[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (13)

Search Parameters:
Keywords = squirrel monkey

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 1767 KiB  
Article
Using Social Network Analysis to Assess ‘Groupness’ in a Mixed-Species Zoo Exhibit of Tufted Capuchins (Sapajus apella) and Squirrel Monkeys (Saimiri sciureus)
by Sophia Daoudi-Simison, Phyllis Lee and Hannah M. Buchanan-Smith
Animals 2024, 14(23), 3360; https://doi.org/10.3390/ani14233360 - 22 Nov 2024
Viewed by 966
Abstract
Mixed-species groups have been recorded in various primates, including tufted capuchin and squirrel monkeys. Measures of their ‘groupness’ are typically based on factors such as group stability, social interactions, proximity, or behavioural coordination. Social network analysis has become a useful tool for quantifying [...] Read more.
Mixed-species groups have been recorded in various primates, including tufted capuchin and squirrel monkeys. Measures of their ‘groupness’ are typically based on factors such as group stability, social interactions, proximity, or behavioural coordination. Social network analysis has become a useful tool for quantifying relationships among group-living individuals. Here, we apply social network analysis to two captive mixed-species groups of tufted capuchins and squirrel monkeys housed at the Living Links to Human Evolution Research Centre, Royal Zoological Society of Scotland, Edinburgh Zoo, UK. We conducted 183 h of focal observations (three hours per individual, excluding co-observations) and calculated association rates using a simple index ratio. Permutation t-tests were used to assess differences in the overall mixed-species network and network metrics according to species. While the two species exhibited some level of association, they formed separate clusters in the mixed-species networks; however, the East group had more balanced group sizes and showed some signs of closer inter-specific social ties compared to the West group. Our data indicate that, in captivity at least, while these groups co-exist in a small, shared space, they do not form cohesive mixed-species groups. We suggest caution in the assumption of mixed-species groups based on shared space only. Full article
(This article belongs to the Section Zoo Animals)
Show Figures

Figure 1

Figure 1
<p>Schematic diagram (approximately to scale) of the Living Links West (WS = west squirrel monkey, WC = west capuchin) and East (ES = east squirrel monkey, EC = east capuchin) enclosures (adapted from [<a href="#B38-animals-14-03360" class="html-bibr">38</a>]).</p>
Full article ">Figure 2
<p>Mixed-species social networks for the (<b>A</b>) West and (<b>B</b>) East groups. Node shape is based on species: capuchins = square and squirrel monkeys = circle. Sex (capuchins: yellow = female; cyan = male; squirrel monkeys: red = female, blue = male). Node size is based on degree centrality, the strength of ties is based on the frequency of interactions between nodes and the distance between ties is based on the geodesic distance calculated as the sum of the weights of ties along the shortest path.</p>
Full article ">Figure 3
<p>Permutation <span class="html-italic">t</span>-test distributions based on 1000 permutations of the network metrics (eigenvector centrality, clustering coefficient, betweenness centrality, and degree centrality) for the West capuchins and squirrel monkeys. Vertical dashed lines represent the observed difference.</p>
Full article ">Figure 4
<p>Permutation <span class="html-italic">t</span>-test distributions based on 1000 permutations of the network metrics (eigenvector centrality, clustering coefficient, betweenness centrality, and degree centrality) for the East capuchins and squirrel monkeys. Vertical dashed lines represent the observed difference.</p>
Full article ">
32 pages, 8533 KiB  
Article
Mammalian Life History: Weaning and Tooth Emergence in a Seasonal World
by B. Holly Smith
Biology 2024, 13(8), 612; https://doi.org/10.3390/biology13080612 - 12 Aug 2024
Viewed by 2213
Abstract
The young of toothed mammals must have teeth to reach feeding independence. How tooth eruption integrates with gestation, birth and weaning is examined in a life-history perspective for 71 species of placental mammals. Questions developed from high-quality primate data are then addressed in [...] Read more.
The young of toothed mammals must have teeth to reach feeding independence. How tooth eruption integrates with gestation, birth and weaning is examined in a life-history perspective for 71 species of placental mammals. Questions developed from high-quality primate data are then addressed in the total sample. Rather than correlation, comparisons focus on equivalence, sequence, the relation to absolutes (six months, one year), the distribution of error and adaptive extremes. These mammals differ widely at birth, from no teeth to all deciduous teeth emerging, but commonalities appear when infants transit to independent feeding. Weaning follows completion of the deciduous dentition, closest in time to emergence of the first permanent molars and well before second molars emerge. Another layer of meaning appears when developmental age is counted from conception because the total time to produce young feeding independently comes up against seasonal boundaries that are costly to cross for reproductive fitness. Mammals of a vast range of sizes and taxa, from squirrel monkey to moose, hold conception-to-first molars in just under one year. Integrating tooth emergence into life history gives insight into living mammals and builds a framework for interpreting the fossil record. Full article
(This article belongs to the Special Issue Evolutionary Insights into Life History)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Distribution of life-history characteristics for 743 nonvolant placental mammals: (<b>a</b>) age of weaning (duration of nursing); and (<b>b</b>) conception to age of weaning (gestation length plus weaning age) or total time to produce young that feed independently. Peaks recognizable in (<b>a</b>) take on new meaning in (<b>b</b>), where a trough between modes separates species that can reproduce more than once a year from those that cannot (barring concurrent pregnancy and nursing). Data from Ernest [<a href="#B59-biology-13-00612" class="html-bibr">59</a>].</p>
Full article ">Figure 2
<p>Age of weaning versus age of completion of the deciduous dentition (<b>a</b>) and age of M<sub>2</sub> emergence (<b>b</b>) with age counted from birth in both. Residuals (in <span class="html-italic">y</span>-direction from the dashed line <span class="html-italic">y</span> = <span class="html-italic">x</span>) shown for n = 19 primate species as a fine line. Weaning occurs after young have a complete deciduous dentition, but typically well before M<sub>2</sub> is in place. For <span class="html-italic">Pongo</span>, tooth emergence datum is precise at left (<b>a</b>), but only approximated at right (<b>b</b>) with an unfilled symbol. <span class="html-italic">Tarsiidae</span> and <span class="html-italic">Indriidae</span> omitted in (<b>a</b>) for scale, although both wean infants after deciduous teeth are emerged (their symbols remain in key).</p>
Full article ">Figure 3
<p>Age of weaning versus age of emergence of M<sub>1</sub>, with age counted from birth (<b>a</b>) and conception (<b>b</b>) for n = 21 primate species. Residuals (in <span class="html-italic">y</span>-direction from the dashed line <span class="html-italic">y</span> = <span class="html-italic">x</span>) shown for each datum as a fine line. Horizontal dotted lines mark limits for primates (<b>a</b>) or doubling of maternal investment (<b>b</b>). Twinning, litters and marked allocare occur in species that invest less than one year in their offspring. <span class="html-italic">Pongo</span> and <span class="html-italic">Homo</span> are diametrically opposed in tooth emergence vs weaning.</p>
Full article ">Figure 4
<p>Age of M<sub>1</sub> emergence on size, measured by female body weight (<b>a</b>) and adult brain weight (<b>b</b>) for n = 21 living and n = 2 extinct primate species. Stairsteps in <span class="html-italic">y</span>-data show that similar ages of tooth emergence span a large range of body weights, especially near 4.5 months (dotted line). Brain size has a tighter linear relationship with tooth emergence, although echoes of the lateral spread in (<b>a</b>) remain in (<b>b</b>).</p>
Full article ">Figure 5
<p>Weaning takes place after completion of the deciduous dentition in 47 out of 50 species in (<b>a</b>) and before emergence of M<sub>2</sub> in 47 out of 52 in (<b>b</b>). Age is counted from conception and residuals (in <span class="html-italic">y</span>-direction from the line <span class="html-italic">y</span> = <span class="html-italic">x</span>) are shown as fine lines. Boundary cases: (<b>a</b>) suines and <span class="html-italic">Eira</span> wean early relative to teeth, (<b>b</b>) <span class="html-italic">Ursus</span>, <span class="html-italic">Pongo</span>, <span class="html-italic">Mephitis</span> and <span class="html-italic">Cheirogaleus</span> wean late relative to teeth (M<sub>2</sub> datum for <span class="html-italic">Pongo</span> uncertain).</p>
Full article ">Figure 6
<p>‘Vole to elephant’ plots for the correspondence between age of weaning and M<sub>1</sub> emergence for 67 mammal species with age counted from birth, (<b>a</b>) and from conception, (<b>b</b>). Residuals in <span class="html-italic">y</span>-direction from the line <span class="html-italic">y</span> = <span class="html-italic">x</span> are shown as fine lines. Maximum for nonvolant placental mammals dotted in (<b>a</b>); doubling of maternal investment dotted in (<b>b</b>). Except for apes, slow-growing mammals wean early relative to the M<sub>1</sub> emergence.</p>
Full article ">Figure 7
<p>Age of M<sub>1</sub> emergence counted from conception relative to size, as measured by adult body weight (<b>a</b>), and adult brain weight (<b>b</b>) for 67 mammal species. Dotted lines represent doublings in time invested to raise young with first permanent molars. Some species that seem extreme in (<b>a</b>) are less so in (<b>b</b>), but the crowd with investment of ca 11 months remains.</p>
Full article ">Figure 8
<p>Distributions of estimators of the length of maternal investment, the time used to gestate and raise young to feeding independence: age of M<sub>1</sub> emergence at top (<b>a</b>,<b>b</b>) and age of weaning at bottom (<b>c</b>,<b>d</b>); age is counted from birth at left (<b>a</b>,<b>c</b>) and from conception at right (<b>b</b>,<b>d</b>) for 67 species with complete data. When age is counted from conception (<b>right</b>), extreme peak values between six months and one year suggest seasonal boundaries have shaped maternal investment. Paired <span class="html-italic">t</span>-tests cannot distinguish (<b>a</b>) from (<b>c</b>), at <span class="html-italic">p</span> = 0.78, or (<b>b</b>) from (<b>d</b>), at <span class="html-italic">p</span> = 0.15.</p>
Full article ">Figure 9
<p>Seven stages of sub-adult life history of mandibular teeth of <span class="html-italic">Trachypithcus</span> sp., redrawn and modified from Ingicco et al. [<a href="#B191-biology-13-00612" class="html-bibr">191</a>], mapped against expectations for feeding primates. Teeth in lighter gray are just cutting the gum and shading represents dentin exposure. Findings suggest that three morphological divisions correspond with three stages of feeding: individuals without a full deciduous dentition remain supplemented with milk (infants); the transition to all solid food takes place in the period around the appearance of M<sub>1</sub> (dashed lines) and individuals with M<sub>2</sub> emerging are fully independent feeders (juveniles). Permanent I1-P4 replace deciduous predecessors during independent feeding, completing the adult dentition.</p>
Full article ">
8 pages, 260 KiB  
Communication
Occurrence of Clostridium perfringens in Wild Mammals in the Amazon Biome
by Hanna Gabriela da Silva Oliveira, Ananda Iara de Jesus Sousa, Isabela Paduá Zanon, Cinthia Távora de Albuquerque Lopes, Rodrigo Otavio Silveira Silva, Sheyla Farhayldes Souza Domingues and Felipe Masiero Salvarani
Animals 2024, 14(9), 1333; https://doi.org/10.3390/ani14091333 - 29 Apr 2024
Viewed by 946
Abstract
The objective of this study was to evaluate the occurrence of Clostridium perfringens in stool samples and swabs collected from wild mammals in the Amazon biome. Sixty-five faecal and swab samples were collected in situ and ex situ from 16 species and three [...] Read more.
The objective of this study was to evaluate the occurrence of Clostridium perfringens in stool samples and swabs collected from wild mammals in the Amazon biome. Sixty-five faecal and swab samples were collected in situ and ex situ from 16 species and three genera of wild mammals, some of which were in good health and some of which had diarrhoea. After pre-enrichment, the samples were plated on selective agar for C. perfringens. Characteristic colonies were subjected to multiplex PCR for the detection of genes encoding the main C. perfringens toxins (alpha, beta, epsilon, and iota toxin and enterotoxin). Among the 65 samples, 40 (61.5%) were positive for the gene encoding the alpha toxin and were classified as type A, 36 of which were asymptomatic animals and four were diarrheal. No other toxinotypes were found. The findings of this study suggest that C. perfringens type A is commonly found in mammal species of the Amazon biome. This seems to be the first study to identify C. perfringens type A in species such as B. variegatus (common ground sloth), C. didactylus (two-toed sloth), P. flavus (Jupará), T. tetradactyla (anteater), S. collinsi (squirrel monkey), S. niger (black marmoset), and S. apella (Guyana capuchin) and in the genus Didelphis sp. (opossum). Full article
(This article belongs to the Special Issue Exotic Mammal Care and Medicine)
12 pages, 1341 KiB  
Article
Measurement of Salivary Cortisol in Two New World Primate Species
by Martina Stocker, Eoin P. O’Sullivan, Rupert Palme, Eva Millesi and Ruth Sonnweber
Biology 2023, 12(9), 1181; https://doi.org/10.3390/biology12091181 - 29 Aug 2023
Cited by 1 | Viewed by 1658
Abstract
Glucocorticoids (GCs) are mammalian steroid hormones involved in a variety of physiological processes, including metabolism, the immune response, and cardiovascular functions. Due to their link to the physiological stress response, GC measurement is a valuable tool for conservation and welfare assessment in animal [...] Read more.
Glucocorticoids (GCs) are mammalian steroid hormones involved in a variety of physiological processes, including metabolism, the immune response, and cardiovascular functions. Due to their link to the physiological stress response, GC measurement is a valuable tool for conservation and welfare assessment in animal populations. GC levels can be measured from different matrices, such as urine and feces. Moreover, especially in captive settings, measuring GCs from saliva samples proved particularly useful as those samples can be collected non-invasively and easily from trained animals. Salivary GC levels can be measured using a variety of analytical methods, such as enzyme immunoassays. However, it is crucial to validate the analytical method for each specific application and species when using a new matrix. Using high-pressure liquid chromatography and a cortisol enzyme immunoassay, we show that the main glucocorticoids secreted in the saliva of squirrel monkeys and brown capuchin monkeys are cortisol and cortisone. Our biological validation found the expected salivary cortisol level to decline throughout the day. Our findings support the reliability of salivary cortisol measurements and their potential to be used as a valid tool in research and welfare assessment for these non-human primates. Full article
(This article belongs to the Special Issue Research on Primate Endocrinology)
Show Figures

Figure 1

Figure 1
<p>Research cubicles at Living Links Research Centre, Edinburgh Zoo. The green squares on the left indicate the exits towards the outdoor enclosure and on the right towards the indoor enclosure. All exits can be closed with sliding doors. Between each cubicle, partitioning slides can be inserted to separate individual monkeys.</p>
Full article ">Figure 2
<p>High-performance liquid chromatography (reverse phase) separation of immunoreactive substances in a salivary pool sample of (<b>a</b>) squirrel monkeys and (<b>b</b>) capuchin monkeys when measured with the cortisol EIA.</p>
Full article ">Figure 3
<p>Parallelism between the standard curve (solid lines) and serial dilutions of salivary cortisol (dashed lines) of capuchin monkeys (orange lines) and squirrel monkeys (blue lines). The <span class="html-italic">y</span>-axis shows the optical density, the <span class="html-italic">x</span>-axis shows log-transformed levels of cortisol [ng/mL]. For both species, standard curves and serial dilution curves are parallel to each other.</p>
Full article ">Figure 4
<p>Diurnal patterns of salivary cortisol levels (log-transformed) plotted against time of the day for capuchin monkeys (orange) and squirrel monkeys (blue). Individual dots represent individual data points. The lines are linear regression lines with 95% confidence intervals as calculated from the model. Salivary cortisol levels decrease throughout the day in both capuchin and squirrel monkeys, as expected due to the circadian rhythm in salivary cortisol excretion patterns.</p>
Full article ">
21 pages, 3291 KiB  
Article
Zika Virus Infection Damages the Testes in Pubertal Common Squirrel Monkeys (Saimiri collinsi)
by Gabriela da Costa Benchimol, Josye Bianca Santos, Ana Sophia da Costa Lopes, Karol Guimarães Oliveira, Eviny Sayuri Trindade Okada, Bianca Nascimento de Alcantara, Washington Luiz Assunção Pereira, Danuza Leite Leão, Ana Cristina Carneiro Martins, Liliane Almeida Carneiro, Aline Amaral Imbeloni, Sheila Tetsume Makiama, Luiz Paulo Printes Albarelli de Castro, Leandro Nassar Coutinho, Lívia Medeiros Neves Casseb, Pedro Fernando da Costa Vasconcelos, Sheyla Farhayldes Souza Domingues, Daniele Barbosa de Almeida Medeiros and Sarah Raphaella Rocha de Azevedo Scalercio
Viruses 2023, 15(3), 615; https://doi.org/10.3390/v15030615 - 23 Feb 2023
Viewed by 2430
Abstract
During the Zika virus (ZIKV) outbreak and after evidence of its sexual transmission was obtained, concerns arose about the impact of the adverse effects of ZIKV infection on human fertility. In this study, we evaluated the clinical-laboratory aspects and testicular histopathological patterns of [...] Read more.
During the Zika virus (ZIKV) outbreak and after evidence of its sexual transmission was obtained, concerns arose about the impact of the adverse effects of ZIKV infection on human fertility. In this study, we evaluated the clinical-laboratory aspects and testicular histopathological patterns of pubertal squirrel monkeys (Saimiri collinsi) infected with ZIKV, analyzing the effects at different stages of infection. The susceptibility of S. collinsi to ZIKV infection was confirmed by laboratory tests, which detected viremia (mean 1.63 × 106 RNA copies/µL) and IgM antibody induction. Reduced fecal testosterone levels, severe testicular atrophy and prolonged orchitis were observed throughout the experiment by ultrasound. At 21 dpi, testicular damage associated with ZIKV was confirmed by histopathological and immunohistochemical (IHC) analyses. Tubular retraction, the degeneration and necrosis of somatic and germ cells in the seminiferous tubules, the proliferation of interstitial cells and an inflammatory infiltrate were observed. ZIKV antigen was identified in the same cells where tissue injuries were observed. In conclusion, squirrel monkeys were found to be susceptible to the Asian variant of ZIKV, and this model enabled the identification of multifocal lesions in the seminiferous tubules of the infected group evaluated. These findings may suggest an impact of ZIKV infection on male fertility. Full article
(This article belongs to the Special Issue Zika Viruses: State-of-the-Art Research in Brazil)
Show Figures

Figure 1

Figure 1
<p>Experimental design of the study. The upper portion of the figure shows the timeline of sample collection and biometric data from the animals evaluated; the periods were divided into (1) Pre-inoculation, (2) Acute and (3) Convalescence phases of infection, each with its corresponding color according to the legend on the right of the figure; on the ruler we can see the days marked in red on which biological material was collected, the day of inoculation and euthanasia of the animals, as well as what was collected and evaluated, and on which days they were evaluated. In the lower portion of the figure, we have a flowchart showing to which methodology each of the samples was forwarded and the culmination of the entire study.</p>
Full article ">Figure 2
<p>Noninvasive evaluation of experimental ZIKA infection in male <span class="html-italic">S. collinsi</span>. (<b>A</b>) Measurement of rectal temperature, showing feverish period of infected group between 3 to 10 dpi. The dashed line represents the temperature cut-off based in control group data. The (<b>B</b>) RNA viral load in blood by RT-qPCR and (<b>C</b>) IgM antibody profile of the infected group detected from 14 dpi. The dashed line represents the cut-off of ELISA assay. Group evaluation, mean and min-max values: (<b>D</b>) body weight, (<b>E</b>) testicular volume, (<b>F</b>) gonadosomatic index, and (<b>G</b>) testosterone by stages of infection, highlighting the decrease of testicular volume, GSI and testosterone values during acute phase of infection in G1.</p>
Full article ">Figure 3
<p>Ultrasonographic analysis at 21 dpi. The left testicle of animal AT-003 (G1) showed normal homogeneity (<b>A</b>) and the left testicle of animal AT-163 (G2) presented hypoechoicity in all its extensions (<b>B</b>). The testicular echogenicity of G1 and G2 was evaluated at the indicated day (<b>C</b>) and by stages of ZIKV infection (<b>D</b>). Mean and min-max values.</p>
Full article ">Figure 4
<p>Postmortem evaluation of experimental ZIKA infection in male <span class="html-italic">S. collinsi</span>. (<b>A</b>) Control animal AT-001, macroscopy of the right and left testes. (<b>B</b>) Infected animal AT-156, macroscopy showing a slight difference in size between the right and left testes. (<b>C</b>) Postmortem GSI graph at 21 dpi of each animal. (<b>D</b>) Graph of the detection of ZIKV RNA copies/mg in a pool of both testes of each infected animal. (<b>E</b>) Graph of Pearson’s correlation: demonstrating a significant inverse and very strong correlation between GSI X ZIKV tissue variables, R-value = −0.9986 and <span class="html-italic">p</span> = 0.033.</p>
Full article ">Figure 5
<p>Microscopic evaluation of <span class="html-italic">S. collinsi</span>. (<b>A</b>) Integrity of testicular tissue from control animal AT-001 (100×). (<b>B</b>) Seminiferous tubule of the control animal, in a higher magnification lens, showing sperm (arrow), score 10 (400×). (<b>C</b>) AT-005, infected with ZIKV, showing diffuse tubular edema and tubular retraction (star). Highlighted (circle) interstitial cell proliferation associated with inflammatory infiltrate (100×). (<b>D</b>) Perivascular inflammatory infiltrate (circle), with the presence of lymphocytes and neutrophils. The seminiferous tubule of an infected animal, showing a decrease in the number of germ cells, which are degenerated (arrowhead) (400×). (<b>E</b>) AT-163 infected animal, showing proliferation of Leydig cells and degenerated germ cells. (<b>F</b>) Infected animal, showing seminiferous tubule containing degenerating spermatogenic cells (hollow arrows), presence of pyknotic nuclei and cytoplasmic macrovacuolization (arrowhead), score 4. In the extra tubular area, there is an increase in the proliferation of Leydig cells (black arrows) (400×). (<b>G</b>) Light micrograph showing IHC staining with peroxidase using anti-ZIKV polyclonal antibodies. Seminiferous tubule from infected animal AT-005, showing ZIKV antigen labeling on both the germ cells of the seminiferous tubule (arrow) and the Leydig cells (circle). (<b>H</b>) Graph showing the quantification of viral antigen per group in the testicular tissue of the control and infected animals. ** <span class="html-italic">p</span> &lt; 0.03. (<b>I</b>) Graph showing the quantification of ZIKV antigen per animal based on the IHC test. In blue is the animal AT-163, in green the AT-156 and in red the AT-005; the filled columns represent the right testis and the empty columns the left testis.</p>
Full article ">
12 pages, 1661 KiB  
Article
Experimental Yellow Fever in the Squirrel Monkey (Saimiri spp.): Hematological, Biochemical, and Immunological Findings
by Milene S. Ferreira, Lívia C. Martins, Karla F. L. de Melo, Wellington B. da Silva, Aline A. Imbeloni, José Augusto P. C. Muniz, Camille F. de Oliveira, Maria Nazaré O. Freitas, Éder B. dos Santos, Liliane L. Chagas, Márcia B. M. Luz, Luiz A. D. de Queiroz, Robert B. Tesh and Pedro F. C. Vasconcelos
Viruses 2023, 15(3), 613; https://doi.org/10.3390/v15030613 - 23 Feb 2023
Viewed by 1671
Abstract
Between 2016 and 2018, Brazil experienced the largest sylvatic epidemic of yellow fever virus (YFV). Despite to the magnitude and rapid spread of the epidemic, little is known about YFV dispersion. The study evaluated whether the squirrel monkey is a good model for [...] Read more.
Between 2016 and 2018, Brazil experienced the largest sylvatic epidemic of yellow fever virus (YFV). Despite to the magnitude and rapid spread of the epidemic, little is known about YFV dispersion. The study evaluated whether the squirrel monkey is a good model for yellow fever (YF) studies. Methods: Ten animals were infected with 1 × 106 PFU/mL of YFV, with one negative control. Blood samples were collected daily during the first 7 days and at 10, 20 and 30 days post infection (dpi) for detection of viral load and cytokines by RT-qPCR; measurements of AST, ALT, urea and creatinine were taken; IgM/IgG antibodies were detected by ELISA, and hemagglutination inhibition and neutralization tests were performed. The animals exhibited fever, flushed appearance, vomiting and petechiae, and one animal died. Viremia was detected between 1 and 10 dpi, and IgM/IgG antibodies appeared between 4 and 30 dpi. The levels of AST, ALT and urea increased. The immune responses were characterized by expression of S100 and CD11b cells; endothelial markers (VCAM-1, ICAM-1 and VLA-4), cell death and stress (Lysozyme and iNOS); and pro-inflammatory cytokines (IL-8, TNF-α, and IFN-γ) and anti-inflammatory cytokines (IL-10 and TGF-β). The squirrel monkeys showed changes similar to those described in humans with YF, and are a good experimental model for the study of YF. Full article
(This article belongs to the Section Human Virology and Viral Diseases)
Show Figures

Figure 1

Figure 1
<p>Detection of viral genome and anti-YFV antibodies in squirrel monkeys (<span class="html-italic">Saimiri</span> spp.) infected with yellow fever virus. (<b>A</b>) Detection of the viral genome by real-time RT-PCR; (<b>B</b>) detection of anti-YFV IgM antibodies by enzyme immune assay; (<b>C</b>) detection of total antibodies by HI technique.</p>
Full article ">Figure 2
<p>Hematological profile of squirrel monkeys (<span class="html-italic">Saimiri</span> spp.) infected with the yellow fever virus. (<b>A</b>) Leukogram, leukocytes, monocytes, lymphocytes, and neutrophils were measured. (<b>B</b>) Erythrogram, red blood cells, hemoglobin and hematocrit were evaluated. (<b>C</b>) Coagulogram, platelets, prothrombin time (PT), and activated partial thromboplastin time (TPPA) were verified. The data were transformed into a ratio where values were considered before and after infection. Normal values appear between 0.75 and 1.5 (gray band). The relevant differences are highlighted by the arrows in the figure (↓ = decrease &lt; 0.75 and ↑ = increase &gt; 1.5 in the baseline).</p>
Full article ">Figure 3
<p>Biochemical tests of hepatic and renal function of squirrel monkeys (<span class="html-italic">Saimiri</span> spp.) infected with the yellow fever virus. (<b>A</b>,<b>B</b>) Relative ratio of the enzymes aspartate aminotransferase (AST) and alanine aminotransferase (ALT); (<b>C</b>,<b>D</b>) relative ratio of blood urea and creatinine. The data were transformed into a ratio where values were considered before and after infection. Normal values appear between 0.75 and 1.5 (gray band). The relevant differences are highlighted by the arrows in the figure ((↓ = decrease &lt; 0.75 and ↑ = increase &gt; 1.5 in the base).</p>
Full article ">Figure 4
<p>Correlation of viral load with serum profile of the cellular phenotype markers (S100 and CD11b) (<b>A</b>,<b>B</b>), stress and cell death (iNOS and lysozyme) (<b>C</b>,<b>D</b>), of squirrel monkeys (<span class="html-italic">Saimiri</span> spp.) infected with the yellow fever virus. Performing each sample in technical and biological triplicate increased the reliability of the generated data.</p>
Full article ">Figure 5
<p>Correlation of viral load with serum profile of endothelial markers (VCAM-1, ICAM-1 and VLA-4) (<b>A</b>–<b>C</b>), in squirrel monkeys (<span class="html-italic">Saimiri</span> spp.) infected with yellow fever virus. Evaluating each sample in technical and biological triplicate increased the reliability of the generated data.</p>
Full article ">Figure 6
<p>Correlation of viral load with serum profile of the proinflammatory (IL-8, TNF-α and INF-γ) (<b>A</b>–<b>C</b>) and anti-inflammatory cytokines (IL-10 and TGF-β) (<b>D</b>,<b>E</b>), in squirrel monkeys (<span class="html-italic">Saimiri</span> spp.) infected with yellow fever virus. Evaluating each sample in technical and biological triplicate increased the reliability of the generated data.</p>
Full article ">
16 pages, 5259 KiB  
Article
Experimental Yellow Fever in Squirrel Monkey: Characterization of Liver In Situ Immune Response
by Milene S. Ferreira, Jorge R. Sousa, Pedro S. Bezerra Júnior, Valíria D. Cerqueira, Carlos A. Oliveira Júnior, Gabriela R. C. Rivero, Paulo H. G. Castro, Gilmara A. Silva, José Augusto P. C. Muniz, Eliana V. P. da Silva, Samir M. M. Casseb, Carla Pagliari, Lívia C. Martins, Robert B. Tesh, Juarez A. S. Quaresma and Pedro F. C. Vasconcelos
Viruses 2023, 15(2), 551; https://doi.org/10.3390/v15020551 - 16 Feb 2023
Cited by 1 | Viewed by 2208
Abstract
Non-human primates contribute to the spread of yellow fever virus (YFV) and the establishment of transmission cycles in endemic areas, such as Brazil. This study aims to investigate virological, histopathological and immunohistochemical findings in livers of squirrel monkeys (Saimiri spp.) infected with [...] Read more.
Non-human primates contribute to the spread of yellow fever virus (YFV) and the establishment of transmission cycles in endemic areas, such as Brazil. This study aims to investigate virological, histopathological and immunohistochemical findings in livers of squirrel monkeys (Saimiri spp.) infected with the YFV. Viremia occurred 1–30 days post infection (dpi) and the virus showed a predilection for the middle zone (Z2). The livers were jaundiced with subcapsular and hemorrhagic multifocal petechiae. Apoptosis, lytic and coagulative necrosis, steatosis and cellular edema were also observed. The immune response was characterized by the expression of S100, CD11b, CD57, CD4 and CD20; endothelial markers; stress and cell death; pro and anti-inflammatory cytokines, as well as Treg (IL-35) and IL-17 throughout the experimental period. Lesions during the severe phase of the disease were associated with excessive production of apoptotic pro-inflammatory cytokines, such as IFN-γ and TNF-α, released by inflammatory response cells (CD4+ and CD8+ T lymphocytes) and associated with high expression of molecules of adhesion in the inflammatory foci observed in Z2. Immunostaining of the local endothelium in vascular cells and the bile duct was intense, suggesting a fundamental role in liver damage and in the pathogenesis of the disease. Full article
(This article belongs to the Section Viral Immunology, Vaccines, and Antivirals)
Show Figures

Figure 1

Figure 1
<p>Gross macroscopy, viral load, histopathology and immunohistochemical assay in hepatic parenchyma of squirrel monkey (<span class="html-italic">Saimiri</span> spp.) infected with YFV South American genotype I. (<b>A</b>) Macroscopic representation of liver infected with YFV at 6 dpi showing an icteric and hemorrhagic pattern compared to a normal uninfected liver. (<b>B</b>) Evolution of viral kinetics highlighting viremic peak at 4 dpi. (<b>C</b>) Quantification of cell expressing specific YFV antigens by Immunohistochemical assay (IHC) in the 4 hepatic compartments analyzed demonstrating a wide predominance in Z2 during the acute phase of infection. (<b>D</b>) Representative histopathology photographs showing the involvement of Z3, with emphasis to lytic necrosis (asterisks), intense hemorrhage (black circle), positive immunohistochemistry for presence of specific YFV antigens in hepatocytes (red circle). (<b>E</b>) Histopathology showing the major involvement of Z2 with lytic necrosis area (black circle), positive immunohistochemistry showing large amounts of YFV antigens in hepatocytes (red circle). (<b>F</b>) Representative histopathology photographs of Z1, showing areas of periportal lytic necrosis and hemorrhages (black cycle), positive immunohistochemical for defection of specific YFV antigens and a periportal infiltrate is highlighted (red circle) and Kupffer cells (arrow). (<b>G</b>) Representative histopathology photographs of portal tract (PT) showing inflammatory infiltrate (black circle), positive immunohistochemistry for YFV antigens periportal infiltrate (red circle) and in the bile duct (black arrow); (<b>D</b>–<b>G</b>) Negative control for YFV antigens in Z3, Z2, Z1, and PT.</p>
Full article ">Figure 2
<p>Quantitative immunohistochemical analysis for specific antibodies for immune cells in Z3, Z2, Z1 and PT cells in hepatic parenchyma of squirrel monkeys (<span class="html-italic">Saimiri</span> spp.) infected with YFV. Circles represent areas expressing higher immunostaining cells in different hepatic areas. (<b>A</b>) Expression of S100. (<b>F</b>) Immunostaining for S100 in Kupffer cells (F-Z3), inflammatory infiltrate (F-Z2), hepatocytes (F-Z1) and bile duct (F-PT). (<b>B</b>) Expression of CD11b. (<b>G</b>) Immunostaining for CD11b macrophages in inflammatory infiltrate (G-Z3), (G-Z2), (G-Z1), (G-PT). (<b>C</b>) Expression of CD57 NK cell. (<b>H</b>) Immunostaining for CD57 in inflammatory infiltrate in all hepatic compartments (H-Z3), (H-Z2), (H-Z1), (H-PT). (<b>D</b>) CD4 expression. (<b>I</b>) Immunostaining for CD4 T lymphocyte in the inflammatory infiltrate in all acini areas (I-Z3), (I-Z2), (I-Z1), (I-PT). (<b>E</b>) Expression of CD20 (B cell). (<b>J</b>) Absence of Immunostaining for CD20 in all acini areas (J-Z3), (J-Z2), (J-Z1), (J-PT). (<b>E</b>–<b>J</b>) obtained after 6 dpi.</p>
Full article ">Figure 3
<p>Quantitative immunohistochemical analysis of selected endothelial cell markers in zones Z3, Z2, Z1 and PT in hepatic parenchyma of squirrel monkeys (<span class="html-italic">Saimiri</span> spp.) infected with YFV. Circles represent areas expressing higher immunostaining cells in different hepatic areas. (<b>A</b>) Expression of VCAM-1. (<b>D</b>) Immunostaining for VCAM-1 in the inflammatory infiltrate (D-Z3), in Kupffer cells (D-Z2), (D-Z1) and bile duct (D-PT). (<b>B</b>) Expression of ICAM-1. (<b>E</b>) Immunostaining for ICAM-1 in hepatocytes (E-Z3), (E-Z2), (E-Z1) in Kupffer cells, (E-PT) in the bile duct. (<b>C</b>) VLA-4 expression. (<b>F</b>) Immunostaining for VLA-4 in endothelial and Kupffer (F-Z3) cells, (F-Z2) in inflammatory infiltrate, (F-Z1), (F-PT) in 6 dpi bile duct.</p>
Full article ">Figure 4
<p>Quantitative immunohistochemical analysis of specific markers in Z3, Z2, Z1 and PT in the hepatic parenchyma of squirrel monkeys (<span class="html-italic">Saimiri</span> spp.) infected with YFV. Circles represent areas expressing higher immunostaining cells in different hepatic areas. (<b>A</b>) Caspase 3 expression. (<b>E</b>) Immunostaining for Caspase 3 in hepatocytes (E-Z3), (E-Z2), (E-Z1) and bile duct (E-PT). (<b>B</b>) MLKL expression. (<b>F</b>) Immunostaining for MLKL in hepatocytes and inflammatory infiltrate (F-Z3), (F-Z2) inflammatory infiltrate, (F-Z1) in hepatocytes, (F-PT) in hepatocytes. (<b>C</b>) Expression of iNOS. (<b>G</b>) Immunostaining for iNOS in inflammatory infiltrate (G-Z3), (G-Z2) in hepatocytes, (G-Z1) hepatocytes, (G-PT) bile duct and inflammatory infiltrate. (<b>D</b>) Lysozyme Expression. (<b>H</b>) In inflammatory infiltrate (E-Z3), (E-Z2), (H-Z1) in Kupffer cells, (H-PT) in inflammatory infiltrate at 6 dpi.</p>
Full article ">Figure 5
<p>Quantitative immunohistochemical analysis of selected pro- inflammatory cytokines in zones Z3, Z2, Z1 and PT in the hepatic parenchyma of squirrel monkeys (<span class="html-italic">Saimiri</span> spp.) infected with YFV. Circles represent areas expressing higher immunostaining cells in different hepatic areas. (<b>A</b>) IFN-γ expression. (<b>F</b>)-Immunostaining for IFN-γ in hepatocytes (F-Z3), and in inflammatory infiltrate (F-Z2), (F-Z1), (F-PT). (<b>B</b>) Expression of IFN-β. (<b>G</b>) Immunostaining for IFN-β in hepatocytes (F-Z3), (F-Z2) in inflammatory infiltrate, (F-Z1) in hepatocytes, (F-PT). (<b>C</b>) Expression of TNF-α. (<b>H</b>) Immunostaining for TNF-α in inflammatory infiltrate (F-Z3), (F-Z2), (F-Z1), (F-PT). (<b>D</b>) Expression of IL-8. (<b>I</b>) Immunostaining for IL-8 in hepatocytes (F-Z3), (F-Z2), (F-Z1), (F-PT). (<b>E</b>) Expression of IL-17. (<b>J</b>) Immunostaining for IL-17 in hepatocytes (J-Z3), (J-Z2), (J-Z1), (J-PT) in 6 dpi.</p>
Full article ">Figure 6
<p>Quantitative immunohistochemical analysis of selected anti- inflammatory cytokines in zones Z3, Z2, Z1 and PT in hepatic parenchyma of squirrel monkeys (<span class="html-italic">Saimiri</span> spp.) infected with YFV. Circles represent areas expressing higher immunostaining cells in different hepatic areas. (<b>A</b>) Expression of IL-4. (<b>E</b>) Immunostaining for IL-4 in hepatocytes (E-Z3), (E-Z2), (E-Z1) in inflammatory infiltrate area (E-PT). (<b>B</b>) Expression of IL-10. (<b>F</b>) Immunostaining for IL-10 in hepatocytes (F-Z3), (F-Z2), (F-Z1), (F-PT). (<b>C</b>) Expression of IL-35. (<b>G</b>) Immunostaining for IL-35 in hepatocytes in all examined hepatic tissues (G-Z3), (G-Z2), (G-Z1), (G-PT). (<b>D</b>) Expression of TGF-β. (<b>H</b>) Immunostaining for TGF-β in the inflammatory infiltrate (H-Z3), (H-Z2), and in hepatocytes (H-Z1), (H-PT) 6 dpi.</p>
Full article ">Figure 7
<p>Integrated network and possible mechanism of in situ immune response in the hepatic parenchyma of non-human primates (<span class="html-italic">Saimiri</span> spp.) infected with the yellow fever virus. After the infection, YFV is recognized by dendritic cells (DC), initiating the immune response by triggering immune cells that will produce the mechanisms to activate different local cells in liver and the production of cytokines and chemokines to drive the organism defenses. Both innate and adaptive immune cells have an important role in triggering the adequate response and the Treg cells are directly involved in the regulation of an equilibrated immunologic response in efforts to control the damage caused by YFV in the hepatic tissue.</p>
Full article ">
13 pages, 1888 KiB  
Article
Cebidae Alu Element Alignments and a Complex Non-Human Primate Radiation
by Jessica M. Storer, Jerilyn A. Walker, Morgan A. Brown and Mark A. Batzer
Life 2022, 12(10), 1655; https://doi.org/10.3390/life12101655 - 20 Oct 2022
Viewed by 1687
Abstract
Phylogenetic relationships among Cebidae species of platyrrhine primates are presently under debate. Studies prior to whole genome sequence (WGS) availability utilizing unidirectional Alu repeats linked Callithrix and Saguinus as sister taxa, based on a limited number of genetic markers and specimens, while the [...] Read more.
Phylogenetic relationships among Cebidae species of platyrrhine primates are presently under debate. Studies prior to whole genome sequence (WGS) availability utilizing unidirectional Alu repeats linked Callithrix and Saguinus as sister taxa, based on a limited number of genetic markers and specimens, while the relative positions of Cebus, Saimiri and Aotus remained controversial. Multiple WGS allowed computational detection of Alu-genome junctions, however random mutation and evolutionary decay of these short-read segments prevented phylogenetic resolution. In this study, WGS for four Cebidae genomes of marmoset, squirrel monkey, owl monkey and capuchin were analyzed for full-length Alu elements and each locus was compared to the other three genomes in all possible combinations using orthologous region sequence alignments. Over 2000 candidates were aligned and subjected to visual inspection. Approximately 34% passed inspection and were considered shared in their respective category, 48% failed due to the target being present in all four genomes, having N’s in the sequence or other sequence quality anomalies, and 18% were determined to represent near parallel insertions (NP). Wet bench locus specific PCR confirmed the presence of shared Alu insertions in all phylogenetically informative categories, providing evidence of extensive incomplete lineage sorting (ILS) and an abundance of Alu proliferation during the complex radiation of Cebidae taxa. Full article
(This article belongs to the Special Issue Genomic Impact of Transposable Elements)
Show Figures

Figure 1

Figure 1
<p>Distribution and genome comparison of shared and lineage-specific <span class="html-italic">Alu</span> insertions. Alignments were categorized into pre-defined groups (See Materials and Methods) and then broadly characterized into three groups: LS (lineage-specific) in orange indicates the percentage of the <span class="html-italic">Alu</span> insertions that were not found in orthologous position in any other genome. CMOS in blue indicates the percentage of the elements from the BLAT analysis that were shared by all four Cebidae genomes. (<b>A</b>) marmoset (<b>B</b>) squirrel monkey (<b>C</b>) owl monkey (<b>D</b>) capuchin monkey. The small gray pie slices are the percentages of <span class="html-italic">Alu</span> insertions within any of the other ten pre-defined shared categories.</p>
Full article ">Figure 2
<p>PCR analysis of phylogenetically informative <span class="html-italic">Alu</span> element categories. (<b>A</b>) CMO_blat_2; (<b>B</b>) CMS_blat_1; (<b>C</b>) COS_blat_4; (<b>D</b>) MOS_blat_2; (<b>E</b>) CM_blat_2; (<b>F</b>) CO_blat_4; (<b>G</b>) MS_blat_4; (<b>H</b>) CS_blat_2; (<b>I</b>) MO _blat_2; (<b>J</b>) OS_blat_4. Lanes: 1—100 bp ladder; 2—TLE (negative control); 3—Human (HeLa); 4—Chimpanzee; 5—African green monkey; 6—Wooly monkey; 7—White-bellied spider monkey; 8—Black-handed spider monkey; 9—Bolivian red howler monkey; 10—Common marmoset; 11—Pygmy marmoset; 12—Goeldi’s marmoset; 13—Red-chested mustached tamarin; 14—Geoffroys saddle-back tamarin; 15–17—Capuchin monkey; 18—Squirrel monkey; 19—Owl monkey; 20—Northern white-faced saki; 21—Bolivian gray titi; 22—100 bp ladder. Scientific names of the primates are indicated below the gel images. Letters on the right side of the gel image correspond with those found in <a href="#life-12-01655-t001" class="html-table">Table 1</a>. Loci names, PCR primers and DNA samples are available in <a href="#app1-life-12-01655" class="html-app">Supplementary File S1</a>.</p>
Full article ">Figure 3
<p><span class="html-italic">Alu</span> subfamily distribution of post-alignment shared <span class="html-italic">Alu</span> insertions for each of ten pre-defined categories. CS: shared by capuchin and squirrel monkey to the exclusion of marmoset and owl monkey has the most elements. <span class="html-italic">Alu</span>Ta15 and derived younger subfamilies dominate recent <span class="html-italic">Alu</span> expansion in the two-genome categories, while four-way sequence alignments in which an <span class="html-italic">Alu</span> insertion appeared to be shared in three genomes and absent from the fourth are more broadly represented by both older <span class="html-italic">Alu</span>S and younger <span class="html-italic">Alu</span>Ta subfamilies.</p>
Full article ">Figure 4
<p>Genome alignments showing examples of predicted shared <span class="html-italic">Alu</span> insertions and post-alignment inspection results. (<b>A</b>) Locus CMS-20 the target <span class="html-italic">Alu</span> (start position in gray highlight) is shared by capuchin, marmoset, and squirrel monkey to the exclusion of owl monkey (precise pre-integration site) with matching flanking sequence and TDSs in yellow highlight. (<b>B</b>) Locus CMS-11 was predicted to be the same as in A, however upon inspection of the alignment the owl monkey sequence displays portions of the target <span class="html-italic">Alu</span> sequence, both after the start position (gray highlight) and before the TSDs (yellow highlight), while lacking the insertion sequence only between positions 652–784. Thus, the target <span class="html-italic">Alu</span> is actually shared by all four CMOS. (<b>C</b>) Locus CM-32 was predicted to be shared by capuchin and marmoset, to the exclusion of owl monkey and squirrel monkey. The target <span class="html-italic">Alu</span> from the capuchin genome [Cebus imitator_1.0] starts at position 641 (gray highlight), is flanked by TSDs in yellow highlight, while owl monkey and squirrel monkey display precise pre-integration sites. However, the marmoset sequence has a different <span class="html-italic">Alu</span> insertion, a near parallel insertion (NP) starting at position 610 (aqua highlight) with TSDs in bright green highlight.</p>
Full article ">Figure 4 Cont.
<p>Genome alignments showing examples of predicted shared <span class="html-italic">Alu</span> insertions and post-alignment inspection results. (<b>A</b>) Locus CMS-20 the target <span class="html-italic">Alu</span> (start position in gray highlight) is shared by capuchin, marmoset, and squirrel monkey to the exclusion of owl monkey (precise pre-integration site) with matching flanking sequence and TDSs in yellow highlight. (<b>B</b>) Locus CMS-11 was predicted to be the same as in A, however upon inspection of the alignment the owl monkey sequence displays portions of the target <span class="html-italic">Alu</span> sequence, both after the start position (gray highlight) and before the TSDs (yellow highlight), while lacking the insertion sequence only between positions 652–784. Thus, the target <span class="html-italic">Alu</span> is actually shared by all four CMOS. (<b>C</b>) Locus CM-32 was predicted to be shared by capuchin and marmoset, to the exclusion of owl monkey and squirrel monkey. The target <span class="html-italic">Alu</span> from the capuchin genome [Cebus imitator_1.0] starts at position 641 (gray highlight), is flanked by TSDs in yellow highlight, while owl monkey and squirrel monkey display precise pre-integration sites. However, the marmoset sequence has a different <span class="html-italic">Alu</span> insertion, a near parallel insertion (NP) starting at position 610 (aqua highlight) with TSDs in bright green highlight.</p>
Full article ">Figure 5
<p>PCR analyses of <span class="html-italic">Alu</span> elements shared in three of four cebid genera and also absent in Atelidae and Pitheciidae. Lanes: 1—100 bp DNA ladder; 2—blank; 3—TLE (negative control); 4—Human (HeLa); 5—<span class="html-italic">Pan troglodytes</span> (common chimpanzee); 6—<span class="html-italic">Chlorocebus aethiops</span> (African green monkey); 7—<span class="html-italic">Lagothrix lagotricha</span> (woolly monkey); 8—<span class="html-italic">Ateles belzebuth</span> (white bellied spider monkey); 9—<span class="html-italic">Ateles geoffroyi</span> (black-handed spider monkey); 10—<span class="html-italic">Alouatta sara</span> (Bolivian red howler monkey; 11—<span class="html-italic">Callithrix jacchus</span> (common marmoset); 12—<span class="html-italic">Callithrix pygmea</span> (Pygmy marmoset); 13—<span class="html-italic">Callimico goeldii</span> (Goeldi’s marmoset); 14—<span class="html-italic">Saguinus labiatus</span> (red-chested mustached tamarin); 15—<span class="html-italic">Saguinus fuscicollis nigrifrons</span> (Geoffroys saddle-back tamarin); 16–18 <span class="html-italic">Sapajus apella</span> (tufted capuchin); 19—100 bp DNA ladder; 20—blank; 21—<span class="html-italic">Saimiri s. sciureus</span> (common squirrel monkey); 22—<span class="html-italic">Aotus trivirgatus</span> (Three striped owl monkey); 23—<span class="html-italic">Pithecia p. pithecia</span> (Northern white-faced saki); 24—<span class="html-italic">Callicebus d. donacophilus</span> (Bolivian gray titi monkey). (<b>A</b>) COS #51, <span class="html-italic">Alu</span> is present capuchin, owl monkey and squirrel monkey (~790 bp DNA fragment lanes 16–18, 21–22) and absent in marmosets and tamarins (~465 bp DNA fragment lanes 11–15). This <span class="html-italic">Alu</span>Sc supports callitrichines (marmosets and tamarins) as basal within Cebidae. (<b>B</b>) CMS #64, <span class="html-italic">Alu</span> is present in capuchin, marmosets, tamarins and squirrel monkey (~800 bp DNA fragment lanes 11–18, 21) and absent in owl monkey (~496 bp DNA fragment lane 22). This <span class="html-italic">Alu</span>Sc supports owl monkey (<span class="html-italic">Aotus</span>) as basal within Cebidae. (<b>C</b>) CMO #29, <span class="html-italic">Alu</span> is present in capuchin, marmosets, tamarins and owl monkey (~860 bp DNA fragment lanes 11–18, 22) and absent in squirrel monkey (~525 bp DNA fragment lane 21). This <span class="html-italic">Alu</span>Ta7 supports squirrel monkey as basal within Cebidae. (<b>D</b>) MOS #5, <span class="html-italic">Alu</span> is present in marmosets, tamarins, owl monkey and squirrel monkey (~880 bp DNA fragment lanes 11–15, 21–22) and absent in capuchins (~564 bp DNA fragment lanes 16–18). This <span class="html-italic">Alu</span>Ta7 supports capuchin as basal within Cebidae.</p>
Full article ">
13 pages, 3428 KiB  
Review
Monkeypox: Re-Emerging Zoonotic Threat
by Rajeev Ranjan and Jitendra Kumar Biswal
Zoonotic Dis. 2022, 2(4), 234-246; https://doi.org/10.3390/zoonoticdis2040019 - 18 Oct 2022
Cited by 5 | Viewed by 5503
Abstract
Monkeypox (MPX) is a relatively unknown and minor resurgent viral zoonotic disease caused by the monkeypox virus (MPXV). The disease can spread from person to person or from animal to person. The disease is most prevalent in the tropical rainforests of West and [...] Read more.
Monkeypox (MPX) is a relatively unknown and minor resurgent viral zoonotic disease caused by the monkeypox virus (MPXV). The disease can spread from person to person or from animal to person. The disease is most prevalent in the tropical rainforests of West and Central Africa. The first MPXV outbreak was recorded in a monkey during 1958 as a small pox-like disease causing flu-like symptoms, such as chills and fever, as well as a rash, and the first MPXV case in a human was in a 9-month-old child in the Democratic Republic of the Congo on 1 September 1970. There were 16,016 laboratory confirmed cases of MPXV infection and five deaths reported in 75 countries/territories/areas across all six WHO Regions as of 22 July 2022. MPXV has a wide host range, including humans, squirrels, mice, rabbits, hamsters, porcupines, non-human primates (orangutans, chimps, sooty mangabeys, cynomolgus monkeys), black-tailed prairie dogs, African brush-tailed porcupines, rats, and shrews. MPXV replicates at the site of inoculation, the respiratory or oropharyngeal mucosa, and spreads to other organs, such as the skin, lungs, and gastrointestinal tract, where clinical signs and symptoms of the disease manifest. Before the rash appears, most patients have prominent lymphadenopathy, which distinguishes human MPX from small pox. This is followed by macules, papules, vesicles, pustules, umbilication, scabbing, and desquamation. Laboratory tools, such as virus isolation, PCR-based assays, haemagglutination inhibition assays, electron microscopy, ELISA, Western blotting, or immunohistochemistry, have been used to confirm diagnoses. Following a confirmatory diagnosis, tecovirimat, an FDA-approved antiviral drug, is currently available to treat severe cases of MPXV infection, along with symptomatic and supportive therapies. Physical and close contact activities, such as sleeping in the same room or on the same bed as the infected person, intimate contact with an infected partner, living in the same house as infected people, and sharing the same cups and plates, must be avoided to prevent the spread of the disease. Vaccination with vaccinia virus against monkeypox is approximately 85% effective and may protect against MPXV infection if administered within 4 days and up to 14 days (without showing any symptoms) after initial contact with a confirmed monkeypox case. Full article
(This article belongs to the Special Issue Feature Papers of Zoonotic Diseases 2021–2022)
Show Figures

Figure 1

Figure 1
<p>Timeline of monkeypox outbreaks globally at various interval [<xref ref-type="bibr" rid="B26-zoonoticdis-02-00019">26</xref>,<xref ref-type="bibr" rid="B27-zoonoticdis-02-00019">27</xref>,<xref ref-type="bibr" rid="B28-zoonoticdis-02-00019">28</xref>,<xref ref-type="bibr" rid="B29-zoonoticdis-02-00019">29</xref>,<xref ref-type="bibr" rid="B30-zoonoticdis-02-00019">30</xref>] Data used for making timeline from Centers for Disease Control and Prevention, 2016, Formenty et al. (2010), Learned et al. (2005), International Federation of Red Cross and Red Crescent Societies (2016), Damon et al. 2006.</p>
Full article ">Figure 2
<p>Geographic distribution of confirmed cases of monkeypox reported to or identified by WHO from official public sources from 1 January 2022 to 7 August 17:00 CEST (adopted from WHO official site, <uri>https://www.who.int/publications/m/item/multi-country-outbreak-of-monkeypox--external-situation-report--3---10-august-2022</uri>) accessed on 13 August 2022 [<xref ref-type="bibr" rid="B6-zoonoticdis-02-00019">6</xref>].</p>
Full article ">Figure 3
<p>Number of confirmed monkeypox cases and deaths reported as on 7 August 17:00 CEST by WHO (Numerical value taken from WHO official site).</p>
Full article ">Figure 4
<p>Transmission of human monkeypox in endemic and non-endemic settings, where monkeypox transmission leads to an outbreak of this disease. Zoonotic factors are very important in endemic settings, while in non-endemic settings, transmission could be the result of transferring infected animals from one place to another. Human-to-human transmission occurs both in endemic and non-endemic settings (adopted form Titanji et al., 2022 [<xref ref-type="bibr" rid="B59-zoonoticdis-02-00019">59</xref>] with minor modification).</p>
Full article ">Figure 5
<p>Mode of transmission of monkeypox [<xref ref-type="bibr" rid="B6-zoonoticdis-02-00019">6</xref>].</p>
Full article ">Figure 6
<p>Pathogenesis of monkeypox virus.</p>
Full article ">Figure 7
<p>Frequency of symptoms in reported cases of monkeypox globally, as of 22 July 2022 (<italic>n</italic> = 9099) (data were taken from WHO official site) [<xref ref-type="bibr" rid="B75-zoonoticdis-02-00019">75</xref>].</p>
Full article ">
11 pages, 1670 KiB  
Article
Experimental Bovine Spongiform Encephalopathy in Squirrel Monkeys: The Same Complex Proteinopathy Appearing after Very Different Incubation Times
by Pedro Piccardo, Juraj Cervenak, Wilfred Goldmann, Paula Stewart, Kitty L. Pomeroy, Luisa Gregori, Oksana Yakovleva and David M. Asher
Pathogens 2022, 11(5), 597; https://doi.org/10.3390/pathogens11050597 - 20 May 2022
Viewed by 2924
Abstract
Incubation periods in humans infected with transmissible spongiform encephalopathy (TSE) agents can exceed 50 years. In humans infected with bovine spongiform encephalopathy (BSE) agents, the effects of a “species barrier,” often observed when TSE infections are transmitted from one species to another, would [...] Read more.
Incubation periods in humans infected with transmissible spongiform encephalopathy (TSE) agents can exceed 50 years. In humans infected with bovine spongiform encephalopathy (BSE) agents, the effects of a “species barrier,” often observed when TSE infections are transmitted from one species to another, would be expected to increase incubation periods compared with transmissions of same infectious agents within the same species. As part of a long-term study investigating the susceptibility to BSE of cell cultures used to produce vaccines, we inoculated squirrel monkeys (Saimiri sp., here designated SQ) with serial dilutions of a bovine brain suspension containing the BSE agent and monitored them for as long as ten years. Previously, we showed that SQ infected with the original “classical” BSE agent (SQ-BSE) developed a neurological disease resembling that seen in humans with variant CJD (vCJD). Here, we report the final characterization of the SQ-BSE model. We observed an unexpectedly marked difference in incubation times between two animals inoculated with the same dilution and volume of the same C-BSE bovine brain extract on the same day. SQ-BSE developed, in addition to spongiform changes and astrogliosis typical of TSEs, a complex proteinopathy with severe accumulations of protease-resistant prion protein (PrPTSE), hyperphosphorylated tau (p-tau), ubiquitin, and α-synuclein, but without any amyloid plaques or β-amyloid protein (Aβ) typical of Alzheimer’s disease. These results suggest that PrPTSE enhanced the accumulation of several key proteins characteristically seen in human neurodegenerative diseases. The marked variation in incubation periods in the same experimental TSE should be taken into account when modeling the epidemiology of human TSEs. Full article
(This article belongs to the Special Issue Human Prion Disease)
Show Figures

Figure 1

Figure 1
<p>Western blot of brain extracts from bovine BSE (lanes 1–2), human vCJD (lanes 3–4), SQ-BSE 735 (lanes 5–6), SQ-BSE 736 (lanes 7–8) and from SQ 659 uninfected control (lanes 9–10). Total PrP (brain extracts with no proteinase K [PK] digestion) are shown in lanes 1, 3, 5, 7 and 9; brain extracts treated with PK are shown in lanes 2, 4, 6, 8 and 10. Western blots were probed with PrP monoclonal antibody 6D11.</p>
Full article ">Figure 2
<p>Comparative neuropathology of two SQ-BSE with extremely different incubation times. Squirrel monkeys inoculated with classical BSE (SQ-BSE) developed TSE and a complex proteinopathy (<b>A</b>–<b>H</b>). SQ-BSE 735, incubation period 3.3 years (39.6 months) (<b>A</b>,<b>C</b>,<b>E</b>,<b>G</b>); SQ-BSE 736 incubation period 8.1 years (94.8 months) (<b>B</b>,<b>D</b>,<b>F</b>,<b>H</b>). Moderate (<b>A</b>) or severe (<b>B</b>) spongiform degeneration in frontal cortex. Sections stained with hematoxylin-eosin. Moderate (<b>C</b>) or severe (<b>D</b>) PrP<sup>TSE</sup> in frontal cortex. Moderate (<b>E</b>) or severe (<b>F</b>) p-tau immunopositivity in frontal cortex. (<b>A</b>,<b>B</b>) sections stained with hematoxylin-eosin; (<b>C</b>,<b>D</b>) sections immunostained with anti-PrP antibody 6H4; (<b>E</b>,<b>F</b>) sections immunostained with anti-tau antibody AT8. Panels A–F, 20× magnification. (<b>G</b>,<b>H</b>) sections of frontal cortex immunostained with 4D6 antibody against α-synuclein showing small granular accumulations (arrows), 40× magnifications and further enlarged in squared areas.</p>
Full article ">
16 pages, 3283 KiB  
Article
Toxoplasmosis in Zoo Animals: A Retrospective Pathology Review of 126 Cases
by Daniela Denk, Simon De Neck, Shannon Khaliq and Mark F. Stidworthy
Animals 2022, 12(5), 619; https://doi.org/10.3390/ani12050619 - 1 Mar 2022
Cited by 16 | Viewed by 5511
Abstract
Toxoplasma gondii is an extremely successful zoonotic protozoan parasite that has been demonstrated in a wide range of endo- and poikilothermic species. Although infection is widespread amongst domestic animals, overt disease other than abortion in small ruminants is sporadic. This survey evaluates toxoplasmosis [...] Read more.
Toxoplasma gondii is an extremely successful zoonotic protozoan parasite that has been demonstrated in a wide range of endo- and poikilothermic species. Although infection is widespread amongst domestic animals, overt disease other than abortion in small ruminants is sporadic. This survey evaluates toxoplasmosis in zoo animals based on a systematic review of pathology archive material (n = 33,506 submissions) over a 16-year study period. A total of 126 submissions, deriving from 32 zoos, two educational facilities and two private owners, were included in the study, based on gross lesions, cytological, histological and immunohistological diagnosis of toxoplasmosis. Clinical history, signalment, annual distribution and post-mortem findings were evaluated. A total of 31 species (mammalian 97%/avian 3%) were represented in the study material. Ring-tailed lemurs, slender tailed meerkats, Pallas’ cats, and squirrel monkeys were most affected. An unusual outbreak occurred in Asian small-clawed otters, in which toxoplasmosis has not been reported to date. Clinically, animals over 12 months of age presented with non-specific symptoms (anorexia, weight loss, lethargy, debilitation), neurological, gastrointestinal or respiratory signs and sudden death. Systemic disease predominated, with a propensity for encephalitis in meerkats and Pallas’ cats and systemic disease involving lymphoid tissues in ring-tailed lemurs. Cases in the UK occurred year-round, with species-specific peaks and increases between August and November. This study reinforces the importance of toxoplasmosis as a significant cause of sporadic and epizootic mortalities in a wide range of zoo animals. Feral cat control is crucial to reduce infection pressure. Full article
(This article belongs to the Special Issue Pathology in Zoo Animals and Conservation)
Show Figures

Figure 1

Figure 1
<p>Overview of affected ages. Infection is most common in animals over 12 months of age.</p>
Full article ">Figure 2
<p>Pale grey dotted line indicates a statistically significant rising annual trend in the per-centage of total cases submitted with a clinical suspicion of toxoplasmosis (<span class="html-italic">p</span> &lt; 0.05, Mann–Kendall test, Real Statistics Rel 7.10, Microsoft Excel). There is no rising trend in the overall number of confirmed toxoplasmosis cases over the same period (<span class="html-italic">p</span> &gt; 0.05, Mann–Kendall test).</p>
Full article ">Figure 3
<p>(<b>A</b>,<b>B</b>): Slender-tailed meerkat, 3 years, female, liver. A: Amongst hepatocytes, histiocytes and a background of erythrocytes there is a large cluster of intracytoplasmic <span class="html-italic">Toxoplasma</span> tachyzoites (arrow head); impression smear cytology, Leishman’s stain, ×100. (<b>B</b>): Large areas of coagulative necrosis efface hepatic parenchyma. Organisms are difficult to identify: H&amp;E, ×20. (<b>C</b>): Pallas’ cat, 9 years, female, brain: Moderate to marked mixed mononuclear encephalitis with perivascular cuffing and intralesional protozoa. H&amp;E, ×10, inset ×60. (<b>D</b>): Eurasian badger, adult, female, lung: Severe coalescent necrotising pneumonia. Intralesional intracellular tachyzoites (arrow heads) are present, and the cytoplasm of macrophages contain intralesional bacteria (arrow) (identified as mycobacteria on special stains). H&amp;E, ×2, inset ×100.</p>
Full article ">Figure 4
<p>(<b>A</b>): Ring-tailed lemur, 2 years, male, mesenteric lymph node. Severe confluent necrotising lymphadenitis. H&amp;E, ×2. (<b>B</b>): Speckled mousebird, &gt;7 years, male, spleen. Multifocal to coalescing necrotising splenitis with myriads of intralesional <span class="html-italic">T. gondii</span> protozoa, confirmed by immunohistochemistry (inset). H&amp;E, ×2 and IHC, ×60. (<b>C</b>): Snow leopard, 18 years, female, tongue. Glossal myofibres are separated by histiocytic and lesser neutrophilic infiltrates, amongst which protozoal tachyzoites (arrow head) are identified. (<b>D</b>): Alpaca, 14 years, male, abdominal mass. Numerous clusters of intracellular (arrow heads) and extracellular (arrow) tachyzoites are present amongst sheets of mononuclear inflammation, embedded on fibrovascular stroma. H&amp;E, ×60.</p>
Full article ">Figure 5
<p><span class="html-italic">Toxoplasma</span>-related lesions were evident in a wide range of tissues, presented in descending order.</p>
Full article ">Figure 6
<p>Tissue distribution of <span class="html-italic">Toxoplasma</span> lesions in ring tailed lemurs (<span class="html-italic">n</span> = 31), slender tailed meerkats (<span class="html-italic">n</span> = 30), Pallas’ cats (<span class="html-italic">n</span> = 13), squirrel monkeys (<span class="html-italic">n</span> = 13), Asian small-clawed otters (<span class="html-italic">n</span> = 5) and Patagonian maras (<span class="html-italic">n</span> = 5). The Y axis values demonstrate the frequency of <span class="html-italic">Toxoplasma</span>-induced lesions.</p>
Full article ">Figure 7
<p>Annual distribution of case numbers between March 2003 and May 2019.</p>
Full article ">Figure 8
<p>Monthly distribution of case numbers over the study period.</p>
Full article ">Figure 9
<p>Monthly distribution of case numbers in ring-tailed lemurs, slender-tailed meerkats, Pallas’ cats, and squirrel monkeys. Pallas’ cat cases were not recorded between January and April.</p>
Full article ">
32 pages, 5924 KiB  
Article
The Intriguing Effects of Substituents in the N-Phenethyl Moiety of Norhydromorphone: A Bifunctional Opioid from a Set of “Tail Wags Dog” Experiments
by Meining Wang, Thomas C. Irvin, Christine A. Herdman, Ramsey D. Hanna, Sergio A. Hassan, Yong-Sok Lee, Sophia Kaska, Rachel Saylor Crowley, Thomas E. Prisinzano, Sarah L. Withey, Carol A. Paronis, Jack Bergman, Saadet Inan, Ellen B. Geller, Martin W. Adler, Theresa A. Kopajtic, Jonathan L. Katz, Aaron M. Chadderdon, John R. Traynor, Arthur E. Jacobson and Kenner C. Riceadd Show full author list remove Hide full author list
Molecules 2020, 25(11), 2640; https://doi.org/10.3390/molecules25112640 - 6 Jun 2020
Cited by 11 | Viewed by 4392
Abstract
(−)-N-Phenethyl analogs of optically pure N-norhydromorphone were synthesized and pharmacologically evaluated in several in vitro assays (opioid receptor binding, stimulation of [35S]GTPγS binding, forskolin-induced cAMP accumulation assay, and MOR-mediated β-arrestin recruitment assays). “Body” and “tail” interactions with opioid [...] Read more.
(−)-N-Phenethyl analogs of optically pure N-norhydromorphone were synthesized and pharmacologically evaluated in several in vitro assays (opioid receptor binding, stimulation of [35S]GTPγS binding, forskolin-induced cAMP accumulation assay, and MOR-mediated β-arrestin recruitment assays). “Body” and “tail” interactions with opioid receptors (a subset of Portoghese’s message-address theory) were used for molecular modeling and simulations, where the “address” can be considered the “body” of the hydromorphone molecule and the “message” delivered by the substituent (tail) on the aromatic ring of the N-phenethyl moiety. One compound, N-p-chloro-phenethynorhydromorphone ((7aR,12bS)-3-(4-chlorophenethyl)-9-hydroxy-2,3,4,4a,5,6-hexahydro-1H-4,12-methanobenzofuro[3,2-e]isoquinolin-7(7aH)-one, 2i), was found to have nanomolar binding affinity at MOR and DOR. It was a potent partial agonist at MOR and a full potent agonist at DOR with a δ/μ potency ratio of 1.2 in the ([35S]GTPγS) assay. Bifunctional opioids that interact with MOR and DOR, the latter as agonists or antagonists, have been reported to have fewer side-effects than MOR agonists. The p-chlorophenethyl compound 2i was evaluated for its effect on respiration in both mice and squirrel monkeys. Compound 2i did not depress respiration (using normal air) in mice or squirrel monkeys. However, under conditions of hypercapnia (using air mixed with 5% CO2), respiration was depressed in squirrel monkeys. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Conserved polar/charged residues (in red; numbering as in MOR) that interacted with the -O and -OH groups of the <span class="html-italic">N</span>-phenethylnorhydromorphone body through H-bonds; the body was also stabilized by close packing with four conserved nonpolar residues (blue) through hydrophobic forces. Although not in direct interactions, a few non-conserved residues (green) were seen to interact with the ligands indirectly through short water chains. These residues may thus be important to modulate the behavior of MOR and DOR independently, which may be accomplished through specific substituents that can engage them more directly. Frequencies of contacts were deemed statistically relevant if they were observed at least 25% of the time, except for the critical -NH---D147 distance (MOR) and -NH---D128 distance (DOR) that was required to persist for at least 75% of the time for the conformer to be considered (see details of the analysis in <a href="#app1-molecules-25-02640" class="html-app">Table S2</a>).</p>
Full article ">Figure 2
<p>Critical tail-OR interactions of <span class="html-italic">p</span>-F (<b>2e</b>) and -Cl (<b>2i</b>) substituents. (<b>A</b>) <span class="html-italic">p</span>-F (weak partial DOR agonist) vs. <span class="html-italic">p</span>-Cl (potent full DOR agonist); only one of the two conformers of each ligands are shown (see text); (<b>B</b>) <span class="html-italic">p</span>-Cl (potent partial MOR agonists); the two conformers shown. The red lines represent frequent, statistically significant interactions that were obtained from the dynamic simulations. Sequence numbering as in the corresponding ORs.</p>
Full article ">Figure 3
<p>Effects of morphine and <b>2i</b> on respiratory rate in mice. After acclimation in observation boxes, mice were injected with either saline, morphine 10 mg/kg, or <b>2i</b> and connected to a throat sensor. Five min later, the recording was started and respiratory rate was measured from 6 min to 45 min post-injection (<b>A</b>). Area under the curve (AUC) was calculated from 6 min to 45 min. Morphine significantly reduced respiratory rate compared to saline (<b>B</b>). Data are expressed as mean ± standard error of the mean (SEM.) (<span class="html-italic">n</span> = 6–8) (**** <span class="html-italic">p</span> &lt; 0.0001). One-way ANOVA followed by Dunnett’s multiple comparison test.</p>
Full article ">Figure 4
<p>Effects of <b>2i</b> on tail withdrawal latency from different temperatures of water (filled symbols) and operant behavior disruption (open symbols) in squirrel monkeys (<span class="html-italic">n</span> = 4). Data are expressed as mean ± SEM (<span class="html-italic">n</span> = 4).</p>
Full article ">Figure 5
<p>Effects in squirrel monkeys of <b>2i</b> and morphine on respiratory rate (bottom panels) or on minute volume (top panels) in the presence of normal air (open symbols) or air mixed with 5% CO<sub>2</sub> (filled symbols). <b>2i</b> and morphine significantly reduced minute volume in 5% CO<sub>2</sub> without significantly altering respiratory rate. Data are expressed as mean ± SEM. (<span class="html-italic">n</span> = 4); (*) indicates the difference from saline (<span class="html-italic">p</span> ≤ 0.01; one-way ANOVA followed by Dunnett’s multiple comparison test.</p>
Full article ">Scheme 1
<p>Prototypical 4,5-epoxymorphinan agonists and antagonists.</p>
Full article ">Scheme 2
<p>Synthesis of N-substituted hydromorphones.</p>
Full article ">
550 KiB  
Review
Recent Advances in BLV Research
by Pierre-Yves Barez, Alix De Brogniez, Alexandre Carpentier, Hélène Gazon, Nicolas Gillet, Gerónimo Gutiérrez, Malik Hamaidia, Jean-Rock Jacques, Srikanth Perike, Sathya Neelature Sriramareddy, Nathalie Renotte, Bernard Staumont, Michal Reichert, Karina Trono and Luc Willems
Viruses 2015, 7(11), 6080-6088; https://doi.org/10.3390/v7112929 - 24 Nov 2015
Cited by 71 | Viewed by 12610
Abstract
Different animal models have been proposed to investigate the mechanisms of Human T-lymphotropic Virus (HTLV)-induced pathogenesis: rats, transgenic and NOD-SCID/γcnull (NOG) mice, rabbits, squirrel monkeys, baboons and macaques. These systems indeed provide useful information but have intrinsic limitations such as lack of disease [...] Read more.
Different animal models have been proposed to investigate the mechanisms of Human T-lymphotropic Virus (HTLV)-induced pathogenesis: rats, transgenic and NOD-SCID/γcnull (NOG) mice, rabbits, squirrel monkeys, baboons and macaques. These systems indeed provide useful information but have intrinsic limitations such as lack of disease relevance, species specificity or inadequate immune response. Another strategy based on a comparative virology approach is to characterize a related pathogen and to speculate on possible shared mechanisms. In this perspective, bovine leukemia virus (BLV), another member of the deltaretrovirus genus, is evolutionary related to HTLV-1. BLV induces lymphoproliferative disorders in ruminants providing useful information on the mechanisms of viral persistence, genetic determinants of pathogenesis and potential novel therapies. Full article
(This article belongs to the Special Issue Recent Advances in HTLV Research 2015)
Show Figures

Figure 1

Figure 1
<p>Schematic structure of (<b>a</b>) the bovine leukemia virus (BLV) genome and (<b>b</b>) the viral particle.</p>
Full article ">
Back to TopTop