Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 296
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 181(6): 1263-1275.e16, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32437658

RESUMEN

Very low-carbohydrate, high-fat ketogenic diets (KDs) induce a pronounced shift in metabolic fuel utilization that elevates circulating ketone bodies; however, the consequences of these compounds for host-microbiome interactions remain unknown. Here, we show that KDs alter the human and mouse gut microbiota in a manner distinct from high-fat diets (HFDs). Metagenomic and metabolomic analyses of stool samples from an 8-week inpatient study revealed marked shifts in gut microbial community structure and function during the KD. Gradient diet experiments in mice confirmed the unique impact of KDs relative to HFDs with a reproducible depletion of bifidobacteria. In vitro and in vivo experiments showed that ketone bodies selectively inhibited bifidobacterial growth. Finally, mono-colonizations and human microbiome transplantations into germ-free mice revealed that the KD-associated gut microbiota reduces the levels of intestinal pro-inflammatory Th17 cells. Together, these results highlight the importance of trans-kingdom chemical dialogs for mediating the host response to dietary interventions.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Microbioma Gastrointestinal/fisiología , Intestinos/inmunología , Intestinos/microbiología , Células Th17/inmunología , Células Th17/fisiología , Adolescente , Adulto , Animales , Dieta Alta en Grasa/métodos , Dieta Cetogénica/métodos , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Microbiota/inmunología , Microbiota/fisiología , Persona de Mediana Edad , Células Th17/microbiología , Adulto Joven
2.
Immunity ; 57(10): 2280-2295.e6, 2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-39299238

RESUMEN

Toll/interleukin-1/resistance (TIR)-domain proteins with enzymatic activity are essential for immunity in plants, animals, and bacteria. However, it is not known how these proteins function in pathogen sensing in animals. We discovered that the lone enzymatic TIR-domain protein in the nematode C. elegans (TIR-1, homolog of mammalian sterile alpha and TIR motif-containing 1 [SARM1]) was strategically expressed on the membranes of a specific intracellular compartment called lysosome-related organelles. The positioning of TIR-1 on lysosome-related organelles enables intestinal epithelial cells in the nematode C. elegans to survey for pathogen effector-triggered host damage. A virulence effector secreted by the bacterial pathogen Pseudomonas aeruginosa alkalinized and condensed lysosome-related organelles. This pathogen-induced morphological change in lysosome-related organelles triggered TIR-1 multimerization, which engaged its intrinsic NAD+ hydrolase (NADase) activity to activate the p38 innate immune pathway and protect the host against microbial intoxication. Thus, TIR-1 is a guard protein in an effector-triggered immune response, which enables intestinal epithelial cells to survey for pathogen-induced host damage.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Inmunidad Innata , Lisosomas , Pseudomonas aeruginosa , Animales , Caenorhabditis elegans/inmunología , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/inmunología , Proteínas de Caenorhabditis elegans/genética , Pseudomonas aeruginosa/inmunología , Lisosomas/metabolismo , Lisosomas/inmunología , Inmunidad Innata/inmunología , Intestinos/inmunología , Infecciones por Pseudomonas/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Interacciones Huésped-Patógeno/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Receptores Acoplados a Proteínas G
3.
Immunol Rev ; 323(1): 126-137, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38491842

RESUMEN

Group 3 innate lymphoid cells (ILC3s) are tissue-resident immune lymphocytes that critically regulate intestinal homeostasis, organogenesis, and immunity. ILC3s possess the capacity to "sense" the inflammatory environment within tissues, especially in the context of pathogen challenges that imprints durable non-antigen-specific changes in ILC3 function. As such, ILC3s become a new actor in the emerging field of trained innate immunity. Here, we summarize recent discoveries regarding ILC3 responses to bacterial challenges and the role these encounters play in triggering trained innate immunity. We further discuss how signaling events throughout ILC3 ontogeny potentially control the development and function of trained ILC3s. Finally, we highlight the open questions surrounding ILC3 "training" the answers to which may reveal new insights into innate immunity. Understanding the fundamental concepts behind trained innate immunity could potentially lead to the development of new strategies for improving immunity-based modulation therapies for inflammation, infectious diseases, and cancer.


Asunto(s)
Inmunidad Innata , Linfocitos , Transducción de Señal , Humanos , Animales , Linfocitos/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Homeostasis , Inflamación/inmunología , Microbioma Gastrointestinal/inmunología , Intestinos/inmunología
4.
Immunity ; 49(1): 151-163.e5, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-29980437

RESUMEN

The intestinal barrier is vulnerable to damage by microbiota-induced inflammation that is normally restrained through mechanisms promoting homeostasis. Such disruptions contribute to autoimmune and inflammatory diseases including inflammatory bowel disease. We identified a regulatory loop whereby, in the presence of the normal microbiota, intestinal antigen-presenting cells (APCs) expressing the chemokine receptor CX3CR1 reduced expansion of intestinal microbe-specific T helper 1 (Th1) cells and promoted generation of regulatory T cells responsive to food antigens and the microbiota itself. We identified that disruption of the microbiota resulted in CX3CR1+ APC-dependent inflammatory Th1 cell responses with increased pathology after pathogen infection. Colonization with microbes that can adhere to the epithelium was able to compensate for intestinal microbiota loss, indicating that although microbial interactions with the epithelium can be pathogenic, they can also activate homeostatic regulatory mechanisms. Our results identify a cellular mechanism by which the microbiota limits intestinal inflammation and promotes tissue homeostasis.


Asunto(s)
Receptor 1 de Quimiocinas CX3C/metabolismo , Microbioma Gastrointestinal/inmunología , Mucosa Intestinal/inmunología , Sistema Mononuclear Fagocítico/inmunología , Linfocitos T Reguladores/inmunología , Células TH1/inmunología , Animales , Presentación de Antígeno , Adhesión Bacteriana/inmunología , Modelos Animales de Enfermedad , Femenino , Homeostasis , Tolerancia Inmunológica , Inmunidad Mucosa , Inflamación/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Interleucina-10/inmunología , Interleucina-10/metabolismo , Mucosa Intestinal/microbiología , Masculino , Ratones , Células RAW 264.7
5.
Semin Immunol ; 70: 101819, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37632991

RESUMEN

The enteric nervous system is an autonomous neuronal circuit that regulates many processes far beyond the peristalsis in the gastro-intestinal tract. This circuit, consisting of enteric neurons and enteric glial cells, can engage in many intercellular interactions shaping the homeostatic microenvironment in the gut. Perhaps the most well documented interactions taking place, are the intestinal neuro-immune interactions which are essential for the fine-tuning of oral tolerance. In the context of intestinal disease, compelling evidence demonstrates both protective and detrimental roles for this bidirectional neuro-immune signaling. This review discusses the different immune cell types that are recognized to engage in neuronal crosstalk during intestinal health and disease. Highlighting the molecular pathways involved in the neuro-immune interactions might inspire novel strategies to target intestinal disease.


Asunto(s)
Sistema Nervioso Entérico , Enfermedades Intestinales , Humanos , Neuroinmunomodulación , Homeostasis
6.
Eur J Immunol ; 54(6): e2350631, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38556632

RESUMEN

The intestinal barrier is mainly formed by a monolayer of epithelial cells, which forms a physical barrier to protect the gut tissues from external insults and provides a microenvironment for commensal bacteria to colonize while ensuring immune tolerance. Moreover, various immune cells are known to significantly contribute to intestinal barrier function by either directly interacting with epithelial cells or by producing immune mediators. Fulfilling this function of the gut barrier for mucosal homeostasis requires not only the intrinsic regulation of intestinal epithelial cells (IECs) but also constant communication with immune cells and gut microbes. The reciprocal interactions between IECs and immune cells modulate mucosal barrier integrity. Dysregulation of barrier function could lead to dysbiosis, inflammation, and tumorigenesis. In this overview, we provide an update on the characteristics and functions of IECs, and how they integrate their functions with tissue immune cells and gut microbiota to establish gut homeostasis.


Asunto(s)
Células Epiteliales , Microbioma Gastrointestinal , Homeostasis , Mucosa Intestinal , Humanos , Homeostasis/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Animales , Microbioma Gastrointestinal/inmunología , Células Epiteliales/inmunología , Comunicación Celular/inmunología , Tolerancia Inmunológica/inmunología
7.
Eur J Immunol ; 54(10): e2451080, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39072720

RESUMEN

Although the functions of tyrosine phosphatases in T-cell biology have been extensively studied, our knowledge on the contribution of serine/threonine phosphatases in T cells remains poor. Protein phosphatase 2A (PP2A) is one of the most abundantly expressed serine/threonine phosphatases. It is important in thymocyte development and CD4+ T-cell differentiation. Utilizing a genetic model in which its catalytic subunit alpha isoform (PP2A Cα) is deleted in T cells, we investigated its contribution to CD8+ T-cell homeostasis and effector functions. Our results demonstrate that T-cell intrinsic PP2A Cα is critically required for CD8+ T-cell homeostasis in secondary lymphoid organs and intestinal mucosal site. Importantly, PP2A Cα-deficient CD8+ T cells exhibit reduced proliferation and survival. CD8+ T-cell antibacterial response is strictly dependent on PP2A Cα. Expression of Bcl2 transgene rescues CD8+ T-cell homeostasis in spleens, but not in intestinal mucosal site, nor does it restore defective antibacterial responses. Finally, proteomics and phosphoproteomics analyses reveal potential targets dependent on PP2A Cα, including mTORC1 and AKT. Thus, PP2A Cα is a key modulator of CD8+ T-cell homeostasis and effector functions.


Asunto(s)
Linfocitos T CD8-positivos , Homeostasis , Proteína Fosfatasa 2 , Linfocitos T CD8-positivos/inmunología , Animales , Homeostasis/inmunología , Ratones , Proteína Fosfatasa 2/metabolismo , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/inmunología , Ratones Endogámicos C57BL , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Ratones Noqueados , Proliferación Celular , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/inmunología , Activación de Linfocitos/inmunología , Ratones Transgénicos
8.
J Biol Chem ; 299(12): 105414, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37918806

RESUMEN

The proteins that coordinate the complex transcriptional networks of aging have not been completely documented. Protein 14-3-3zeta is an adaptor protein that coordinates signaling and transcription factor networks, but its function in aging is not fully understood. Here, we showed that the protein expression of 14-3-3zeta gradually increased during aging. High levels of 14-3-3zeta led to shortened lifespan and imbalance of intestinal immune homeostasis in Drosophila, but the decrease in 14-3-3zeta protein levels by RNAi was able to significantly promote the longevity and intestinal immune homeostasis of fruit flies. Importantly, we demonstrate that adult-onset administration of TIC10, a compound that reduces the aging-related AKT and extracellular signal-regulated kinase (ERK) signaling pathways, rescues the shortened lifespan of 14-3-3zeta-overexpressing flies. This finding suggests that 14-3-3zeta plays a critical role in regulating the aging process. Our study elucidates the role of 14-3-3zeta in natural aging and provides the rationale for subsequent 14-3-3zeta-based antiaging research.


Asunto(s)
Proteínas 14-3-3 , Envejecimiento , Proteínas de Drosophila , Drosophila melanogaster , Intestinos , Animales , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Envejecimiento/genética , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/inmunología , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Longevidad , Transducción de Señal , Intestinos/inmunología
9.
Immunology ; 172(1): 1-20, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38174581

RESUMEN

Irritable bowel syndrome (IBS), one of the most prevalent functional gastrointestinal disorders, is characterized by recurrent abdominal pain and abnormal defecation habits, resulting in a severe healthcare burden worldwide. The pathophysiological mechanisms of IBS are multi-factorially involved, including food antigens, visceral hypersensitivity reactions, and the brain-gut axis. Numerous studies have found that gut microbiota and intestinal mucosal immunity play an important role in the development of IBS in crosstalk with multiple mechanisms. Therefore, based on existing evidence, this paper elaborates that the damage and activation of intestinal mucosal immunity and the disturbance of gut microbiota are closely related to the progression of IBS. Combined with the application prospect, it also provides references for further in-depth exploration and clinical practice.


Asunto(s)
Microbioma Gastrointestinal , Síndrome del Colon Irritable , Humanos , Microbioma Gastrointestinal/fisiología , Intestinos , Mucosa Intestinal , Inmunidad Mucosa
10.
BMC Immunol ; 25(1): 71, 2024 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-39449131

RESUMEN

Immune thrombocytopenia (ITP) is a prevalent hemorrhage condition that causes notable immune-related abnormalities. Recently discovered data has shown that the intestinal flora plays a crucial role in maintaining a balanced immune system. Furthermore, an imbalance in gut flora has the potential to increase the possibility of developing ITP. Moreover, some studies reported a strong link between inflammatory bowel disease (IBD) and ITP. In this review, we described the significance of gut immunity in ITP. In addition, we explored the associations between gut flora and ITP as well as IBD and ITP. Finally, we examined the effectiveness of existing therapies that regulate gut homeostasis and their impact on the prognosis of patients with ITP.


Asunto(s)
Microbioma Gastrointestinal , Homeostasis , Enfermedades Inflamatorias del Intestino , Púrpura Trombocitopénica Idiopática , Humanos , Púrpura Trombocitopénica Idiopática/inmunología , Microbioma Gastrointestinal/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Animales , Intestinos/inmunología
11.
Eur J Immunol ; 53(10): e2250270, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37366299

RESUMEN

Mucosal barrier integrity and pathogen clearance is a complex process influenced by both Th17 and Treg cells. Previously, we had described the DNA methylation profile of Th17 cells and identified Zinc finger protein (Zfp)362 to be uniquely demethylated. Here, we generated Zfp362-/- mice to unravel the role of Zfp362 for Th17 cell biology. Zfp362-/- mice appeared clinically normal, showed no phenotypic alterations in the T-cell compartment, and upon colonization with segmented filamentous bacteria, no effect of Zfp362 deficiency on Th17 cell differentiation was observed. By contrast, Zfp362 deletion resulted in increased frequencies of colonic Foxp3+ Treg cells and IL-10+ and RORγt+ Treg cell subsets in mesenteric lymph nodes. Adoptive transfer of naïve CD4+ T cells from Zfp362-/- mice into Rag2-/- mice resulted in a significantly lower weight loss when compared with controls receiving cells from Zfp362+/+ littermates. However, this attenuated weight loss did not correlate with alterations of Th17 cells but instead was associated with an increase of effector Treg cells in mesenteric lymph nodes. Together, these results suggest that Zfp362 plays an important role in promoting colonic inflammation; however, this function is derived from constraining the effector function of Treg cells rather than directly promoting Th17 cell differentiation.


Asunto(s)
Linfocitos T Reguladores , Células Th17 , Ratones , Animales , Linfocitos T Reguladores/metabolismo , Células Th17/metabolismo , Diferenciación Celular , Inflamación/metabolismo , Pérdida de Peso , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo
12.
Cytokine ; 183: 156749, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39236431

RESUMEN

In humans and mice, the induction of interleukin (IL)-17 expression enhances epithelial barrier integrity through the secretion of antimicrobial peptides (AMP), thereby improving antibacterial defense. However, it is unclear whether IL-17 has similar antibacterial effects in chickens by modulating the expression of AMPs, such as avian beta-defensins (also known as gallinacins) and cathelicidins. This study evaluated the in vivo effects of inoculating 20-day-old broiler chickens with two doses of a plasmid encoding chicken IL-17 (pCDNA3.1/rchIL-17-V5-HIS TOPO plasmid [pCDNA3.1-IL-17]; 5 or 10 µg/bird). On day 23 of age, all broilers, except those in the negative control group, were orally challenged with a virulent Clostridium perfringens strain for three days. To investigate IL-17-mediated effects against C. perfringens infection, the expression of avian beta-defensin 1 (avBD1), avBD2, avBD4, avBD6, cathelicidins, and inducible nitric oxide synthase (iNOS) genes were quantified, and gross necrotic enteritis (NE) lesion scores were assessed in the small intestine. The results showed that broilers receiving the higher dose of pCDNA3.1-IL-17 (10 µg) had significantly lower NE lesion scores compared to those receiving the lower dose (5 µg), the vector control, and the positive control groups. Furthermore, the expression of all avian beta-defensins and cathelicidin genes was detectable across all groups, regardless of treatment and time points. IL-17 treatment led to significantly higher expression of avBD1, avBD2, avBD4, avBD6, cathelicidin, and iNOS in the duodenum, jejunum, and ileum compared to control chickens. In C. perfringens-infected chickens, the expression of avBD1, avBD2, avBD4, cathelicidin, and iNOS in the ileum was significantly higher than in control chickens. Pre-treatment with the higher dose of pCDNA3.1-IL-17 (10 µg) in infected chickens was associated with reduced NE lesion severity and increased expression of avBD1, avBD2, cathelicidin, and iNOS in the ileum, but not avBD4 and avBD6. These findings provide new insights into the potential effect of IL-17 and reduction in NE lesion severity by modulating AMP expression which may be involved in mediating protective immunity against intestinal infection with C. perfringens.


Asunto(s)
Pollos , Clostridium perfringens , Enteritis , Interleucina-17 , Intestino Delgado , beta-Defensinas , Animales , Pollos/microbiología , Interleucina-17/metabolismo , Interleucina-17/genética , Enteritis/microbiología , Enteritis/inmunología , Enteritis/veterinaria , Enteritis/metabolismo , Intestino Delgado/metabolismo , Intestino Delgado/microbiología , Intestino Delgado/inmunología , beta-Defensinas/metabolismo , beta-Defensinas/genética , Enfermedades de las Aves de Corral/microbiología , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/metabolismo , Catelicidinas , Péptidos Antimicrobianos/genética , Péptidos Antimicrobianos/metabolismo , Necrosis , Modelos Animales de Enfermedad , Infecciones por Clostridium/veterinaria , Infecciones por Clostridium/inmunología , Péptidos Catiónicos Antimicrobianos/metabolismo , Péptidos Catiónicos Antimicrobianos/genética , Regulación de la Expresión Génica/efectos de los fármacos
13.
J Nutr ; 154(5): 1711-1721, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38367809

RESUMEN

BACKGROUND: Glutamine (Gln) has an important effect on the growth performance and immune function of piglets. However, the effect of Gln on intestinal immunity in piglets through modulating the signaling pathways of the helper T cells 17 (Th17)/regulatory T cells (Treg) immune response has not been reported. OBJECTIVE: This study aimed to determine the effect of Gln on piglet growth performance and immune stress response and its mechanism in piglets. METHODS: Twenty-four weaned piglets were randomly assigned to 4 treatments with 6 replicates each, using a 2 × 2 factorial arrangement: diet (basal diet or 1% Gln diet) and immunological challenge [saline or lipopolysaccharide (LPS)]. After 21 d, half of the piglets on the basal diet and 1% Gln diet received the intraperitoneal injection of LPS and the other half received the same volume of normal saline. RESULTS: The results showed that Gln increased average daily feed intake and average daily weight gain in comparison with the control group (P < 0.05). Dietary Gln increased the villus height, villus height-to-crypt depth ratio, and the abundance of Bacteroidetes, Lactobacillus sp., and Ruminococcus sp. while reducing the abundance of Firmicutes, Clostridium sensu stricto 1 sp., and Terrisporobacter sp. (P < 0.05). Furthermore, Gln increased the concentration of short-chain fatty acids in the colon and the expression of genes of interleukin (IL)-10, transforming growth factor-beta-1, forkhead box P3 while downregulating the expression of genes of IL-6, IL-8, IL-1ß, tumor necrosis factor-α, IL-17A, IL-21, signal transducer and activator of transcription 3, and rar-related orphan receptor c in ileum (P < 0.05). Correlation analysis demonstrated a strong association between colonic microbiota, short-chain fatty acids, and ileal inflammatory cytokines. CONCLUSIONS: These results suggest that dietary Gln could improve growth performance and attenuate LPS-challenged intestinal inflammation by modulating microbiota and the Th17/Treg immune response signaling pathway in piglets.


Asunto(s)
Suplementos Dietéticos , Microbioma Gastrointestinal , Glutamina , Lipopolisacáridos , Transducción de Señal , Linfocitos T Reguladores , Células Th17 , Animales , Glutamina/farmacología , Glutamina/administración & dosificación , Porcinos , Microbioma Gastrointestinal/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Alimentación Animal/análisis , Dieta/veterinaria
14.
Fish Shellfish Immunol ; 154: 109941, 2024 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-39368523

RESUMEN

The preceding study observed that yeast ß-glucan supplementation enhanced intestinal health and augmented disease resistance in pearl gentian grouper (Epinephelus lanceolatus♂ × Epinephelus fuscoguttatus♀), which occurred concurrently with the activation of the nuclear factor kappa B (NFκB) signaling pathway. Thus, we hypothesized that ß-glucan improves intestinal health in grouper by modulating the NFκB pathway. Accordingly, the present study examined the effects of NFκB pathway disruption using a specific inhibitor on the intestinal health of pearl gentian grouper that had been injected with ß-glucan. The experimental groups were as follows, (1) CD group: PBS injected; (2) ßG group: ß-glucan injected at a dose of 80 mg/kg; (3) PDTC group: NFκB inhibitor PDTC injected at a dose of 30 mg/kg; (4) ßG + PDTC group: a combination of ß-glucan (80 mg/kg) and PDTC (30 mg/kg) injected together. The results demonstrated that ß-glucan-induced increases in mRNA expression levels of NFκB inhibitor α (iκbα) and p65, the degradation and phosphorylation of IκBα, and the phosphorylation of NFκB p65 were significantly inhibited following NFκB inhibition using PDTC in the intestine of grouper. The PDTC injection resulted in a significant reduction in the ß-glucan-induced increase in mucin levels. The ß-glucan-induced elevation of alkaline phosphatase (AKP) activity, component 3 (C3) content, and inflammatory factors were significantly suppressed following NFκB inhibition. The ßG + PDTC treatment resulted in a restoration of catalase (CAT) enzyme activity to the level observed in the CD treatment, while total antioxidant capacity (T-AOC) was decreased to the level of the ßG treatment. The ß-glucan-induced downregulation of caspase8 (casp8) was reversed following NFκB inhibition, as well as the mRNA levels of casp3 and casp9 being elevated to a greater extent. In conclusion, the ß-glucan-regulated intestinal immunity in grouper may be mediated by the NFκB pathway. Furthermore, the inhibitory effect of ß-glucan on apoptosis and oxidative stress may not be related to the NFκB signaling pathway.

15.
Fish Shellfish Immunol ; 153: 109790, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39059563

RESUMEN

Grass carp (Ctenopharyngodon idella) is an intensively cultured and economically important herbivorous fish species in China, but its culture is often impacted by Aeromonas pathogens such as Aeromonas hydrophila and Aeromonas veronii. In this study, healthy grass carp were separately infected with A. hydrophila or A. veronii for 12, 24, 48 or 72 h. The results showed that the mRNA expression levels of intestinal inflammatory factors (tnf-α, il-1ß and il-8), complement factors (c3 and c4), antimicrobial peptides (hepcidin, nk-lysin and ß-defensin-1), immunoglobulins (igm and igt), and immune pathway-related signaling molecules (tlr1, tlr2, tlr4, myd88, irak4, irak1, traf6, nf-κb p65 and ap-1) were differentially upregulated in response to A. hydrophila and A. veronii challenge. Additionally, the expression levels of the intestinal pro-apoptotic genes tnfr1, tnfr2, tradd, caspase-8, caspase-3 and bax were significantly increased, whereas the expression of the inhibitory factor bcl-2 was significantly downregulated, indicating that Aeromonas infection significantly induced apoptosis in the intestine of grass carp. Moreover, the expression of intestinal tight junction proteins (occludin, zo-1, claudin b and claudin c) was significantly decreased after infection with Aeromonas. Histopathological analysis indicated the Aeromonas challenge caused severe damage to the intestinal villi with adhesions and detachment of intestinal villi accompanied by severe inflammatory cell infiltration at 12 h and 72 h. The 16S rRNA sequencing results showed that Aeromonas infection significantly altered the structure of the intestinal microflora of the grass carp at the phylum (Proteobacteria, Fusobacteria, Bacteroidetes and Firmicutes) and genus (Proteus, Cetobacterium, Bacteroides, and Aeromonas) levels. Take together, the findings of this study revealed that Aeromonas infection induces an intestinal immune response, triggers cell apoptosis, destroys physical barriers and alters microflora structure in the intestine of juvenile grass carp; the results will help to reveal the pathogenesis of intestinal bacterial diseases in grass carp.


Asunto(s)
Aeromonas hydrophila , Aeromonas veronii , Carpas , Enfermedades de los Peces , Microbioma Gastrointestinal , Infecciones por Bacterias Gramnegativas , Inmunidad Innata , Intestinos , Animales , Carpas/inmunología , Infecciones por Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/veterinaria , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/microbiología , Aeromonas hydrophila/fisiología , Intestinos/inmunología , Intestinos/microbiología , Aeromonas veronii/fisiología , Aeromonas/fisiología , Aeromonas/inmunología , Proteínas de Peces/genética , Proteínas de Peces/inmunología
16.
Fish Shellfish Immunol ; 151: 109741, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38964436

RESUMEN

Decay-accelerating factor (DAF) is an essential member of the complement regulatory protein family that plays an important role in immune response and host homeostasis in mammals. However, the immune function of DAF has not been well characterized in bony fish. In this study, a complement regulatory protein named CiDAF was firstly characterized from Ctenopharyngodon idella and its potential roles were investigated in intestine following bacterial infection. Similar to mammalian DAFs, CiDAF has multiple complement control protein (CCP) functional domains, suggesting the evolutionary conservation of DAFs. CiDAF was broadly expressed in all tested tissues, with a relatively high expression level detected in the spleen and kidney. In vivo immune challenge experiments revealed that CiDAF strongly responded to bacterial pathogens (Aeromonas hydrophila and Aeromonas veronii) and PAMPs (lipopolysaccharide (LPS) or muramyl dipeptide (MDP)) challenges. In vitro RNAi experiments indicated that knockdown of CiDAF could upregulate the expression of complement genes (C4b, C5 and C7) and inflammatory cytokines (TNF-α, IL-1ß and IL-8). Moreover, 2000 ng/mL of CiDAF agonist progesterone effectively alleviated LPS- or MDP-induced intestinal inflammation by regulating expression of complement factors, TLR/PepT1 pathway genes and inflammatory cytokines. Overall, these findings revealed that CiDAF may act as a negative regulator of intestinal complement pathway and immune response to bacterial challenge in grass carp.


Asunto(s)
Carpas , Enfermedades de los Peces , Proteínas de Peces , Infecciones por Bacterias Gramnegativas , Inmunidad Innata , Intestinos , Animales , Carpas/inmunología , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Enfermedades de los Peces/inmunología , Inmunidad Innata/genética , Infecciones por Bacterias Gramnegativas/inmunología , Infecciones por Bacterias Gramnegativas/veterinaria , Intestinos/inmunología , Regulación de la Expresión Génica/inmunología , Filogenia , Perfilación de la Expresión Génica/veterinaria , Aeromonas hydrophila/fisiología , Secuencia de Aminoácidos , Alineación de Secuencia/veterinaria , Proteínas del Sistema Complemento/inmunología
17.
Fish Shellfish Immunol ; 146: 109362, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38218423

RESUMEN

A 56-d feeding trial was conducted to evaluate the influences of Rhodiola rosea L. on digestive enzyme activities, intestinal barrier, inflammatory response, and microbiota dysbiosis in Lateolabrax maculatus juveniles (9.37 ± 0.03 g) fed with high-carbohydrate diets. Six diets were designed: a control diet (20% corn starch, Control), high-carbohydrate diet (30% corn starch, HC1), and four high-carbohydrate diets supplemented with Rhodiola rosea L. at 30, 60, 90 and 120 mg/kg (HC2, HC3, HC4 and HC5, respectively). Compared with the control group, the HC1 diet remarkably increased α-amylase, lipase, and chymotrypsin activities in the intestine (p < 0.05), as well as the mRNA levels of Claudin-15, NF-κB, TNF-α, IL-1ß, and IL-8 (p < 0.05) and the relative abundance of Proteobacteria and Photobacterium in the intestine, which belong to the phylum and genus level, respectively. But the opposite trend was found in muscular thickness and villus lengths (p < 0.05), the mRNA levels of Occludin, ZO-1, and TGF-ß (p < 0.05), at the level of phylum and genus level in the HC1 group, and the relative abundance of Firmicutes, Bacteroidetes, and Bacillus in the intestine compared with the control group. Intestinal chymotrypsin activity was significantly higher in the HC3 group and intestinal muscular thickness and villus lengths were also significantly higher in the HC2, HC3, HC4, and HC5 groups compared to the HC1 group (p < 0.05). In addition, Occludin mRNA expression in the intestine was significantly increased in the HC2, HC4, and HC5 groups compared to the HC1 group. ZO-1 and TGF-ß mRNA expression in the intestine were significantly increased in the HC2, HC3, HC4, and HC5 groups compared to the HC1 group (p < 0.05). At the phylum level, the relative abundance of Firmicutes and Bacteroidetes was higher in the intestine in the HC2, HC3, HC4, and HC5 groups than that in the HC1 group. On the contrary, intestinal lipase and chymotrypsin activities were significantly decreased in the HC2 group compared to the HC1 group, respectively (p < 0.05). The Claudin-15, NF-κB, TNF-α, IL-1ß, and IL-8 mRNA expression in the intestine were significantly decreased in the HC2, HC3, HC4, and HC5 groups compared to the HC1 group (p < 0.05). Besides, at the genus level, compared to the HC1 group, the relative abundance of Photobacterium in the intestine and the diversity of the intestinal microbiota in the HC2, HC3, HC4, and HC5 groups were all decreased. In conclusion, these results demonstrated that the addition of Rhodiola rosea L. in high-carbohydrate diets can improve intestinal digestive enzyme activities, inflammatory response and intestinal barrier-related gene expression, and microbiota dysbiosis in L. maculatus. The suitable supplemental level of Rhodiola rosea L. in high-carbohydrate diets of L. maculatus is 60 mg/kg.


Asunto(s)
Microbiota , Rhodiola , Animales , FN-kappa B , Factor de Necrosis Tumoral alfa , Quimotripsina , Disbiosis , Interleucina-8 , Ocludina , Intestinos/fisiología , Dieta/veterinaria , Peces , Lipasa , ARN Mensajero , Almidón , Factor de Crecimiento Transformador beta , Alimentación Animal/análisis
18.
BMC Vet Res ; 20(1): 276, 2024 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-38926724

RESUMEN

BACKGROUND: Trace elements play a crucial role in fish nutrition, with zinc (Zn) being one of the most important elements. BIO-sourced zinc nanoparticles were synthesized using the green microalga Pediastrum boryanum (BIO-ZnNPs, 29.35 nm). 30 or 60 mg/ kg dry feed of the BIO-ZnNPs (BIO-ZnNPs30 and BIO-ZnNPs60) were mixed with the Nile tilapia (Oreochromis niloticus) basal diet and fed to the fish for 8 weeks to evaluate their impact on fish growth, digestion, intestinal integrity, antioxidative status, and immunity. RESULTS: A significant enhancement was observed in all investigated parameters, except for the serum protein profile. BIO-ZnNPs at 60 mg/kg feed elevated the activities of reduced glutathione (GSH) and catalase (CAT), enzymatic antioxidants, but did not induce oxidative stress as reflected by no change in MDA level. Fish intestinal immunity was improved in a dose-dependent manner, in terms of improved morphometry and a higher count of acid mucin-producing goblet cells. Interleukin-8 (IL-8) was upregulated in BIO-ZnNPs30 compared to BIO-ZnNPs60 and control fish groups, while no significant expressions were noted in tumor necrosis factor-alpha (TNFα), nuclear factor kappa B (NFkB), and Caspase3 genes. CONCLUSION: Overall, BIO-ZnNPs inclusion at 60 mg/kg feed showed the most advantage in different scenarios, compared to BIO-ZnNPs at 30 mg/kg feed. The positive effects on growth and intestinal health suggest that BIO-ZnNPs supplementation of aquafeeds has many benefits for farmed fish.


Asunto(s)
Alimentación Animal , Cíclidos , Dieta , Intestinos , Zinc , Animales , Zinc/farmacología , Zinc/administración & dosificación , Alimentación Animal/análisis , Cíclidos/inmunología , Cíclidos/crecimiento & desarrollo , Intestinos/efectos de los fármacos , Intestinos/inmunología , Dieta/veterinaria , Suplementos Dietéticos , Nanopartículas del Metal , Antioxidantes , Chlorophyta/química , Microalgas
19.
Int J Mol Sci ; 25(2)2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-38255801

RESUMEN

Iron is a vital trace element that plays an important role in humans and other organisms. It plays an active role in the growth, development, and reproduction of bacteria, such as Bifidobacteria. Iron deficiency or excess can negatively affect bacterial hosts. Studies have reported a major role of iron in the human intestine, which is necessary for maintaining body homeostasis and intestinal barrier function. Organisms can maintain their normal activities and regulate some cancer cells in the body by regulating iron excretion and iron-dependent ferroptosis. In addition, iron can modify the interaction between hosts and microorganisms by altering their growth and virulence or by affecting the immune system of the host. Lactic acid bacteria such as Lactobacillus acidophilus (L. acidophilus), Lactobacillus rhamnosus (L. rhamnosus), and Lactobacillus casei (L. casei) were reported to increase trace elements, protect the host intestinal barrier, mitigate intestinal inflammation, and regulate immune function. This review article focuses on the two aspects of the iron and gut and generally summarizes the mechanistic role of iron ions in intestinal immunity and the remodeling of gut microbiota.


Asunto(s)
Microbioma Gastrointestinal , Oligoelementos , Humanos , Hierro , Homeostasis , Iones , Lactobacillus acidophilus
20.
J Cell Physiol ; 238(7): 1465-1477, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37210730

RESUMEN

Follicular regulatory T (Tfr) cells are a novel and unique subset of effector regulatory T (Treg) cells that are located in germinal centers (GCs). Tfr cells express transcription profiles that are characteristic of both follicular helper T (Tfh) cells and Treg cells and negatively regulate GC reactions, including Tfh cell activation and cytokine production, class switch recombination and B cell activation. Evidence also shows that Tfr cells have specific characteristics in different local immune microenvironments. This review focuses on the regulation of Tfr cell differentiation and function in unique local immune microenvironments, including the intestine and tumor.


Asunto(s)
Intestinos , Linfocitos T Reguladores , Microambiente Tumoral , Centro Germinal , Intestinos/citología , Intestinos/inmunología , Linfocitos T Colaboradores-Inductores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA