Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
1.
Poult Sci ; 103(9): 103972, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38936074

ABSTRACT

White Leghorn chickens from a common founder population have been divergently selected for high (HAS) or low (LAS) antibody responses to sheep red blood cells (SRBC) for 49 generations resulting in 2 diverse lines for this trait. Much has been studied in these two lines; however, the impact of these selection pressures on cytokine and chemokine expression is not fully understood. The purpose of this study is to determine if selection for antibody response to SRBC impacts cytokine and chemokine expression in peripheral blood leukocytes (PBL) and spleen from HAS and LAS chickens. Total RNA was isolated from PBL and spleen after which mRNA expression of cytokines (IL4, IL6, IL10, TGF-ß4) and chemokines (CXCL8, CCL4) were determined by quantitative real-time RT-PCR (qRT-PCR). The data were analyzed using Student's t test comparing HAS and LAS (P < 0.05) and are reported as corrected 40-CT. PBL and spleen samples were analyzed separately. With respect to PBL, expression of IL6 was higher (P < 0.05) in PBL isolated from LAS chickens compared to those from the HAS line whereas there were no differences (P > 0.05) in IL4, IL10, CXCL8, CCL4, or TGF-ß4. The cytokine and chemokine mRNA expression profiles were different in the spleen between the two lines. IL4 and CXCL8 expression were higher (P < 0.05) in spleen samples from HAS chickens than LAS. The expression of IL6, IL10, CCL4, or TGF-ß4 in the spleens did not differ (P > 0.05) between the lines. The data indicate that selection for specific antibody responses to SRBC impacts the cytokine and chemokine expression profile in PBL and spleens but in different ways in HAS and LAS. These studies provide insight into the influence that selection pressures for antibody responses have on different immune response components, specifically cytokines and chemokines typically involved in the innate response.

2.
Poult Sci ; 103(2): 103278, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38052127

ABSTRACT

It has been hypothesized that reducing the bioenergetic costs of gut inflammation as an explanation for the effect of antibiotic growth promoters (AGPs) on animal efficiency, framing some observations but not explaining the increase in growth rate or the prevention of infectious diseases. The host's ability to adapt to alterations in environmental conditions and to maintain health involves managing all physiological interactions that regulate homeostasis. Thus, metabolic pathways are vital in regulating physiological health as the energetic demands of the host guides most biological functions. Mitochondria are not only the metabolic heart of the cell because of their role in energy metabolism and oxidative phosphorylation, but also a central hub of signal transduction pathways that receive messages about the health and nutritional states of cells and tissues. In response, mitochondria direct cellular and tissue physiological alterations throughout the host. The endosymbiotic theory suggests that mitochondria evolved from prokaryotes, emphasizing the idea that these organelles can be affected by some antibiotics. Indeed, therapeutic levels of several antibiotics can be toxic to mitochondria, but subtherapeutic levels may improve mitochondrial function and defense mechanisms by inducing an adaptive response of the cell, resulting in mitokine production which coordinates an array of adaptive responses of the host to the stressor(s). This adaptive stress response is also observed in several bacteria species, suggesting that this protective mechanism has been preserved during evolution. Concordantly, gut microbiome modulation by subinhibitory concentration of AGPs could be the result of direct stimulation rather than inhibition of determined microbial species. In eukaryotes, these adaptive responses of the mitochondria to internal and external environmental conditions, can promote growth rate of the organism as an evolutionary strategy to overcome potential negative conditions. We hypothesize that direct and indirect subtherapeutic AGP regulation of mitochondria functional output can regulate homeostatic control mechanisms in a manner similar to those involved with disease tolerance.


Subject(s)
Gastrointestinal Microbiome , Poultry , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Chickens , Mitochondria/metabolism
3.
Animals (Basel) ; 13(18)2023 Sep 06.
Article in English | MEDLINE | ID: mdl-37760224

ABSTRACT

The gastrointestinal ecosystem involves interactions between the host, gut microbiota, and external environment. To colonize the gut of poultry, Salmonella must surmount barriers levied by the intestine including mucosal innate immune responses and microbiota-mediated niche restrictions. Accordingly, comprehending Salmonella intestinal colonization in poultry requires an understanding of how the pathogen interacts with the intestinal ecosystem. In chickens, the paratyphoid Salmonella have evolved the capacity to survive the initial immune response and persist in the avian ceca for months without triggering clinical signs. The persistence of a Salmonella infection in the avian host involves both host defenses and tolerogenic defense strategies. The initial phase of the Salmonella-gut ecosystem interaction is characteristically an innate pro-inflammatory response that controls bacterial invasion. The second phase is initiated by an expansion of the T regulatory cell population in the cecum of Salmonella-infected chickens accompanied by well-defined shifts in the enteric neuro-immunometabolic pathways that changes the local phenotype from pro-inflammatory to an anti-inflammatory environment. Thus, paratyphoid Salmonella in chickens have evolved a unique survival strategy that minimizes the inflammatory response (disease resistance) during the initial infection and then induces an immunometabolic reprogramming in the cecum that alters the host defense to disease tolerance that provides an environment conducive to drive asymptomatic carriage of the bacterial pathogen.

4.
Poult Sci ; 102(10): 102982, 2023 10.
Article in English | MEDLINE | ID: mdl-37598552

Subject(s)
Chickens , Poultry , Animals
5.
Poult Sci ; 102(10): 102732, 2023 10.
Article in English | MEDLINE | ID: mdl-37556983

Subject(s)
Chickens , Poultry , Animals
6.
Microorganisms ; 11(7)2023 Jul 19.
Article in English | MEDLINE | ID: mdl-37513010

ABSTRACT

Salmonella enterica is a group of facultative, gram-negative bacteria. Recently, new evidence indicated that Salmonella could reprogram the host metabolism to increase energy or metabolites available for intracellular replication. In this study, using a chicken-specific kinomic immunometabolism peptide array analysis, we found that infection by S. Enteritidis induced significant phosphorylation changes in many key proteins of the glycolytic pathway in chicken macrophage HD-11 cells, indicating a shift in glycolysis caused by Salmonella infection. Nitric oxide production and changes of glycolysis and mitochondrial oxidative phosphorylation (OXPHOS) represented by extracellular acidification rate (ECAR) and oxygen consumption rate (OCR), respectively, were measured in chicken macrophages infected with three Salmonella strains (S. Enteritidis, S. Heidelberg, and S. Senftenberg). The infection reduced glycolysis and enhanced OXPHOS in chicken macrophages as indicated by changes of ECAR and OCR. Salmonella strains differentially affected macrophage polarization and glycolysis. Among three strains tested, S. Enteritidis was most effective in downregulating glycolysis and promoting M2 polarization as measured by ECAR, ORC, and NO production; while S. Senftenberg did not alter glycolysis and may promote M1 polarization. Our results suggested that downregulation of host cell glycolysis and increase of M2 polarization of macrophages may contribute to increased intracellular survival of S. Enteritidis.

7.
Poult Sci ; 102(9): 102849, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37454643

ABSTRACT

The intestinal wall has on its surface, protrusions called villi that are responsible for the absorption of nutrients. Commonly, these structures have their dimensions measured to related more area surface with better absorption. However, the measurement of these villi neglects the inflammation and the presence of immature cells that increase the surface area but affect negatively the absorption and compromise the animal performance. The measurements of villi/crypt are traditional tools in animal research; however, they may overlook alterations that impact the mucosal functionality. This study aimed to compare the morphometry of the intestinal villi/crypt with the I See Inside (ISI) scoring methodology, exploring their correlation with zootechnical performance. Therefore, broilers were grouped as nonchallenged (NC) and challenged with Eimeria (CH) and jejunum samples were collected at 22 d for histological analysis. The same villi were submitted to the ISI methodology, which is based on the scoring of 8 parameters related to the inflammatory process, and the measurements of villus height (VH), villus width (VW), crypt depth (CD), crypt width (CW), VH:CD ratio and villi absorptive surface (VAS). The CH group presented higher ISI total score, VW, CD, CW and lower VH, VH:CD, and VAS in comparison to the NC group. While the villi/crypt morphometry did not exhibit correlations with performance, the presence of Eimeria oocysts and the ISI total score was positively correlated (P < 0.05) with the feed conversion ratio (FCR), demonstrating a statistical interaction between high ISI scores and worse performance. In conclusion, a larger villus is not related to better intestinal functionality when this enlargement is unleashed by the immune processes occurring inside. The scoring system that evaluates the type of alteration observed has a direct impact on the animal's zootechnical performance which is not observed with the single metric surface evaluation.


Subject(s)
Coccidiosis , Eimeria , Poultry Diseases , Animals , Chickens , Coccidiosis/veterinary , Diet , Intestinal Mucosa/pathology , Animal Feed/analysis , Dietary Supplements/analysis
8.
Microorganisms ; 11(3)2023 Feb 28.
Article in English | MEDLINE | ID: mdl-36985187

ABSTRACT

Using a previously characterized and described abdominal model to define the avian immune response to Salmonella intra-abdominal challenge in chickens, we have adapted this technique for the study of chickens' immune response to a Campylobacter intra-abdominal challenge. The intra-abdominal Campylobacter infection model facilitates the characterization of peripheral blood leukocyte dynamics and abdominal cell infiltrates. Day-of-hatch Leghorn chickens were injected intra-abdominally (IA) with Campylobacter jejuni [(CJ)1 × 108 colony-forming units (CFUs)]. Changes in peripheral blood leukocyte numbers and abdominal cell infiltrates were monitored at 0, 4, 8, and 24 h post-injection. Peripheral blood leukocyte numbers were also determined for 2 h post-injection. For mortality studies, birds were injected intra-abdominally with 1 × 108 CFUs CJ and mortalities were recorded for 72 h post-injection. In the peripheral blood of CJ-injected chicks, total white blood cell (WBC) numbers began increasing by 2 h post-injection, peaking at 4 h post-injection with the predominant cell type being polymorphonuclear leukocytes (heterophils). Total WBCs declined after 8 h and this decline continued at 24 h, with total WBC numbers approaching control values. The injection of CJ into the abdominal cavity caused a rapid rise in abdominal cell infiltrates with the predominant infiltrating leukocytes being heterophils. Peak abdominal heterophil infiltrates were observed at 8 h post-injection, declining only slightly by 24 h post-injection. Mortality in the CJ challenge groups reached 37%. Mortality in the Salmonella enteritidis positive control groups were greater than 50%. The data suggest that Campylobacter infection does stimulate the innate immune response in chickens when administered IA, however, the immune response and infection is not characterized with the high levels of mortality observed with a Salmonella infection. These data provide a basis for a more definitive characterization of chickens' immune response to Campylobacter and a model to evaluate intervention strategies to prevent the infection and colonization of poultry.

9.
Poult Sci ; 102(4): 102531, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36805406

ABSTRACT

Addition of vitamins and antioxidants has been long associated with increased immunity and are commonly used in the poultry industry; however, less is known regarding their use in broiler breeder hens. The objective of this study was to determine if feeding a complex of protected biofactors and antioxidants composed of vitamins and fermentation extracts to broiler breeder hens conferred resistance against Salmonella enterica serovar Enteritidis (S. Enteritidis) in the progeny chicks. Three-day-old chicks from control- and supplement-fed hens were challenged with S. Enteritidis and necropsied 4- and 11-days postchallenge (dpc) to determine if there were differences in invasion and colonization. Serum and jejunum were evaluated for various cytokine and chemokine production. Fewer (P = 0.002) chicks from supplement-fed hens had detectable S. Enteritidis in the ceca (32.6%) compared to chicks from control-fed hens (64%). By 11 dpc, significantly (P < 0.001) fewer chicks from supplement-fed hens were positive for S. Enteritidis (liver [36%]; ceca [16%]) compared to chicks from the control hens (liver [76%]; ceca [76%]). The recoverable S. Enteritidis in the cecal content was also lower (P = 0.01) at 11 dpc. In additional to the differences in invasion and colonization, cytokine and chemokine production were distinct between the 2 groups of chicks. Chicks from supplement-fed hens had increased production of IL-16, IL-6, MIP-3α, and RANTES in the jejunum while IL-16 and MIP-1ß were higher in the serum of chicks from the control-fed hens. By 11 dpc, production of IFN-γ was decreased in the jejunum of chicks from supplement-fed hens. Collectively, these data demonstrate adding a protected complex of biofactors and antioxidants to the diet of broiler breeder hens offers a measure of transgenerational protection to the progeny against S. Enteritidis infection and reduces colonization that is mediated, in part, by a robust and distinct cytokine and chemokine response locally at the intestine and systemically in the blood.


Subject(s)
Poultry Diseases , Salmonella Infections, Animal , Animals , Female , Salmonella enteritidis , Chickens , Antioxidants , Interleukin-16 , Diet/veterinary , Vitamins , Salmonella Infections, Animal/prevention & control , Poultry Diseases/prevention & control
11.
Animals (Basel) ; 12(21)2022 Nov 04.
Article in English | MEDLINE | ID: mdl-36359160

ABSTRACT

The complex interaction between the intestinal mucosa, the gut microbiota, and the diet balances the host physiological homeostasis and is fundamental for the maximal genetic potential of production animals. However, factors such as chemical and physical characteristics of the diet and/or environmental stressors can continuously affect this balance, potentially inducing a state of chronic low-grade inflammation in the gut, where inflammatory parameters are present and demanding energy, but not in enough intensity to provoke clinical manifestations. It's vital to expand the understanding of inflammation dynamics and of how they compromise the function activity and microscopic morphology of the intestinal mucosa. These morphometric alterations are associated with the release of structural and functional cellular components into the feces and the blood stream creating measurable biomarkers to track this condition. Moreover, the identification of novel, immunometabolic biomarkers can provide dynamic and predictors of low-grade chronic inflammation, but also provide indicators of successful nutritional or feed additive intervention strategies. The objective of this paper is to review the mechanisms of low-grade inflammation, its effects on animal production and sustainability, and the biomarkers that could provide early diagnosis of this process and support studies of useful interventional strategies.

12.
Front Cell Infect Microbiol ; 12: 899395, 2022.
Article in English | MEDLINE | ID: mdl-35846741

ABSTRACT

Poultry is a major source of human foodborne illness caused by broad host range Salmonella serovars (paratyphoid), and developing cost-effective, pre-harvest interventions to reduce these pathogens would be valuable to the industry and consumer. Host responses to infectious agents are often regulated through phosphorylation. However, proteomic mechanisms of Salmonella acute infection biology and host responses to the bacteria have been limited concentrating predominately on the genomic responses of the host to infection. Our recent development of chicken-specific peptide arrays for kinome analysis of host phosphorylation-based cellular signaling responses provided us with the opportunity to develop a more detailed understanding of the early (4-24 h post-infection) host-pathogen interactions during the initial colonization of the cecum by Salmonella. Using the chicken-specific kinomic immune peptide array, biological pathway analysis showed infection with S. Enteritidis increased signaling related to the innate immune response, relative to the non-infected control ceca. Notably, the acute innate immune signaling pathways were characterized by increased peptide phosphorylation (activation) of the Toll-like receptor and NOD-like receptor signaling pathways, the activation of the chemokine signaling pathway, and the activation of the apoptosis signaling pathways. In addition, Salmonella infection induced a dramatic alteration in the phosphorylation events of the JAK-STAT signaling pathway. Lastly, there is also significant activation of the T cell receptor signaling pathway demonstrating the initiation of the acquired immune response to Salmonella infection. Based on the individual phosphorylation events altered by the early Salmonella infection of the cecum, certain conclusions can be drawn: (1) Salmonella was recognized by both TLR and NOD receptors that initiated the innate immune response; (2) activation of the PPRs induced the production of chemokines CXCLi2 (IL-8) and cytokines IL-2, IL-6, IFN-α, and IFN-γ; (3) Salmonella infection targeted the JAK-STAT pathway as a means of evading the host response by targeting the dephosphorylation of JAK1 and TYK2 and STAT1,2,3,4, and 6; (4) apoptosis appears to be a host defense mechanism where the infection with Salmonella induced both the intrinsic and extrinsic apoptotic pathways; and (5) the T cell receptor signaling pathway activates the AP-1 and NF-κB transcription factor cascades, but not NFAT.


Subject(s)
Poultry Diseases , Salmonella Infections, Animal , Animals , Cecum/microbiology , Chickens , Humans , Janus Kinases , Poultry Diseases/microbiology , Proteomics , Receptors, Antigen, T-Cell , STAT Transcription Factors , Salmonella Infections, Animal/microbiology , Salmonella enteritidis , Signal Transduction
13.
Poult Sci ; 101(4): 101753, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35240358

ABSTRACT

Necrotic enteritis (NE) is a devastating disease that has seen a resurgence of cases following the removal of antibiotics from feed resulting in financial loss and significant animal health concerns across the poultry industry. The objective was to evaluate the efficacy of a microencapsulated blend of organic (25% citric and 16.7% sorbic) acids and botanicals (1.7% thymol and 1% vanillin [AviPlusP]) to reduce clinical NE and determine the signaling pathways associated with any changes. Day-of-hatch by-product broiler breeder chicks were randomly assigned to a control (0) or supplemented (500 g/MT) diet (n = 23-26) and evaluated in a NE challenge model (n = 3). Birds were administered 2X cocci vaccine on d 14 and challenged with a cocktail of Clostridium perfringens strains (107) on d 17 to 19. On d 20 to 21 birds were weighed, euthanized, and scored for NE lesions. Jejunal tissue was collected for kinome analysis using an immuno-metabolism peptide array (n = 5; 15/treatment) to compare tissue from supplement-fed birds to controls. Mortality and weight were analyzed using Student's t test and lesion scores analyzed using F-test two-sample for variances (P < 0.05). The kinome data was analyzed using PIIKA2 peptide array analysis software and fold-change between control and treated groups determined. Mortality in the supplemented group was 47.4% and 70.7% in controls (P = 0.004). Lesions scores were lower (P = 0.006) in supplemented birds (2.47) compared to controls (3.3). Supplement-fed birds tended (P = 0.19) to be heavier (848.6 g) than controls (796.2 g). Kinome analysis showed T cell receptor, TNF and NF-kB signaling pathways contributed to the improvements seen in the supplement-fed birds. The following peptides were significant (P < 0.05) in all 3 pathways: CHUK, MAP3K14, MAP3K7, and NFKB1 indicating their importance. Additionally, there were changes to IL6, IL10, and IFN- γ mRNA expression in tissue between control- and supplement-fed chickens. In conclusion, the addition of a microencapsulated blend of organic acids and botanicals to a broiler diet reduced the clinical signs of NE that was mediated by specific immune-related pathways.


Subject(s)
Clostridium Infections , Enteritis , Poultry Diseases , Animals , Acids , Animal Feed/analysis , Chickens , Clostridium Infections/prevention & control , Clostridium Infections/veterinary , Clostridium perfringens , Diet/veterinary , Enteritis/drug therapy , Enteritis/prevention & control , Enteritis/veterinary , Necrosis/prevention & control , Necrosis/veterinary , Organic Chemicals , Poultry Diseases/prevention & control , Signal Transduction
14.
Poult Sci ; 101(3): 101673, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35104729

ABSTRACT

In the intestine, host-derived factors are genetically hardwired and difficult to modulate. However, the intestinal microbiome is more plastic and can be readily modulated by dietary factors. Further, it is becoming more apparent that the microbiome can potentially impact poultry physiology by participating in digestion, the absorption of nutrients, shaping of the mucosal immune response, energy homeostasis, and the synthesis or modulation of several potential bioactive metabolites. These activities are dependent on the quantity and quality of the microbiota alongside its metabolic potential, which are dictated in large part by diet. Thus, diet-induced microbiota alterations may be harnessed to induce changes in host physiology, including disease development and progression. In this regard, the gut microbiome is malleable and renders the gut microbiome a candidate 'organ' for the possibility of precision nutrition to induce precision microbiomics-the use of the gut microbiome as a biomarker to predict responsiveness to specific dietary constituents to generate precision diets and interventions for optimal poultry performance and health. However, it is vital to identify the causal relationships and mechanisms by which dietary components and additives affect the gut microbiome which then ultimately influence avian physiology. Further, an improved understanding of the spatial and functional relationships between the different sections of the avian gut and their regional microbiota will provide a better understanding of the role of the diet in regulating the intestinal microbiome.


Subject(s)
Animal Nutritional Physiological Phenomena , Chickens , Diet , Gastrointestinal Microbiome , Animals , Chickens/microbiology , Diet/veterinary , Nutritional Status
15.
Poult Sci ; 101(3): 101642, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35016046

ABSTRACT

Intestinal organoids (IO), known as "mini-guts", derived from intestinal crypts, are self-organizing three-dimensional (3D) multicellular ex vivo models that recapitulate intestine epithelial structure and function and have been widely used for studying intestinal physiology, pathophysiology, molecular mechanisms of host-pathogen interactions, and intestinal disease in mammals. However, studies on avian IO are limited and the development of long-term cultures of IO model for poultry research is lacking. Therefore, the objectives of this study were to generate crypt-derived organoids from chicken intestines and to optimize conditions for cell growth and enrichments, passages, and cryopreservation. Crypts were collected from the small intestines of birds at embryonic d-19 and ceca from layer and broiler chickens with ages ranging from d 1 to 20 wk, embedded in a basement membrane matrix, and cultured with organoid growth media (OGM) prepared in house. The crypt-derived organoids were successfully grown and propagated to form 3D spheres like structures that were cultured for up to 3 wk. Organoids were formed on d one, budding appeared on d 3, and robust budding was observed on d 7 and beyond. For cryopreservation, dissociated organoids were resuspended in a freezing medium. The characteristics of IO upon extended passages and freeze-thaw cycles were analyzed using reverse transcription (RT)-PCR, immunoblotting, and live cell imaging. Immunoblotting and RT-PCR using E-cadherin (the marker for epithelial cells), leucine-rich repeat-containing G protein-coupled receptor 5 (LGR5, the marker for stem cells), chromogranin A (the marker for enteroendocrine cells), lysozyme (the marker for Paneth cells), and mucin (the biomarker for goblet cells) confirmed that IO were composed of heterogeneous cell populations, including epithelial cells, stem cells, enteroendocrine cells, Paneth cells, and goblet cells. Furthermore, OGM supplemented with both valproic acid and CHIR99021, a glycogen synthase kinase 3ß inhibitor and a histone deacetylase inhibitor, increased the size of the avian IO (P < 0.001). To the best of our knowledge, this is the first comprehensive report for establishing long-term, organoid culture models from small intestines and ceca of layer and broiler chickens. This model will facilitate elucidation of the mechanisms impacting host-pathogen interactions, eventually leading to the discovery of pathogen intervention strategies in poultry.


Subject(s)
Intestinal Mucosa , Organoids , Animals , Cell Differentiation/physiology , Chickens , Intestinal Mucosa/metabolism , Intestines , Organoids/physiology , Paneth Cells
16.
Adv Exp Med Biol ; 1354: 145-159, 2022.
Article in English | MEDLINE | ID: mdl-34807441

ABSTRACT

The chicken gastrointestinal tract (GIT) has a complex, biodiverse microbial community of ~ 9 million bacterial genes plus archaea and fungi that links the host diet to its health. This microbial population contributes to host physiology through metabolite signaling while also providing local and systemic nutrients to multiple organ systems. In a homeostatic state, the host-microbial interaction is symbiotic; however, physiological issues are associated with dysregulated microbiota. Manipulating the microbiota is a therapeutic option, and the concept of adding beneficial bacteria to the intestine has led to probiotic and prebiotic development. The gut microbiome is readily changeable by diet, antibiotics, pathogenic infections, and host- and environmental-dependent events. The intestine performs key roles of nutrient absorption, tolerance of beneficial microbiota, yet responding to undesirable microbes or microbial products and preventing translocation to sterile body compartments. During homeostasis, the immune system is actively preventing or modulating the response to known or innocuous antigens. Manipulating the microbiota through nutrition, modulating host immunity, preventing pathogen colonization, or improving intestinal barrier function has led to novel methods to prevent disease, but also resulted in improved body weight, feed conversion, and carcass yield in poultry. This review highlights the importance of adding different feed additives to the diets of poultry in order to manipulate and enhance health and productivity of flocks.


Subject(s)
Gastrointestinal Microbiome , Probiotics , Animals , Anti-Bacterial Agents/pharmacology , Gastrointestinal Tract , Poultry , Prebiotics/analysis
17.
Commun Biol ; 4(1): 1359, 2021 12 03.
Article in English | MEDLINE | ID: mdl-34862463

ABSTRACT

Salmonella enterica persist in the chicken gut by suppressing inflammatory responses via expansion of intestinal regulatory T cells (Tregs). In humans, T cell activation is controlled by neurochemical signaling in Tregs; however, whether similar neuroimmunological signaling occurs in chickens is currently unknown. In this study, we explore the role of the neuroimmunological axis in intestinal Salmonella resistance using the drug reserpine, which disrupts intracellular storage of catecholamines like norepinephrine. Following reserpine treatment, norepinephrine release was increased in both ceca explant media and Tregs. Similarly, Salmonella killing was greater in reserpine-treated explants, and oral reserpine treatment reduced the level of intestinal Salmonella Typhimurium and other Enterobacteriaceae in vivo. These antimicrobial responses were linked to an increase in antimicrobial peptide and IL-2 gene expression as well as a decrease in CTLA-4 gene expression. Globally, reserpine treatment led to phosphorylative changes in epidermal growth factor receptor (EGFR), mammalian target of rapamycin (mTOR), and the mitogen-associated protein kinase 2(MEK2). Exogenous norepinephrine treatment alone increased Salmonella resistance, and reserpine-induced antimicrobial responses were blocked using beta-adrenergic receptor inhibitors, suggesting norepinephrine signaling is crucial in this mechanism. Furthermore, EGF treatment reversed reserpine-induced antimicrobial responses, whereas mTOR inhibition increased antimicrobial activities, confirming the roles of metabolic signaling in these responses. Finally, MEK1/2 inhibition suppressed reserpine, norepinephrine, and mTOR-induced antimicrobial responses. Overall, this study demonstrates a central role for MEK1/2 activity in reserpine induced neuro-immunometabolic signaling and subsequent antimicrobial responses in the chicken intestine, providing a means of reducing bacterial colonization in chickens to improve food safety.


Subject(s)
Chickens , Disease Resistance/drug effects , Enterobacteriaceae Infections/veterinary , Enterobacteriaceae/physiology , Poultry Diseases/microbiology , Reserpine/pharmacology , Signal Transduction , Animals , Enterobacteriaceae Infections/microbiology , Intestines/immunology , Intestines/microbiology , Salmonella Infections, Animal/microbiology , Salmonella typhimurium/physiology
18.
PLoS One ; 16(11): e0260280, 2021.
Article in English | MEDLINE | ID: mdl-34843525

ABSTRACT

Poultry infected with Salmonella mount an immune response initially, however the immune responses eventually disappear leading the bird to be a carrier of Salmonella. The hypothesis of this study is that Salmonella infection induces T regulatory cell numbers and cytokine production and suppress host T cells locally in the gut to escape the host immune responses. An experiment was conducted to comparatively analyze the effect of S. enterica ser. Enteritidis (S. Enteritidis) and S. enterica ser. Heidelberg (S. Heidelberg) infection on CD4+CD25+ T regulatory cell properties in chickens. A total of 144 broiler chicks were randomly distributed into three experimental groups of non-infected control, S. Enteritidis infected and S. Heidelberg infected groups. Chickens were orally inoculated with PBS (control) or 5x106 CFU/mL of either S. Enteritidis or S. Heidelberg at 3 d of age. Each group was replicated in six pens with eight chickens per pen. Chickens infected with S. Enteritidis had 6.2, 5.4, and 3.8 log10 CFU/g, and chickens infected with S. Heidelberg had 7.1, 4.8, and 4.1 log10 CFU/g Salmonella in the cecal contents at 4, 11, and 32 dpi, respectively. Both S. Enteritidis and S. Heidelberg were recovered from the liver and spleen 4 dpi. At 4, 11, and 32 dpi, chickens infected with S. Enteritidis and S. Heidelberg had increased CD4+CD25+ cell numbers as well as IL-10 mRNA transcription of CD4+CD25+ cells compared to that in the control group. CD4+CD25+ cells from S. Enteritidis- and S. Heidelberg-infected chickens and restimulated with 1 µg antigen in vitro, had higher (P < 0.05) IL-10 mRNA transcription than the CD4+CD25+ cells from the non-infected controls Though at 4dpi, chickens infected with S. Enteritidis and S. Heidelberg had a significant (P < 0.05) increase in CD4+CD25- IL-2, IL-1ß, and IFNγ mRNA transcription, the CD4+CD25- IL-2, IL-1ß, and IFNγ mRNA transcription, were comparable to that in the control group at 11 and 32dpi identifying that the host inflammatory response against Salmonella disappears at 11 dpi. It can be concluded that S. Enteritidis and S. Heidelberg infection at 3 d of age induces a persistent infection through inducing CD4+CD25+ cells and altering the IL-10 mRNA transcription of CD4+CD25+ cell numbers and cytokine production in chickens between 3 to 32 dpi allowing chickens to become asymptomatic carriers of Salmonella after 18 dpi.


Subject(s)
Avian Proteins/immunology , CD4 Antigens/immunology , Chickens/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Poultry Diseases/immunology , Salmonella Infections, Animal/immunology , Salmonella enteritidis/immunology , Animals , Chickens/microbiology , Host-Pathogen Interactions , Immunity , Poultry Diseases/microbiology , Salmonella Infections, Animal/microbiology , Salmonella enteritidis/physiology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/microbiology
19.
Front Physiol ; 12: 707757, 2021.
Article in English | MEDLINE | ID: mdl-34366895

ABSTRACT

This study was conducted to distinguish the effects of heat stress (HS) and feed intake (FI) on broiler chicken's physiological responses. Day-old male Cobb 500 broilers (n = 672) were allocated to three treatments: (1) control (CTL): birds raised under normal temperature (23°C) from day 29 to 42; (2) cyclic heat stress (CHS): birds exposed to high temperatures (8 h/day at 35°C; from 9:30 am to 5:30 pm) from day 29 to 42; (3) pair-fed (PF): birds raised under thermoneutral condition but fed the same amount of feed as CHS from day 29 to 42. On day 42, 15 birds/pen were processed, to measure carcass and meat yields. To measure blood parameters and gut integrity (using fluorescein isothiocyanate-dextran), on day 42, CHS birds were sampled before (Pre-CHS) and 2 h after (Post-CHS) the temperature increased. Furthermore, after sampling CTL birds, they were exposed to 2h heat and sampled (acute heat stress, AHS). Data were analyzed using one-way ANOVA (JMP Pro15) and significance between treatments identified by LSD (P < 0.05). BW and relative carcass yield were significantly higher in CTL compared to CHS and PF. Compared to CHS, PF had significantly higher BW and lower relative carcass yield. Breast yield was significantly higher for CTL and PF, while leg quarters and wings yield were significantly lower compared to CHS. Gut barrier integrity was significantly altered in Post-CHS and AHS compared to CTL. mRNA abundances of tumor necrosis factor-α, C-C motif chemokine ligand-20, heat shock protein (HSP)-27, and HSP70 were significantly higher in Post-CHS and AHS compared to CTL. AHS had significantly higher mRNA abundances of CARD domain containing (NLRC)-3 and NLRC5 inflammasomes, and lower superoxide dismutase (SOD)-1 and SOD2 abundance compared with CTL. PF had significantly higher liver weight (% BW) compared to all other groups; while abdominal fat was significantly higher in Pre-CHS compared to CTL, PF, and AHS. Together, these data indicate that the negative effects of HS are partially due to reduced FI. However, the negative effect of HS on gut integrity, average daily gain, feed conversion ratio, and meat yield are direct and independent of the reduced FI during the HS. Thus, warrant investigating the underlying mechanisms in future research.

20.
Front Immunol ; 12: 676628, 2021.
Article in English | MEDLINE | ID: mdl-34054868

ABSTRACT

For poultry producers, chronic low-grade intestinal inflammation has a negative impact on productivity by impairing nutrient absorption and allocation of nutrients for growth. Understanding the triggers of chronic intestinal inflammation and developing a non-invasive measurement is crucial to managing gut health in poultry. In this study, we developed two novel models of low-grade chronic intestinal inflammation in broiler chickens: a chemical model using dextran sodium sulfate (DSS) and a dietary model using a high non-starch polysaccharide diet (NSP). Further, we evaluated the potential of several proteins as biomarkers of gut inflammation. For these experiments, the chemical induction of inflammation consisted of two 5-day cycles of oral gavage of either 0.25mg DSS/ml or 0.35mg DSS/ml; whereas the NSP diet (30% rice bran) was fed throughout the experiment. At four times (14, 22, 28 and 36-d post-hatch), necropsies were performed to collect intestinal samples for histology, and feces and serum for biomarkers quantification. Neither DSS nor NSP treatments affected feed intake or livability. NSP-fed birds exhibited intestinal inflammation through 14-d, which stabilized by 36-d. On the other hand, the cyclic DSS-treatment produced inflammation throughout the entire experimental period. Histological examination of the intestine revealed that the inflammation induced by both models exhibited similar spatial and temporal patterns with the duodenum and jejunum affected early (at 14-d) whereas the ileum was compromised by 28-d. Calprotectin (CALP) was the only serum protein found to be increased due to inflammation. However, fecal CALP and Lipocalin-2 (LCN-2) concentrations were significantly greater in the induced inflammation groups at 28-d. This experiment demonstrated for the first time, two in vivo models of chronic gut inflammation in chickens, a DSS and a nutritional NSP protocols. Based on these models we observed that intestinal inflammation begins in the upper segments of small intestine and moved to the lower region over time. In the searching for a fecal biomarker for intestinal inflammation, LCN-2 showed promising results. More importantly, calprotectin has a great potential as a novel biomarker for poultry measured both in serum and feces.


Subject(s)
Dextran Sulfate/adverse effects , Diet, Carbohydrate Loading/adverse effects , Diet, Carbohydrate Loading/veterinary , Gastroenteritis/blood , Gastroenteritis/chemically induced , Poultry Diseases/blood , Poultry Diseases/chemically induced , Animal Feed , Animals , Biomarkers/metabolism , Chickens , Chronic Disease , Dextran Sulfate/administration & dosage , Dietary Fiber/adverse effects , Disease Models, Animal , Feces/chemistry , Gastroenteritis/immunology , Intestinal Mucosa/immunology , Leukocyte L1 Antigen Complex/metabolism , Lipocalin-2/metabolism , Male , Oryza/adverse effects , Poultry Diseases/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...