1
|
Cosme D, Soares-da-Silva P, Magro F. Effect of Toll-like receptor-2, -4, -5, -7, and NOD2 stimulation on potassium channel conductance in intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2022; 323:G410-G419. [PMID: 36040119 DOI: 10.1152/ajpgi.00139.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Disproportionate activation of pattern recognition receptors plays a role in inflammatory bowel disease (IBD) pathophysiology. Diarrhea is a hallmark symptom of IBD, resulting at least in part from an electrolyte imbalance that may be caused by changes in potassium channel activity. We evaluated the impact of Toll-like receptors (TLRs) and nucleotide-binding oligomerization domain 2 (NOD2) stimulation on potassium conductance of the basolateral membrane in human intestinal epithelial cells (IECs) and the role of potassium channels through electrophysiological assays under short-circuit current in Ussing chambers. TLRs and NOD2 were stimulated using specific agonists, and potassium channels were selectively blocked using triarylmethane-34 (TRAM-34), adenylyl-imidodiphosphate (AMP-PNP), and BaCl2. Potassium conductance of the basolateral membrane decreased upon activation of TLR2, TLR4, and TLR7 in T84 cells (means ± SE, -11.2 ± 4.5, -40.4 ± 7.2, and -19.4 ± 5.9, respectively) and in Caco-2 cells (-13.1 ± 5.7, -55.7 ± 7.4, and -29.1 ± 7.2, respectively). In contrast, activation of TLR5 and NOD2 increased basolateral potassium conductance, both in T84 cells (18.0 ± 4.1 and 18.4 ± 2.8, respectively) and in Caco-2 cells (21.2 ± 8.4 and 16.0 ± 3.6, respectively). TRAM-34 and AMP-PNP induced a decrease in basolateral potassium conductance upon TLR4 stimulation in both cell lines. Both KCa3.1- and Kir6-channels appear to be important mediators of this effect in IECs and could be potential targets for therapeutic agent development.NEW & NOTEWORTHY This study highlights that PRRs stimulation directly influences K+-channel conductance in IECs. TLR-2, -4, -7 stimulation decreased K+ conductance, whereas TLR5 and NOD2 stimulation had the opposite effect, leading to an increase of it instead. This study reports for the first time that KCa3.1- and Kir6-channels play a role in K+ transport pathways triggered by TLR4 stimulation. These findings suggest that KCa3.1- and Kir6-channels modulation may be a potential target for new therapeutic agents in IBD.
Collapse
Affiliation(s)
- Dina Cosme
- Unit of Pharmacology and Therapeutics, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal.,MedInUP, Center for Drug Discovery and Innovative Medicines, Porto, Portugal
| | - Patrício Soares-da-Silva
- Unit of Pharmacology and Therapeutics, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal.,MedInUP, Center for Drug Discovery and Innovative Medicines, Porto, Portugal
| | - Fernando Magro
- Unit of Pharmacology and Therapeutics, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal.,Department of Gastroenterology, São João Hospital University Centre, Porto, Portugal.,Center for Health Technology and Services Research, Porto, Portugal.,Clinical Pharmacology Unit, São João Hospital University Centre, Porto, Portugal.,Portuguese Inflammatory Bowel Disease Group, Porto, Portugal
| |
Collapse
|
2
|
Abstract
Inflammatory bowel disease (IBD) as a chronic inflammation in colon and small intestine has two subtypes: ulcerative colitis (UC) and Crohn's disease (CD). Genome studies have shown that UC and CD are related to microRNAs (miRNAs) expression in addition to environmental factors. This article reviews important researches that have recently been done on miRNAs roles in CD and UC disease. First, miRNA is introduced and its biogenesis and function are discussed. Afterward, roles of miRNAs in inflammatory processes involved in IBD are showed. Finally, this review proposes some circulating and tissue-specific miRNAs, which are useful for CD and UC fast diagnosis and grade prediction. As a conclusion, miRNAs are efficient diagnostic molecules especially in IBD subtypes discrimination and can be used by microarray and real time PCR methods for disease detection and classification.
Collapse
|
3
|
Involvement of eIF2α in halofuginone-driven inhibition of TGF-β1-induced EMT. J Biosci 2020. [DOI: 10.1007/s12038-020-00042-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
4
|
Volz NB, Hanna DL, Stintzing S, Zhang W, Yang D, Cao S, Ning Y, Matsusaka S, Sunakawa Y, Berger MD, Cremolini C, Loupakis F, Falcone A, Lenz HJ. Polymorphisms within Immune Regulatory Pathways Predict Cetuximab Efficacy and Survival in Metastatic Colorectal Cancer Patients. Cancers (Basel) 2020; 12:2947. [PMID: 33065994 PMCID: PMC7601940 DOI: 10.3390/cancers12102947] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 10/03/2020] [Accepted: 10/06/2020] [Indexed: 12/19/2022] Open
Abstract
Cetuximab, an IgG1 EGFR-directed antibody, promotes antibody-dependent cell-mediated cytotoxicity. We hypothesized that single-nucleotide polymorphisms (SNPs) in immune regulatory pathways may predict outcomes in patients with metastatic colorectal cancer treated with cetuximab-based regimens. A total of 924 patients were included: 105 received cetuximab in IMCL-0144 and cetuximab/irinotecan in GONO-ASL608LIOM01 (training cohort), 225 FOLFIRI/cetuximab in FIRE-3 (validation cohort 1), 74 oxaliplatin/cetuximab regimens in JACCRO CC-05/06 (validation cohort 2), and 520 FOLFIRI/bevacizumab in FIRE-3 and TRIBE (control cohorts). Twelve SNPs in five genes (IDO1; PD-L1; PD-1; CTLA-4; CD24) were evaluated by PCR-based direct sequencing. We analyzed associations between genotype and clinical outcomes. In the training cohort; patients with the CD24 rs52812045 A/A genotype had a significantly shorter median PFS and OS than those with the G/G genotype (PFS 1.3 vs. 3.6 months; OS 2.3 vs. 7.8 months) in univariate (PFS HR 3.62; p = 0.001; OS HR 3.27; p = 0.0004) and multivariate (PFS HR 3.18; p = 0.009; OS HR 4.93; p = 0.001) analyses. Similarly; any A allele carriers in the JACCRO validation cohort had a significantly shorter PFS than G/G carriers (9.2 vs. 11.8 months; univariate HR 1.90; p = 0.011; multivariate HR 2.12; p = 0.018). These associations were not demonstrated in the control cohorts. CD24 genetic variants may help select patients with metastatic colorectal cancer most likely to benefit from cetuximab-based therapy.
Collapse
Affiliation(s)
- Nico B. Volz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA; (N.B.V.); (D.L.H.); (S.S.); (W.Z.); (Y.N.); (S.M.); (Y.S.); (M.D.B.)
- Department of Emergency Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Diana L. Hanna
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA; (N.B.V.); (D.L.H.); (S.S.); (W.Z.); (Y.N.); (S.M.); (Y.S.); (M.D.B.)
| | - Sebastian Stintzing
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA; (N.B.V.); (D.L.H.); (S.S.); (W.Z.); (Y.N.); (S.M.); (Y.S.); (M.D.B.)
- Department of Medicine III, University Hospital LMU Munich, 80539 Munich, Germany
| | - Wu Zhang
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA; (N.B.V.); (D.L.H.); (S.S.); (W.Z.); (Y.N.); (S.M.); (Y.S.); (M.D.B.)
| | - Dongyun Yang
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA; (D.Y.); (S.C.)
| | - Shu Cao
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA; (D.Y.); (S.C.)
| | - Yan Ning
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA; (N.B.V.); (D.L.H.); (S.S.); (W.Z.); (Y.N.); (S.M.); (Y.S.); (M.D.B.)
| | - Satoshi Matsusaka
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA; (N.B.V.); (D.L.H.); (S.S.); (W.Z.); (Y.N.); (S.M.); (Y.S.); (M.D.B.)
| | - Yu Sunakawa
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA; (N.B.V.); (D.L.H.); (S.S.); (W.Z.); (Y.N.); (S.M.); (Y.S.); (M.D.B.)
| | - Martin D. Berger
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA; (N.B.V.); (D.L.H.); (S.S.); (W.Z.); (Y.N.); (S.M.); (Y.S.); (M.D.B.)
| | - Chiara Cremolini
- U.O. Oncologia Medica 2—Aziendo Ospedaliero-Universitaria Pisana, 56126 Pisa, Italy; (C.C.); (F.L.); (A.F.)
| | - Fotios Loupakis
- U.O. Oncologia Medica 2—Aziendo Ospedaliero-Universitaria Pisana, 56126 Pisa, Italy; (C.C.); (F.L.); (A.F.)
| | - Alfredo Falcone
- U.O. Oncologia Medica 2—Aziendo Ospedaliero-Universitaria Pisana, 56126 Pisa, Italy; (C.C.); (F.L.); (A.F.)
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA 90033, USA; (N.B.V.); (D.L.H.); (S.S.); (W.Z.); (Y.N.); (S.M.); (Y.S.); (M.D.B.)
| |
Collapse
|
5
|
Leber A, Hontecillas R, Zoccoli-Rodriguez V, Bienert C, Chauhan J, Bassaganya-Riera J. Activation of NLRX1 by NX-13 Alleviates Inflammatory Bowel Disease through Immunometabolic Mechanisms in CD4 + T Cells. THE JOURNAL OF IMMUNOLOGY 2019; 203:3407-3415. [PMID: 31694910 DOI: 10.4049/jimmunol.1900364] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 10/08/2019] [Indexed: 02/06/2023]
Abstract
Inflammatory bowel disease (IBD) is a complex autoimmune disease with dysfunction in pattern-recognition responses, including within the NLR family. Nucleotide-binding oligomerization domain, leucine rich repeat containing X1 (NLRX1) is a unique NLR with regulatory and anti-inflammatory functions resulting in protection from IBD in mouse models. NX-13 is an orally active, gut-restricted novel drug candidate that selectively targets and activates the NLRX1 pathway locally in the gut. In vitro and in vivo efficacy of NLRX1 activation by NX-13 was examined. Oral treatment with NX-13 alleviates disease severity, colonic leukocytic infiltration, and cytokine markers of inflammation in three mouse models of IBD (dextran sulfate sodium, Mdr1a-/-, and CD45RBhi adoptive transfer). Treatment of naive CD4+ T cells with NX-13 in vitro decreases differentiation into Th1 and Th17 subsets with increased oxidative phosphorylation and decreased NF-κB activation and reactive oxygen species. With stimulation by PMA/ionomycin, TNF-α, or H2O2, PBMCs from ulcerative colitis patients treated with NX-13 had decreased NF-κB activity, TNF-α+ and IFN-γ+ CD4+ T cells and overall production of IL-6, MCP1, and IL-8. NX-13 activates NLRX1 to mediate a resistance to both inflammatory signaling and oxidative stress in mouse models and human primary cells from ulcerative colitis patients with effects on NF-κB activity and oxidative phosphorylation. NX-13 is a promising oral, gut-restricted NLRX1 agonist for treating IBD.
Collapse
Affiliation(s)
- Andrew Leber
- Landos Biopharma, Inc., Blacksburg, VA 24060; and.,BioTherapeutics, Inc., Blacksburg, VA 24060
| | - Raquel Hontecillas
- Landos Biopharma, Inc., Blacksburg, VA 24060; and.,BioTherapeutics, Inc., Blacksburg, VA 24060
| | | | | | | | - Josep Bassaganya-Riera
- Landos Biopharma, Inc., Blacksburg, VA 24060; and .,BioTherapeutics, Inc., Blacksburg, VA 24060
| |
Collapse
|
6
|
Stahl M, Tremblay S, Montero M, Vogl W, Xia L, Jacobson K, Menendez A, Vallance BA. The Muc2 mucin coats murine Paneth cell granules and facilitates their content release and dispersion. Am J Physiol Gastrointest Liver Physiol 2018; 315:G195-G205. [PMID: 29698056 PMCID: PMC6139647 DOI: 10.1152/ajpgi.00264.2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Paneth cells are a key subset of secretory epithelial cells found at the base of small intestinal crypts. Unlike intestinal goblet cells, which secrete the mucin Muc2, Paneth cells are best known for producing an array of antimicrobial factors. We unexpectedly identified Muc2 staining localized around Paneth cell granules. Electron microscopy (EM) confirmed an electron lucent halo around these granules, which was lost in Paneth cells from Muc2-deficient (-/-) mice. EM and immunostaining for lysozyme revealed that Muc2-/- Paneth cells contained larger, more densely packed granules within their cytoplasm, and we detected defects in the transcription of key antimicrobial genes in the ileal tissues of Muc2-/- mice. Enteroids derived from the small intestine of wild-type and Muc2-/- mice revealed phenotypic differences in Paneth cells similar to those seen in vivo. Moreover, lysozyme-containing granule release from Muc2-/- enteroid Paneth cells was shown to be impaired. Surprisingly, Paneth cells within human ileal and duodenal tissues were found to be Muc2 negative. Thus Muc2 plays an important role in murine Paneth cells, suggesting links in function with goblet cells; however human Paneth cells lack Muc2, highlighting that caution should be applied when linking murine to human Paneth cell functions. NEW & NOTEWORTHY We demonstrate for the first time that murine Paneth cell granules possess a halo comprised of the mucin Muc2. The presence of Muc2 exerts an impact on Paneth cell granule size and number and facilitates the release and dispersal of antimicrobials into the mucus layer. Interestingly, despite the importance of Muc2 in murine Paneth cell function, our analysis of Muc2 in human intestinal tissues revealed no trace of Muc2 expression by human Paneth cells.
Collapse
Affiliation(s)
- Martin Stahl
- 1Division of Gastroenterology, Department of Pediatrics, British Columbia Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Sarah Tremblay
- 2Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, University of Sherbrooke, Quebec, Canada
| | - Marinieve Montero
- 1Division of Gastroenterology, Department of Pediatrics, British Columbia Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Wayne Vogl
- 3Department of Cellular and Physiological Sciences, Faculty of Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Lijun Xia
- 4Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma
| | - Kevan Jacobson
- 1Division of Gastroenterology, Department of Pediatrics, British Columbia Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
| | - Alfredo Menendez
- 2Department of Microbiology and Infectious Diseases, Faculty of Medicine and Health Sciences, University of Sherbrooke, Quebec, Canada
| | - Bruce A. Vallance
- 1Division of Gastroenterology, Department of Pediatrics, British Columbia Children’s Hospital and the University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
7
|
Balasubramanian I, Gao N. From sensing to shaping microbiota: insights into the role of NOD2 in intestinal homeostasis and progression of Crohn's disease. Am J Physiol Gastrointest Liver Physiol 2017; 313:G7-G13. [PMID: 28450278 PMCID: PMC5538831 DOI: 10.1152/ajpgi.00330.2016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 04/06/2017] [Accepted: 04/20/2017] [Indexed: 01/31/2023]
Abstract
NOD2 was the first susceptibility gene identified for Crohn's disease (CD), one of the major forms of inflammatory bowel disease (IBD). The field of NOD2 research has opened up many questions critical to understanding the complexities of microbiota-host interactions. In addition to sensing its specific bacterial components as a cytosolic pattern recognition receptor, NOD2 also appears to shape the colonization of intestinal microbiota. Activated NOD2 triggers downstream signaling cascades exampled by the NF-κB pathway to induce antimicrobial activities, however, defective or loss of NOD2 functions incur a similarly activated inflammatory response. Additional studies have identified the involvement of NOD2 in protection against non-microbiota-related intestinal damages as well as extraintestinal infections. We survey recent molecular and genetic studies of NOD2-mediated bacterial sensing and immunological modulation, and integrate evidence to suggest a highly reciprocal but still poorly understood cross talk between enteric microbiota and host cells.
Collapse
Affiliation(s)
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Newark, New Jersey
| |
Collapse
|
8
|
Cui S, Chang PY. Current understanding concerning intestinal stem cells. World J Gastroenterol 2016; 22:7099-7110. [PMID: 27610020 PMCID: PMC4988314 DOI: 10.3748/wjg.v22.i31.7099] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 05/21/2016] [Accepted: 06/15/2016] [Indexed: 02/06/2023] Open
Abstract
In mammals, the intestinal epithelium is a tissue that contains two distinct pools of stem cells: active intestinal stem cells and reserve intestinal stem cells. The former are located in the crypt basement membrane and are responsible for maintaining epithelial homeostasis under intact conditions, whereas the latter exhibit the capacity to facilitate epithelial regeneration after injury. These two pools of cells can convert into each other, maintaining their quantitative balance. In terms of the active intestinal stem cells, their development into functional epithelium is precisely controlled by the following signaling pathways: Wnt/β-catenin, Ras/Raf/Mek/Erk/MAPK, Notch and BMP/Smad. However, mutations in some of the key regulator genes associated with these signaling pathways, such as APC, Kras and Smad4, are also highly associated with gut malformations. At this point, clarifying the biological characteristics of intestinal stem cells will increase the feasibility of preventing or treating some intestinal diseases, such as colorectal cancer. Moreover, as preclinical data demonstrate the therapeutic effects of colon stem cells on murine models of experimental colitis, the prospects of stem cell-based regenerative treatments for ulcerous lesions in the gastrointestinal tract will be improved all the same.
Collapse
|
9
|
Lu P, Hontecillas R, Abedi V, Kale S, Leber A, Heltzel C, Langowski M, Godfrey V, Philipson C, Tubau-Juni N, Carbo A, Girardin S, Uren A, Bassaganya-Riera J. Modeling-Enabled Characterization of Novel NLRX1 Ligands. PLoS One 2015; 10:e0145420. [PMID: 26714018 PMCID: PMC4694766 DOI: 10.1371/journal.pone.0145420] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Accepted: 12/03/2015] [Indexed: 12/11/2022] Open
Abstract
Nucleotide-binding domain and leucine-rich repeat containing (NLR) family are intracellular sentinels of cytosolic homeostasis that orchestrate immune and inflammatory responses in infectious and immune-mediated diseases. NLRX1 is a mitochondrial-associated NOD-like receptor involved in the modulation of immune and metabolic responses. This study utilizes molecular docking approaches to investigate the structure of NLRX1 and experimentally assesses binding to naturally occurring compounds from several natural product and lipid databases. Screening of compound libraries predicts targeting of NLRX1 by conjugated trienes, polyketides, prenol lipids, sterol lipids, and coenzyme A-containing fatty acids for activating the NLRX1 pathway. The ligands of NLRX1 were identified by docking punicic acid (PUA), eleostearic acid (ESA), and docosahexaenoic acid (DHA) to the C-terminal fragment of the human NLRX1 (cNLRX1). Their binding and that of positive control RNA to cNLRX1 were experimentally determined by surface plasmon resonance (SPR) spectroscopy. In addition, the ligand binding sites of cNLRX1 were predicted in silico and validated experimentally. Target mutagenesis studies demonstrate that mutation of 4 critical residues ASP677, PHE680, PHE681, and GLU684 to alanine resulted in diminished affinity of PUA, ESA, and DHA to NLRX1. Consistent with the regulatory actions of NLRX1 on the NF-κB pathway, treatment of bone marrow derived macrophages (BMDM)s with PUA and DHA suppressed NF-κB activity in a NLRX1 dependent mechanism. In addition, a series of pre-clinical efficacy studies were performed using a mouse model of dextran sodium sulfate (DSS)-induced colitis. Our findings showed that the regulatory function of PUA on colitis is NLRX1 dependent. Thus, we identified novel small molecules that bind to NLRX1 and exert anti-inflammatory actions.
Collapse
Affiliation(s)
- Pinyi Lu
- The Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, 24061, United States of America
- Nutritional Immunology and Molecular Medicine Laboratory (www.nimml.org), Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, 24061, United States of America
| | - Raquel Hontecillas
- The Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, 24061, United States of America
- Nutritional Immunology and Molecular Medicine Laboratory (www.nimml.org), Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, 24061, United States of America
| | - Vida Abedi
- The Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, 24061, United States of America
- Nutritional Immunology and Molecular Medicine Laboratory (www.nimml.org), Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, 24061, United States of America
| | - Shiv Kale
- The Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, 24061, United States of America
- Nutritional Immunology and Molecular Medicine Laboratory (www.nimml.org), Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, 24061, United States of America
| | - Andrew Leber
- The Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, 24061, United States of America
- Nutritional Immunology and Molecular Medicine Laboratory (www.nimml.org), Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, 24061, United States of America
| | - Chase Heltzel
- The Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, 24061, United States of America
- Nutritional Immunology and Molecular Medicine Laboratory (www.nimml.org), Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, 24061, United States of America
| | - Mark Langowski
- The Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, 24061, United States of America
- Nutritional Immunology and Molecular Medicine Laboratory (www.nimml.org), Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, 24061, United States of America
| | - Victoria Godfrey
- The Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, 24061, United States of America
- Nutritional Immunology and Molecular Medicine Laboratory (www.nimml.org), Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, 24061, United States of America
| | - Casandra Philipson
- BioTherapeutics, 1800 Kraft Drive, Suite 200, Blacksburg, Virginia, 24060, United States of America
| | - Nuria Tubau-Juni
- The Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, 24061, United States of America
- Nutritional Immunology and Molecular Medicine Laboratory (www.nimml.org), Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, 24061, United States of America
| | - Adria Carbo
- BioTherapeutics, 1800 Kraft Drive, Suite 200, Blacksburg, Virginia, 24060, United States of America
| | - Stephen Girardin
- Laboratory of Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Aykut Uren
- Georgetown University Medical Center, Washington, District of Columbia, 20057, United States of America
| | - Josep Bassaganya-Riera
- The Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, 24061, United States of America
- Nutritional Immunology and Molecular Medicine Laboratory (www.nimml.org), Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, 24061, United States of America
- * E-mail:
| |
Collapse
|
10
|
Chang PY, Jin X, Jiang YY, Wang LX, Liu YJ, Wang J. Mensenchymal stem cells can delay radiation-induced crypt death: impact on intestinal CD44(+) fragments. Cell Tissue Res 2015; 364:331-44. [PMID: 26613604 PMCID: PMC4846698 DOI: 10.1007/s00441-015-2313-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 10/13/2015] [Indexed: 12/20/2022]
Abstract
Intestinal stem cells are primitive cells found within the intestinal epithelium that play a central role in maintaining epithelial homeostasis through self-renewal and commitment into functional epithelial cells. Several markers are available to identify intestinal stem cells, such as Lgr5, CD24 and EphB2, which can be used to sort intestinal stem cells from mammalian gut. Here, we identify and isolate intestinal stem cells from C57BL/6 mice by using a cell surface antigen, CD44. In vitro, some CD44+ crypt cells are capable of forming “villus-crypt”–like structures (organoids). A subset strongly positive for CD44 expresses high levels of intestinal stem-cell-related genes, including Lgr5, Bmi1, Hopx, Lrig1, Ascl2, Smoc2 and Rnf43. Cells from this subset are more capable of developing into organoids in vitro, compared with the subset weakly positive for CD44. However, the organoids are sensitive to ionizing irradiation. We investigate the specific roles of mesenchymal stem cells in protecting organoids against radiation-induced crypt death. When co-cultured with mesenchymal stem cells, the crypt domains of irradiated organoids possess more proliferative cells and fewer apoptotic cells than those not co-cultured with mesenchymal stem cells. Cd44v6 continues to be expressed in the crypt domains of irradiated organoids co-cultured with mesenchymal stem cells. Our results indicate specific roles of mesenchymal stem cells in delaying radiation-induced crypt death in vitro.
Collapse
Affiliation(s)
- Peng-Yu Chang
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun, 130021, People's Republic of China.,Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130000, People's Republic of China
| | - Xing Jin
- Ever Union Biotechology, Tianjin, 300162, People's Republic of China
| | - Yi-Yao Jiang
- Department of Cardiac Surgery, TEDA International Cardiovascular Hospital, Tianjin, 300000, People's Republic of China
| | - Li-Xian Wang
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences, Tianjin, 300300, People's Republic of China
| | - Yong-Jun Liu
- Alliancells Bioscience, Tianjin, 300300, People's Republic of China.
| | - Jin Wang
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130000, People's Republic of China.
| |
Collapse
|
11
|
Huang XL, Xu DH, Wang GP, Zhang S, Yu CG. Associations between CD24 gene polymorphisms and inflammatory bowel disease: A meta-analysis. World J Gastroenterol 2015; 21:6052-6059. [PMID: 26019472 PMCID: PMC4438042 DOI: 10.3748/wjg.v21.i19.6052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 12/20/2014] [Accepted: 02/12/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To evaluate the relationships between CD24 gene polymorphisms and the risk of inflammatory bowel disease (IBD), including ulcerative colitis (UC) and Crohn’s disease (CD).
METHODS: The PubMed, Web of Science and Cochrane Library databases were searched (up to May 30, 2014). The search terms “CD24”, “inflammatory bowel disease”, “Crohn’s disease”, “Ulcerative colitis”, “IBD”, “CD” or “UC”; and “polymorphism”, “mutation” or “variant” were used. Association studies were limited to the English language, but no limitations in terms of race, ethnicity or geographic area were employed. Stata SE12 software was used to calculate the pooled odds ratios (ORs) with 95% confidence intervals (CIs). P < 0.05 was considered statistically significant. The information was independently extracted from each eligible study by two investigators. Two common polymorphisms, C170T (rs8734) and TG1527del (rs3838646), in the CD24 gene were assessed.
RESULTS: A total of three case-control studies including 2342 IBD patients and 1965 healthy controls were involved in this meta-analysis. The patients and controls were from Caucasian cohorts. The three articles included in this meta-analysis all conformed to Hardy-Weinberg equilibrium. This meta-analysis revealed that there were no significant associations between the two CD24 polymorphisms and the risk for IBD (all P > 0.05). However, in a disease subgroup analysis, we found that the CD24 C170T polymorphism was associated with an increased risk of UC in a dominant model (OR = 1.79, 95%CI: 1.15-2.77, P = 0.009) and an additive model (OR = 1.87, 95%CI: 1.19-2.93, P = 0.007), but this relationship was not present for CD. The CD24 TG1570del polymorphism was significantly associated with CD in the additive model (OR = 1.24, 95%CI: 1.01-1.52, P = 0.037).
CONCLUSION: Our findings provide evidence that the CD24 C170T polymorphism might contribute to the susceptibility to UC, and the CD24 TG1527del polymorphism might be associated with the risk of CD.
Collapse
|
12
|
Shanahan MT, Carroll IM, Gulati AS. Critical design aspects involved in the study of Paneth cells and the intestinal microbiota. Gut Microbes 2014; 5:208-14. [PMID: 24637592 PMCID: PMC4063846 DOI: 10.4161/gmic.27466] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Paneth cells are long-lived secretory cells that reside in the base of the crypts of Lieberkühn of the small intestine. They produce an arsenal of molecules that are involved in numerous biological processes, ranging from the control of gut microbial populations to supporting the intestinal stem cell niche. Because of these important functions, Paneth cell abnormalities are becoming implicated in a variety of disease processes. As such, it is necessary to establish parameters that will allow for the comprehensive study of Paneth cells in health and disease. In this addendum, we highlight critical design aspects involved in the study of Paneth cells and their downstream effects on the intestinal microbiota. The importance of this approach is demonstrated by our recent findings that Nod2 does not regulate mouse Paneth cell antimicrobial function, in contrast to previous reports. This work defines key issues to consider when studying Paneth cells in mouse systems.
Collapse
Affiliation(s)
- Michael T Shanahan
- Department of Medicine; Division of Gastroenterology and Hepatology; University of North Carolina at Chapel Hill; Chapel Hill, NC USA
| | - Ian M Carroll
- Department of Medicine; Division of Gastroenterology and Hepatology; University of North Carolina at Chapel Hill; Chapel Hill, NC USA
| | - Ajay S Gulati
- Department of Pediatrics; Division of Gastroenterology; University of North Carolina at Chapel Hill; Chapel Hill, NC USA,Correspondence to: Ajay S Gulati,
| |
Collapse
|
13
|
Lipinski S, Rosenstiel P. Debug Your Bugs - How NLRs Shape Intestinal Host-Microbe Interactions. Front Immunol 2013; 4:479. [PMID: 24409180 PMCID: PMC3873519 DOI: 10.3389/fimmu.2013.00479] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 12/09/2013] [Indexed: 12/14/2022] Open
Abstract
The host's ability to discriminate friend and foe and to establish a precise homeostasis with its associated microbiota is crucial for its survival and fitness. Among the mediators of intestinal host-microbe interactions, NOD-like receptor (NLR) proteins take center stage. They are present in the epithelial lining and innate immune cells that constantly monitor microbial activities at the intestinal barrier. Dysfunctional NLRs predispose to intestinal inflammation as well as sensitization to extra-intestinal immune-mediated diseases and are linked to the alteration of microbial communities. Here, we review advances in our understanding of their reciprocal relationship in the regulation of intestinal homeostasis and implications for intestinal health.
Collapse
Affiliation(s)
- Simone Lipinski
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| |
Collapse
|
14
|
Philpott DJ, Sorbara MT, Robertson SJ, Croitoru K, Girardin SE. NOD proteins: regulators of inflammation in health and disease. Nat Rev Immunol 2013; 14:9-23. [PMID: 24336102 DOI: 10.1038/nri3565] [Citation(s) in RCA: 482] [Impact Index Per Article: 40.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Entry of bacteria into host cells is an important virulence mechanism. Through peptidoglycan recognition, the nucleotide-binding oligomerization domain (NOD) proteins NOD1 and NOD2 enable detection of intracellular bacteria and promote their clearance through initiation of a pro-inflammatory transcriptional programme and other host defence pathways, including autophagy. Recent findings have expanded the scope of the cellular compartments monitored by NOD1 and NOD2 and have elucidated the signalling pathways that are triggered downstream of NOD activation. In vivo, NOD1 and NOD2 have complex roles, both during bacterial infection and at homeostasis. The association of alleles that encode constitutively active or constitutively inactive forms of NOD2 with different diseases highlights this complexity and indicates that a balanced level of NOD signalling is crucial for the maintenance of immune homeostasis.
Collapse
Affiliation(s)
- Dana J Philpott
- 1] Department of Immunology, University of Toronto, Toronto M5S 1A8, Canada. [2]
| | - Matthew T Sorbara
- 1] Department of Immunology, University of Toronto, Toronto M5S 1A8, Canada. [2]
| | | | - Kenneth Croitoru
- Institute of Medical Science, Department of Medicine, University of Toronto, Toronto M5S 1A8, Canada
| | - Stephen E Girardin
- 1] Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto M5S 1A8, Canada. [2]
| |
Collapse
|