy Cramer
- Department of Sociology, Anthropology, and General Studies, American Public University System, Charles Town, WV, USA
| | - Dongzhu Ma
- Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Willem G Coetzer
- Department of Genetics, University of the Free State, Bloemfontein, South Africa
| | - J Paul Grobler
- Department of Genetics, University of the Free State, Bloemfontein, South Africa
| | - Trudy R Turner
- Department of Genetics, University of the Free State, Bloemfontein, South Africa
- Department of Anthropology, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Nelson Freimer
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA
| | - Ivona Pandrea
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Cristian Apetrei
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
30
|
Chiou KL, Montague MJ, Goldman EA, Watowich MM, Sams SN, Song J, Horvath JE, Sterner KN, Ruiz-Lambides AV, Martínez MI, Higham JP, Brent LJN, Platt ML, Snyder-Mackler N. Rhesus macaques as a tractable physiological model of human ageing. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190612. [PMID: 32951555 DOI: 10.1098/rstb.2019.0612] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Research in the basic biology of ageing is increasingly identifying mechanisms and modifiers of ageing in short-lived organisms such as worms and mice. The ultimate goal of such work is to improve human health, particularly in the growing segment of the population surviving into old age. Thus far, few interventions have robustly transcended species boundaries in the laboratory, suggesting that changes in approach are needed to avoid costly failures in translational human research. In this review, we discuss both well-established and alternative model organisms for ageing research and outline how research in nonhuman primates is sorely needed, first, to translate findings from short-lived organisms to humans, and second, to understand key aspects of ageing that are unique to primate biology. We focus on rhesus macaques as a particularly promising model organism for ageing research owing to their social and physiological similarity to humans as well as the existence of key resources that have been developed for this species. As a case study, we compare gene regulatory signatures of ageing in the peripheral immune system between humans and rhesus macaques from a free-ranging study population in Cayo Santiago. We show that both mRNA expression and DNA methylation signatures of immune ageing are broadly shared between macaques and humans, indicating strong conservation of the trajectory of ageing in the immune system. We conclude with a review of key issues in the biology of ageing for which macaques and other nonhuman primates may uniquely contribute valuable insights, including the effects of social gradients on health and ageing. We anticipate that continuing research in rhesus macaques and other nonhuman primates will play a critical role in conjunction with the model organism and human biodemographic research in ultimately improving translational outcomes and extending health and longevity in our ageing population. This article is part of the theme issue 'Evolution of the primate ageing process'.
Collapse
Affiliation(s)
- Kenneth L Chiou
- Department of Psychology, University of Washington, Seattle, WA 98195, USA.,Department of Pathology, Nathan Shock Center of Excellence in the Basic Biology of Aging, University of Washington, Seattle, WA 98195, USA.,Center for Evolution and Medicine, Arizona State University, Tempe, AZ 85281, USA
| | - Michael J Montague
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Marina M Watowich
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Sierra N Sams
- Department of Psychology, University of Washington, Seattle, WA 98195, USA
| | - Jeff Song
- Department of Biological and Biomedical Sciences, North Carolina Central University, Durham, NC 27707, USA
| | - Julie E Horvath
- Department of Biological and Biomedical Sciences, North Carolina Central University, Durham, NC 27707, USA.,Research and Collections Section, North Carolina Museum of Natural Sciences, Raleigh, NC 27601, USA.,Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA.,Department of Evolutionary Anthropology, Duke University, Durham, NC 27708, USA
| | - Kirstin N Sterner
- Department of Anthropology, University of Oregon, Eugene, OR 97403, USA
| | - Angelina V Ruiz-Lambides
- Caribbean Primate Research Center, Unit of Comparative Medicine, University of Puerto Rico, San Juan, PR 00936, USA
| | - Melween I Martínez
- Caribbean Primate Research Center, Unit of Comparative Medicine, University of Puerto Rico, San Juan, PR 00936, USA
| | - James P Higham
- Department of Anthropology, New York University, New York, NY 10003, USA.,New York Consortium in Evolutionary Primatology, New York, NY, USA
| | - Lauren J N Brent
- Centre for Research in Animal Behaviour, University of Exeter, Exeter EX4 4QG, UK
| | - Michael L Platt
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Psychology, University of Pennsylvania, Philadelphia, PA 19104, USA.,Department of Marketing, Wharton School of Business, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Noah Snyder-Mackler
- Department of Psychology, University of Washington, Seattle, WA 98195, USA.,Department of Pathology, Nathan Shock Center of Excellence in the Basic Biology of Aging, University of Washington, Seattle, WA 98195, USA.,Department of Biology, University of Washington, Seattle, WA 98195, USA.,Center for Studies in Demography and Ecology, University of Washington, Seattle, WA 98195, USA.,Center for Evolution and Medicine, Arizona State University, Tempe, AZ 85281, USA.,School of Life Sciences, Arizona State University, Tempe, AZ 85281, USA
| |
Collapse
|
31
|
Hartman AL, Nambulli S, McMillen CM, White AG, Tilston-Lunel NL, Albe JR, Cottle E, Dunn MD, Frye LJ, Gilliland TH, Olsen EL, O’Malley KJ, Schwarz MM, Tomko JA, Walker RC, Xia M, Hartman MS, Klein E, Scanga CA, Flynn JL, Klimstra WB, McElroy AK, Reed DS, Duprex WP. SARS-CoV-2 infection of African green monkeys results in mild respiratory disease discernible by PET/CT imaging and shedding of infectious virus from both respiratory and gastrointestinal tracts. PLoS Pathog 2020; 16:e1008903. [PMID: 32946524 PMCID: PMC7535860 DOI: 10.1371/journal.ppat.1008903] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/05/2020] [Accepted: 08/19/2020] [Indexed: 01/19/2023] Open
Abstract
Vaccines are urgently needed to combat the global coronavirus disease 2019 (COVID-19) pandemic, and testing of candidate vaccines in an appropriate non-human primate (NHP) model is a critical step in the process. Infection of African green monkeys (AGM) with a low passage human isolate of SARS-CoV-2 by aerosol or mucosal exposure resulted in mild clinical infection with a transient decrease in lung tidal volume. Imaging with human clinical-grade 18F-fluoro-2-deoxy-D-glucose positron emission tomography (18F-FDG PET) co-registered with computed tomography (CT) revealed pulmonary lesions at 4 days post-infection (dpi) that resolved over time. Infectious virus was shed from both respiratory and gastrointestinal (GI) tracts in all animals in a biphasic manner, first between 2-7 dpi followed by a recrudescence at 14-21 dpi. Viral RNA (vRNA) was found throughout both respiratory and gastrointestinal systems at necropsy with higher levels of vRNA found within the GI tract tissues. All animals seroconverted simultaneously for IgM and IgG, which has also been documented in human COVID-19 cases. Young AGM represent an species to study mild/subclinical COVID-19 disease and with possible insights into live virus shedding. Future vaccine evaluation can be performed in AGM with correlates of efficacy being lung lesions by PET/CT, virus shedding, and tissue viral load.
Collapse
Affiliation(s)
- Amy L. Hartman
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Sham Nambulli
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Cynthia M. McMillen
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Alexander G. White
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Natasha Louise Tilston-Lunel
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Joseph R. Albe
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Emily Cottle
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Matthew D. Dunn
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - L. James Frye
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Theron H. Gilliland
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Emily L. Olsen
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Katherine J. O’Malley
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Madeline M. Schwarz
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Jaime A. Tomko
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Reagan C. Walker
- Division of Laboratory Animal Resources, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Mengying Xia
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Matthew S. Hartman
- Department of Radiology, Allegheny Health Network, Pittsburgh, Pennsylvania, United States of America
| | - Edwin Klein
- Division of Laboratory Animal Resources, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Charles A. Scanga
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - JoAnne L. Flynn
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - William B. Klimstra
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Anita K. McElroy
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Pediatrics, Division of Pediatric Infectious Disease, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Douglas S. Reed
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - W. Paul Duprex
- Center for Vaccine Research, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
32
|
Suárez-Bonnet A, Priestnall SL, Ramírez GA, Molín J, Jaber JR. Aberrant Expression of Cell Cycle Regulator 14-3-3-σ and E-Cadherin in a Metastatic Cholangiocarcinoma in a Vervet Monkey (Chlorocebus pygerythrus). J Comp Pathol 2020; 179:25-30. [PMID: 32958143 DOI: 10.1016/j.jcpa.2020.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/04/2020] [Accepted: 07/01/2020] [Indexed: 01/20/2023]
Abstract
We present a unique case of metastatic cholangiocarcinoma with concurrent abdominal cestodiasis in an African green monkey (Chlorocebus pygerythrus) that presented with respiratory insufficiency and abdominal discomfort. There were multiple white-grey masses in the liver and colonic serosa alongside intra-abdominal parasitic cysts. Histopathologically, the liver masses were composed of poorly-differentiated epithelial cells that formed densely cellular solid areas and trabeculae. The neoplastic cells were strongly immunopositive for CK7 but negative for Hep-Par1 antigen, which confirmed a diagnosis of cholangiocarcinoma. Interestingly, there was strong and diffuse neoexpression in the tumour of the cell cycle regulator 14-3-3σ, which is not constitutively expressed in normal liver. There was aberrantly strong expression of E-cadherin, a key cell-cell adhesion protein, in neoplastic cells with evidence of cytoplasmic internalization. This is the first immunohistochemical analysis of 14-3-3σ and E-cadherin in a liver neoplasm in an animal species and the use of these markers requires further investigation in animal liver neoplasms.
Collapse
Affiliation(s)
- A Suárez-Bonnet
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, Hatfield, UK.
| | - S L Priestnall
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, Hatfield, UK
| | - G A Ramírez
- Department of Animal Science, Universitat de Lleida, Lleida, Spain
| | - J Molín
- Department of Animal Science, Universitat de Lleida, Lleida, Spain
| | - J R Jaber
- Morphology Department, Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain
| |
Collapse
|
33
|
Schmitt CA, Bergey CM, Jasinska AJ, Ramensky V, Burt F, Svardal H, Jorgensen MJ, Freimer NB, Grobler JP, Turner TR. ACE2 and TMPRSS2 variation in savanna monkeys (Chlorocebus spp.): Potential risk for zoonotic/anthroponotic transmission of SARS-CoV-2 and a potential model for functional studies. PLoS One 2020; 15:e0235106. [PMID: 32574196 PMCID: PMC7310727 DOI: 10.1371/journal.pone.0235106] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 06/08/2020] [Indexed: 01/04/2023] Open
Abstract
The COVID-19 pandemic, caused by the coronavirus SARS-CoV-2, has devastated health infrastructure around the world. Both ACE2 (an entry receptor) and TMPRSS2 (used by the virus for spike protein priming) are key proteins to SARS-CoV-2 cell entry, enabling progression to COVID-19 in humans. Comparative genomic research into critical ACE2 binding sites, associated with the spike receptor binding domain, has suggested that African and Asian primates may also be susceptible to disease from SARS-CoV-2 infection. Savanna monkeys (Chlorocebus spp.) are a widespread non-human primate with well-established potential as a bi-directional zoonotic/anthroponotic agent due to high levels of human interaction throughout their range in sub-Saharan Africa and the Caribbean. To characterize potential functional variation in savanna monkey ACE2 and TMPRSS2, we inspected recently published genomic data from 245 savanna monkeys, including 163 wild monkeys from Africa and the Caribbean and 82 captive monkeys from the Vervet Research Colony (VRC). We found several missense variants. One missense variant in ACE2 (X:14,077,550; Asp30Gly), common in Ch. sabaeus, causes a change in amino acid residue that has been inferred to reduce binding efficiency of SARS-CoV-2, suggesting potentially reduced susceptibility. The remaining populations appear as susceptible as humans, based on these criteria for receptor usage. All missense variants observed in wild Ch. sabaeus populations are also present in the VRC, along with two splice acceptor variants (at X:14,065,076) not observed in the wild sample that are potentially disruptive to ACE2 function. The presence of these variants in the VRC suggests a promising model for SARS-CoV-2 infection and vaccine and therapy development. In keeping with a One Health approach, characterizing actual susceptibility and potential for bi-directional zoonotic/anthroponotic transfer in savanna monkey populations may be an important consideration for controlling COVID-19 epidemics in communities with frequent human/non-human primate interactions that, in many cases, may have limited health infrastructure.
Collapse
Affiliation(s)
- Christopher A. Schmitt
- Department of Anthropology, Boston University, Boston, Massachusetts, United States of America
| | - Christina M. Bergey
- Department of Genetics, Rutgers University, New Brunswick, New Jersey, United States of America
| | - Anna J. Jasinska
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior,University of California—Los Angeles, Los Angeles, California, United States of America
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
- Eye on Primates, Los Angeles, California, United States of America
| | - Vasily Ramensky
- Federal State Institution “National Medical Research Center for Therapy and Preventive Medicine” of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Felicity Burt
- Division of Medical Virology, National Health Laboratory Service, Bloemfontein, Free State, South Africa
- Division of Virology, Faculty of Health Sciences, University of the Free State, Bloemfontein, Free State, South Africa
| | - Hannes Svardal
- Department of Biology, University of Antwerp, Antwerp, Belgium
- Naturalis Biodiversity Center, Leiden, The Netherlands
| | - Matthew J. Jorgensen
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, United States of America
| | - Nelson B. Freimer
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior,University of California—Los Angeles, Los Angeles, California, United States of America
| | - J. Paul Grobler
- Department of Genetics, University of the Free State, Bloemfontein, Free State, South Africa
| | - Trudy R. Turner
- Department of Genetics, University of the Free State, Bloemfontein, Free State, South Africa
- Department of Anthropology, University of Wisconsin–Milwaukee, Milwaukee, Wisconsin, United States of America
| |
Collapse
|
34
|
Rajeev S, Bolfa P, Shiokawa K, Beierschmitt A, Palmour R. Leptospira Infection in African Green Monkeys in an Endemic Area: An Opportunity for Comparative Studies in a Natural Environment. Pathogens 2020; 9:E474. [PMID: 32560055 PMCID: PMC7350389 DOI: 10.3390/pathogens9060474] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/11/2020] [Accepted: 06/11/2020] [Indexed: 12/12/2022] Open
Abstract
This study was performed to investigate the potential asymptomatic Leptospira reservoir status among African green monkeys (AGMs) in the Caribbean island of Saint Kitts, and whether there is any renal pathology associated with Leptospira exposure. Forty-eight percent of AGMs tested were positive for Leptospira antibodies by the microscopic agglutination test. Leptospira DNA was detected in 4% of kidney samples tested using a lipl32 gene based PCR. We observed minimal to severe microscopic renal lesions in 85% of the AGM kidneys evaluated. The majority of the AGMs (n = 26) had only minimal to mild interstitial nephritis and a few (n = 3) had moderate to severe lesions. The presence of interstitial nephritis was not significantly associated with Leptospira exposure. The presence of infected AGMs in a small surface limited geographic region may pose zoonotic threat to humans and animals. The impact of Leptospira infection in renal pathology in AGMs warrants further investigation. AGMs residing in a natural setting in an insular, surface limited Leptospira endemic geographic region may offer opportunities for comparative studies to advance the field of leptospirosis. Due to their similarity to humans, such studies in AGMs may also provide translational opportunities to advance Leptospira research.
Collapse
Affiliation(s)
- Sreekumari Rajeev
- School of Veterinary Medicine, Ross University, Basseterre KN 0101, Saint Kitts and Nevis; (P.B.); (K.S.); (A.B.)
| | - Pompei Bolfa
- School of Veterinary Medicine, Ross University, Basseterre KN 0101, Saint Kitts and Nevis; (P.B.); (K.S.); (A.B.)
| | - Kanae Shiokawa
- School of Veterinary Medicine, Ross University, Basseterre KN 0101, Saint Kitts and Nevis; (P.B.); (K.S.); (A.B.)
| | - Amy Beierschmitt
- School of Veterinary Medicine, Ross University, Basseterre KN 0101, Saint Kitts and Nevis; (P.B.); (K.S.); (A.B.)
- Behavioral Science Foundation, Basseterre KN 0101, Saint Kitts and Nevis;
| | - Roberta Palmour
- Behavioral Science Foundation, Basseterre KN 0101, Saint Kitts and Nevis;
- Departments of Human Genetics and Psychiatry, Faculty of Medicine, McGill University, Montréal, QC H3A 1A1, Canada
| |
Collapse
|
35
|
Jasinska AJ. Resources for functional genomic studies of health and development in nonhuman primates. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2020; 171 Suppl 70:174-194. [PMID: 32221967 DOI: 10.1002/ajpa.24051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 01/22/2020] [Accepted: 02/26/2020] [Indexed: 01/01/2023]
Abstract
Primates display a wide range of phenotypic variation underlaid by complex genetically regulated mechanisms. The links among DNA sequence, gene function, and phenotype have been of interest from an evolutionary perspective, to understand functional genome evolution and its phenotypic consequences, and from a biomedical perspective to understand the shared and human-specific roots of health and disease. Progress in methods for characterizing genetic, transcriptomic, and DNA methylation (DNAm) variation is driving the rapid development of extensive omics resources, which are now increasingly available from humans as well as a growing number of nonhuman primates (NHPs). The fast growth of large-scale genomic data is driving the emergence of integrated tools and databases, thus facilitating studies of gene functionality across primates. This review describes NHP genomic resources that can aid in exploration of how genes shape primate phenotypes. It focuses on the gene expression trajectories across development in different tissues, the identification of functional genetic variation (including variants deleterious for protein function and regulatory variants modulating gene expression), and DNAm profiles as an emerging tool to understand the process of aging. These resources enable comparative functional genomics approaches to identify species-specific and primate-shared gene functionalities associated with health and development.
Collapse
Affiliation(s)
- Anna J Jasinska
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, California, USA.,Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland.,Eye on Primates, Los Angeles, California, USA
| |
Collapse
|
36
|
Li W, Qiang X, Qin S, Huang Y, Hu Y, Bai B, Hou J, Gao R, Zhang X, Mi Z, Fan H, Ye H, Tong Y, Mao P. Virome diversity analysis reveals novel enteroviruses and a human picobirnavirus in stool samples from African green monkeys with diarrhea. INFECTION GENETICS AND EVOLUTION 2020; 82:104279. [PMID: 32165243 PMCID: PMC7102571 DOI: 10.1016/j.meegid.2020.104279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/05/2020] [Accepted: 03/07/2020] [Indexed: 12/12/2022]
Abstract
It is important to identify viruses in animals because most infectious diseases in humans are caused by viruses of zoonotic origin. African green monkey is a widely used non-human primate model in biomedical investigations. In this study, total RNAs were extracted from stool samples of 10 African green monkeys with diarrhea. High-throughput sequencing was used to characterize viromes. PCR and Sanger sequencing were used to determine the full genome sequences. Great viral diversity was observed. The dominant viruses were enteroviruses and picobirnaviruses. Six enterovirus genomes and a picobirnavirus RNA-dependent RNA polymerase sequence were characterized. Five enteroviruses belonged to two putative new genotypes of species Enterovirus J. One enterovirus belonged to EV-A92. The picobirnavirus RNA-dependent RNA polymerase sequence had the highest nucleotide similarity (93.48%) with human picobirnavirus isolate GPBV6C2. The present study helped to identify the potential zoonotic viruses in African green monkeys. Further investigations are required to elucidate their pathogenic roles in animals and humans.
Collapse
Affiliation(s)
- Wenjuan Li
- Chinese PLA Medical School, Beijing 100853, China; Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Xin Qiang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Si Qin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yong Huang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yan Hu
- Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Bingke Bai
- Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Jun Hou
- Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Rong Gao
- Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China
| | - Xianglilan Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Zhiqiang Mi
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Hang Fan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China.
| | - Huahu Ye
- Laboratory Animal Center of the Academy of Military Medical Sciences, Beijing 100071, China.
| | - Yigang Tong
- BAIC-SM, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Panyong Mao
- Chinese PLA Medical School, Beijing 100853, China; Fifth Medical Center of Chinese PLA General Hospital, Beijing 100039, China.
| |
Collapse
|
37
|
Jasinska AJ, Pandrea I, He T, Benjamin C, Newton M, Lee JC, Freimer NB, Coppola G, Jentsch JD. Immunosuppressive effect and global dysregulation of blood transcriptome in response to psychosocial stress in vervet monkeys (Chlorocebus sabaeus). Sci Rep 2020; 10:3459. [PMID: 32103041 PMCID: PMC7044305 DOI: 10.1038/s41598-020-59934-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 02/03/2020] [Indexed: 11/09/2022] Open
Abstract
Psychosocial stressors - life events that challenge social support and relationships - represent powerful risk factors for human disease; included amongst these events are relocation, isolation and displacement. To evaluate the impact of a controlled psychosocial stressor on physiology and underlying molecular pathways, we longitudinally studied the influence of a 28-day period of quarantine on biomarkers of immune signalling, microbial translocation, glycaemic health and blood transcriptome in the wild-born vervet monkey. This event caused a coordinated, mostly transient, reduction of circulating levels of nine immune signalling molecules. These were paralleled by a massive dysregulation of blood transcriptome, including genes implicated in chronic pathologies and immune functions. Immune and inflammatory functions were enriched among the genes downregulated in response to stress. An upregulation of genes involved in blood coagulation, platelet activation was characteristic of the rapid response to stress induction. Stress also decreased neutrophils and increased CD4 + T cell proportions in blood. This model of psychosocial stress, characterised by an immune dysregulation at the transcriptomic, molecular and cellular levels, creates opportunities to uncover the underlying mechanisms of stress-related diseases with an immune component, including cardiovascular diseases and susceptibility to infections.
Collapse
Affiliation(s)
- Anna J Jasinska
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, The University of California Los Angeles, California, USA.
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland.
| | - Ivona Pandrea
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Tianyu He
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Cassandra Benjamin
- St. Kitts Biomedical Research Foundation, St. Kitts, West Indies, Saint Kitts and Nevis
| | - Maurice Newton
- St. Kitts Biomedical Research Foundation, St. Kitts, West Indies, Saint Kitts and Nevis
| | - Jen Chieh Lee
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, The University of California Los Angeles, California, USA
| | - Nelson B Freimer
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, The University of California Los Angeles, California, USA
| | - Giovanni Coppola
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, The University of California Los Angeles, California, USA
- Department of Neurology, The University of California Los Angeles, California, USA
| | - James D Jentsch
- Department of Psychology, Binghamton University, Binghamton, NY, 13902, USA
| |
Collapse
|
38
|
Jasinska AJ, Rostamian D, Davis AT, Kavanagh K. Transcriptomic Analysis of Cell-free Fetal RNA in the Amniotic Fluid of Vervet Monkeys ( Chlorocebus sabaeus). Comp Med 2020; 70:67-74. [PMID: 31969210 PMCID: PMC7024774 DOI: 10.30802/aalas-cm-19-000037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 04/22/2019] [Accepted: 05/15/2019] [Indexed: 12/15/2022]
Abstract
NHP are important translational models for understanding the genomic underpinnings of growth, development, fetal programming, and predisposition to disease, with potential for the development of early health biomarkers. Understanding how prenatal gene expression is linked to pre- and postnatal health and development requires methods for assessing the fetal transcriptome. Here we used RNAseq methodology to analyze the expression of cell-free fetal RNA in the amniotic fluid supernatant (AFS) of vervet monkeys. Despite the naturally high level of degradation of free-floating RNA, we detected more than 10,000 gene transcripts in vervet AFS. The most highly expressed genes were H19, IGF2, and TPT1, which are involved in embryonic growth and glycemic health. We noted global similarities in expression profiles between vervets and humans, with genes involved in embryonic growth and glycemic health among the genes most highly expressed in AFS. Our study demonstrates both the feasibility and usefulness of prenatal transcriptomic profiles, by using amniocentesis procedures to obtain AFS and cell-free fetal RNA from pregnant vervets.
Collapse
Affiliation(s)
- Anna J Jasinska
- Center for Neurobehavioral Genetics, University of California-Los Angeles, Los Angeles, California; Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland;,
| | - Dalar Rostamian
- Center for Neurobehavioral Genetics, University of California-Los Angeles, Los Angeles, California
| | - Ashley T Davis
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Kylie Kavanagh
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina; Department of Biomedicine, University of Tasmania, Hobart, Australia
| |
Collapse
|
39
|
Ngqaneka T, Khoza S, Magwebu ZE, Chauke CG. Mutational analysis of BFSP1, CRYBB1, GALK1, and GJA8 in captive-bred vervet monkeys (Chlorocebus aethiops). J Med Primatol 2020; 49:79-85. [PMID: 31975409 DOI: 10.1111/jmp.12455] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 11/08/2019] [Accepted: 12/06/2019] [Indexed: 11/28/2022]
Abstract
BACKGROUND Congenital cataract has been reported in a colony of captive-bred vervet monkeys (Chlorocebus aethiops). METHODS Molecular tools such as genotyping and gene expression were used to identify mutations associated with congenital cataract in this vervet colony. Beaded filament structural protein 1 (BFSP1), beta-crystallin B1 (CRYBB1), galactokinase1 (GALK1), and gap junction alpha-8 protein (GJA8) were screened, sequenced, and analyzed for mutations in 24 vervet monkeys (control and cataract). RESULTS Five missense sequence variants were identified (V147E, A167P, L212F, N55K, and T247A), three of which were found to be potentially disease-causing. Furthermore, downregulation was observed in BFSP1, CRYBB1, and GALK1 genes. CONCLUSION This study reports two cases of incomplete penetrance and/or uniparental disomy (L212F and T247A) in BSFP1. Mutations in BSFP1 together with three mutations in GALK1 and GJA8 were predicted to be disease-causing.
Collapse
Affiliation(s)
- Thobile Ngqaneka
- Primate Unit and Delft Animal Centre (PUDAC), South African Medical Research Council, Tygerberg, South Africa
| | - Sanele Khoza
- Primate Unit and Delft Animal Centre (PUDAC), South African Medical Research Council, Tygerberg, South Africa
| | - Zandisiwe E Magwebu
- Primate Unit and Delft Animal Centre (PUDAC), South African Medical Research Council, Tygerberg, South Africa
| | - Chesa G Chauke
- Primate Unit and Delft Animal Centre (PUDAC), South African Medical Research Council, Tygerberg, South Africa
| |
Collapse
|
40
|
Simons ND, Eick GN, Ruiz-Lopez MJ, Hyeroba D, Omeja PA, Weny G, Zheng H, Shankar A, Frost SDW, Jones JH, Chapman CA, Switzer WM, Goldberg TL, Sterner KN, Ting N. Genome-Wide Patterns of Gene Expression in a Wild Primate Indicate Species-Specific Mechanisms Associated with Tolerance to Natural Simian Immunodeficiency Virus Infection. Genome Biol Evol 2019; 11:1630-1643. [PMID: 31106820 PMCID: PMC6561381 DOI: 10.1093/gbe/evz099] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2019] [Indexed: 12/12/2022] Open
Abstract
Over 40 species of nonhuman primates host simian immunodeficiency viruses (SIVs). In natural hosts, infection is generally assumed to be nonpathogenic due to a long coevolutionary history between host and virus, although pathogenicity is difficult to study in wild nonhuman primates. We used whole-blood RNA-seq and SIV prevalence from 29 wild Ugandan red colobus (Piliocolobus tephrosceles) to assess the effects of SIV infection on host gene expression in wild, naturally SIV-infected primates. We found no evidence for chronic immune activation in infected individuals, suggesting that SIV is not immunocompromising in this species, in contrast to human immunodeficiency virus in humans. Notably, an immunosuppressive gene, CD101, was upregulated in infected individuals. This gene has not been previously described in the context of nonpathogenic SIV infection. This expands the known variation associated with SIV infection in natural hosts and may suggest a novel mechanism for tolerance of SIV infection in the Ugandan red colobus.
Collapse
Affiliation(s)
| | - Geeta N Eick
- Department of Anthropology, University of Oregon
| | | | - David Hyeroba
- College of Veterinary Medicine, Animal Resources, and Bio-Security, Makerere University, Kampala, Uganda
| | - Patrick A Omeja
- Makerere University Biological Field Station, Fort Portal, Uganda
| | - Geoffrey Weny
- Makerere University Biological Field Station, Fort Portal, Uganda
| | - HaoQiang Zheng
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV, Hepatitis, STD and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Anupama Shankar
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV, Hepatitis, STD and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Simon D W Frost
- Department of Veterinary Medicine, University of Cambridge, United Kingdom
| | - James H Jones
- Department of Earth System Science, Woods Institute for the Environment, Stanford University
| | - Colin A Chapman
- Makerere University Biological Field Station, Fort Portal, Uganda
- Department of Anthropology, McGill School of Environment, McGill University, Montreal, Quebec, Canada
| | - William M Switzer
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV, Hepatitis, STD and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Tony L Goldberg
- Department of Pathobiological Sciences, University of Wisconsin-Madison
- Global Health Institute, University of Wisconsin-Madison
| | | | - Nelson Ting
- Department of Anthropology, University of Oregon
- Institute of Ecology and Evolution, University of Oregon
| |
Collapse
|
41
|
Nagpal R, Shively CA, Register TC, Craft S, Yadav H. Gut microbiome-Mediterranean diet interactions in improving host health. F1000Res 2019; 8:699. [PMID: 32704349 PMCID: PMC7359750 DOI: 10.12688/f1000research.18992.1] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/10/2019] [Indexed: 12/22/2022] Open
Abstract
The gut microbiota plays a fundamental role in host health and disease. Host diet is one of the most significant modulators of the gut microbial community and its metabolic activities. Evidence demonstrates that dietary patterns such as the 'Western diet' and perturbations in gut microbiome (dysbiosis) have strong associations with a wide range of human diseases, including obesity, metabolic syndrome, type-2 diabetes and cardiovascular diseases. However, consumption of Mediterranean-style diets is considered healthy and associated with the prevention of cardiovascular and metabolic diseases, colorectal cancers and many other diseases. Such beneficial effects of the Mediterranean diet might be attributed to high proportion of fibers, mono- and poly-unsaturated fatty acids, antioxidants and polyphenols. Concurrent literature has demonstrated beneficial modulation of the gut microbiome following a Mediterranean-style diet in humans as well as in experimental animal models such as rodents. We recently demonstrated similar positive changes in the gut microbiome of non-human primates consuming a Mediterranean-style diet for long term (30 months). Therefore, it is rational to speculate that this positive modulation of the gut microbiome diversity, composition and function is one of the main factors intermediating the health effects of Mediterranean diet on the host. The present perspective discusses the evidences that the Mediterranean diet induces gut microbiome modulation in rodents, non-human primates and human subjects, and discusses the potential role of gut microbiota and microbial metabolites as one of the fundamental catalysts intermediating various beneficial health effects of Mediterranean diet on the host.
Collapse
Affiliation(s)
- Ravinder Nagpal
- Division of Internal Medicine - Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, 27101, USA
- Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, North Carolina, 27101, USA
| | - Carol A. Shively
- Department of Pathology - Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, 27101, USA
| | - Thomas C. Register
- Department of Pathology - Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, 27101, USA
| | - Suzanne Craft
- Department of Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, 27101, USA
| | - Hariom Yadav
- Division of Internal Medicine - Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, 27101, USA
- Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, North Carolina, 27101, USA
| |
Collapse
|
42
|
Multivariate profiling of African green monkey and rhesus macaque T lymphocytes. Sci Rep 2019; 9:4834. [PMID: 30886198 PMCID: PMC6423277 DOI: 10.1038/s41598-019-41209-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 02/27/2019] [Indexed: 12/22/2022] Open
Abstract
The complexity of immune responses limits the usefulness of univariate methods in answering complex immunology questions. To demonstrate the utility of a multivariate approach, we employ such approach to compare T cells of African green monkeys (AGMs) and rhesus macaques (RMs). Among the most prominent distinguishing features we found were lower CD3 and higher CD28 surface expression in AGMs compared to RMs. After in vitro stimulation, a larger proportion of AGM T cells secreted cytokines, especially those producing more than one cytokine (i.e. multifunctional cells). To find out whether multifunctional responses associate with protection in other species, we compared T cells of cynomolgus macaques (CMs) infected with wild-type Simian Immunodeficiency Virus (SIV) to those of CMs infected (vaccinated) with a replication-defective virus. Wild-type SIV infection in macaques leads to simian Acquired Immunodeficiency Syndrome (AIDS), which does not happen in animals previously vaccinated with a replication-defective virus. Interestingly, after in vitro stimulation, multifunctional cells were more abundant among T cells of vaccinated CMs. Our results propose T-cell multifunctionality as a potentially useful marker of immunity, although additional verification is needed. Finally, we hope our multivariate model and its associated validation methods will inform future studies in the field of immunology.
Collapse
|
43
|
A nonhuman primate model of early Alzheimer's disease pathologic change: Implications for disease pathogenesis. Alzheimers Dement 2018; 15:93-105. [PMID: 30467082 PMCID: PMC6383152 DOI: 10.1016/j.jalz.2018.06.3057] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 06/18/2018] [Accepted: 06/28/2018] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Nonhuman primates may serve as excellent models of sporadic age-associated brain β-amyloid deposition and Alzheimer's disease pathologic changes. We examined whether a vervet nonhuman primate model recapitulated pathologic, physiologic, and behavioral features of early Alzheimer's disease. METHODS Nine middle-aged (mean = 11.2 years) and nine aged (mean = 21.7 years) female vervet/African green monkeys underwent cerebrospinal fluid collection, gait speed measurement, and neuroimaging before neuropathologic assessment. RESULTS β-amyloid plaques were identified in all aged vervets and paired helical filament tau immunoreactivity was observed in all animals. Cerebrospinal fluid β-amyloid42 and gait speed correlated negatively with age and plaque density. Greater plaque and paired helical filament tau burden predicted reduced volumes and CMRg in several brain regions. DISCUSSION We observed a coordinated set of relationships among neuropathologic, cerebrospinal fluid, imaging, and behavioral modalities consistent with early Alzheimer's disease. Our results support future use of the vervet model to explore disease mechanisms, biomarkers, and novel therapeutic strategies.
Collapse
|
44
|
Wilson QN, Wells M, Davis AT, Sherrill C, Tsilimigras MCB, Jones RB, Fodor AA, Kavanagh K. Greater Microbial Translocation and Vulnerability to Metabolic Disease in Healthy Aged Female Monkeys. Sci Rep 2018; 8:11373. [PMID: 30054517 PMCID: PMC6063974 DOI: 10.1038/s41598-018-29473-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 07/06/2018] [Indexed: 12/14/2022] Open
Abstract
Monkeys demonstrate gastrointestinal barrier dysfunction (leaky gut) as evidenced by higher biomarkers of microbial translocation (MT) and inflammation with ageing despite equivalent health status, and lifelong diet and environmental conditions. We evaluated colonic structural, microbiomic and functional changes in old female vervet monkeys (Chlorocebus aethiops sabeus) and how age-related leaky gut alters responses to Western diet. We additionally assessed serum bovine immunoglobulin therapy to lower MT burden. MT was increased in old monkeys despite comparable histological appearance of the ascending colon. Microbiome profiles from 16S sequencing did not show large differences by age grouping, but there was evidence for higher mucosal bacterial loads using qPCR. Innate immune responses were increased in old monkeys consistent with higher MT burdens. Western diet challenge led to elevations in glycemic and hepatic biochemistry values only in old monkeys, and immunoglobulin therapy was not effective in reducing MT markers or improving metabolic health. We interpret these findings to suggest that ageing may lead to lower control over colonization at the mucosal surface, and reduced clearance of pathogens resulting in MT and inflammation. Leaky gut in ageing, which is not readily rescued by innate immune support with immunoglobulin, primes the liver for negative consequences of high fat, high sugar diets.
Collapse
Affiliation(s)
- Quentin N Wilson
- Wake Forest School of Medicine, Department of Pathology, Winston-Salem, USA
| | - Magan Wells
- Wake Forest School of Medicine, Department of Pathology, Winston-Salem, USA
| | - Ashley T Davis
- Wake Forest School of Medicine, Department of Pathology, Winston-Salem, USA
| | - Christina Sherrill
- Wake Forest School of Medicine, Department of Pathology, Winston-Salem, USA
| | - Matthew C B Tsilimigras
- University of North Carolina at Charlotte, Department of Bioinformatics and Genomics, Charlotte, USA
| | - Roshonda B Jones
- University of North Carolina at Charlotte, Department of Bioinformatics and Genomics, Charlotte, USA
| | - Anthony A Fodor
- University of North Carolina at Charlotte, Department of Bioinformatics and Genomics, Charlotte, USA
| | - Kylie Kavanagh
- Wake Forest School of Medicine, Department of Pathology, Winston-Salem, USA.
| |
Collapse
|
45
|
Turner TR, Schmitt CA, Cramer JD, Lorenz J, Grobler JP, Jolly CJ, Freimer NB. Morphological variation in the genus Chlorocebus: Ecogeographic and anthropogenically mediated variation in body mass, postcranial morphology, and growth. AMERICAN JOURNAL OF PHYSICAL ANTHROPOLOGY 2018; 166:682-707. [PMID: 29577231 PMCID: PMC6039265 DOI: 10.1002/ajpa.23459] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 02/28/2018] [Accepted: 03/01/2018] [Indexed: 01/23/2023]
Abstract
OBJECTIVES Direct comparative work in morphology and growth on widely dispersed wild primate taxa is rarely accomplished, yet critical to understanding ecogeographic variation, plastic local variation in response to human impacts, and variation in patterns of growth and sexual dimorphism. We investigated population variation in morphology and growth in response to geographic variables (i.e., latitude, altitude), climatic variables (i.e., temperature and rainfall), and human impacts in the vervet monkey (Chlorocebus spp.). METHODS We trapped over 1,600 wild vervets from across Sub-Saharan Africa and the Caribbean, and compared measurements of body mass, body length, and relative thigh, leg, and foot length in four well-represented geographic samples: Ethiopia, Kenya, South Africa, and St. Kitts & Nevis. RESULTS We found significant variation in body mass and length consistent with Bergmann's Rule in adult females, and in adult males when excluding the St. Kitts & Nevis population, which was more sexually dimorphic. Contrary to Rensch's Rule, although the South African population had the largest average body size, it was the least dimorphic. There was significant, although very small, variation in all limb segments in support for Allen's Rule. Females in high human impact areas were heavier than those with moderate exposures, while those in low human impact areas were lighter; human impacts had no effect on males. CONCLUSIONS Vervet monkeys appear to have adapted to local climate as predicted by Bergmann's and, less consistently, Allen's Rule, while also responding in predicted ways to human impacts. To better understand deviations from predicted patterns will require further comparative work in vervets.
Collapse
Affiliation(s)
- Trudy R. Turner
- Department of Anthropology, University of Wisconsin –
Milwaukee, Milwaukee, WI 53201, USA
- Department of Genetics, University of the Free State, Bloemfontein,
FS, South Africa
| | - Christopher A. Schmitt
- Department of Anthropology, Boston University, Boston, MA 02215,
USA
- Center for Neurobehavioral Genetics, University of California
– Los Angeles, Los Angeles, CA 90095, USA
| | - Jennifer Danzy Cramer
- Department of Sociology, Anthropology and Women's Studies,
American Military University and American Public University, Charles Town, WV 25414,
USA
| | - Joseph Lorenz
- Department of Anthropology and Museum Studies, Central Washington
University, Ellensburg, WA 98926, USA
| | - J. Paul Grobler
- Department of Genetics, University of the Free State, Bloemfontein,
FS, South Africa
| | - Clifford J. Jolly
- CSHO, Department of Anthropology, New York University, and NYCEP,
New York, NY 10003, USA
| | - Nelson B. Freimer
- Center for Neurobehavioral Genetics, University of California
– Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
46
|
Zhou X, Wang J, Fa Y, Ye H. Signature microRNA expression profile is associated with spontaneous hypertension in African green monkey. Clin Exp Hypertens 2018; 41:287-291. [PMID: 29787292 DOI: 10.1080/10641963.2018.1469646] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
Chlorocebus aethiops sabaeus, the African Green monkey (AGM), has been proved to exhibit renal vascular remodeling and spontaneous hypertension. However, little is known about the roles of microRNAs (miRNAs) in this process.Using small RNA deep sequencing, we compared the plasma miRNA expression patterns between hypertensive (HT) AGMs and normotensive (NT) AGMs. Expression of miRNAs (miR-122, miR-339, miR-296-5p) was validated independently in plasma samples from 10 HT AGMs and 10 NT AGMs (fold changes are 2.0, 1.6, 2.7 respectively; all P< 0.001). Potential BP (blood pressure)-regulating mRNA targets were predicted by TargetScan and confirmed in the Vero cells. We report for the first time a circulating miRNA profile for AGM. miRNAs, such as miR-122, miR-339, miR-296-5p, may be involved in renal pathologies and spontaneous hypertension of AGM.
Collapse
Affiliation(s)
- Xiaojun Zhou
- a Laboratory Animal Center , Academy of Military Medical Sciences , Beijing , China
| | - Jin Wang
- a Laboratory Animal Center , Academy of Military Medical Sciences , Beijing , China
| | - Yunzhi Fa
- a Laboratory Animal Center , Academy of Military Medical Sciences , Beijing , China
| | - Huahu Ye
- a Laboratory Animal Center , Academy of Military Medical Sciences , Beijing , China
| |
Collapse
|
47
|
Althouse BM, Guerbois M, Cummings DAT, Diop OM, Faye O, Faye A, Diallo D, Sadio BD, Sow A, Faye O, Sall AA, Diallo M, Benefit B, Simons E, Watts DM, Weaver SC, Hanley KA. Role of monkeys in the sylvatic cycle of chikungunya virus in Senegal. Nat Commun 2018. [PMID: 29535306 PMCID: PMC5849707 DOI: 10.1038/s41467-018-03332-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Arboviruses spillover into humans either as a one-step jump from a reservoir host species into humans or as a two-step jump from the reservoir to an amplification host species and thence to humans. Little is known about arbovirus transmission dynamics in reservoir and amplification hosts. Here we elucidate the role of monkeys in the sylvatic, enzootic cycle of chikungunya virus (CHIKV) in the region around Kédougou, Senegal. Over 3 years, 737 monkeys were captured, aged using anthropometry and dentition, and tested for exposure to CHIKV by detection of neutralizing antibodies. Infant monkeys were positive for CHIKV even when the virus was not detected in a concurrent survey of mosquitoes and when population immunity was too high for monkeys alone to support continuous transmission. We conclude that monkeys in this region serve as amplification hosts of CHIKV. Additional efforts are needed to identify other hosts capable of supporting continuous circulation.
Collapse
Affiliation(s)
- Benjamin M Althouse
- Institute for Disease Modeling, Bellevue, 98005, WA, USA. .,Information School, University of Washington, Seattle, 98105, WA, USA. .,Department of Biology, New Mexico State University, Las Cruces, 88003, NM, USA.
| | - Mathilde Guerbois
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, 77555, TX, USA
| | - Derek A T Cummings
- Emerging Pathogens Institute, University of Florida, Gainesville, 32608, FL, USA
| | | | | | | | | | | | | | - Oumar Faye
- Institut Pasteur de Dakar, Dakar, Senegal
| | | | | | - Brenda Benefit
- Department of Anthropology, New Mexico State University, Las Cruces, 88003, NM, USA
| | - Evan Simons
- Department of Anthropology, New Mexico State University, Las Cruces, 88003, NM, USA
| | - Douglas M Watts
- Office of Research and Sponsored Projects, University of Texas at El Paso, El Paso, 79968, TX, USA.,Center for Biodefense and Emerging Infectious Diseases and Department of Pathology, University of Texas Medical Branch, Galveston, 77555, TX, USA
| | - Scott C Weaver
- Institute for Human Infections and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, 77555, TX, USA
| | - Kathryn A Hanley
- Department of Biology, New Mexico State University, Las Cruces, 88003, NM, USA
| |
Collapse
|
48
|
Obesity and obesogenic growth are both highly heritable and modified by diet in a nonhuman primate model, the African green monkey (Chlorocebus aethiops sabaeus). Int J Obes (Lond) 2017; 42:765-774. [PMID: 29211707 PMCID: PMC5984074 DOI: 10.1038/ijo.2017.301] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 11/10/2017] [Accepted: 11/19/2017] [Indexed: 01/14/2023]
Abstract
Objective: In humans, the ontogeny of obesity throughout the life course and the genetics underlying it has been historically difficult to study. We compared, in a non-human primate model, the lifelong growth trajectories of obese and non-obese adults to assess the heritability of and map potential genomic regions implicated in growth and obesity. Study population: A total of 905 African green monkeys, or vervets (Chlorocebus aethiops sabaeus) (472 females, 433 males) from a pedigreed captive colony. Methods: We measured fasted body weight (BW), crown-to-rump length (CRL), body-mass index (BMI) and waist circumference (WC) from 2000 to 2015. We used a longitudinal clustering algorithm to detect obesogenic growth, and logistic growth curves implemented in nonlinear mixed effects models to estimate three growth parameters. We used maximum likelihood variance decomposition methods to estimate the genetic contributions to obesity-related traits and growth parameters, including a test for the effects of a calorie-restricted dietary intervention. We used multipoint linkage analysis to map implicated genomic regions. Results: All measurements were significantly influenced by sex, and with the exception of WC, also influenced by maternal and post-natal diet. Chronic obesity outcomes were significantly associated with a pattern of extended growth duration with slow growth rates for BW. After accounting for environmental influences, all measurements were found to have a significant genetic component to variability. Linkage analysis revealed several regions suggested to be linked to obesity-related traits that are also implicated in human obesity and metabolic disorders. Conclusions: As in humans, growth patterns in vervets have a significant impact on adult obesity and are largely under genetic control with some evidence for maternal and dietary programming. These results largely mirror findings from human research, but reflect shorter developmental periods, suggesting that the vervet offers a strong genetic model for elucidating the ontogeny of human obesity.
Collapse
|
49
|
Svardal H, Jasinska AJ, Apetrei C, Coppola G, Huang Y, Schmitt CA, Jacquelin B, Ramensky V, Müller-Trutwin M, Antonio M, Weinstock G, Grobler JP, Dewar K, Wilson RK, Turner TR, Warren WC, Freimer NB, Nordborg M. Ancient hybridization and strong adaptation to viruses across African vervet monkey populations. Nat Genet 2017; 49:1705-1713. [PMID: 29083404 PMCID: PMC5709169 DOI: 10.1038/ng.3980] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 09/27/2017] [Indexed: 12/16/2022]
Abstract
Vervet monkeys are among the most widely distributed nonhuman primates, show considerable phenotypic diversity, and have long been an important biomedical model for a variety of human diseases and in vaccine research. Using whole-genome sequencing data from 163 vervets sampled from across Africa and the Caribbean, we find high diversity within and between taxa and clear evidence that taxonomic divergence was reticulate rather than following a simple branching pattern. A scan for diversifying selection across taxa identifies strong and highly polygenic selection signals affecting viral processes. Furthermore, selection scores are elevated in genes whose human orthologs interact with HIV and in genes that show a response to experimental simian immunodeficiency virus (SIV) infection in vervet monkeys but not in rhesus macaques, suggesting that part of the signal reflects taxon-specific adaptation to SIV.
Collapse
Affiliation(s)
- Hannes Svardal
- Gregor Mendel Institute, Austrian Academy of Sciences, Vienna Biocenter (VBC), Vienna, Austria
| | - Anna J Jasinska
- Center for Neurobehavioral Genetics, University of California Los Angeles, Los Angeles, USA
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Cristian Apetrei
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Giovanni Coppola
- Center for Neurobehavioral Genetics, University of California Los Angeles, Los Angeles, USA
- Department of Neurology, University of California Los Angeles, USA
| | - Yu Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China
| | | | | | - Vasily Ramensky
- Center for Neurobehavioral Genetics, University of California Los Angeles, Los Angeles, USA
- Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | | | - Martin Antonio
- Medical Research Council (MRC), The Gambia Unit, The Gambia
| | - George Weinstock
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
| | - J Paul Grobler
- Department of Genetics, University of the Free State, Bloemfontein, South Africa
| | - Ken Dewar
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Richard K Wilson
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, USA
- Department of Anthropology, University of Wisconsin-Milwaukee, Milwaukee, USA
| | - Trudy R Turner
- Department of Genetics, University of the Free State, Bloemfontein, South Africa
| | - Wesley C Warren
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, USA
| | - Nelson B Freimer
- Center for Neurobehavioral Genetics, University of California Los Angeles, Los Angeles, USA
| | - Magnus Nordborg
- Gregor Mendel Institute, Austrian Academy of Sciences, Vienna Biocenter (VBC), Vienna, Austria
| |
Collapse
|
50
|
Jasinska AJ, Zelaya I, Service SK, Peterson CB, Cantor RM, Choi OW, DeYoung J, Eskin E, Fairbanks LA, Fears S, Furterer AE, Huang YS, Ramensky V, Schmitt CA, Svardal H, Jorgensen MJ, Kaplan JR, Villar D, Aken BL, Flicek P, Nag R, Wong ES, Blangero J, Dyer TD, Bogomolov M, Benjamini Y, Weinstock GM, Dewar K, Sabatti C, Wilson RK, Jentsch JD, Warren W, Coppola G, Woods RP, Freimer NB. Genetic variation and gene expression across multiple tissues and developmental stages in a nonhuman primate. Nat Genet 2017; 49:1714-1721. [PMID: 29083405 PMCID: PMC5714271 DOI: 10.1038/ng.3959] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 08/29/2017] [Indexed: 12/12/2022]
Abstract
By analyzing multitissue gene expression and genome-wide genetic variation data in samples from a vervet monkey pedigree, we generated a transcriptome resource and produced the first catalog of expression quantitative trait loci (eQTLs) in a nonhuman primate model. This catalog contains more genome-wide significant eQTLs per sample than comparable human resources and identifies sex- and age-related expression patterns. Findings include a master regulatory locus that likely has a role in immune function and a locus regulating hippocampal long noncoding RNAs (lncRNAs), whose expression correlates with hippocampal volume. This resource will facilitate genetic investigation of quantitative traits, including brain and behavioral phenotypes relevant to neuropsychiatric disorders.
Collapse
Affiliation(s)
- Anna J. Jasinska
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Ivette Zelaya
- Interdepartmental Program in Bioinformatics, University of California Los Angeles, Los Angeles CA, USA
| | - Susan K. Service
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Christine B. Peterson
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston TX, USA
| | - Rita M. Cantor
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA,USA
| | - Oi-Wa Choi
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Joseph DeYoung
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Eleazar Eskin
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA,USA
- Department of Computer Science, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lynn A. Fairbanks
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Scott Fears
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Allison E. Furterer
- Interdepartmental Graduate Program in Neuroscience, University of California Los Angeles, Los Angeles CA, USA
| | - Yu S. Huang
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Vasily Ramensky
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Christopher A. Schmitt
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | | | | | - Jay R. Kaplan
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Diego Villar
- University of Cambridge, Cancer Research UK Cambridge Institute, Cambridge, UK
| | - Bronwen L. Aken
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Paul Flicek
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Rishi Nag
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Emily S. Wong
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - John Blangero
- South Texas Diabetes and Obesity Institute, UTHSCSA/UTRGV, Brownsville, TX, USA
| | - Thomas D. Dyer
- South Texas Diabetes and Obesity Institute, UTHSCSA/UTRGV, Brownsville, TX, USA
| | - Marina Bogomolov
- Faculty of Industrial Engineering and Management, Technion, Haifa, Israel
| | - Yoav Benjamini
- Department of Statistics and Operation Research, Tel Aviv University, Tel Aviv, Israel
| | | | - Ken Dewar
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Chiara Sabatti
- Department of Biomedical Data Science, Stanford University, Stanford, California, USA
- Department of Statistics, Stanford University, Stanford, California, USA
| | - Richard K. Wilson
- The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - J. David Jentsch
- Department of Psychology, University of California, Los Angeles, Los Angeles, California, USA
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Wesley Warren
- The McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Giovanni Coppola
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles CA, USA
| | - Roger P. Woods
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles CA, USA
| | - Nelson B. Freimer
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA,USA
| |
Collapse
|