Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
1.
Sci Rep ; 13(1): 14950, 2023 09 11.
Article in English | MEDLINE | ID: mdl-37696929

ABSTRACT

The environment has an important effect on the gut microbiota-an essential part of the host's health-and is strongly influenced by the dietary pattern of the host as these together shape the composition and functionality of the gut microbiota in humans and other animals. This study compared the gut microbiota of Macaca fascicularis fascicularis and M. f. aurea in mangrove and island populations using 16S rRNA gene sequencing on a nanopore platform to investigate the effect of the environment and/or diet. The results revealed that the M. f. fascicularis populations that received anthropogenic food exhibited a higher richness and evenness of gut microbiota than the M. f. aurea populations in different habitats. Firmicutes and Bacteroidetes were the two most abundant bacterial phyla in the gut microbiota of both these subspecies; however, the relative abundance of these phyla was significantly higher in M. f. aurea than in M. f. fascicularis. This variation in the gut microbiota between the two subspecies in different habitats mostly resulted from the differences in their diets. Moreover, the specific adaptation of M. f. aurea to different environments with a different food availability had a significant effect on their microbial composition.


Subject(s)
Diet , Ecosystem , Gastrointestinal Microbiome , Macaca fascicularis , Animals , Gastrointestinal Microbiome/genetics , Macaca fascicularis/genetics , Macaca fascicularis/microbiology , RNA, Ribosomal, 16S/genetics , Species Specificity
2.
J Vet Med Sci ; 85(4): 528-531, 2023 Apr 22.
Article in English | MEDLINE | ID: mdl-36878554

ABSTRACT

Aeromonas hydrophila is a facultative anaerobic gram-negative bacterium regarded as an opportunistic pathogen in animals. A 17-year-old female crab-eating macaque (Macaca fascicularis) died after experiencing anorexia and depression for several days. The carcass was severely emaciated, and the sternum was exposed under subcutaneous lesions in the thorax. Many abnormal pathological lesions were found, including tracheal inflammation, pulmonary inflammatory emphysema, yellowish discoloration of the liver, enlargement of the gall bladder, necrosis of the heart, congested bilateral kidneys, and enlargement of the adrenal glands. The stomach was empty, mucosal ulcerations were found, and the duodenum was congested. Giemsa staining revealed rod-shaped organisms in the whole blood smear and major organs, which were identified as A. hydrophila. The animal had experienced stress, and decreased immune system function possibly contributed to the infection.


Subject(s)
Aeromonas hydrophila , Gram-Negative Bacterial Infections , Macaca fascicularis , Animals , Female , Aeromonas hydrophila/isolation & purification , Death, Sudden/etiology , Death, Sudden/veterinary , Gram-Negative Bacterial Infections/diagnosis , Gram-Negative Bacterial Infections/veterinary , Macaca fascicularis/microbiology , Macaca fascicularis/psychology , Animals, Zoo/microbiology , Animals, Zoo/psychology , Stress, Psychological/complications
4.
Fungal Genet Biol ; 144: 103468, 2020 11.
Article in English | MEDLINE | ID: mdl-32980453

ABSTRACT

Cynomolgus macaque (Macaca fascicularis) is currently a common animal model for biomedical research. The National Primate Research Center of Thailand, Chulalongkorn University (NPRCT-CU) translocated wild-borne macaques to reared colony for research purposes. At present, no studies focus on fungal microbiome (Mycobiome) of this macaque. The functional roles of mycobiome and fungal pathogens have not been elucidated. Thus, this study aimed to investigate and compare oral and fecal mycobiome between wild and captive macaques by using high-throughput sequencing on internal transcribed spacer 2 (ITS2) rDNA. The results showed that the mycobiome of wild macaque has greater alpha diversity. The fecal mycobiome has more limited alpha diversity than those in oral cavity. The community is mainly dominated by saprophytic yeast in Kasachstania genus which is related to aiding metabolic function in gut. The oral microbiome of most captive macaques presented the Cutaneotrichosporon suggesting the fungal transmission through skin-oral contact within the colony. The potential pathogens that would cause harmful transmission in reared colonies were not found in either group of macaques but the pathogen prevention and animal care is still important to be concerned. In conclusion, the results of gut mycobiome analysis in Thai cynomolgus macaques provide us with the basic information of oral and fecal fungi and for monitoring macaque's health status for animal care of research use.


Subject(s)
DNA, Ribosomal Spacer/genetics , Fungi/genetics , Macaca fascicularis/microbiology , Mycobiome/genetics , Animals , Feces/microbiology , Fungi/classification , Fungi/isolation & purification , Genetic Variation/genetics , High-Throughput Nucleotide Sequencing , Macaca fascicularis/genetics , Mouth/microbiology
5.
Anaerobe ; 64: 102236, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32623046

ABSTRACT

Clostridium perfringens is ubiquitous in the environment and the gastrointestinal tract of warm-blooded animals. While part of the gut microbiome, abnormal growth of C. perfringens causes histotoxic, neurologic, and enteric diseases in a variety of animal species, including humans, due to the production of toxins. There is extremely limited information on C. perfringens infection in non-human primates. Presently, 10 strains were successfully isolated from 126 monkeys and confirmed by molecular and biochemical analyses. All isolates were genotype A based on molecular analysis. Alpha toxin was identified in all isolates. Beta 2 toxin was detected in only three isolates. No other toxins, including enterotoxin, beta, iota, epsilon, and net B toxin, were identified in any isolate. All isolates were highly susceptible to ß-lactam antibiotics. Double hemolysis and lecithinase activity were commonly observed in all strains. Biofilm formation, which can increase antibiotic resistance, was identified in 90% of the isolates. The data are the first report the prevalence and characteristics of C. perfringens isolated from captive cynomolgus monkeys.


Subject(s)
Bacterial Toxins/genetics , Biofilms/growth & development , Clostridium perfringens/drug effects , Clostridium perfringens/genetics , Drug Resistance, Multiple, Bacterial , Macaca fascicularis/microbiology , Animals , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Clostridium Infections/microbiology , Clostridium Infections/veterinary , Clostridium perfringens/isolation & purification , DNA, Bacterial/genetics , Feces/microbiology , Female , Genotype , Male , Phylogeny , Prevalence , RNA, Ribosomal, 16S/genetics , beta-Lactams/pharmacology
7.
Aging (Albany NY) ; 11(24): 12080-12096, 2019 12 14.
Article in English | MEDLINE | ID: mdl-31837260

ABSTRACT

Age can significantly affect human physiology and disease risk. Recent studies have shown that age may affect the composition and function of the gut microbiota, but the underlying mechanisms remain largely unknown. Non-human primates are an ideal model for uncovering how age shapes the gut microbiota, as their microbial composition is highly similar to that of humans and is not easily affected by confounding factors. Here, using the 16S rRNA and metagenomic sequencing methods, we characterized the microbial phenotypes of 16 female cynomolgus macaques from three age groups (young, adult and old). Our findings revealed significant differences in microbial composition among the three groups. With increased age, the relative abundances of Veillonellaceae, Coriobacteriaceae and Succinivibrionaceae were significantly increased, Ruminococcaceae and Rikenellaceae were significantly decreased at the family level. Functional enrichment showed that genes that differed among the three groups were mainly involved in arginine biosynthesis, purine metabolism and microbial polysaccharides metabolism. Moreover, CAZymes corresponding to polysaccharide degrading activities were also observed among the three groups. In conclusion, we characterized the composition and function of the gut microbiota at different ages, and our findings provide a new entry point for understanding the effects of age on the human body.


Subject(s)
Aging/physiology , Bacteria/classification , Gastrointestinal Microbiome , Macaca fascicularis/microbiology , Amino Acids/metabolism , Animals , Bacteria/enzymology , Bacteria/genetics , Carbohydrates/chemistry , Female , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/genetics
8.
J Vet Sci ; 20(3): e19, 2019 May.
Article in English | MEDLINE | ID: mdl-31161737

ABSTRACT

Microorganisms play important roles in obesity; however, the role of the gut microbiomes in obesity is controversial because of the inconsistent findings. This study investigated the gut microbiome communities in obese and lean groups of captive healthy cynomolgus monkeys reared under strict identical environmental conditions, including their diet. No significant differences in the relative abundance of Firmicutes, Bacteroidetes and Prevotella were observed between the obese and lean groups, but a significant difference in Spirochetes (p < 0.05) was noted. Microbial diversity and richness were similar, but highly variable results in microbial composition, diversity, and richness were observed in individuals, irrespective of their state of obesity. Distinct clustering between the groups was not observed by principal coordinate analysis using an unweighted pair group method. Higher sharedness values (95.81% ± 2.28% at the genus level, and 79.54% ± 5.88% at the species level) were identified among individual monkeys. This paper reports the association between the gut microbiome and obesity in captive non-human primate models reared under controlled environments. The relative proportion of Firmicutes and Bacteroidetes as well as the microbial diversity known to affect obesity were similar in the obese and lean groups of monkeys reared under identical conditions. Therefore, obesity-associated microbial changes reported previously appear to be associated directly with environmental factors, particularly diet, rather than obesity.


Subject(s)
Bacterial Physiological Phenomena , Feces/microbiology , Gastrointestinal Microbiome , Macaca fascicularis/microbiology , Monkey Diseases/microbiology , Obesity/veterinary , Animals , Bacteria/classification , Bacteria/genetics , Biodiversity , Female , Obesity/microbiology , RNA, Ribosomal, 16S/genetics
9.
Zool Res ; 40(2): 89-93, 2019 Mar 18.
Article in English | MEDLINE | ID: mdl-30127329

ABSTRACT

Rhesus macaques (Macaca mulatta) and cynomolgus macaques (Macaca fascicularis) are frequently used in establishing animal models for human diseases. To determine the differences in gut microbiota between these species, rectal swabs from 20 rhesus macaques and 21 cynomolgus macaques were collected, and the microbial composition was examined by deep sequencing of the 16S rRNA gene. We found that the rectal microbiota of cynomolgus macaques exhibited significantly higher alpha diversity than that of rhesus macaques, although the observed number of operational taxonomic units (OTUs) was almost the same. The dominant taxa at both the phylum and genus levels were similar between the two species, although the relative abundances of these dominant taxa were significantly different between them. Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt) showed significant differences in the functional components between the microbiota of the two species, in particular the lipopolysaccharide (LPS) synthesis proteins. The above data indicated significant differences in microbial composition and function between these two closely related macaque species, which should be taken into consideration in the future selection of these animals for disease models.


Subject(s)
Bacteria/genetics , Gastrointestinal Microbiome , Macaca fascicularis/microbiology , Macaca mulatta/microbiology , Rectum/microbiology , Animals , Bacteria/classification , Genome, Bacterial , Genomics , Species Specificity
10.
ISME J ; 12(1): 77-86, 2018 01.
Article in English | MEDLINE | ID: mdl-28885626

ABSTRACT

Helicobacter suis is the second most prevalent Helicobacter species in the stomach of humans suffering from gastric disease. This bacterium mainly inhabits the stomach of domesticated pigs, in which it causes gastric disease, but it appears to be absent in wild boars. Interestingly, it also colonizes the stomach of asymptomatic rhesus and cynomolgus monkeys. The origin of modern human-, pig- or non-human primate-associated H. suis strains in these respective host populations was hitherto unknown. Here we show that H. suis in pigs possibly originates from non-human primates. Our data suggest that a host jump from macaques to pigs happened between 100 000 and 15 000 years ago and that pig domestication has had a significant impact on the spread of H. suis in the pig population, from where this pathogen occasionally infects humans. Thus, in contrast to our expectations, H. suis appears to have evolved in its main host in a completely different way than its close relative Helicobacter pylori in humans.


Subject(s)
Helicobacter Infections/microbiology , Helicobacter Infections/veterinary , Helicobacter heilmannii/isolation & purification , Macaca fascicularis/microbiology , Macaca mulatta/microbiology , Swine Diseases/microbiology , Animals , Animals, Domestic/microbiology , Helicobacter heilmannii/classification , Helicobacter heilmannii/genetics , Helicobacter heilmannii/growth & development , Humans , Phylogeny , Stomach/microbiology , Swine
11.
J Med Primatol ; 46(5): 232-238, 2017 10.
Article in English | MEDLINE | ID: mdl-28488364

ABSTRACT

BACKGROUND: Rhesus and cynomologus macaques are valuable animal models for the study of human immunodeficiency virus (HIV) prevention strategies. However, for such studies focused on the vaginal route of infection, differences in vaginal environment may have deterministic impact on the outcome of such prevention, providing the rationale for this study. METHODS: We tested the vaginal environment of rhesus and cynomolgus macaques longitudinally to characterize the normal microflora based on Nugent scores and pH. This evaluation was extended after colonization of the vaginal space with Lactobacilli in an effort to recreate NHP models representing the healthy human vaginal environment. RESULTS AND CONCLUSION: Nugent scores and pH differed significantly between species, although data from both species were suggestive of stable bacterial vaginosis. Colonization with Lactobacilli was successful in both species leading to lower Nugent score and pH, although rhesus macaques appeared better able to sustain Lactobacillus spp over time.


Subject(s)
Lactobacillus/physiology , Macaca fascicularis/microbiology , Macaca mulatta/microbiology , Vagina/microbiology , Animals , Disease Models, Animal , Female , HIV Infections/prevention & control , Humans , Reference Values
12.
PLoS One ; 12(3): e0171906, 2017.
Article in English | MEDLINE | ID: mdl-28273087

ABSTRACT

BACKGROUND: New interventions for tuberculosis are urgently needed. Non-human primate (NHP) models provide the most relevant pre-clinical models of human disease and play a critical role in vaccine development. Models utilising Asian cynomolgus macaque populations are well established but the restricted genetic diversity of the Mauritian cynomolgus macaques may be of added value. METHODS: Mauritian cynomolgus macaques were exposed to a range of doses of M. tuberculosis delivered by aerosol, and the outcome was assessed using clinical, imaging and pathology-based measures. RESULTS: All macaques developed characteristic clinical signs and disease features of tuberculosis (TB). Disease burden and the ability to control disease were dependent on exposure dose. Mauritian cynomolgus macaques showed less variation in pulmonary disease burden and total gross pathology scores within exposure dose groups than either Indian rhesus macaques or Chinese cynomolgus macaques. CONCLUSIONS: The genetic homogeneity of Mauritian cynomolgus macaques makes them a potentially useful model of human tuberculosis.


Subject(s)
Macaca fascicularis/microbiology , Mycobacterium tuberculosis/physiology , Tuberculosis/pathology , Animals , Enzyme-Linked Immunospot Assay , Interferon-gamma/blood , Interferon-gamma/metabolism , Kidney/pathology , Liver/pathology , Lung/diagnostic imaging , Lung/microbiology , Lung/pathology , Macaca fascicularis/immunology , Magnetic Resonance Imaging , Radiography, Thoracic , Severity of Illness Index
13.
Helicobacter ; 22(2)2017 Apr.
Article in English | MEDLINE | ID: mdl-27558281

ABSTRACT

BACKGROUND: Helicobacter suis (H. suis) is the most prevalent gastric non-H. pylori Helicobacter species in humans. This bacterium mainly colonizes the stomach of pigs, but it has also been detected in the stomach of nonhuman primates. The aim of this study was to obtain better insights into potential differences between pig- and primate-associated H. suis strains in virulence and pathogenesis. MATERIALS AND METHODS: In vitro-isolated H. suis strains obtained from pigs, cynomolgus monkeys (Macaca fascicularis), and rhesus monkeys (Macaca mulatta) were used for intragastric inoculation of BALB/c mice and Mongolian gerbils. Nine weeks and six months later, samples of the stomach of inoculated and control animals were taken for PCR analysis and histopathological examination. RESULTS: The cynomolgus monkey-associated H. suis strain only colonized the stomach of mice, but not of Mongolian gerbils. All other H. suis strains colonized the stomach in both rodent models. In all colonized animals, severe gastric inflammation was induced. Gastric lymphoid follicles and destruction of the antral epithelium were observed in infected gerbils, but not in mice. Infection with both pig- and primate-associated H. suis strains evoked a similar marked Th17 response in mice and gerbils, accompanied by increased CXCL-13 expression levels. CONCLUSIONS: Apart from the cynomolgus monkey-associated strain which was unable of colonizing the stomach of Mongolian gerbils, no substantial differences in virulence were found in rodent models between in vitro-cultured pig-associated, cynomolgus monkey-associated and rhesus monkey-associated H. suis strains. The experimental host determines the outcome of the immune response against H. suis infection, rather than the original host.


Subject(s)
Gastritis/pathology , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter heilmannii/isolation & purification , Helicobacter heilmannii/pathogenicity , Animals , Chemokine CXCL13/analysis , Disease Models, Animal , Gastric Mucosa/pathology , Gene Expression Profiling , Gerbillinae , Histocytochemistry , Macaca fascicularis/microbiology , Macaca mulatta/microbiology , Mice, Inbred BALB C , Polymerase Chain Reaction , Swine/microbiology , Th17 Cells/immunology , Virulence
14.
Article in English | MEDLINE | ID: mdl-25766142

ABSTRACT

Tularemia is a zoonotic disease caused by Francisella tularensis, which is transmitted to humans most commonly by contact with infected animals, tick bites, or inhalation of aerosolized bacteria. F. tularensis is highly infectious via the aerosol route; inhalation of as few as 10-50 organisms can cause pneumonic tularemia. Left untreated, the pneumonic form has more than >30% case-fatality rate but with early antibiotic intervention can be reduced to 3%. This study compared tularemia disease progression across three species of nonhuman primates [African green monkey (AGM), cynomolgus macaque (CM), and rhesus macaque (RM)] following aerosolized F. tularensis Schu S4 exposure. Groups of the animals exposed to various challenge doses were observed for clinical signs of infection and blood samples were analyzed to characterize the disease pathogenesis. Whereas the AGMs and CMs succumbed to disease following challenge doses of 40 and 32 colony forming units (CFU), respectively, the RM lethal dose was 276,667 CFU. Following all challenge doses that caused disease, the NHPs experienced weight loss, bacteremia, fever as early as 4 days post exposure, and tissue burden. Necrotizing-to-pyogranulomatous lesions were observed most commonly in the lung, lymph nodes, spleen, and bone marrow. Overall, the CM model consistently manifested pathological responses similar to those resulting from inhalation of F. tularensis in humans and thereby most closely emulates human tularemia disease. The RM model displayed a higher tolerance to infection and survived exposures of up to 15,593 CFU of aerosolized F. tularensis.


Subject(s)
Chlorocebus aethiops , Disease Models, Animal , Francisella tularensis/pathogenicity , Macaca fascicularis , Macaca mulatta , Tularemia , Aerosols , Animals , Bacterial Load , Chlorocebus aethiops/microbiology , Disease Progression , Lung/immunology , Lung/microbiology , Lung/pathology , Macaca fascicularis/microbiology , Macaca mulatta/microbiology , Spleen/microbiology , Spleen/pathology , Tularemia/microbiology , Tularemia/pathology
15.
Cold Spring Harb Perspect Med ; 4(12): a018564, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25213189

ABSTRACT

Nonhuman primates have emerged as an excellent model of human tuberculosis, in large part because they recapitulate the full spectrum of infection outcome and pathology seen in humans. Several variables inherent to the nonhuman primate models of tuberculosis are discussed in this review, including the monkey species, Mycobacterium tuberculosis strains, and routes of infection, all of which can influence the model to be chosen for various studies. New technologies for studying the microbiology, immunology, and pathogenesis of tuberculosis in nonhuman primates have greatly expanded the capabilities of this model for basic and translational studies, including the development and testing of new treatment and prevention strategies for tuberculosis.


Subject(s)
Disease Models, Animal , Primate Diseases/microbiology , Tuberculosis, Pulmonary/physiopathology , Animals , Antitubercular Agents/therapeutic use , Callithrix/microbiology , Coinfection , HIV Infections/microbiology , Humans , Macaca fascicularis/microbiology , Macaca mulatta/microbiology , Primate Diseases/physiopathology , Tuberculosis Vaccines/therapeutic use , Tuberculosis, Pulmonary/drug therapy
16.
PLoS Pathog ; 9(8): e1003501, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23966854

ABSTRACT

Dominant Vγ2Vδ2 T-cell subset exist only in primates, and recognize phosphoantigen from selected pathogens including M. tuberculosis(Mtb). In vivo function of Vγ2Vδ2 T cells in tuberculosis remains unknown. We conducted mechanistic studies to determine whether earlier expansion/differentiation of Vγ2Vδ2 T cells during Mtb infection could increase immune resistance to tuberculosis in macaques. Phosphoantigen/IL-2 administration specifically induced major expansion and pulmonary trafficking/accumulation of phosphoantigen-specific Vγ2Vδ2 T cells, significantly reduced Mtb burdens and attenuated tuberculosis lesions in lung tissues compared to saline/BSA or IL-2 controls. Expanded Vγ2Vδ2 T cells differentiated into multifunctional effector subpopulations capable of producing anti-TB cytokines IFNγ, perforin and granulysin, and co-producing perforin/granulysin in lung tissue. Mechanistically, perforin/granulysin-producing Vγ2Vδ2 T cells limited intracellular Mtb growth, and macaque granulysin had Mtb-bactericidal effect, and inhibited intracellular Mtb in presence of perforin. Furthermore, phosphoantigen/IL2-expanded Vγ2Vδ2 T effector cells produced IL-12, and their expansion/differentiation led to enhanced pulmonary responses of peptide-specific CD4+/CD8+ Th1-like cells. These results provide first in vivo evidence implicating that early expansion/differentiation of Vγ2Vδ2 T effector cells during Mtb infection increases resistance to tuberculosis. Thus, data support a rationale for conducting further studies of the γδ T-cell-targeted treatment of established TB, which might ultimately help explore single or adjunctive phosphoantigen expansion of Vγ2Vδ2 T-cell subset as intervention of MDR-tuberculosis or HIV-related tuberculosis.


Subject(s)
Interleukin-2/administration & dosage , Lung/immunology , Macaca fascicularis/microbiology , Mycobacterium tuberculosis/physiology , Phosphoproteins/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocytes/cytology , Tuberculosis/prevention & control , Animals , Bronchoalveolar Lavage , Cell Differentiation/drug effects , Flow Cytometry , Fluorescent Antibody Technique , Interleukin-2/pharmacology , Lung/metabolism , Lung/microbiology , Macaca fascicularis/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/microbiology , Tuberculosis/immunology , Tuberculosis/microbiology
17.
PLoS One ; 8(6): e64212, 2013.
Article in English | MEDLINE | ID: mdl-23755118

ABSTRACT

Little is known about the role of gut microbiota in response to live oral vaccines against enteric pathogens. We examined the effect of immunization with an oral live-attenuated Shigella dysenteriae 1 vaccine and challenge with wild-type S. dysenteriae 1 on the fecal microbiota of cynomolgus macaques using 16 S rRNA analysis of fecal samples. Multi-dimensional cluster analysis identified different bacterial community types within macaques from geographically distinct locations. The fecal microbiota of Mauritian macaques, observed to be genetically distinct, harbored a high-diversity community and responded differently to Shigella immunization, as well as challenge compared to the microbiota in non-Mauritian macaques. While both macaque populations exhibited anti-Shigella antibody responses, clinical shigellosis was observed only among non-Mauritian macaques. These studies highlight the importance of further investigation into the possible protective role of the microbiota against enteric pathogens and consideration of host genetic backgrounds in conducting vaccine studies.


Subject(s)
Dysentery, Bacillary/prevention & control , Macaca fascicularis/microbiology , Microbiota/genetics , Shigella dysenteriae/immunology , Vaccination , Administration, Oral , Animals , Antibodies, Bacterial/blood , Dysentery, Bacillary/microbiology , Feces/microbiology , Female , Gastrointestinal Tract/microbiology , Genetic Variation , Host-Pathogen Interactions , Male , Molecular Typing , Phylogeny , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/genetics , Shigella Vaccines/administration & dosage , Shigella dysenteriae/physiology , Vaccines, Attenuated/administration & dosage
18.
J Infect Dis ; 207(8): 1242-52, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-22891286

ABSTRACT

BACKGROUND: Recent advances in rational adjuvant design and antigen selection have enabled a new generation of vaccines with potential to treat and prevent infectious disease. The aim of this study was to assess whether therapeutic immunization could impact the course of Mycobacterium tuberculosis infection with use of a candidate tuberculosis vaccine antigen, ID93, formulated in a synthetic nanoemulsion adjuvant, GLA-SE, administered in combination with existing first-line chemotherapeutics rifampicin and isoniazid. METHODS: We used a mouse model of fatal tuberculosis and the established cynomolgus monkey model to design an immuno-chemotherapeutic strategy to increase long-term survival and reduce bacterial burden, compared with standard antibiotic chemotherapy alone. RESULTS: This combined approach induced robust and durable pluripotent antigen-specific T helper-1-type immune responses, decreased bacterial burden, reduced the duration of conventional chemotherapy required for survival, and decreased M. tuberculosis-induced lung pathology, compared with chemotherapy alone. CONCLUSIONS: These results demonstrate the ability of therapeutic immunization to significantly enhance the efficacy of chemotherapy against tuberculosis and other infectious diseases, with implications for treatment duration, patient compliance, and more optimal resource allocation.


Subject(s)
Antigens, Bacterial/immunology , Antitubercular Agents/pharmacology , Mycobacterium tuberculosis/immunology , Rifampin/pharmacology , Tuberculosis Vaccines/therapeutic use , Tuberculosis, Pulmonary/therapy , Adjuvants, Immunologic/administration & dosage , Animals , Antigens, Bacterial/administration & dosage , Antitubercular Agents/immunology , Bacterial Proteins/immunology , Chemotherapy, Adjuvant/methods , Disease Models, Animal , Female , Isoniazid/administration & dosage , Isoniazid/pharmacology , Lung/immunology , Lung/microbiology , Lung/pathology , Macaca fascicularis/immunology , Macaca fascicularis/microbiology , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/pathogenicity , Rifampin/administration & dosage , Secondary Prevention , Survival Analysis , Time Factors , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/drug therapy , Tuberculosis, Pulmonary/immunology , Vaccination
19.
Am J Pathol ; 181(2): 508-14, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22721647

ABSTRACT

In an attempt to contain Mycobacterium tuberculosis, host immune cells form a granuloma as a physical and immunological barrier. To date, the contribution of humoral immunity, including antibodies and specific functions of B cells, to M. tuberculosis infection in humans remains largely unknown. Recent studies in mice show that humoral immunity can alter M. tuberculosis infection outcomes. M. tuberculosis infection in cynomolgus macaques recapitulates essentially all aspects of human tuberculosis. As a first step toward understanding the importance of humoral immunity to control of M. tuberculosis infection in primates, we characterized the B-cell and plasma-cell populations in infected animals and found that B cells are present primarily in clusters within the granuloma. The B-cell clusters are in close proximity to peripheral node addressin-positive cells and contain cells positive for Ki-67, a proliferation marker. Granuloma B cells also express CXCR5 and have elevated HLA-DR expression. Tissues containing M. tuberculosis bacilli had higher levels of M. tuberculosis-specific IgG, compared with uninvolved tissue from the same monkeys. Plasma cells detected within the granuloma produced mycobacteria-specific antibodies. Together, these data demonstrate that B cells are present and actively secreting antibodies specific for M. tuberculosis antigens at the site of infection, including lung granulomas and thoracic lymph nodes. These antibodies likely have the capacity to modulate local control of infection in tissues.


Subject(s)
B-Lymphocytes/immunology , Granuloma/immunology , Lymphocyte Activation/immunology , Macaca fascicularis/immunology , Macaca fascicularis/microbiology , Mycobacterium tuberculosis/immunology , Tuberculosis, Pulmonary/pathology , Animals , Antibody Formation/immunology , Antigens, Bacterial/immunology , Antigens, CD20/metabolism , B-Lymphocytes/pathology , Cell Aggregation/immunology , Germinal Center/immunology , Germinal Center/pathology , Humans , Immunoglobulin G/immunology , Lung/immunology , Lung/microbiology , Lung/pathology , Mice , Plasma Cells/immunology , Species Specificity , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology
20.
PLoS One ; 6(9): e24250, 2011.
Article in English | MEDLINE | ID: mdl-21935390

ABSTRACT

Costimulatory molecules play a central role in the development of cellular immunity. Understanding how costimulatory pathways can be directed to positively influence the immune response may be critical for the generation of an effective HIV vaccine. Here, we evaluated the ability of intravenous administration of a blocking monoclonal antibody (mAb) directed against the negative costimulatory molecule CTLA-4, and an agonist mAb directed against the positive costimulatory molecule 4-1BB, either alone or in combination, to augment intramuscular SIV DNA immunizations. We then tested the ability these of these responses to impact a high-dose SIVmac251 challenge. Following immunization, the groups infused with the anti-4-1BB mAb exhibited enhanced IFN-γ responses compared to the DNA vaccine only group. Interestingly, although CTLA-4 blockade alone did not enhance IFN-γ responses it did increase the proliferative capacity of the CD4(+) and CD8(+) T cells. The combination of both mAbs enhanced the magnitude of the polyfunctional CD8(+) T cell response. Following challenge, the group that received both mAbs exhibited a significant, ∼2.0 log, decrease in plasma viral load compared to the naïve group the included complete suppression of viral load in some animals. Furthermore, the use of the CTLA-4 blocking antibody resulted in significantly higher viral loads during chronic infection compared to animals that received the 4-1BB mAb, likely due to the higher CD4(+) T cell proliferative responses which were driven by this adjuvant following immunization. These novel studies show that these adjuvants induce differential modulation of immune responses, which have dramatically different consequences for control of SIV replication, suggesting important implications for HIV vaccine development.


Subject(s)
Macaca fascicularis/microbiology , Macaca fascicularis/virology , Simian Immunodeficiency Virus/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Vaccines, DNA/immunology , Adjuvants, Immunologic , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/metabolism , Female , Flow Cytometry , Male , Simian Immunodeficiency Virus/pathogenicity , Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...