Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 41
Filtrer
1.
Front Bioeng Biotechnol ; 11: 1191162, 2023.
Article de Anglais | MEDLINE | ID: mdl-37288353

RÉSUMÉ

Uric acid disequilibrium is implicated in chronic hyperuricemia-related diseases. Long-term monitoring and lowering of serum uric acid levels may be crucial for diagnosis and effective management of these conditions. However, current strategies are not sufficient for accurate diagnosis and successful long-term management of hyperuricemia. Moreover, drug-based therapeutics can cause side effects in patients. The intestinal tract plays an important role in maintaining healthy serum acid levels. Hence, we investigated the engineered human commensal Escherichia coli as a novel method for diagnosis and long-term management of hyperuricemia. To monitor changes in uric acid concentration in the intestinal lumen, we developed a bioreporter using the uric acid responsive synthetic promoter, pucpro, and uric acid binding Bacillus subtilis PucR protein. Results demonstrated that the bioreporter module in commensal E. coli can detect changes in uric acid concentration in a dose-dependent manner. To eliminate the excess uric acid, we designed a uric acid degradation module, which overexpresses an E. coli uric acid transporter and a B. subtilis urate oxidase. Strains engineered with this module degraded all the uric acid (250 µM) found in the environment within 24 h, which is significantly lower (p < 0.001) compared to wild type E. coli. Finally, we designed an in vitro model using human intestinal cell line, Caco-2, which provided a versatile tool to study the uric acid transport and degradation in an environment mimicking the human intestinal tract. Results showed that engineered commensal E. coli reduced (p < 0.01) the apical uric acid concentration by 40.35% compared to wild type E. coli. This study shows that reprogramming E. coli holds promise as a valid alternative synthetic biology therapy to monitor and maintain healthy serum uric acid levels.

2.
ACS Biomater Sci Eng ; 9(9): 5123-5135, 2023 09 11.
Article de Anglais | MEDLINE | ID: mdl-36399014

RÉSUMÉ

The etiology of inflammatory bowel diseases (IBDs) frequently results in the uncontrolled inflammation of intestinal epithelial linings and the local environment. Here, we hypothesized that interferon-driven immunomodulation could promote anti-inflammatory effects. To test this hypothesis, we engineered probiotic Escherichia coli Nissle 1917 (EcN) to produce and secrete a type III interferon, interferon lambda 1 (IFNL1), in response to nitric oxide (NO), a well-known colorectal inflammation marker. We then validated the anti-inflammatory effects of the engineered EcN strains in two in vitro models: a Caco-2/Jurkat T cell coculture model and a scaffold-based 3D coculture IBD model that comprises intestinal epithelial cells, myofibroblasts, and T cells. The IFNL1-expressing EcN strains upregulated Foxp3 expression in T cells and thereafter reduced the production of pro-inflammatory cytokines such as IL-13 and -33, significantly ameliorating inflammation. The engineered strains also rescued the integrity of the inflamed epithelial cell monolayer, protecting epithelial barrier integrity even under inflammation. In the 3D coculture model, IFNL1-expressing EcN treatment enhanced the population of regulatory T cells and increased anti-inflammatory cytokine IL-10. Taken together, our study showed the anti-inflammatory effects of IFNL1-expressing probiotics in two in vitro IBD models, demonstrating their potential as live biotherapeutics for IBD immunotherapy.


Sujet(s)
Maladies inflammatoires intestinales , Probiotiques , Humains , Cellules Caco-2 , Interféron lambda , Escherichia coli , Maladies inflammatoires intestinales/traitement médicamenteux , Cytokines/métabolisme , Cytokines/usage thérapeutique , Inflammation , Anti-inflammatoires/métabolisme , Anti-inflammatoires/usage thérapeutique , Probiotiques/pharmacologie , Probiotiques/usage thérapeutique
3.
Nat Commun ; 13(1): 3834, 2022 07 04.
Article de Anglais | MEDLINE | ID: mdl-35787625

RÉSUMÉ

Clostridioides difficile infection (CDI) results in significant morbidity and mortality in hospitalised patients. The pathogenesis of CDI is intrinsically related to the ability of C. difficile to shuffle between active vegetative cells and dormant endospores through the processes of germination and sporulation. Here, we hypothesise that dysregulation of microbiome-mediated bile salt metabolism contributes to CDI and that its alleviation can limit the pathogenesis of CDI. We engineer a genetic circuit harbouring a genetically encoded sensor, amplifier and actuator in probiotics to restore intestinal bile salt metabolism in response to antibiotic-induced microbiome dysbiosis. We demonstrate that the engineered probiotics limited the germination of endospores and the growth of vegetative cells of C. difficile in vitro and further significantly reduced CDI in model mice, as evidenced by a 100% survival rate and improved clinical outcomes. Our work presents an antimicrobial strategy that harnesses the host-pathogen microenvironment as the intervention target to limit the pathogenesis of infection.


Sujet(s)
Clostridioides difficile , Infections à Clostridium , Probiotiques , Animaux , Antibactériens/pharmacologie , Acides et sels biliaires/métabolisme , Clostridioides difficile/génétique , Infections à Clostridium/traitement médicamenteux , Infections à Clostridium/prévention et contrôle , Souris , Spores bactériens/métabolisme
4.
Infect Immun ; 90(6): e0004122, 2022 06 16.
Article de Anglais | MEDLINE | ID: mdl-35579465

RÉSUMÉ

Secretory IgA (SIgA) is the most abundant antibody type in intestinal secretions where it contributes to safeguarding the epithelium from invasive pathogens like the Gram-negative bacterium, Salmonella enterica serovar Typhimurium (STm). For example, we recently reported that passive oral administration of the recombinant monoclonal SIgA antibody, Sal4, to mice promotes STm agglutination in the intestinal lumen and restricts bacterial invasion of Peyer's patch tissues. In this report, we sought to recapitulate Sal4-mediated protection against STm in human Enteroids and human intestinal organoids (HIOs) as models to decipher the molecular mechanisms by which antibodies function in mucosal immunity in the human gastrointestinal tract. We confirm that Enteroids and HIO-derived monolayers are permissive to STm infection, dependent on HilD, the master transcriptional regulator of the SPI-I type three secretion system (T3SS). Stimulation of M-like cells in both Enteroids and HIOs by the addition of RANKL further enhanced STm invasion. The apical addition of Sal4 mouse IgA, as well as recombinant human Sal4 dimeric IgA (dIgA) and SIgA resulted a dose-dependent reduction in bacterial invasion. Moreover, basolateral application of Sal4 dIgA to Enteroid and HIO monolayers gave rise to SIgA in the apical compartment via a pathway dependent on expression of the polymeric immunoglobulin receptor (pIgR). The resulting Sal4 SIgA was sufficient to reduce STm invasion of Enteroid and HIO epithelial cell monolayers by ~20-fold. Recombinant Sal4 IgG was also transported in the Enteroid and HIOs, but to a lesser degree and via a pathway dependent on the neonatal Fc receptor (FCGRT). The models described lay the foundation for future studies into detailed mechanisms of IgA and IgG protection against STm and other pathogens.


Sujet(s)
Immunoglobuline A , Organoïdes , Animaux , Humains , Immunoglobuline A/métabolisme , Immunoglobuline A sécrétoire , Immunoglobuline G/métabolisme , Muqueuse intestinale/métabolisme , Souris , Organoïdes/métabolisme , Salmonella typhimurium , Transcytose
5.
EMBO Mol Med ; 12(4): e11621, 2020 04 07.
Article de Anglais | MEDLINE | ID: mdl-32153125

RÉSUMÉ

The human PXR (pregnane X receptor), a master regulator of drug metabolism, has essential roles in intestinal homeostasis and abrogating inflammation. Existing PXR ligands have substantial off-target toxicity. Based on prior work that established microbial (indole) metabolites as PXR ligands, we proposed microbial metabolite mimicry as a novel strategy for drug discovery that allows exploiting previously unexplored parts of chemical space. Here, we report functionalized indole derivatives as first-in-class non-cytotoxic PXR agonists as a proof of concept for microbial metabolite mimicry. The lead compound, FKK6 (Felix Kopp Kortagere 6), binds directly to PXR protein in solution, induces PXR-specific target gene expression in cells, human organoids, and mice. FKK6 significantly represses pro-inflammatory cytokine production cells and abrogates inflammation in mice expressing the human PXR gene. The development of FKK6 demonstrates for the first time that microbial metabolite mimicry is a viable strategy for drug discovery and opens the door to underexploited regions of chemical space.


Sujet(s)
Mimétisme moléculaire , Récepteur du prégnane X/composition chimique , Animaux , Cellules cultivées , Cytokines , Humains , Inflammation , Intestins , Ligands , Souris , Organoïdes
6.
Tissue Eng Part A ; 25(17-18): 1225-1241, 2019 09.
Article de Anglais | MEDLINE | ID: mdl-30652526

RÉSUMÉ

IMPACT STATEMENT: This study is significant because it demonstrates an attempt to design a scaffold specifically for small intestine using a novel fabrication method, resulting in an architecture that resembles intestinal villi. In addition, we use the versatile polymer poly(glycerol sebacate) (PGS) for artificial intestine, which has tunable mechanical and degradation properties that can be harnessed for further fine-tuning of scaffold design. Moreover, the utilization of PGS allows for future development of growth factor and drug delivery from the scaffolds to promote artificial intestine formation.


Sujet(s)
Intestins/cytologie , Ingénierie tissulaire/méthodes , Structures d'échafaudage tissulaires/composition chimique , Animaux , Matériaux biocompatibles/composition chimique , Décanoate/composition chimique , Glycérol/analogues et dérivés , Glycérol/composition chimique , Immunohistochimie , Mâle , Souris , Souris de lignée C57BL , Microscopie électronique à balayage , Polymères/composition chimique , Suidae
7.
Bioeng Transl Med ; 3(3): 209-221, 2018 Sep.
Article de Anglais | MEDLINE | ID: mdl-30377661

RÉSUMÉ

For therapies targeting diseases of the gastrointestinal tract, we and others envision probiotic bacteria that synthesize and excrete biotherapeutics at disease sites. Toward this goal, we have engineered commensal E. coli that selectively synthesize and secrete a model biotherapeutic in the presence of nitric oxide (NO), an intestinal biomarker for Crohn's disease (CD). This is accomplished by co-expressing the pore forming protein TolAIII with the biologic, granulocyte macrophage-colony stimulating factor (GM-CSF). We have additionally engineered these bacteria to accumulate at sites of elevated NO by engineering their motility circuits and controlling pseudotaxis. Importantly, because we have focused on in vitro test beds, motility and biotherapeutics production are spatiotemporally characterized. Together, the targeted recognition, synthesis, and biomolecule delivery comprises a "smart" probiotics platform that may have utility in the treatment of CD. Further, this platform could be modified to accommodate other pursuits by swapping the promoter and therapeutic gene to reflect other disease biomarkers and treatments, respectively.

8.
Tissue Eng Part C Methods ; 24(6): 331-345, 2018 06.
Article de Anglais | MEDLINE | ID: mdl-29638197

RÉSUMÉ

Short bowel syndrome (SBS) is a major cause of morbidity and mortality in the pediatric population, for which treatment options are limited. To develop novel approaches for the treatment of SBS, we now focus on the development of a tissue-engineered intestine (also known as an "artificial intestine"), in which intestinal stem cells are cultured onto an absorbable bioscaffold, followed by implantation into the host. To enhance the translational potential of these preclinical studies, we have developed three clinically relevant models in neonatal piglets, which approximate the size of the human infant and were evaluated after implantation and establishment of intestinal continuity over the long term. The models included (1) a staged, multioperation approach; (2) a single operation with a de-functionalized loop of small intestine; and (3) a single operation with an intestinal bypass. The first model had complications related to multiple operations in a short time period, including surgical site infections and wound hernias. The second model avoided wound complications, but was associated with high ostomy output, local skin breakdown, and systemic dehydration with associated electrolyte imbalances. The third model was the most effective, although resulted in stoma prolapse. In summary, we have now developed and evaluated three operative methods for the long-term evaluation of the artificial intestine in the piglet, and conclude that a single operation with a de-functionalized loop of small intestine may be an optimal approach for evaluation over the long term.


Sujet(s)
Modèles animaux de maladie humaine , Intestin grêle/chirurgie , Intestin grêle/transplantation , Infection de plaie opératoire/thérapie , Ingénierie tissulaire , Animaux , Animaux nouveau-nés , Intestin grêle/anatomopathologie , Suidae
9.
Pediatr Res ; 83(1-2): 249-257, 2018 01.
Article de Anglais | MEDLINE | ID: mdl-28937976

RÉSUMÉ

Short bowel syndrome is a major cause of morbidity and mortality in children. Despite decades of experience in the management of short bowel syndrome, current therapy is primarily supportive. Definitive treatment often requires intestinal transplantation, which is associated with significant morbidity and mortality. In order to develop novel approaches to the treatment of short bowel syndrome, we and others have focused on the development of an artificial intestine, by placing intestinal stem cells on a bioscaffold that has an absorptive surface resembling native intestine, and taking advantage of neovascularization to develop a blood supply. This review will explore recent advances in biomaterials, vascularization, and progress toward development of a functional epithelium and mesenchymal niche, highlighting both success and ongoing challenges in the field.


Sujet(s)
Intestin grêle/chirurgie , Syndrome de l'intestin court/chirurgie , Ingénierie tissulaire , Animaux , Matériaux biocompatibles/composition chimique , Prolifération cellulaire , Enfant , Système nerveux entérique/physiologie , Humains , Souris , Péristaltisme , Polymères/composition chimique , Cellules souches/cytologie , Structures d'échafaudage tissulaires/composition chimique
10.
Parasit Vectors ; 10(1): 606, 2017 Dec 15.
Article de Anglais | MEDLINE | ID: mdl-29246169

RÉSUMÉ

BACKGROUND: Human hookworm larvae arrest development until they enter an appropriate host. This makes it difficult to access the larvae for studying larval development or host-parasite interactions. While there are in vivo and in vitro animal models of human hookworm infection, there is currently no human, in vitro model. While animal models have provided much insight into hookworm biology, there are limitations to how closely this can replicate human infection. Therefore, we have developed a human, in vitro model of the initial phase of hookworm infection using intestinal epithelial cell culture. RESULTS: Co-culture of the human hookworm Ancylostoma ceylanicum with the mucus-secreting, human intestinal epithelial cell line HT-29-MTX resulted in activation of infective third-stage larvae, as measured by resumption of feeding. Larvae were maximally activated by direct contact with fully differentiated HT-29-MTX intestinal epithelial cells. HT-29-MTX cells treated with A. ceylanicum larvae showed differential gene expression of several immunity-related genes. CONCLUSIONS: Co-culture with HT-29-MTX can be used to activate A. ceylanicum larvae. This provides an opportunity to study the interaction of activated larvae with the human intestinal epithelium.


Sujet(s)
Ancylostoma/croissance et développement , Cellules épithéliales/parasitologie , Interactions hôte-pathogène , Animaux , Techniques de coculture , Cellules HT29 , Humains , Larve/croissance et développement , Modèles biologiques
11.
Sci Rep ; 7(1): 12515, 2017 10 02.
Article de Anglais | MEDLINE | ID: mdl-28970586

RÉSUMÉ

The development of in vitro artificial small intestines that realistically mimic in vivo systems will enable vast improvement of our understanding of the human gut and its impact on human health. Synthetic in vitro models can control specific parameters, including (but not limited to) cell types, fluid flow, nutrient profiles and gaseous exchange. They are also "open" systems, enabling access to chemical and physiological information. In this work, we demonstrate the importance of gut surface topography and fluid flow dynamics which are shown to impact epithelial cell growth, proliferation and intestinal cell function. We have constructed a small intestinal bioreactor using 3-D printing and polymeric scaffolds that mimic the 3-D topography of the intestine and its fluid flow. Our results indicate that TEER measurements, which are typically high in static 2-D Transwell apparatuses, is lower in the presence of liquid sheer and 3-D topography compared to a flat scaffold and static conditions. There was also increased cell proliferation and discovered localized regions of elevated apoptosis, specifically at the tips of the villi, where there is highest sheer. Similarly, glucose was actively transported (as opposed to passive) and at higher rates under flow.


Sujet(s)
Organes artificiels , Muqueuse intestinale/croissance et développement , Intestin grêle/croissance et développement , Impression tridimensionnelle , Biomimétique , Bioréacteurs , Cellules Caco-2 , Prolifération cellulaire/génétique , Cellules épithéliales/composition chimique , Humains , Muqueuse intestinale/composition chimique , Intestin grêle/composition chimique , Ingénierie tissulaire , Structures d'échafaudage tissulaires/tendances
12.
Nat Commun ; 8: 15028, 2017 04 11.
Article de Anglais | MEDLINE | ID: mdl-28398304

RÉSUMÉ

Bacteria can be genetically engineered to kill specific pathogens or inhibit their virulence. We previously developed a synthetic genetic system that allows a laboratory strain of Escherichia coli to sense and kill Pseudomonas aeruginosa in vitro. Here, we generate a modified version of the system, including a gene encoding an anti-biofilm enzyme, and use the probiotic strain Escherichia coli Nissle 1917 as host. The engineered probiotic shows in vivo prophylactic and therapeutic activity against P. aeruginosa during gut infection in two animal models (Caenorhabditis elegans and mice). These findings support the further development of engineered microorganisms with potential prophylactic and therapeutic activities against gut infections.


Sujet(s)
Escherichia coli/génétique , Gastroentérite/thérapie , Micro-organismes génétiquement modifiés , Probiotiques/usage thérapeutique , Infections à Pseudomonas/thérapie , Pseudomonas aeruginosa/pathogénicité , Animaux , Caenorhabditis elegans , Modèles animaux de maladie humaine , Femelle , Gastroentérite/microbiologie , Génie génétique/méthodes , Souris , Souris de lignée ICR , Infections à Pseudomonas/microbiologie , Virulence
13.
Free Radic Biol Med ; 105: 110-131, 2017 04.
Article de Anglais | MEDLINE | ID: mdl-28040473

RÉSUMÉ

The intestine is the site of digestion and forms a critical interface between the host and the outside world. This interface is composed of host epithelium and a complex microbiota which is "connected" through an extensive web of chemical and biological interactions that determine the balance between health and disease for the host. This biology and the associated chemical dialogues occur within a context of a steep oxygen gradient that provides the driving force for a variety of reduction and oxidation (redox) reactions. While some redox couples (e.g., catecholics) can spontaneously exchange electrons, many others are kinetically "insulated" (e.g., biothiols) allowing the biology to set and control their redox states far from equilibrium. It is well known that within cells, such non-equilibrated redox couples are poised to transfer electrons to perform reactions essential to immune defense (e.g., transfer from NADH to O2 for reactive oxygen species, ROS, generation) and protection from such oxidative stresses (e.g., glutathione-based reduction of ROS). More recently, it has been recognized that some of these redox-active species (e.g., H2O2) cross membranes and diffuse into the extracellular environment including lumen to transmit redox information that is received by atomically-specific receptors (e.g., cysteine-based sulfur switches) that regulate biological functions. Thus, redox has emerged as an important modality in the chemical signaling that occurs in the intestine and there have been emerging efforts to develop the experimental tools needed to probe this modality. We suggest that electrochemistry provides a unique tool to experimentally probe redox interactions at a systems level. Importantly, electrochemistry offers the potential to enlist the extensive theories established in signal processing in an effort to "reverse engineer" the molecular communication occurring in this complex biological system. Here, we review our efforts to develop this electrochemical tool for in vitro redox-probing.


Sujet(s)
Oxygène/métabolisme , Animaux , Dysbiose/métabolisme , Dysbiose/microbiologie , Électrochimie , Microbiome gastro-intestinal , Tube digestif/métabolisme , Tube digestif/microbiologie , Homéostasie , Humains , Interactions microbiennes , Oxydoréduction , Stress oxydatif , Espèces réactives de l'oxygène/métabolisme , Transduction du signal
14.
Curr Opin Organ Transplant ; 21(2): 178-85, 2016 Apr.
Article de Anglais | MEDLINE | ID: mdl-26867049

RÉSUMÉ

PURPOSE OF REVIEW: This article discusses the current state of the art in artificial intestine generation in the treatment of short bowel syndrome. RECENT FINDINGS: Short bowel syndrome defines the condition in which patients lack sufficient intestinal length to allow for adequate absorption of nutrition and fluids, and thus need parenteral support. Advances toward the development of an artificial intestine have improved dramatically since the first attempts in the 1980s, and the last decade has seen significant advances in understanding the intestinal stem cell niche, the growth of complex primary intestinal stem cells in culture, and fabrication of the biomaterials that can support the growth and differentiation of these stem cells. There has also been recent progress in understanding the role of the microbiota and the immune cells on the growth of intestinal cultures on scaffolds in animal models. Despite recent progress, there is much work to be done before the development of a functional artificial intestine for short bowel syndrome is successfully achieved. SUMMARY: Continued concerted efforts by cell biologists, bioengineers, and clinician-scientists will be required for the development of an artificial intestine as a clinical treatment modality for short bowel syndrome.


Sujet(s)
Intestins/transplantation , Syndrome de l'intestin court/chirurgie , Animaux , Techniques de culture cellulaire , Humains , Intestins/vascularisation , Ingénierie tissulaire , Résultat thérapeutique
15.
Methods Mol Biol ; 1350: 469-76, 2016.
Article de Anglais | MEDLINE | ID: mdl-26820874

RÉSUMÉ

A technique is described for synthesizing and transfecting double stranded RNA (dsRNA) for RNA interference (RNAi) in Sf-21 cell culture. Transfection with dsRNA only requires an hour and the cells usually recover within 12 h. Suggestions for designing dsRNA are included in the methods. Furthermore, websites are provided for rapid and effective dsRNA design. Three kits are essential for using the described methods: RNAqueous®-4PCR, Megascript™ T7 kit, and the Superscript™ III kit from Life Technologies, Inc.


Sujet(s)
Techniques génétiques , Insectes/cytologie , Interférence par ARN , Animaux , Lignée cellulaire , ARN double brin/génétique , Transfection
16.
Regen Med ; 11(1): 45-61, 2016 Jan.
Article de Anglais | MEDLINE | ID: mdl-26395928

RÉSUMÉ

AIMS: To investigate the growth and differentiation of intestinal stem cells on a novel tubular scaffold in vitro and in vivo. MATERIALS & METHODS: Intestinal progenitor cells from mice or humans were cultured with myofibroblasts, macrophages and/or bacteria, and evaluated in mice via omental implantation. Mucosal regeneration was evaluated in dogs after rectal mucosectomy followed by scaffold implantation. RESULTS: Intestinal progenitor cells differentiated into crypt-villi structures on the scaffold. Differentiation and scaffold coverage was enhanced by coculture with myofibroblasts, macrophages and probiotic bacteria, while the implanted scaffolds enhanced mucosal regeneration in the dog rectum. CONCLUSION: Intestinal stem cell growth and differentiation on a novel tubular scaffold is enhanced through addition of cellular and microbial components, as validated in mice and dogs.


Sujet(s)
Différenciation cellulaire , Intestin grêle/cytologie , Cellules souches/cytologie , Structures d'échafaudage tissulaires/composition chimique , Animaux , Vaisseaux sanguins/anatomopathologie , Prolifération cellulaire , Techniques de coculture , Modèles animaux de maladie humaine , Chiens , Escherichia coli/physiologie , Inflammation/anatomopathologie , Acide lactique/composition chimique , Lactobacillus/physiologie , Souris de lignée C57BL , Microvillosités/ultrastructure , Acide polyglycolique/composition chimique , Copolymère d'acide poly(lactique-co-glycolique) , Niche de cellules souches , Transplantation de cellules souches
17.
Biotechnol Bioeng ; 113(3): 492-500, 2016 Mar.
Article de Anglais | MEDLINE | ID: mdl-26370067

RÉSUMÉ

Intestinal inflammation has been implicated in a number of diseases, including diabetes, Crohn's disease, and irritable bowel syndrome. Important components of inflammation are interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α), which are elevated both on the luminal and submucosal sides of the intestinal epithelial barrier in several diseases. Here, we developed a novel Escherichia coli based detection system for IFN-γ and TNF-α comprised of a chimeric protein and a simple signal transduction construct, which could be deployed on the luminal side of the intestine. OmpA of E. coli was engineered to detect IFN-γ or TNF-α through the replacement of extracellular loops with peptide fragments from OprF of P. aeruginosa. OmpA/OprF chimeras were developed, capable of binding IFN-γ or TNF-α. The specific peptide fragments that bind IFN-γ were identified. IFN-γ or TNF-α binding the OmpA/OprF chimera induced the pspA promoter, driving ß-galactosidase production. The OmpA/OprF chimera had a detection limit of 300 pM for IFN-γ and 150 pM for TNF-α. This work will further the development of bacteria based therapeutics for the treatment of inflammatory diseases of the gut.


Sujet(s)
Protéines de la membrane externe bactérienne/métabolisme , Protéines bactériennes/métabolisme , Escherichia coli/métabolisme , Interféron gamma/métabolisme , Récepteurs artificiels/métabolisme , Facteur de nécrose tumorale alpha/métabolisme , Protéines de la membrane externe bactérienne/génétique , Protéines bactériennes/génétique , Escherichia coli/génétique , Récepteurs artificiels/génétique , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme
18.
Diabetes ; 64(5): 1794-803, 2015 May.
Article de Anglais | MEDLINE | ID: mdl-25626737

RÉSUMÉ

The inactive full-length form of GLP-1(1-37) stimulates conversion of both rat and human intestinal epithelial cells into insulin-secreting cells. We investigated whether oral administration of human commensal bacteria engineered to secrete GLP-1(1-37) could ameliorate hyperglycemia in a rat model of diabetes by reprogramming intestinal cells into glucose-responsive insulin-secreting cells. Diabetic rats were fed daily with human lactobacilli engineered to secrete GLP-1(1-37). Diabetic rats fed GLP-1-secreting bacteria showed significant increases in insulin levels and, additionally, were significantly more glucose tolerant than those fed the parent bacterial strain. These rats developed insulin-producing cells within the upper intestine in numbers sufficient to replace ∼25-33% of the insulin capacity of nondiabetic healthy rats. Intestinal tissues in rats with reprogrammed cells expressed MafA, PDX-1, and FoxA2. HNF-6 expression was observed only in crypt epithelia expressing insulin and not in epithelia located higher on the villous axis. Staining for other cell markers in rats treated with GLP-1(1-37)-secreting bacteria suggested that normal function was not inhibited by the close physical proximity of reprogrammed cells. These results provide evidence of the potential for a safe and effective nonabsorbed oral treatment for diabetes and support the concept of engineered commensal bacterial signaling to mediate enteric cell function in vivo.


Sujet(s)
Diabète/thérapie , Cellules épithéliales/cytologie , Génie génétique , Glucose/pharmacologie , Insuline/métabolisme , Lactobacillus/physiologie , Animaux , Lignée cellulaire , Reprogrammation cellulaire , Diabète expérimental/thérapie , Cellules épithéliales/métabolisme , Femelle , Régulation de l'expression des gènes , Glucagon-like peptide 1/génétique , Glucagon-like peptide 1/métabolisme , Humains , Sécrétion d'insuline , Muqueuse intestinale/cytologie , Lactobacillus/génétique , Rats
19.
J Agric Food Chem ; 62(40): 9760-8, 2014 Oct 08.
Article de Anglais | MEDLINE | ID: mdl-25265934

RÉSUMÉ

There are many unresolved questions concerning the health benefits of dietary antioxidants due in part to the complexity of the materials and mechanisms of action. We applied a new electrochemical method and report new observations for one of the richest sources of dietary antioxidants. We observed that the insoluble fraction of clove is redox-active and can be rapidly and repeatedly switched between oxidized and reduced states. Also, the radical scavenging antioxidant properties of insoluble clove are largely independent of this reversible redox activity, which is similar to observations made with the natural phenolic melanin. In contrast to melanin, insoluble clove was observed to have little pro-oxidant activity (as measured by H2O2 generation) irrelevant to whether it was poised in an oxidized or reduced state. These results suggest that dietary antioxidants, even when insoluble and nonabsorbed, can undergo important redox interactions in the intestinal tract.


Sujet(s)
Antioxydants/analyse , Électrochimie/méthodes , Syzygium/composition chimique , Antioxydants/composition chimique , Antioxydants/pharmacologie , Chitosane/composition chimique , Peroxyde d'hydrogène/métabolisme , Oxydoréduction , Solubilité
20.
Drug Discov Today ; 19(10): 1587-94, 2014 Oct.
Article de Anglais | MEDLINE | ID: mdl-24853950

RÉSUMÉ

In vitro human small intestine models play a crucial part in preclinical drug development. Although conventional 2D systems possess many advantages, such as facile accessibility and high-throughput capability, they can also provide misleading results due to their relatively poor recapitulation of in vivo physiology. Significant progress has recently been made in developing 3D human small intestine models, suggesting that more-reliable preclinical results could be obtained by recreating the 3D intestinal microenvironment in vitro. Although there are still many challenges, 3D human small intestine models have the potential to facilitate drug screening and drug development.


Sujet(s)
Intestin grêle/métabolisme , Modèles biologiques , Animaux , Évaluation préclinique de médicament , Effets secondaires indésirables des médicaments , Humains , Pharmacocinétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE