Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.352
Filter
1.
Nat Commun ; 15(1): 3784, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38710716

ABSTRACT

Probiotic and engineered microbe-based therapeutics are an emerging class of pharmaceutical agents. They represent a promising strategy for treating various chronic and inflammatory conditions by interacting with the host immune system and/or delivering therapeutic molecules. Here, we engineered a targeted probiotic yeast platform wherein Saccharomyces boulardii is designed to bind to abundant extracellular matrix proteins found within inflammatory lesions of the gastrointestinal tract through tunable antibody surface display. This approach enabled an additional 24-48 h of probiotic gut residence time compared to controls and 100-fold increased probiotic concentrations within the colon in preclinical models of ulcerative colitis in female mice. As a result, pharmacodynamic parameters including colon length, colonic cytokine expression profiles, and histological inflammation scores were robustly improved and restored back to healthy levels. Overall, these studies highlight the potential for targeted microbial therapeutics as a potential oral dosage form for the treatment of inflammatory bowel diseases.


Subject(s)
Colitis, Ulcerative , Colon , Disease Models, Animal , Extracellular Matrix , Probiotics , Saccharomyces boulardii , Animals , Probiotics/administration & dosage , Female , Mice , Extracellular Matrix/metabolism , Colitis, Ulcerative/therapy , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/pathology , Colon/microbiology , Colon/metabolism , Colon/pathology , Mice, Inbred C57BL , Colitis/therapy , Colitis/microbiology , Colitis/pathology , Cytokines/metabolism , Humans
2.
Clin Transl Sci ; 17(5): e13821, 2024 May.
Article in English | MEDLINE | ID: mdl-38742709

ABSTRACT

Inflammatory bowel disease (IBD) is characterized by a chronically dysregulated immune response in the gastrointestinal tract. Bone marrow multipotent mesenchymal stromal cells have an important immunomodulatory function and support regeneration of inflamed tissue by secretion of soluble factors as well as through direct local differentiation. CXCR4 is the receptor for CXCL12 (SDF-1, stromal-derived factor-1) and has been shown to be the main chemokine receptor, required for homing of MSCs. Increased expression of CXCL12 by inflamed intestinal tissue causes constitutive inflammation by attracting lymphocytes but can also be used to direct MSCs to sites of injury/inflammation. Trypsin is typically used to dissociate MSCs into single-cell suspensions but has also been shown to digest surface CXCR4. Here, we assessed the regenerative effects of CXCR4high and CXCR4low MSCs in an immune-deficient mouse model of DSS-induced colitis. We found that transplantation of MSCs resulted in clinical improvement and histological recovery of intestinal epithelium. In contrary to our expectations, the levels of CXCR4 on transplanted MSCs did not affect their regenerative supporting potential, indicating that paracrine effects of MSCs may be largely responsible for their regenerative/protective effects.


Subject(s)
Colitis , Disease Models, Animal , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Mice, Inbred C57BL , Receptors, CXCR4 , Regeneration , Animals , Receptors, CXCR4/metabolism , Receptors, CXCR4/genetics , Mesenchymal Stem Cells/metabolism , Colitis/chemically induced , Colitis/pathology , Colitis/immunology , Colitis/therapy , Colitis/metabolism , Mesenchymal Stem Cell Transplantation/methods , Mice , Dextran Sulfate , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/immunology , Chemokine CXCL12/metabolism , Chemokine CXCL12/genetics , Bone Marrow Cells/metabolism
3.
J Int Med Res ; 52(5): 3000605241241000, 2024 May.
Article in English | MEDLINE | ID: mdl-38749910

ABSTRACT

Ileostomy diverts the flow of feces, which can result in malnutrition in the distal part of the intestine. The diversity of the gut microbiota consequently decreases, ultimately leading to intestinal dysbiosis and dysfunction. This condition can readily result in diversion colitis (DC). Potential treatment strategies include interventions targeting the gut microbiota. In this case study, we effectively treated a patient with severe DC by ileostomy and allogeneic fecal microbiota transplantation (FMT). A 69-year-old man presented with a perforated malignant tumor in the descending colon and an iliac abscess. He underwent laparoscopic radical sigmoid colon tumor resection and prophylactic ileostomy. Follow-up colonoscopy 3 months postoperatively revealed diffuse intestinal mucosal congestion and edema along with granular inflammatory follicular hyperplasia, leading to a diagnosis of severe DC. After two rounds of allogeneic FMT, both the intestinal mucosal bleeding and edema significantly improved, as did the diversity of the gut microbiota. The positive outcome of allogeneic FMT in this case highlights the potential advantages that this procedure can offer patients with DC. However, few studies have focused on allogeneic FMT, and more in-depth research is needed to gain a better understanding.


Subject(s)
Colitis , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Ileostomy , Humans , Male , Aged , Fecal Microbiota Transplantation/methods , Colitis/microbiology , Colitis/therapy , Transplantation, Homologous/methods , Treatment Outcome , Colonoscopy
4.
Int J Biol Macromol ; 269(Pt 1): 132063, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38705329

ABSTRACT

Probiotic therapy has emerged as a promising approach for the treatment of gastrointestinal diseases, offering advantages in terms of safety and convenience. However, oral probiotics encounter significant challenges, including exposure to a hostile gastric environment with low pH, bile salts, elevated levels of reactive oxygen species (ROS), and damage to the protective mucus layer. These factors reduce probiotic survival rates and limit their physiological activity. To address these challenges, we developed a layer-by-layer coated probiotics with curcumin-loaded liposome and polymer. Through DSS-induced colitis mice experiments, we demonstrated that the coated probiotics exhibited an improved survival rate in the gastrointestinal tract and enhanced adhesion to the intestinal mucosa. Furthermore, multi-layered coated probiotics exhibited remarkable efficacy in alleviating colitis by efficiently repairing the gut barrier, modulating gut microbial homeostasis, and reducing bacterial motility at sites of colonic inflammation. Our innovative approach holds promise for effectively treating gastrointestinal diseases.


Subject(s)
Chitosan , Colitis , Dextran Sulfate , Liposomes , Probiotics , Animals , Probiotics/administration & dosage , Probiotics/pharmacology , Colitis/chemically induced , Colitis/therapy , Colitis/drug therapy , Liposomes/chemistry , Mice , Chitosan/chemistry , Chitosan/pharmacology , Curcumin/pharmacology , Curcumin/chemistry , Disease Models, Animal , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Gastrointestinal Microbiome/drug effects
5.
Int Immunopharmacol ; 133: 112092, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38626548

ABSTRACT

BACKGROUND: Endometrial regenerative cells (ERCs) have been proven to be an effective strategy for attenuating experimental colitis, but the complex in vivo microenvironment such as oxidative stress may largely limit and weaken ERC efficacy. Melatonin (MT) works as an anti-oxidative agent in a variety of preclinical diseases, and has been identified to promote mesenchymal stem cell-mediated therapeutic effects in different diseases. However, the ability of MT to enhance ERC-mediated effects in colitis is currently poorly understood. METHODS: Menstrual blood was collected from healthy female volunteers to obtain ERCs and identified. In vitro, H2O2-induced oxidative stress was introduced to test if MT could prevent ERCs from damage through detection of intracellular reactive oxidative species (ROS) and apoptosis assay. In vivo, dextran sodium sulfate (DSS)-induced acute colitis was treated by ERCs and MT-primed ERCs, therapeutic effects were assayed by the disease activity index (DAI), histological features, and macrophage and CD4+ T cell in the spleen and colon, and cytokine profiles in the sera and colon were also measured. RESULTS: In vitro, ERCs that underwent MT-precondition were found to possess more anti-oxidative potency in comparison to naïve ERCs, which were characterized by decreased apoptosis rate and intracellular ROS under H2O2 stimulation. In vivo, MT pretreatment can significantly enhance the therapeutic effects of ERCs in the attenuation of experimental colitis, including decreased DAI index and damage score. In addition, MT pretreatment was found to promote ERC-mediated inhibition of Th1, Th17, and M1 macrophage and pro-inflammatory cytokines, increase of Treg, and immunomodulation of cytokines in the spleen and colon. CONCLUSIONS: MT pretreatment facilitates the promotion of cell viability under oxidative stress in vitro, while also enhancing ERC-mediated therapeutic effects in experimental colitis.


Subject(s)
Colitis , Dextran Sulfate , Endometrium , Melatonin , Oxidative Stress , Melatonin/therapeutic use , Melatonin/pharmacology , Animals , Female , Colitis/chemically induced , Colitis/therapy , Colitis/drug therapy , Humans , Endometrium/pathology , Endometrium/drug effects , Oxidative Stress/drug effects , Mice , Mice, Inbred C57BL , Hydrogen Peroxide/metabolism , Cytokines/metabolism , Disease Models, Animal , Apoptosis/drug effects , Cells, Cultured , Antioxidants/therapeutic use , Antioxidants/pharmacology , Colon/pathology , Colon/drug effects , Reactive Oxygen Species/metabolism , Adult , Regeneration/drug effects , Macrophages/immunology , Macrophages/drug effects
6.
Biomaterials ; 308: 122564, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38581763

ABSTRACT

Probiotic-based therapies have shown great potential in the prevention and treatment of many diseases by positively regulating intestinal flora homeostasis. However, the efficacy of oral probiotics is severely limited due to the loss of bioactivity, short intestinal retention time, and insufficient therapeutic effect. Here, based on droplet microfluidics, we developed a hydrogel microsphere with colonic targeting and mucoadhesive capabilities as a multifunctional delivery platform, which can be used for co-delivery of probiotics (Escherichia coli Nissle 1917, EcN) and auxiliary molecules (indole-3-propionic acid, IPA), achieving synergistic therapeutic effects. In vivo studies shown that the integrated multifunctional microspheres can significantly reduce intestinal inflammation, repair intestinal barrier function, enhance probiotic colonization in the intestine, and modulate disordered intestinal flora, demonstrating enhanced therapeutic effects in a mouse model of colitis. This work reveals that microfluidic-based smart droplet microspheres can provide a versatile platform for advanced microbial therapies.


Subject(s)
Microspheres , Probiotics , Probiotics/administration & dosage , Animals , Administration, Oral , Mice , Escherichia coli , Colitis/therapy , Microfluidics/methods , Mice, Inbred C57BL , Drug Delivery Systems/methods , Hydrogels/chemistry , Indoles/chemistry , Gastrointestinal Microbiome/drug effects , Humans
7.
EBioMedicine ; 103: 105128, 2024 May.
Article in English | MEDLINE | ID: mdl-38653187

ABSTRACT

BACKGROUND: The use of mesenchymal stem cells (MSCs) has recently emerged as a promising new therapeutic strategy for many diseases including perianal fistulizing Crohn's disease (CD). Whether hUC-MSCs can promote the healing of luminal ulcer in CD has not been studied so far. METHODS: The model of TNBS-induced colitis in rats was used to confirm the efficacy of hUC-MSCs in the treatment of CD. Then, seventeen CD patients refractory to or unsuitable for currently available therapies were enrolled and received once submucosal local injection through colonoscopy combined with once intravenous drip on the next day. All patients received a 24-week follow-up. Clinical and laboratory assessments were monitored at baseline, week 4, 8, 12, and 24. Endoscopic evaluations were conducted at baseline and week 12. Mucosal specimens were obtained at the margin of lesions by endoscopy biopsies and used for RNA sequencing. Two hUC-MSCs co-culture systems were established in vitro, one with the mucosa specimens and the other with M1 macrophages induced from THP1. The expressions of genes representing inflammation (TNFα, IL-6, and IL-1ß) and intestinal barrier function (ZO1, CLAUDIN1, and CDH1) were tested by RT-PCR. FINDINGS: hUC-MSCs treatment increased body weight and decreased disease activity index (DAI), colon macroscopic damage index (CMDI), and histopathological score (HPS) of rats with TNBS-induced colitis. The results of the clinical study also showed that this mode of hUC-MSCs application was associated with regression of intestinal ulceration. Eight patients (47%) got endoscopic responses (SES-CD improvement of ≥50% from baseline) and three patients (17.65%) got mucosal healing (SES-CD is zero), with a parallel improvement of clinical and laboratory parameters without serious adverse events. RNA sequencing showed hUC-MSCs therapy was associated with an upregulation of transcripts linked to intestinal epithelial barrier integrity and a downregulation of inflammatory signaling pathways in the intestinal mucosa, especially the TNF signaling pathway, IL-17 signaling pathway, and TLR signaling pathway. RNA expression of intestinal epithelial tight junction protein (ZO1, CLAUDIN1, and CDH1), and the RNA expression of major intestinal inflammatory factors in CD (IL-1ß, IL-6, and TNFα, p < 0.001 for all) were improved significantly. Moreover, hUC-MSCs could attenuate the polarization of M1 macrophage induced from THP1, thereby decreasing the mRNA expression of IL-1ß, IL-6, and TNFα significantly (p < 0.05 for all). TSG-6 expression was evaluated in hUC-MSCs culture supernatant after treatment with TNFα, IFNγ, and LPS for 48 h. And hUC-MSCs could inhibit the phosphorylation of JAK/STAT1 in the intestinal mucosa of CD patients. INTERPRETATION: hUC-MSCs transplantation alleviated TNBS-induced colitis in rats. In this pilot clinical study, preliminary data suggested that this approach to administering hUC-MSCs might have potential for clinical efficacy and manageable safety in treating refractory CD, potentially providing hope for better outcomes. No serious adverse events were observed. FUNDING: This work was funded by General Program of National Natural Science Foundation of China (Grant No. 82270639), the Scientific research project of Shanghai Municipal Health Committee (Grant No. 202240001), Specialty Feature Construction Project of Shanghai Pudong New Area Health Commission (Grant No. PWZzb2022-05), Shanghai East Hospital Youth Research and Cultivation Foundation program (Grant No. DFPY2022015), Peak Disciplines (Type IV) of Institutions of Higher Learning in Shanghai, Technology Development Project of Pudong Science, Technology and Economic Commission of Shanghai (Grant No. PKJ2021-Y08), Key Disciplines Group Construction Project of Shanghai Pudong New Area Health Commission (Grant No. PWZxq2022-06), Medical discipline Construction Project of Pudong Health Committee of Shanghai (Grant No. PWYgf2021-02) and National Natural Science Foundation of China (Grant No. 82300604).


Subject(s)
Colitis , Crohn Disease , Disease Models, Animal , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Trinitrobenzenesulfonic Acid , Animals , Crohn Disease/therapy , Crohn Disease/metabolism , Mesenchymal Stem Cell Transplantation/methods , Rats , Humans , Male , Female , Adult , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Trinitrobenzenesulfonic Acid/adverse effects , Pilot Projects , Colitis/therapy , Colitis/chemically induced , Colitis/metabolism , Middle Aged , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Treatment Outcome , Cytokines/metabolism
8.
Surg Clin North Am ; 104(3): 545-556, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38677819

ABSTRACT

Clostridioides difficile colitis is an important source of hospital-acquired diarrhea associated with antibiotic use. Symptoms are profuse watery diarrhea, typically following a course of antibiotics; however, some cases of fulminant disease may manifest with shock, ileus, or megacolon. Nonfulminant colitis is treated with oral fidaxomicin. C difficile colitis has a high potential for recurrence, and recurrent episodes are also treated with fidaxomicin. Bezlotoxumab is another medication that may be used in populations at high risk for further recurrence. Fulminant disease is treated with maximal medical therapy and early surgical consultation. Antibiotic stewardship is critical to preventing C difficile colitis.


Subject(s)
Anti-Bacterial Agents , Clostridioides difficile , Clostridium Infections , Colitis , Humans , Clostridium Infections/diagnosis , Clostridium Infections/therapy , Anti-Bacterial Agents/therapeutic use , Colitis/microbiology , Colitis/diagnosis , Colitis/therapy , Fidaxomicin/therapeutic use
9.
J Agric Food Chem ; 72(18): 10355-10365, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38620073

ABSTRACT

The genus Bifidobacterium has been widely used in functional foods for health promotion due to its beneficial effects on human health, especially in the gastrointestinal tract (GIT). In this study, we characterize the anti-inflammatory potential of the probiotic strain Bifidobacterium pseudocatenulatum G7, isolated from a healthy male adult. G7 secretion inhibited inflammatory response in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages. Moreover, oral administration of bacteria G7 alleviated the severity of colonic inflammation in dextran sulfate sodium (DSS)-treated colitis mice, which was evidenced by a decreased disease activity index (DAI) and enhanced structural integrity of the colon. The 16S rRNA gene sequencing result illustrated that the G7 alleviated DSS-induced gut microbiota dysbiosis, accompanied by the modulated bile acids and short-chain fatty acid (SCFA) levels. Overall, our results demonstrated the potential anti-inflammatory effects of Bifidobacterium pseudocatenulatum G7 on both in vitro and in vivo models, which provided a solid foundation for further development of a novel anti-inflammatory probiotic.


Subject(s)
Anti-Inflammatory Agents , Bifidobacterium pseudocatenulatum , Colitis , Gastrointestinal Microbiome , Probiotics , Probiotics/administration & dosage , Probiotics/pharmacology , Mice , Animals , RAW 264.7 Cells , Male , Anti-Inflammatory Agents/administration & dosage , Humans , Colitis/microbiology , Colitis/therapy , Colitis/chemically induced , Bifidobacterium pseudocatenulatum/genetics , Bifidobacterium pseudocatenulatum/chemistry , Mice, Inbred C57BL , Macrophages/immunology , Fatty Acids, Volatile/metabolism , Colon/microbiology , Colon/immunology
10.
Front Immunol ; 15: 1366489, 2024.
Article in English | MEDLINE | ID: mdl-38660314

ABSTRACT

Cancer ranks among the foremost causes of mortality worldwide, posing a significant threat to human lives. The advent of tumor immunotherapy has substantially transformed the therapeutic landscape for numerous advanced malignancies, notably non-small cell lung cancer and melanoma. However, as immune checkpoint inhibitors (ICIs) are increasingly applied in clinical settings, a spectrum of undesired reactions, termed immune-related adverse events (irAEs), has emerged. These adverse reactions are associated with immunotherapy and can result in varying degrees of harm to the human body. Among these reactions, Immune checkpoint inhibitor-induced colitis (ICIIC) stands out as one of the most prevalent clinical adverse events. In contemporary times, traditional Chinese medicine (TCM) has demonstrated remarkable efficacy in addressing various maladies. Consequently, investigating the potential application and mechanisms of Chinese medicine in countering immune checkpoint inhibitor-induced colitis assumes significant importance in the treatment of this condition.


Subject(s)
Colitis , Immune Checkpoint Inhibitors , Medicine, Chinese Traditional , Humans , Immune Checkpoint Inhibitors/adverse effects , Immune Checkpoint Inhibitors/therapeutic use , Colitis/chemically induced , Colitis/immunology , Colitis/therapy , Animals , Drugs, Chinese Herbal/adverse effects , Drugs, Chinese Herbal/therapeutic use , Immunotherapy/adverse effects , Immunotherapy/methods
13.
BMC Immunol ; 25(1): 21, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38637733

ABSTRACT

Helminth-derived proteins have immunomodulatory properties, influencing the host's immune response as an adaptive strategy for helminth survival. Helminth-derived proteins modulate the immune response by inducing anti-inflammatory cytokines, promoting regulatory T-cell development, and ultimately favouring a Th2-biased immune response. This systematic review focused on helminth-derived proteins and explored their impact on reducing inflammatory responses in mouse models of colitis. A systematic search across Medline, EMBASE, Web of Science, and Cochrane Library identified fourteen relevant studies. These studies reported immunomodulatory changes, including increased production of anti-inflammatory cells and cytokines. In mouse models of colitis treated with on helminth-derived proteins, significant improvements in pathological parameters such as body weight, colon length, and microscopic inflammatory scores were observed compared to control groups. Moreover, helminth-derived proteins can enhance the function of Tregs and alleviate the severity of inflammatory conditions. The findings underscore the pivotal role of helminth-derived proteins in immunomodulation, specifically in the axis of cytokine secretion and immune cell polarization. The findings offer new opportunities for treating chronic inflammatory conditions such Crohn's disease.


Subject(s)
Colitis , Helminth Proteins , Animals , Mice , Colitis/therapy , Cytokines/metabolism , Disease Models, Animal , Helminth Proteins/therapeutic use , Helminths , Immune System/metabolism , Immunologic Factors
14.
J Food Sci ; 89(5): 3078-3093, 2024 May.
Article in English | MEDLINE | ID: mdl-38605580

ABSTRACT

Human milk contains a variety of microorganisms that exert benefit for human health. In the current study, we isolated a novel Lactobacillus gasseri strain named Lactobacillus gasseri (L. gasseri) SHMB 0001 from human milk and aimed to evaluate the probiotic characteristics and protective effects on murine colitis of the strain. The results showed that L. gasseri SHMB 0001 possessed promising potential probiotic characteristics, including good tolerance against artificial gastric and intestinal fluids, adhesion to Caco-2 cells, susceptibility to antibiotic, no hemolytic activity, and without signs of toxicity or infection in mice. Administration of L. gasseri SHMB 0001 (1 × 108 CFU per gram of mouse weight per day) reduced weight loss, the disease activity index, and colon shortening in mice during murine colitis conditions. Histopathological analysis revealed that L. gasseri SHMB 0001 treatment attenuated epithelial damage and inflammatory infiltration in the colon. L. gasseri SHMB 0001 treatment increased the expression of colonic occludin and claudin-1 while decreasing the expression of pro-inflammatory cytokine genes. L. gasseri SHMB 0001 modified the composition and structure of the gut microbiota community and partially recovered the Clusters of Orthologous Groups (COG) and Kyoto Encyclopedia of Genes and Genomes (KEGG) metabolic pathways altered by dextran sulfate sodium (DSS). Overall, our results indicated that the human breast milk-derived L. gasseri SHMB 0001 exhibited promising probiotic properties and ameliorative effect on DSS-induced colitis in mice. L. gasseri SHMB 0001 may be applied as a promising probiotic against intestinal inflammation in the future. PRACTICAL APPLICATION: L. gasseri SHMB 0001 isolated from human breast milk showed good tolerance to gastrointestinal environment, safety, and protective effect against DSS-induced mice colitis via enforcing gut barrier, downregulating pro-inflammatory cytokines, and modulating gut microbiota. L. gasseri SHMB 0001 may be a promising probiotic candidate for the treatment of intestinal inflammation.


Subject(s)
Colitis , Dextran Sulfate , Gastrointestinal Microbiome , Lactobacillus gasseri , Milk, Human , Probiotics , Probiotics/pharmacology , Animals , Humans , Mice , Colitis/chemically induced , Colitis/therapy , Colitis/microbiology , Dextran Sulfate/adverse effects , Gastrointestinal Microbiome/drug effects , Caco-2 Cells , Female , Colon/microbiology , Colon/pathology , Colon/metabolism , Cytokines/metabolism , Disease Models, Animal
15.
Stem Cell Res Ther ; 15(1): 69, 2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38454492

ABSTRACT

BACKGROUND: A significant unmet need in inflammatory bowel disease is the lack of anti-fibrotic agents targeting intestinal fibrosis. This study aimed to investigate the anti-fibrogenic properties and mechanisms of the conditioned medium (CM) from human umbilical cord/placenta-derived mesenchymal stem cells (UC/PL-MSC-CM) in a murine intestinal fibrosis model and human primary intestinal myofibroblasts (HIMFs). METHODS: UC/PL-MSC-CM was concentrated 15-fold using a 3 kDa cut-off filter. C57BL/6 mice aged 7 weeks old were randomly assigned to one of four groups: (1) control, (2) dextran sulfate sodium (DSS), (3) DSS + CM (late-phase treatment), and (4) DSS + CM (early-phase treatment). Chronic DSS colitis and intestinal fibrosis was induced by three cycles of DSS administration. One DSS cycle consisted of 7 days of oral DSS administration (1.75%, 2%, and 2.5% DSS), followed by 14 days of drinking water. UC/PL-MSC-CM was intraperitoneally administered in the late phase (from day 50, 10 times) or early phase (from day 29, 10 times) of DSS cycles. HIMFs were treated with TGF-ß1 and co-treated with UC/PL-MSC-CM (10% of culture media) in the cellular model. RESULTS: In the animal study, UC/PL-MSC-CM reduced submucosa/muscularis propria thickness and collagen deposition, which improved intestinal fibrosis in chronic DSS colitis. The UC/PL-MSC-CM significantly reduced the expressions of procollagen1A1 and α-smooth muscle actin, which DSS significantly elevated. The anti-fibrogenic effect was more apparent in the UC-MSC-CM or early-phase treatment model. The UC/PL-MSC-CM reduced procollagen1A1, fibronectin, and α-smooth muscle actin expression in HIMFs in the cellular model. The UC/PL-MSC-CM downregulated fibrogenesis by suppressing RhoA, MRTF-A, and SRF expression. CONCLUSIONS: Human UC/PL-MSC-CM inhibits TGF-ß1-induced fibrogenic activation in HIMFs by blocking the Rho/MRTF/SRF pathway and chronic DSS colitis-induced intestinal fibrosis. Thus, it may be regarded as a novel candidate for stem cell-based therapy of intestinal fibrosis.


Subject(s)
Colitis , Mesenchymal Stem Cells , Humans , Mice , Animals , Transforming Growth Factor beta1/metabolism , Culture Media, Conditioned/pharmacology , Culture Media, Conditioned/metabolism , Actins/metabolism , Mice, Inbred C57BL , Colitis/chemically induced , Colitis/therapy , Colitis/metabolism , Immunologic Factors , Fibrosis , Mesenchymal Stem Cells/metabolism , Umbilical Cord , Dextran Sulfate/toxicity , Disease Models, Animal
16.
Nat Commun ; 15(1): 2769, 2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38553486

ABSTRACT

Multiple neurological disorders are associated with gastrointestinal (GI) symptoms, including autism spectrum disorder (ASD). However, it is unclear whether GI distress itself can modify aspects of behavior. Here, we show that mice that experience repeated colitis have impaired active social engagement, as measured by interactions with a foreign mouse, even though signs of colitis were no longer present. We then tested the hypothesis that individuals with ASD harbor a microbiota that might differentially influence GI health by performing microbiota transplantation studies into male germfree animals, followed by induction of colitis. Animals that harbor a microbiota from ASD individuals have worsened gut phenotypes when compared to animals colonized with microbiotas from familial neurotypical (NT) controls. We identify the enrichment of Blautia species in all familial NT controls and observe an association between elevated abundance of Bacteroides uniformis and reductions in intestinal injury. Oral treatment with either of these microbes reduces colon injury in mice. Finally, provision of a Blautia isolate from a NT control ameliorates gut injury-associated active social engagement in mice. Collectively, our data demonstrate that past intestinal distress is associated with changes in active social behavior in mice that can be ameliorated by supplementation of members of the human microbiota.


Subject(s)
Autism Spectrum Disorder , Colitis , Gastrointestinal Diseases , Microbiota , Humans , Male , Mice , Animals , Autism Spectrum Disorder/therapy , Social Participation , Colitis/therapy , Dietary Supplements
17.
Front Immunol ; 15: 1365457, 2024.
Article in English | MEDLINE | ID: mdl-38529272

ABSTRACT

Background: Inflammatory bowel disease (IBD) greatly affects human quality of life. Mannose has been reported to be used to treat IBD, but the mechanism is currently unknown. Methods: C57/BL mice were used as research subjects, and the mouse acute colitis model was induced using dextran sulfate sodium salt (DSS). After oral administration of mannose, the body weights and disease activity index (DAI) scores of the mice were observed. The colon lengths, histopathological sections, fecal content microbial sequencing, colon epithelial inflammatory genes, and tight junction protein Occludin-1 expression levels were measured. We further used the feces of mice that had been orally administered mannose to perform fecal bacterial transplantation on the mice with DSS-induced colitis and detected the colitis-related indicators. Results: Oral administration of mannose increased body weights and colon lengths and reduced DAI scores in mice with DSS-induced colitis. In addition, it reduced the expression of colon inflammatory genes and the levels of serum inflammatory factors (TNF-α, IL-6, and IL-1ß), further enhancing the expression level of the colonic Occludin-1 protein and alleviating the toxic response of DSS to the intestinal epithelium of the mice. In addition, gut microbial sequencing revealed that mannose increased the abundance and diversity of intestinal flora. Additionally, after using the feces of the mannose-treated mice to perform fecal bacterial transplantation on the mice with DSS-induced colitis, they showed the same phenotype as the mannose-treated mice, and both of them alleviated the intestinal toxic reaction induced by the DSS. It also reduced the expression of intestinal inflammatory genes (TNF-α, IL-6, and IL-1ß) and enhanced the expression level of the colonic Occludin-1 protein. Conclusion: Mannose can treat DSS-induced colitis in mice, possibly by regulating intestinal microorganisms to enhance the intestinal immune barrier function and reduce the intestinal inflammatory response.


Subject(s)
Colitis , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Mice , Humans , Animals , Mannose , Dextran Sulfate/toxicity , Interleukin-6 , Tumor Necrosis Factor-alpha , Occludin/genetics , Quality of Life , Colitis/chemically induced , Colitis/therapy , Colitis/metabolism , Sodium Chloride , Sodium Chloride, Dietary , Body Weight
18.
Am J Physiol Gastrointest Liver Physiol ; 326(5): G607-G621, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38502145

ABSTRACT

Fecal microbiota transplantation (FMT) is a promising therapy for inflammatory bowel disease (IBD) via rectifying gut microbiota. The aim of this study was to identify a mechanism of how specific bacteria-associated immune response contributes to alleviated colitis. Forty donors were divided into high (donor H) and low (donor L) groups according to the diversity and the abundance of Bacteroides and Faecalibacterium by 16S rRNA sequencing. FMT was performed on dextran sulfate sodium (DSS)-induced colitis in mice. Mice with colitis showed significant improvement in intestinal injury and immune imbalance after FMT with group donor H (P < 0.05). Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii were identified as targeted strains in donor feces by real-time PCR and droplet digital PCR. Mice with colitis were treated with mono- or dual-bacterial gavage therapy. Dual-bacterial therapy significantly ameliorated intestinal injury compared with mono-bacterial therapy (P < 0.05). Dual-bacterial therapy increased the M2/M1 macrophage polarization and improved the Th17/Treg imbalance and elevated IL-10 production by Tregs compared with the DSS group (P < 0.05). Metabolomics showed increased abundance of lecithin in the glycerophospholipid metabolism pathway. In conclusion, B. thetaiotaomicron and F. prausnitzii, as the key bacteria in donor feces, alleviate colitis in mice. The mechanism may involve increasing lecithin and regulating IL-10 production of intestinal Tregs.NEW & NOTEWORTHY We demonstrate that donors with high abundance of Bacteroides and Faecalibacterium ameliorate dextran sulfate sodium (DSS)-induced colitis in mice by fecal microbiota transplantation (FMT). The combination therapy of Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii is superior to mono-bacterial therapy in ameliorating colitis in mice, of which mechanism may involve promoting lecithin and inducing IL-10 production of intestinal Tregs.


Subject(s)
Bacteroides thetaiotaomicron , Colitis , Faecalibacterium prausnitzii , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Animals , Colitis/therapy , Colitis/microbiology , Colitis/chemically induced , Colitis/immunology , Mice , Male , Humans , Dextran Sulfate , Mice, Inbred C57BL , Interleukin-10/metabolism , Adult , Female , Feces/microbiology , Disease Models, Animal , Middle Aged
19.
Nutrients ; 16(4)2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38398793

ABSTRACT

Lactobacillus species have been shown to alleviate gut inflammation and oxidative stress. However, the effect of different lactobacilli components on gut inflammation has not been well studied. This study aims to identify the differences in the effect and mechanisms of different forms and components of Limosilactobacillus mucosae (LM) treatment in the alleviation of gut inflammation using a colitis mouse model that is induced by dextran sodium sulfate (DSS). Seventy-two C57BL/6 mice were divided into six groups: control, DSS, live LM+DSS (LM+DSS), heat-killed LM+DSS (HKLM+DSS), LM cell-free supernatant + DSS (LMCS+DSS), and MRS medium + DSS (MRS+DSS). The mice were treated with different forms and components of LM for two weeks before DSS treatment. After that, the mice were sacrificed for an assessment of their levels of inflammatory cytokines, serotonin (5-HT) receptors (HTRs), and tryptophan metabolites. The results showed that, compared to other treatments, LMCS was more effective (p < 0.05) in the alleviation of DSS-induced body weight loss and led to an increase in the disease activity index score. All three forms and components of LM increased (p < 0.05) the levels of indole-3-acetic acid but reduced (p < 0.05) the levels of 5-HT in the colon. HKLM or LMCS reduced (p < 0.05) the percentages of CD3+CD8+ cytotoxic T cells but increased (p < 0.05) the percentages of CD3+CD4+ T helper cells in the spleen. LM or HKLM increased (p < 0.05) abundances of CD4+Foxp3+ regulatory T cells in the spleen. The LM and LMCS treatments reduced (p < 0.05) the expression of the pro-inflammatory cytokines Il6 and Il17a. The mice in the HKLM+DSS group had higher (p < 0.05) mRNA levels of the anti-inflammatory cytokine Il10, the cell differentiation and proliferation markers Lgr5 and Ki67, the 5-HT degradation enzyme Maoa, and HTRs (Htr1a, Htr2a, and Htr2b) in the colon. All three forms and components of LM reduced the phosphorylation of STAT3. The above findings can help to optimize the functionality of probiotics and develop new dietary strategies that aid in the maintenance of a healthy gut.


Subject(s)
Colitis , Serotonin , Animals , Mice , Serotonin/metabolism , Hot Temperature , Mice, Inbred C57BL , Colitis/chemically induced , Colitis/therapy , Lactobacillus/metabolism , Inflammation/metabolism , Cytokines/metabolism , Receptors, Serotonin/metabolism , Immunity , Dextran Sulfate/adverse effects , Disease Models, Animal , Colon/metabolism
20.
Nutrients ; 16(3)2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38337648

ABSTRACT

Exclusive enteral nutrition (EEN) is an established dietary treatment for Crohn's disease (CD) by alleviating inflammation and inducing remission. However, the mechanisms of action of EEN are incompletely understood. As CD is associated with gut microbiome dysbiosis, we investigated the effect of EEN on the microbiome in a rat model of CD-like colitis. The rat model of CD-like colitis was established by an intracolonic instillation of TNBS at 65 mg/kg in 250 µL of 40% ethanol. Sham control rats were instilled with saline. Rats were fed ad libitum with either regular pellet food or EEN treatment with a clear liquid diet (Ensure). Rats were euthanized at 7 days. Fecal pellets were collected from the distal colon for 16S rRNA sequencing analysis of gut microbiota. In addition, colon tissues were taken for histological and molecular analyses in all the groups of rats. EEN administration to TNBS-induced CD rats significantly improved the body weight change, inflammation scores, and disease activity index. The mRNA expression of IL-17A and interferon-γ was significantly increased in the colonic tissue in TNBS rats when fed with regular food. However, EEN treatment significantly attenuated the increase in IL-17A and interferon-γ in TNBS rats. Our 16S rRNA sequencing analysis found that gut microbiota diversity and compositions were significantly altered in TNBS rats, compared to controls. However, EEN treatment improved alpha diversity and increased certain beneficial bacteria such as Lactobacillus and Dubosiella and decreased bacteria such as Bacteroides and Enterorhabdus in CD-like rats, compared to CD-like rats with the regular pellet diet. In conclusion, EEN treatment increases the diversity of gut microbiota and the composition of certain beneficial bacteria. These effects may contribute to the reduced inflammation by EEN in the rat model of CD-like colitis.


Subject(s)
Colitis , Crohn Disease , Gastrointestinal Microbiome , Rats , Animals , Crohn Disease/microbiology , Enteral Nutrition , RNA, Ribosomal, 16S/genetics , Interleukin-17 , Interferon-gamma , Colitis/chemically induced , Colitis/therapy , Bacteria , Inflammation/therapy , Remission Induction
SELECTION OF CITATIONS
SEARCH DETAIL
...