1
|
Pravoverov K, Fatima I, Barman S, Jühling F, Primeaux M, Baumert TF, Singh AB, Dhawan P. IL-22 regulates MASTL expression in intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2024; 327:G123-G139. [PMID: 38771154 DOI: 10.1152/ajpgi.00260.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 05/09/2024] [Accepted: 05/09/2024] [Indexed: 05/22/2024]
Abstract
Microtubule-associated serine-threonine kinase-like (MASTL) has recently been identified as an oncogenic kinase given its overexpression in numerous cancers. Our group has shown that MASTL expression is upregulated in mouse models of sporadic colorectal cancer and colitis-associated cancer (CAC). CAC is one of the most severe complications of chronic inflammatory bowel disease (IBD), but a limited understanding of the mechanisms governing the switch from normal healing to neoplasia in IBD underscores the need for increased research in this area. However, MASTL levels in patients with IBD and its molecular regulation in IBD and CAC have not been studied. This study reveals that MASTL is upregulated by the cytokine interleukin (IL)-22, which promotes proliferation and has important functions in colitis recovery; however, IL-22 can also promote tumorigenesis when chronically elevated. Upon reviewing the publicly available data, we found significantly elevated MASTL and IL-22 levels in the biopsies from patients with late-stage ulcerative colitis compared with controls, and that MASTL upregulation was associated with high IL-22 expression. Our subsequent in vitro studies found that IL-22 increases MASTL expression in intestinal epithelial cell lines, which facilitates IL-22-mediated cell proliferation and downstream survival signaling. Inhibition of AKT activation abrogated IL-22-induced MASTL upregulation. We further found an increased association of carbonic anhydrase IX (CAIX) with MASTL in IL-22-treated cells, which stabilized MASTL expression. Inhibition of CAIX prevented IL-22-induced MASTL expression and cell survival. Overall, we show that IL-22/AKT signaling increases MASTL expression to promote cell survival and proliferation. Furthermore, CAIX associates with and stabilizes MASTL in response to IL-22 stimulation.NEW & NOTEWORTHY MASTL is upregulated in colorectal cancer; however, its role in colitis and colitis-associated cancer is poorly understood. This study is the first to draw a link between MASTL and IL-22, a proinflammatory/intestinal epithelial recovery-promoting cytokine that is also implicated in colon tumorigenesis. We propose that IL-22 increases MASTL protein stability by promoting its association with CAIX potentially via AKT signaling to promote cell survival and proliferation.
Collapse
Affiliation(s)
- Kristina Pravoverov
- Eppley Institute, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Iram Fatima
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Susmita Barman
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Frank Jühling
- Inserm U1110, Université de Strasbourg, Institute for Translational Medicine and Liver Disease (ITM), Strasbourg, France
- Inserm U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Université de Strasbourg, Strasbourg, France
| | - Mark Primeaux
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Thomas F Baumert
- Inserm U1110, Université de Strasbourg, Institute for Translational Medicine and Liver Disease (ITM), Strasbourg, France
- Inserm U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Université de Strasbourg, Strasbourg, France
- IHU Strasbourg and Gastroenterology-Hepatology Service, Strasbourg University Hospitals, Strasbourg, France
- Institut Universitaire de France (IUF), Paris, France
| | - Amar B Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States
- Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska, United States
| | - Punita Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States
- Veteran Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska, United States
| |
Collapse
|
2
|
O'Connell AE, Raveenthiraraj S, Oliveira LFS, Adegboye C, Dasuri VS, Qi W, Khetani RS, Singh A, Sundaram N, Lin J, Nandivada P, Rincón-Cruz L, Goldsmith JD, Thiagarajah JR, Carlone DL, Turner JR, Agrawal PB, Helmrath M, Breault DT. WNT2B Deficiency Causes Enhanced Susceptibility to Colitis Due to Increased Inflammatory Cytokine Production. Cell Mol Gastroenterol Hepatol 2024; 18:101349. [PMID: 38697357 PMCID: PMC11217757 DOI: 10.1016/j.jcmgh.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 04/21/2024] [Accepted: 04/22/2024] [Indexed: 05/05/2024]
Abstract
BACKGROUND & AIMS Humans with WNT2B deficiency have severe intestinal disease, including significant inflammatory injury, highlighting a critical role for WNT2B. We sought to understand how WNT2B contributes to intestinal homeostasis. METHODS We investigated the intestinal health of Wnt2b knock out (KO) mice. We assessed the baseline histology and health of the small intestine and colon, and the impact of inflammatory challenge using dextran sodium sulfate (DSS). We also evaluated human intestinal tissue. RESULTS Mice with WNT2B deficiency had normal baseline histology but enhanced susceptibility to DSS colitis because of an increased early injury response. Although intestinal stem cells markers were decreased, epithelial proliferation was similar to control subjects. Wnt2b KO mice showed an enhanced inflammatory signature after DSS treatment. Wnt2b KO colon and human WNT2B-deficient organoids had increased levels of CXCR4 and IL6, and biopsy tissue from humans showed increased neutrophils. CONCLUSIONS WNT2B is important for regulation of inflammation in the intestine. Absence of WNT2B leads to increased expression of inflammatory cytokines and increased susceptibility to gastrointestinal inflammation, particularly in the colon.
Collapse
Affiliation(s)
- Amy E O'Connell
- Division of Newborn Medicine, Boston Children's Hospital, Boston, Massachusetts; The Manton Center for Orphan Disease Research at Boston Children's Hospital, Boston, Massachusetts; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts.
| | | | | | - Comfort Adegboye
- Division of Newborn Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Venkata Siva Dasuri
- Division of Newborn Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Wanshu Qi
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts
| | | | - Akaljot Singh
- Department of Pediatric, General, and Thoracic Surgery, Cincinnati Children's Hospital, Cincinnati, Ohio; Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital, Cincinnati, Ohio
| | - Nambirajam Sundaram
- Department of Pediatric, General, and Thoracic Surgery, Cincinnati Children's Hospital, Cincinnati, Ohio; Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital, Cincinnati, Ohio
| | - Jasmine Lin
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts
| | - Prathima Nandivada
- Department of Surgery, Boston Children's Hospital, Boston, Massachusetts
| | - Lorena Rincón-Cruz
- Department of Surgery, Boston Children's Hospital, Boston, Massachusetts
| | | | - Jay R Thiagarajah
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts; Division of Gastroenterology, Boston Children's Hospital, Boston, Massachusetts
| | - Diana L Carlone
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts; Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts; Harvard Stem Cell Institute, Cambridge, Massachusetts
| | - Jerrold R Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology and Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Pankaj B Agrawal
- Division of Newborn Medicine, Boston Children's Hospital, Boston, Massachusetts; The Manton Center for Orphan Disease Research at Boston Children's Hospital, Boston, Massachusetts; Department of Pediatrics, Harvard Medical School, Boston, Massachusetts; Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts; Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine and Holtz Children's Hospital, Jackson Health System, Miami, Florida
| | - Michael Helmrath
- Department of Pediatric, General, and Thoracic Surgery, Cincinnati Children's Hospital, Cincinnati, Ohio; Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital, Cincinnati, Ohio
| | - David T Breault
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts; Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts; Harvard Stem Cell Institute, Cambridge, Massachusetts
| |
Collapse
|
3
|
Liu P, Zhang R, Song X, Tian X, Guan Y, Li L, He M, He C, Ding N. RTCB deficiency triggers colitis in mice by influencing the NF-κB and Wnt/β-catenin signaling pathways. Acta Biochim Biophys Sin (Shanghai) 2024; 56:405-413. [PMID: 38425245 PMCID: PMC11292128 DOI: 10.3724/abbs.2023279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/25/2023] [Indexed: 03/02/2024] Open
Abstract
RNA terminal phosphorylase B (RTCB) has been shown to play a significant role in multiple physiological processes. However, the specific role of RTCB in the mouse colon remains unclear. In this study, we employ a conditional knockout mouse model to investigate the effects of RTCB depletion on the colon and the potential molecular mechanisms. We assess the efficiency and phenotype of Rtcb knockout using PCR, western blot analysis, histological staining, and immunohistochemistry. Compared with the control mice, the Rtcb-knockout mice exhibit compromised colonic barrier integrity and prominent inflammatory cell infiltration. In the colonic tissues of Rtcb-knockout mice, the protein levels of TNF-α, IL-8, and p-p65 are increased, whereas the levels of IKKβ and IκBα are decreased. Moreover, the level of GSK3β is increased, whereas the levels of Wnt3a, β-catenin, and LGR5 are decreased. Collectively, our findings unveil a close association between RTCB and colonic tissue homeostasis and demonstrate that RTCB deficiency can lead to dysregulation of both the NF-κB and Wnt/β-catenin signaling pathways in colonic cells.
Collapse
Affiliation(s)
- Peiyan Liu
- />College of Life ScienceShandong Normal UniversityJinan250014China
| | - Ruitao Zhang
- />College of Life ScienceShandong Normal UniversityJinan250014China
| | - Xiaotong Song
- />College of Life ScienceShandong Normal UniversityJinan250014China
| | - Xiaohua Tian
- />College of Life ScienceShandong Normal UniversityJinan250014China
| | - Yichao Guan
- />College of Life ScienceShandong Normal UniversityJinan250014China
| | - Licheng Li
- />College of Life ScienceShandong Normal UniversityJinan250014China
| | - Mei He
- />College of Life ScienceShandong Normal UniversityJinan250014China
| | - Chengqiang He
- />College of Life ScienceShandong Normal UniversityJinan250014China
| | - Naizheng Ding
- />College of Life ScienceShandong Normal UniversityJinan250014China
| |
Collapse
|
4
|
Khanbabei A, Segura L, Petrossian C, Lemus A, Cano I, Frazier C, Halajyan A, Ca D, Loza-Coll M. Experimental validation and characterization of putative targets of Escargot and STAT, two master regulators of the intestinal stem cells in Drosophila melanogaster. Dev Biol 2024; 505:148-163. [PMID: 37952851 DOI: 10.1016/j.ydbio.2023.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 10/15/2023] [Accepted: 10/25/2023] [Indexed: 11/14/2023]
Abstract
Many organs contain adult stem cells (ASCs) to replace cells due to damage, disease, or normal tissue turnover. ASCs can divide asymmetrically, giving rise to a new copy of themselves (self-renewal) and a sister that commits to a specific cell type (differentiation). Decades of research have led to the identification of pleiotropic genes whose loss or gain of function affect diverse aspects of normal ASC biology. Genome-wide screens of these so-called genetic "master regulator" (MR) genes, have pointed to hundreds of putative targets that could serve as their downstream effectors. Here, we experimentally validate and characterize the regulation of several putative targets of Escargot (Esg) and the Signal Transducer and Activator of Transcription (Stat92E, a.k.a. STAT), two known MRs in Drosophila intestinal stem cells (ISCs). Our results indicate that regardless of bioinformatic predictions, most experimentally validated targets show a profile of gene expression that is consistent with co-regulation by both Esg and STAT, fitting a rather limited set of co-regulatory modalities. A bioinformatic analysis of proximal regulatory sequences in specific subsets of co-regulated targets identified additional transcription factors that might cooperate with Esg and STAT in modulating their transcription. Lastly, in vivo manipulations of validated targets rarely phenocopied the effects of manipulating Esg and STAT, suggesting the existence of complex genetic interactions among downstream targets of these two MR genes during ISC homeostasis.
Collapse
Affiliation(s)
- Armen Khanbabei
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Lina Segura
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Cynthia Petrossian
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Aaron Lemus
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Ithan Cano
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Courtney Frazier
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Armen Halajyan
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Donnie Ca
- Department of Biology, California State University, Northridge (CSUN), USA
| | - Mariano Loza-Coll
- Department of Biology, California State University, Northridge (CSUN), USA.
| |
Collapse
|
5
|
Kurup S, Tan C, Kume T. Cardiac and intestinal tissue conduct developmental and reparative processes in response to lymphangiocrine signaling. Front Cell Dev Biol 2023; 11:1329770. [PMID: 38178871 PMCID: PMC10764504 DOI: 10.3389/fcell.2023.1329770] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 12/08/2023] [Indexed: 01/06/2024] Open
Abstract
Lymphatic vessels conduct a diverse range of activities to sustain the integrity of surrounding tissue. Besides facilitating the movement of lymph and its associated factors, lymphatic vessels are capable of producing tissue-specific responses to changes within their microenvironment. Lymphatic endothelial cells (LECs) secrete paracrine signals that bind to neighboring cell-receptors, commencing an intracellular signaling cascade that preludes modifications to the organ tissue's structure and function. While the lymphangiocrine factors and the molecular and cellular mechanisms themselves are specific to the organ tissue, the crosstalk action between LECs and adjacent cells has been highlighted as a commonality in augmenting tissue regeneration within animal models of cardiac and intestinal disease. Lymphangiocrine secretions have been owed for subsequent improvements in organ function by optimizing the clearance of excess tissue fluid and immune cells and stimulating favorable tissue growth, whereas perturbations in lymphatic performance bring about the opposite. Newly published landmark studies have filled gaps in our understanding of cardiac and intestinal maintenance by revealing key players for lymphangiocrine processes. Here, we will expand upon those findings and review the nature of lymphangiocrine factors in the heart and intestine, emphasizing its involvement within an interconnected network that supports daily homeostasis and self-renewal following injury.
Collapse
Affiliation(s)
- Shreya Kurup
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Honors College, University of Illinois at Chicago, Chicago, IL, United States
| | - Can Tan
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Tsutomu Kume
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
6
|
Palsamy K, Chen JY, Skaggs K, Qadeer Y, Connors M, Cutler N, Richmond J, Kommidi V, Poles A, Affrunti D, Powell C, Goldman D, Parent JM. Microglial depletion after brain injury prolongs inflammation and impairs brain repair, adult neurogenesis and pro-regenerative signaling. Glia 2023; 71:2642-2663. [PMID: 37449457 PMCID: PMC10528132 DOI: 10.1002/glia.24444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023]
Abstract
The adult zebrafish brain, unlike mammals, has a remarkable regenerative capacity. Although inflammation in part hinders regeneration in mammals, it is necessary for zebrafish brain repair. Microglia are resident brain immune cells that regulate the inflammatory response. To explore the microglial role in repair, we used liposomal clodronate or colony stimulating factor-1 receptor (csf1r) inhibitor to suppress microglia after brain injury, and also examined regeneration in two genetic mutant lines that lack microglia. We found that microglial ablation impaired telencephalic regeneration after injury. Microglial suppression attenuated cell proliferation at the intermediate progenitor cell amplification stage of neurogenesis. Notably, the loss of microglia impaired phospho-Stat3 (signal transducer and activator of transcription 3) and ß-Catenin signaling after injury. Furthermore, the ectopic activation of Stat3 and ß-Catenin rescued neurogenesis defects caused by microglial loss. Microglial suppression also prolonged the post-injury inflammatory phase characterized by neutrophil accumulation, likely hindering the resolution of inflammation. These findings reveal specific roles of microglia and inflammatory signaling during zebrafish telencephalic regeneration that should advance strategies to improve mammalian brain repair.
Collapse
Affiliation(s)
- Kanagaraj Palsamy
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Jessica Y Chen
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Kaia Skaggs
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
- University of Findlay, Findlay, Ohio, USA
| | - Yusuf Qadeer
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Meghan Connors
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Noah Cutler
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Joshua Richmond
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Vineeth Kommidi
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Allison Poles
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Danielle Affrunti
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
| | - Curtis Powell
- Michigan Neuroscience Institute, Ann Arbor, Michigan, USA
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Daniel Goldman
- Michigan Neuroscience Institute, Ann Arbor, Michigan, USA
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Jack M Parent
- Department of Neurology, University of Michigan, Ann Arbor, Michigan, USA
- Michigan Neuroscience Institute, Ann Arbor, Michigan, USA
- VA Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| |
Collapse
|
7
|
Li M, Li M, Qiao L, Wu C, Xu D, Zhao Y, Zeng X. Role of JAK-STAT signaling pathway in pathogenesis and treatment of primary Sjögren's syndrome. Chin Med J (Engl) 2023; 136:2297-2306. [PMID: 37185152 PMCID: PMC10538906 DOI: 10.1097/cm9.0000000000002539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Indexed: 05/17/2023] Open
Abstract
ABSTRACT Primary Sjögren's syndrome (pSS) is a systemic autoimmune disease with high prevalence and possible poor prognosis. Though the pathogenesis of pSS has not been fully elucidated, B cell hyperactivity is considered as one of the fundamental abnormalities in pSS patients. It has long been identified that Janus kinases-signal transducer and activator of transcription (JAK-STAT) signaling pathway contributes to rheumatoid arthritis and systemic lupus erythematosus. Recently, increasing numbers of studies have provided evidence that JAK-STAT pathway also has an important role in the pathogenesis of pSS via direct or indirect activation of B cells. Signal transducer and activator of transcription 1 (STAT1), STAT3, and STAT5 activated by various cytokines and ribonucleic acid contribute to pSS development, respectively or synergically. These results reveal the potential application of Janus kinase inhibitors for treatment of pSS, which may fundamentally improve the quality of life and prognosis of patients with pSS.
Collapse
Affiliation(s)
- Mucong Li
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology; State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH); Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing 100730, China
| | | | | | | | | | | | | |
Collapse
|
8
|
Yu L, Qi S, Wei G, Rao X, Luo D, Zou M, Mi Y, Zhang C, Li J. Krüppel-like factor 5 activates chick intestinal stem cell and promotes mucosal repair after impairment. Cell Cycle 2023; 22:2142-2160. [PMID: 37950881 PMCID: PMC10732631 DOI: 10.1080/15384101.2023.2278938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 10/30/2023] [Indexed: 11/13/2023] Open
Abstract
The mucosal renewal, which depends on the intestinal stem cell (ISC) activity, is the foundation of mucosal repairment. Importantly, activation of reserve ISCs (rISCs) plays a vital role in initiating mucosal repair after injury. However, the underlying regulatory mechanism of rISCs activation in chickens remains unclear. In this study, immediately after lipopolysaccharide (LPS) challenge, mitochondrial morphological destruction and dysfunction appeared in the crypt, accompanied by decreased epithelial secretion (decreased Muc2 mRNA abundance and LYSOZYME protein level). However, immediately after mucosal injury, the mucosal renewal accelerated, as indicated by the increased BrdU positive rate, proliferating cell nuclear antigen (PCNA) protein level and mRNA abundance of cell cycle markers (Ccnd1, Cdk2). Concerning the ISCs activity, during the early period of injury, there appeared a reduction of active ISCs (aISCs) marker Lgr5 mRNA and protein, and an increasing of rISCs marker Hopx mRNA and protein. Strikingly, upon LPS challenge, increased mRNA transcriptional level of Krüppel-like factor 5 (Klf5) was detected in the crypt. Moreover, under LPS treatment in organoids, the KLF5 inhibitor (ML264) would decrease the mRNA and protein levels of Stat5a and Hopx, the STAT5A inhibitor (AC-4-130) would suppress the Lgr5 mRNA and protein levels. Furthermore, the Dual-Luciferase Reporter assay confirmed that, KLF5 would bind to Hopx promoter and activate the rISCs, STAT5A would trigger Lgr5 promoter and activate the aISCs. Collectively, KLF5 was upregulated during the early period of injury, further activate the rISCs directly and activate aISCs via STAT5A indirectly, thus initiate mucosal repair after injury.
Collapse
Affiliation(s)
- Lingzi Yu
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, P.R. China
| | - Sichao Qi
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, P.R. China
- Hainan Institute of Zhejiang University, Sanya, P.R. China
| | - Guozhen Wei
- Qingliu Animal Husbandry, Veterinary and Aquatic Products Center, Sanming, P.R. China
| | - Xi Rao
- Qingliu Animal Husbandry, Veterinary and Aquatic Products Center, Sanming, P.R. China
| | - Danni Luo
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, P.R. China
| | - Minyao Zou
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, P.R. China
- Hainan Institute of Zhejiang University, Sanya, P.R. China
| | - Yuling Mi
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, P.R. China
| | - Caiqiao Zhang
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, P.R. China
| | - Jian Li
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, P.R. China
| |
Collapse
|
9
|
Gao J, Cao B, Zhao R, Li H, Xu Q, Wei B. Critical Signaling Transduction Pathways and Intestinal Barrier: Implications for Pathophysiology and Therapeutics. Pharmaceuticals (Basel) 2023; 16:1216. [PMID: 37765024 PMCID: PMC10537644 DOI: 10.3390/ph16091216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
The intestinal barrier is a sum of the functions and structures consisting of the intestinal mucosal epithelium, mucus, intestinal flora, secretory immunoglobulins, and digestive juices. It is the first-line defense mechanism that resists nonspecific infections with powerful functions that include physical, endocrine, and immune defenses. Health and physiological homeostasis are greatly dependent on the sturdiness of the intestinal barrier shield, whose dysfunction can contribute to the progression of numerous types of intestinal diseases. Disorders of internal homeostasis may also induce barrier impairment and form vicious cycles during the response to diseases. Therefore, the identification of the underlying mechanisms involved in intestinal barrier function and the development of effective drugs targeting its damage have become popular research topics. Evidence has shown that multiple signaling pathways and corresponding critical molecules are extensively involved in the regulation of the barrier pathophysiological state. Ectopic expression or activation of signaling pathways plays an essential role in the process of shield destruction. Although some drugs, such as molecular or signaling inhibitors, are currently used for the treatment of intestinal diseases, their efficacy cannot meet current medical requirements. In this review, we summarize the current achievements in research on the relationships between the intestinal barrier and signaling pathways. The limitations and future perspectives are also discussed to provide new horizons for targeted therapies for restoring intestinal barrier function that have translational potential.
Collapse
Affiliation(s)
- Jingwang Gao
- Department of General Surgery, Medical School of Chinese PLA, Beijing 100853, China; (J.G.); (R.Z.); (H.L.); (Q.X.)
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China;
| | - Bo Cao
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China;
| | - Ruiyang Zhao
- Department of General Surgery, Medical School of Chinese PLA, Beijing 100853, China; (J.G.); (R.Z.); (H.L.); (Q.X.)
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China;
| | - Hanghang Li
- Department of General Surgery, Medical School of Chinese PLA, Beijing 100853, China; (J.G.); (R.Z.); (H.L.); (Q.X.)
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China;
| | - Qixuan Xu
- Department of General Surgery, Medical School of Chinese PLA, Beijing 100853, China; (J.G.); (R.Z.); (H.L.); (Q.X.)
- Department of General Surgery, First Medical Center, Chinese PLA General Hospital, Beijing 100853, China;
| | - Bo Wei
- Department of General Surgery, Medical School of Chinese PLA, Beijing 100853, China; (J.G.); (R.Z.); (H.L.); (Q.X.)
| |
Collapse
|
10
|
Tan C, Norden PR, Yu W, Liu T, Ujiie N, Lee SK, Yan X, Dyakiv Y, Aoto K, Ortega S, De Plaen IG, Sampath V, Kume T. Endothelial FOXC1 and FOXC2 promote intestinal regeneration after ischemia-reperfusion injury. EMBO Rep 2023; 24:e56030. [PMID: 37154714 PMCID: PMC10328078 DOI: 10.15252/embr.202256030] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 04/07/2023] [Accepted: 04/19/2023] [Indexed: 05/10/2023] Open
Abstract
Intestinal ischemia underlies several clinical conditions and can result in the loss of the intestinal mucosal barrier. Ischemia-induced damage to the intestinal epithelium is repaired by stimulation of intestinal stem cells (ISCs), and paracrine signaling from the vascular niche regulates intestinal regeneration. Here, we identify FOXC1 and FOXC2 as essential regulators of paracrine signaling in intestinal regeneration after ischemia-reperfusion (I/R) injury. Vascular endothelial cell (EC)- and lymphatic EC (LEC)-specific deletions of Foxc1, Foxc2, or both in mice worsen I/R-induced intestinal damage by causing defects in vascular regrowth, expression of chemokine CXCL12 and Wnt activator R-spondin 3 (RSPO3) in blood ECs (BECs) and LECs, respectively, and activation of Wnt signaling in ISCs. Both FOXC1 and FOXC2 directly bind to regulatory elements of the CXCL12 and RSPO3 loci in BECs and LECs, respectively. Treatment with CXCL12 and RSPO3 rescues the I/R-induced intestinal damage in EC- and LEC-Foxc mutant mice, respectively. This study provides evidence that FOXC1 and FOXC2 are required for intestinal regeneration by stimulating paracrine CXCL12 and Wnt signaling.
Collapse
Affiliation(s)
- Can Tan
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Pieter R Norden
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Wei Yu
- Division of Neonatology, Department of PediatricsChildren's Mercy HospitalKansas CityMOUSA
| | - Ting Liu
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Naoto Ujiie
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Sun Kyong Lee
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Xiaocai Yan
- Department of Pediatrics, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Yaryna Dyakiv
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Kazushi Aoto
- Department of BiochemistryHamamatsu University School of MedicineHamamatsuJapan
| | - Sagrario Ortega
- Mouse Genome Editing Unit, Biotechnology ProgramSpanish National Cancer Research CentreMadridSpain
| | - Isabelle G De Plaen
- Department of Pediatrics, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Venkatesh Sampath
- Division of Neonatology, Department of PediatricsChildren's Mercy HospitalKansas CityMOUSA
| | - Tsutomu Kume
- Department of Medicine, Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| |
Collapse
|
11
|
O'Connell AE, Raveenthiraraj S, Adegboye C, Qi W, Khetani RS, Singh A, Sundaram N, Emeonye C, Lin J, Goldsmith JD, Thiagarajah JR, Carlone DL, Turner JR, Agrawal PB, Helmrath M, Breault DT. WNT2B Deficiency Causes Increased Susceptibility to Colitis in Mice and Impairs Intestinal Epithelial Development in Humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.21.537894. [PMID: 37131772 PMCID: PMC10153278 DOI: 10.1101/2023.04.21.537894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Background and aims WNT2B is a canonical Wnt ligand previously thought to be fully redundant with other Wnts in the intestinal epithelium. However, humans with WNT2B deficiency have severe intestinal disease, highlighting a critical role for WNT2B. We sought to understand how WNT2B contributes to intestinal homeostasis. Methods We investigated the intestinal health of Wnt2b knock out (KO) mice. We assessed the impact of inflammatory challenge to the small intestine, using anti-CD3χ antibody, and to the colon, using dextran sodium sulfate (DSS). In addition, we generated human intestinal organoids (HIOs) from WNT2B-deficient human iPSCs for transcriptional and histological analyses. Results Mice with WNT2B deficiency had significantly decreased Lgr5 expression in the small intestine and profoundly decreased expression in the colon, but normal baseline histology. The small intestinal response to anti-CD3χ antibody was similar in Wnt2b KO and wild type (WT) mice. In contrast, the colonic response to DSS in Wnt2b KO mice showed an accelerated rate of injury, featuring earlier immune cell infiltration and loss of differentiated epithelium compared to WT. WNT2B-deficient HIOs showed abnormal epithelial organization and an increased mesenchymal gene signature. Conclusion WNT2B contributes to maintenance of the intestinal stem cell pool in mice and humans. WNT2B deficient mice, which do not have a developmental phenotype, show increased susceptibility to colonic injury but not small intestinal injury, potentially due to a higher reliance on WNT2B in the colon compared to the small intestine.WNT2B deficiency causes a developmental phenotype in human intestine with HIOs showing a decrease in their mesenchymal component and WNT2B-deficient patients showing epithelial disorganization. Data Transparency Statement All RNA-Seq data will be available through online repository as indicated in Transcript profiling. Any other data will be made available upon request by emailing the study authors.
Collapse
|
12
|
Yan X, Liu L, Yao S, Chen Y, Yu Q, Jiang C, Chen W, Chen X, Han S. LncRNA and mRNA profiles of human milk-derived exosomes and their possible roles in protecting against necrotizing enterocolitis. Food Funct 2022; 13:12953-12965. [PMID: 36448375 DOI: 10.1039/d2fo01866g] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Necrotizing enterocolitis (NEC) is one of the most severe diseases commonly afflicting premature infants. Our previous studies suggests that human milk-derived exosomes (HM-Exos) have a potential therapeutic effect on NEC. In this study, we investigate the potentially therapeutic role of HM-Exos in an NEC animal model via comprehensive lncRNA and mRNA expression profiles. A rat model of NEC was induced through hypoxia, hypothermia and formula feeds. We extracted exosomes from the colostrum of healthy lactating mothers and identified their functions in an NEC animal model. Furthermore, high-throughput lncRNA and mRNA sequencings were explored to find the underlying mechanisms. Although both exosomes from term human breast milk (Term-Exos) and exosomes from preterm human breast milk (Pre-Exos) alleviated the severity of NEC, Pre-Exos seemed to better promote the proliferation of intestinal epithelial cells in vivo. We identified a total of 44 differentially expressed lncRNAs and 88 differentially expressed mRNAs between Term-Exos and Pre-Exos. Further GO and KEGG pathway analysis showed that the lncRNA-mRNA network of HM-Exos was associated with the JAK-STAT signaling pathway, bile secretion and the AMPK signaling pathway, which are predicted to be involved in the proliferation of cells. Therefore, this study reveals for the first time the important roles of human milk derived lncRNAs and mRNAs in protecting against necrotizing enterocolitis. These results provide new insight into the development of NEC.
Collapse
Affiliation(s)
- Xiangyun Yan
- Department of Paediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China.
| | - Linjie Liu
- Department of Paediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China.
| | - Shuwen Yao
- Department of Paediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China.
| | - Yanjie Chen
- Department of Paediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China.
| | - Qinlei Yu
- Department of Paediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China.
| | - Chengyao Jiang
- Department of Paediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China.
| | - Wenjuan Chen
- Department of Paediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China.
| | - Xiaohui Chen
- Department of Paediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China.
| | - Shuping Han
- Department of Paediatrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, Jiangsu, China.
| |
Collapse
|
13
|
Subramanian S, Geng H, Du C, Chou PM, Bu HF, Wang X, Swaminathan S, Tan SC, Ridlon JM, De Plaen IG, Tan XD. Feeding mode influences dynamic gut microbiota signatures and affects susceptibility to anti-CD3 mAb-induced intestinal injury in neonatal mice. Am J Physiol Gastrointest Liver Physiol 2022; 323:G205-G218. [PMID: 35819158 PMCID: PMC9394775 DOI: 10.1152/ajpgi.00337.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 07/01/2022] [Accepted: 07/02/2022] [Indexed: 01/31/2023]
Abstract
Feeding modes influence the gut microbiome, immune system, and intestinal barrier homeostasis in neonates; how feeding modes impact susceptibility to neonatal gastrointestinal (GI) diseases is still uncertain. Here, we investigated the impact of dam feeding (DF) and formula feeding (FF) on features of the gut microbiome and physiological inflammation during the first 2 days of postnatal development and on the susceptibility to intestinal injury related to the inflammatory state in neonatal mouse pups. 16S rRNA sequencing data revealed microbiome changes, lower α-diversity, and a distinct pattern of β-diversity including expansion of f_Enterobacteriaceae and f_Enterococcaceae in the ileum of FF pups compared with DF pups by postnatal day (P)2. Together with gut dysbiosis, the FF cohort also had greater ileal mucosa physiological inflammatory activity compared with DF pups by P2 but maintained normal histological features. Interestingly, FF but not DF mouse pups developed necrotizing enterocolitis (NEC)-like intestinal injury within 24 h after anti-CD3 mAb treatment, suggesting that FF influences the susceptibility to intestinal injury in neonates. We further found that NEC-like incidence in anti-CD3 mAb-treated FF neonatal pups was attenuated by antibiotic treatment. Collectively, our data suggest that FF predisposes mouse pups to anti-CD3 mAb-induced intestinal injury due to abnormal f_Enterobacteriaceae and f_Enterococcaceae colonization. These findings advance our understanding of FF-associated microbial colonization and intestinal inflammation, which may help inform the development of new therapeutic approaches to GI diseases like NEC in infants.NEW & NOTEWORTHY This report shows that a feeding mode profoundly affects gut colonization in neonatal mice. Furthermore, our results demonstrate that formula feeding predisposes mouse pups to anti-CD3 mAb-induced necrotizing enterocolitis (NEC)-like intestinal injury upon inadequate microbial colonization. The study suggests the role of the combined presence of formula feeding-associated dysbiosis and mucosal inflammation in the pathogenesis of NEC and provides a new mouse model to study this disease.
Collapse
Affiliation(s)
- Saravanan Subramanian
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Center for Intestinal and Liver Inflammation Research, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Hua Geng
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Center for Intestinal and Liver Inflammation Research, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Chao Du
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Center for Intestinal and Liver Inflammation Research, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Pauline M Chou
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Heng-Fu Bu
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Center for Intestinal and Liver Inflammation Research, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Xiao Wang
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Center for Intestinal and Liver Inflammation Research, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Suchitra Swaminathan
- Division of Rheumatology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Stephanie C Tan
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Center for Intestinal and Liver Inflammation Research, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Jason M Ridlon
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois
| | - Isabelle G De Plaen
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Division of Neonatology, Department of Pediatrics, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Xiao-Di Tan
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Center for Intestinal and Liver Inflammation Research, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Department of Research and Development, Jesse Brown Department of Veterans Affairs Medical Center, Chicago, Illinois
| |
Collapse
|
14
|
Nolan LS, Baldridge MT. Advances in understanding interferon-mediated immune responses to enteric viruses in intestinal organoids. Front Immunol 2022; 13:943334. [PMID: 35935957 PMCID: PMC9354881 DOI: 10.3389/fimmu.2022.943334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/30/2022] [Indexed: 11/16/2022] Open
Abstract
Interferons (IFN) are antiviral cytokines with critical roles in regulating pathogens at epithelial barriers, but their capacity to restrict human enteric viruses has been incompletely characterized in part due to challenges in cultivating some viruses in vitro, particularly human norovirus. Accordingly, advancements in the development of antiviral therapies and vaccine strategies for enteric viral infections have been similarly constrained. Currently emerging is the use of human intestinal enteroids (HIEs) to investigate mechanisms of human enteric viral pathogenesis. HIEs provide a unique opportunity to investigate host-virus interactions using an in vitro system that recapitulates the cellular complexity of the in vivo gastrointestinal epithelium. This approach permits the exploration of intestinal epithelial cell interactions with enteric viruses as well as the innate immune responses mediated by IFNs and IFN-stimulated genes. Here, we describe recent findings related to the production, signaling, and function of IFNs in the response to enteric viral infections, which will ultimately help to reveal important aspects of pathogenesis and facilitate the future development of therapeutics and vaccines.
Collapse
Affiliation(s)
- Lila S. Nolan
- Department of Pediatrics, Division of Newborn Medicine, Washington University School of Medicine, St. Louis Children’s Hospital, St. Louis, MO, United States
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, United States
| | - Megan T. Baldridge
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, United States
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
15
|
Nalapareddy K, Zheng Y, Geiger H. Aging of intestinal stem cells. Stem Cell Reports 2022; 17:734-740. [PMID: 35276089 PMCID: PMC9023768 DOI: 10.1016/j.stemcr.2022.02.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 12/20/2022] Open
Abstract
The intestine is one of the organs that relies on stem cell function for maintaining tissue homeostasis. Recent findings on intestinal aging show that intestinal architecture, such as villus length, crypt size, and cell composition changes in the aged crypts. The correspondent decline in the regenerative capacity of the intestine is mainly due to a decline in intestinal stem cell function upon aging, as the underlying mechanisms of aging intestinal stem cells are beginning to unravel. This review summarizes our current knowledge on stem cell-intrinsic mechanisms of aging of intestinal stem cells and their connection to extrinsic factors, such as niche cells and microbiota and will introduce recent approaches to attenuate or even revert the aging of intestinal stem cells.
Collapse
Affiliation(s)
- Kodandaramireddy Nalapareddy
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, OH 45229, USA
| | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center and University of Cincinnati, OH 45229, USA
| | - Hartmut Geiger
- Institute of Molecular Medicine, Ulm University, Ulm, Germany.
| |
Collapse
|
16
|
Microbes affect gut epithelial cell composition through immune-dependent regulation of intestinal stem cell differentiation. Cell Rep 2022; 38:110572. [PMID: 35354023 PMCID: PMC9078081 DOI: 10.1016/j.celrep.2022.110572] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 12/14/2021] [Accepted: 03/03/2022] [Indexed: 12/29/2022] Open
Abstract
Gut microbes play important roles in host physiology; however, the mechanisms underlying their impact remain poorly characterized. Here, we demonstrate that microbes not only influence gut physiology but also alter its epithelial composition. The microbiota and pathogens both influence intestinal stem cell (ISC) differentiation. Intriguingly, while the microbiota promotes ISC differentiation into enterocytes (EC), pathogens stimulate enteroendocrine cell (EE) fate and long-term accumulation of EEs in the midgut epithelium. Importantly, the evolutionarily conserved Drosophila NFKB (Relish) pushes stem cell lineage specification toward ECs by directly regulating differentiation factors. Conversely, the JAK-STAT pathway promotes EE fate in response to infectious damage. We propose a model in which the balance of microbial pattern recognition pathways, such as Imd-Relish, and damage response pathways, such as JAK-STAT, influence ISC differentiation, epithelial composition, and gut physiology.
Collapse
|
17
|
Jawahar J, McCumber AW, Lickwar CR, Amoroso CR, de la Torre Canny SG, Wong S, Morash M, Thierer JH, Farber SA, Bohannan BJM, Guillemin K, Rawls JF. Starvation causes changes in the intestinal transcriptome and microbiome that are reversed upon refeeding. BMC Genomics 2022; 23:225. [PMID: 35317738 PMCID: PMC8941736 DOI: 10.1186/s12864-022-08447-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/07/2022] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The ability of animals and their microbiomes to adapt to starvation and then restore homeostasis after refeeding is fundamental to their continued survival and symbiosis. The intestine is the primary site of nutrient absorption and microbiome interaction, however our understanding of intestinal adaptations to starvation and refeeding remains limited. Here we used RNA sequencing and 16S rRNA gene sequencing to uncover changes in the intestinal transcriptome and microbiome of zebrafish subjected to long-term starvation and refeeding compared to continuously fed controls. RESULTS Starvation over 21 days led to increased diversity and altered composition in the intestinal microbiome compared to fed controls, including relative increases in Vibrio and reductions in Plesiomonas bacteria. Starvation also led to significant alterations in host gene expression in the intestine, with distinct pathways affected at early and late stages of starvation. This included increases in the expression of ribosome biogenesis genes early in starvation, followed by decreased expression of genes involved in antiviral immunity and lipid transport at later stages. These effects of starvation on the host transcriptome and microbiome were almost completely restored within 3 days after refeeding. Comparison with published datasets identified host genes responsive to starvation as well as high-fat feeding or microbiome colonization, and predicted host transcription factors that may be involved in starvation response. CONCLUSIONS Long-term starvation induces progressive changes in microbiome composition and host gene expression in the zebrafish intestine, and these changes are rapidly reversed after refeeding. Our identification of bacterial taxa, host genes and host pathways involved in this response provides a framework for future investigation of the physiological and ecological mechanisms underlying intestinal adaptations to food restriction.
Collapse
Affiliation(s)
- Jayanth Jawahar
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Alexander W McCumber
- Department of Civil and Environmental Engineering, Duke University, Durham, NC, 27708, USA
| | - Colin R Lickwar
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Caroline R Amoroso
- Department of Evolutionary Anthropology, Duke University, Durham, NC, 27708, USA
| | - Sol Gomez de la Torre Canny
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Sandi Wong
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Margaret Morash
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC, 27710, USA
| | - James H Thierer
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, 21218, USA
- Department of Biology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Steven A Farber
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD, 21218, USA
- Department of Biology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Brendan J M Bohannan
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR, 97403, USA
| | - Karen Guillemin
- Institute of Molecular Biology, University of Oregon, Eugene, OR, 97403, USA
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, NC, 27710, USA.
| |
Collapse
|
18
|
Ma L, Yu J, Zhang H, Zhao B, Zhang J, Yang D, Luo F, Wang B, Jin B, Liu J. Effects of Immune Cells on Intestinal Stem Cells: Prospects for Therapeutic Targets. Stem Cell Rev Rep 2022; 18:2296-2314. [DOI: 10.1007/s12015-022-10347-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2022] [Indexed: 11/29/2022]
|
19
|
Chen MJ, Zhou JY, Chen YJ, Wang XQ, Yan HC, Gao CQ. The in ovo injection of methionine improves intestinal cell proliferation and differentiation in chick embryos by activating the JAK2/STAT3 signaling pathway. ACTA ACUST UNITED AC 2021; 7:1031-1038. [PMID: 34738033 PMCID: PMC8536505 DOI: 10.1016/j.aninu.2021.03.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 01/25/2021] [Accepted: 03/09/2021] [Indexed: 12/15/2022]
Abstract
The intestinal health of chick embryos is vital for their life-long growth, and exogenous nutrition intervention may provide sufficient nutrition for embryonic development. In the present study, we investigated the effect of in ovo injection of L-methionine (L-Met) on the intestinal structure and barrier function of chick embryos. There were 4 groups of treatments: the control (CON) group injected with phosphate-buffered saline (PBS) and the other 3 groups injected with 5, 10, and 20 mg L-Met/egg, respectively. The injection was performed on embryonic day 9 (E9), and intestinal samples were collected on the day of hatching for analysis. The results showed that, compared with the CON group, the groups administered an in ovo injection of L-Met increased relative weights of the duodenum, jejunum, and ileum (P < 0.05). Hematoxylin and eosin (H&E) staining showed that the groups injected with 5, 10, and 20 mg L-Met significantly increased villus height and crypt depth (P < 0.05). Moreover, in ovo injection of 10 mg L-Met also increased the transepithelial electrical resistance (TEER) of the jejunum (P < 0.05). Injection with 10 and 20 mg L-Met increased the expression of the tight junction proteins (ZO-1 and claudin-1) and the fluorescence signal intensity of Ki67 and villin proteins (P < 0.05). Further, the protein expression of phospho-Janus kinase 2 (p-JAK2) and phospho-signal transducer and activator of transcription 3 (p-STAT3) was significantly increased by 10 or 20 mg L-Met injection (P < 0.05). In conclusion, the injection of L-Met, especially at a dose of 10 mg, showed beneficial effects on the intestinal integrity of chick embryos due to the activation of the JAK2/STAT3 signaling pathway. Our results may provide new insights for regulating the intestinal development of embryonic chicks and the rapid growth of chicks after hatching.
Collapse
|
20
|
Lei H, Crawford MS, McCole DF. JAK-STAT Pathway Regulation of Intestinal Permeability: Pathogenic Roles and Therapeutic Opportunities in Inflammatory Bowel Disease. Pharmaceuticals (Basel) 2021; 14:840. [PMID: 34577540 PMCID: PMC8466350 DOI: 10.3390/ph14090840] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 12/15/2022] Open
Abstract
The epithelial barrier forms the interface between luminal microbes and the host immune system and is the first site of exposure to many of the environmental factors that trigger disease activity in chronic inflammatory bowel disease (IBD). Disruption of the epithelial barrier, in the form of increased intestinal permeability, is a feature of IBD and other inflammatory diseases, including celiac disease and type 1 diabetes. Variants in genes that regulate or belong to the JAK-STAT signaling pathway are associated with IBD risk. Inhibitors of the JAK-STAT pathway are now effective therapeutic options in IBD. This review will discuss emerging evidence that JAK inhibitors can be used to improve defects in intestinal permeability and how this plays a key role in resolving intestinal inflammation.
Collapse
Affiliation(s)
| | | | - Declan F. McCole
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA; (H.L.); (M.S.C.)
| |
Collapse
|
21
|
Targeting JAK/STAT signaling pathways in treatment of inflammatory bowel disease. Inflamm Res 2021; 70:753-764. [PMID: 34212215 DOI: 10.1007/s00011-021-01482-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2021] [Indexed: 01/05/2023] Open
Abstract
Janus kinase/signal transduction and transcriptional activator (JAK/STAT) signaling pathway is a transport hub for cytokine secretion and exerts its effects. The activation of JAK/STAT signaling pathway is essential for the regulation of inflammatory responses. Inappropriate activation or deletion of JAK/STAT signaling pathway is the initiator of the inflammatory response. JAK/STAT signaling pathway has been demonstrated to be involved in the process of innate and adaptive immune response to inflammatory bowel disease (IBD). In this review, we discuss the role of the JAK/STAT signaling pathway in the regulation of different cells in IBD, as well as new findings on the involvement of the JAK/STAT signaling pathway in the regulation of the intestinal immune response. The current status of JAK inhibitors in the treatment of IBD is summarized as well. This review highlights natural remedies that can serve as potential JAK inhibitors. These phytochemicals may be useful in the identification of precursor compounds in the process of designing and developing novel JAK inhibitors.
Collapse
|
22
|
Ben Ghezala I, Charkaoui M, Michiels C, Bardou M, Luu M. Small Molecule Drugs in Inflammatory Bowel Diseases. Pharmaceuticals (Basel) 2021; 14:ph14070637. [PMID: 34209234 PMCID: PMC8308576 DOI: 10.3390/ph14070637] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/22/2022] Open
Abstract
Inflammatory bowel diseases (IBDs), mainly represented by Crohn’s disease (CD) and Ulcerative Colitis (UC), are chronic disorders with an unclear pathogenesis. This incurable and iterative intestinal mucosal inflammation requires the life-long use of anti-inflammatory drugs to prevent flares or relapses, which are the major providers of complications, such as small bowel strictures and intestinal perforations. The introduction of tumor necrosis factor (TNF)-alpha inhibitors and other compounds, such as anti-IL12/23 and anti-alpha4/beta7 integrin monoclonal antibodies, has considerably improved the clinical management of IBDs. They are now the standard of care, being the first-line therapy in patients with aggressive disease and in patients with moderate to severe disease with an inadequate response to conventional therapy. However, for approximately one third of all patients, their efficacy remains insufficient by a lack or loss of response due to the formation of anti-drug antibodies or compliance difficulties with parenteral formulations. To address these issues, orally administered Small Molecules Drugs (SMDs) that use a broad range of novel pharmacological pathways, such as JAK inhibitors, sphingosine-1-phosphate receptor modulators, and phosphodiesterase 4 inhibitors, have been developed for CD and UC. This article provides an updated and complete review of the most recently authorized SMDs and SMDs in phase II/III development.
Collapse
Affiliation(s)
- Inès Ben Ghezala
- INSERM, CIC1432, Plurithematic Unit, 21079 Dijon, France; (I.B.G.); (M.B.)
- Clinical Investigation Center, Plurithematic Unit, Dijon Bourgogne University Hospital, 21079 Dijon, France
- Ophthalmology Department, Dijon Bourgogne University Hospital, 21079 Dijon, France
| | - Maëva Charkaoui
- Gastroenterology Department, Dijon Bourgogne University Hospital, 21079 Dijon, France; (M.C.); (C.M.)
| | - Christophe Michiels
- Gastroenterology Department, Dijon Bourgogne University Hospital, 21079 Dijon, France; (M.C.); (C.M.)
| | - Marc Bardou
- INSERM, CIC1432, Plurithematic Unit, 21079 Dijon, France; (I.B.G.); (M.B.)
- Clinical Investigation Center, Plurithematic Unit, Dijon Bourgogne University Hospital, 21079 Dijon, France
- Gastroenterology Department, Dijon Bourgogne University Hospital, 21079 Dijon, France; (M.C.); (C.M.)
| | - Maxime Luu
- INSERM, CIC1432, Plurithematic Unit, 21079 Dijon, France; (I.B.G.); (M.B.)
- Clinical Investigation Center, Plurithematic Unit, Dijon Bourgogne University Hospital, 21079 Dijon, France
- Correspondence:
| |
Collapse
|
23
|
Wisniewski PJ, Nagarkatti M, Nagarkatti PS. Regulation of Intestinal Stem Cell Stemness by the Aryl Hydrocarbon Receptor and Its Ligands. Front Immunol 2021; 12:638725. [PMID: 33777031 PMCID: PMC7988095 DOI: 10.3389/fimmu.2021.638725] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/08/2021] [Indexed: 12/15/2022] Open
Abstract
Maintenance of intestinal homeostasis requires the integration of immunological and molecular processes together with environmental, diet, metabolic and microbial cues. Key to this homeostasis is the proper functioning of epithelial cells originating from intestinal stem cells (ISCs). While local factors and numerous molecular pathways govern the ISC niche, the conduit through which these processes work in concordance is the aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor, whose role in immunoregulation is critical at barrier surfaces. In this review, we discuss how AhR signaling is emerging as one of the critical regulators of molecular pathways involved in epithelial cell renewal. In addition, we examine the putative contribution of specific AhR ligands to ISC stemness and epithelial cell fate.
Collapse
Affiliation(s)
- Paul J Wisniewski
- Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Mitzi Nagarkatti
- Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Prakash S Nagarkatti
- Pathology, Microbiology and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
24
|
Chen Y, Ye Z, Seidler U, Tian D, Xiao F. Microenvironmental regulation of intestinal stem cells in the inflamed intestine. Life Sci 2021; 273:119298. [PMID: 33667519 DOI: 10.1016/j.lfs.2021.119298] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/14/2021] [Accepted: 02/23/2021] [Indexed: 01/21/2023]
Abstract
The rapid renewal of intestinal epithelium during homeostasis requires balanced proliferation and differentiation of intestinal stem cells (ISCs) at the base of crypt. Upon intestinal inflammation, the vigorous expansion of surviving ISCs is responsible for epithelial repair. However, it is not well depicted how ISCs adapt to the inflammatory conditions within intestinal tissue and support epithelial repair. In the intestinal inflammation, niche cells around ISCs along with their secreted niche factors can facilitate the regeneration of ISCs via niche signals. Additionally, the growth of ISCs can respond to inflammatory cells, inflammatory cytokines, and inflammatory signals. Understanding the adaptive mechanism of ISCs in supporting intestinal epithelial regeneration during inflammation is a focus on the treatment for patients with intestinal inflammation. Here, we aim to present an overview of how ISCs adapt to the acute inflammation to support intestinal repair, with a focus on the roles and interaction of niche signals.
Collapse
Affiliation(s)
- Yu Chen
- Department of Gastsroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Zhenghao Ye
- Department of Gastroenterology of Hannover Medical School, Hannover, Germany
| | - Ursula Seidler
- Department of Gastroenterology of Hannover Medical School, Hannover, Germany
| | - Dean Tian
- Department of Gastsroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Fang Xiao
- Department of Gastsroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China.
| |
Collapse
|
25
|
Sanman LE, Chen IW, Bieber JM, Steri V, Trentesaux C, Hann B, Klein OD, Wu LF, Altschuler SJ. Transit-Amplifying Cells Coordinate Changes in Intestinal Epithelial Cell-Type Composition. Dev Cell 2021; 56:356-365.e9. [PMID: 33484640 PMCID: PMC7917018 DOI: 10.1016/j.devcel.2020.12.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 11/09/2020] [Accepted: 12/29/2020] [Indexed: 12/12/2022]
Abstract
Renewing tissues have the remarkable ability to continually produce both proliferative progenitor and specialized differentiated cell types. How are complex milieus of microenvironmental signals interpreted to coordinate tissue-cell-type composition? Here, we investigate the responses of intestinal epithelium to individual and paired perturbations across eight epithelial signaling pathways. Using a high-throughput approach that combines enteroid monolayers and quantitative imaging, we identified conditions that enrich for specific cell types as well as interactions between pathways. Importantly, we found that modulation of transit-amplifying cell proliferation changes the ratio of differentiated secretory to absorptive cell types. These observations highlight an underappreciated role for transit-amplifying cells in the tuning of differentiated cell-type composition.
Collapse
Affiliation(s)
- Laura E Sanman
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ina W Chen
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jake M Bieber
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA; Graduate Program in Bioengineering, University of California, San Francisco and University of California, Berkeley, San Francisco, CA 94158, USA
| | - Veronica Steri
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Preclinical Therapeutics Core, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Coralie Trentesaux
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Byron Hann
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Preclinical Therapeutics Core, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ophir D Klein
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Pediatrics and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lani F Wu
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Steven J Altschuler
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
26
|
Yu L, Xie X, Jiang K, Hong Y, Zhou Z, Mi Y, Zhang C, Li J. Paneth cells mediated the response of intestinal stem cells at the early stage of intestinal inflammation in the chicken. Poult Sci 2020; 100:615-622. [PMID: 33518114 PMCID: PMC7858177 DOI: 10.1016/j.psj.2020.11.055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/17/2020] [Accepted: 11/23/2020] [Indexed: 11/28/2022] Open
Abstract
The rapid renewal and repair of the intestinal mucosa are based on intestinal stem cells (ISC), which are located at the crypt bottom. Paneth cells are an essential component in the crypt, which served as the niche for ISC development. However, in the chicken, how the function of Paneth cells changes during intestinal inflammation is unclear and is the key to understand the mechanism of mucosal repair. In the present study, 36 HyLine White chickens (7 d of age, n = 6) were randomly divided into 1 control and 5 lipopolysaccharide (LPS) injection groups. The chickens were injected (i.p.) with PBS in the control group, however, were injected (i.p.) with LPS (10 mg/kg BW) in the LPS injection groups, which would be sampled at 5 time points (1 h postinjection [hpi], 2 hpi, 4 hpi, 6 hpi, and 8 hpi). Results showed that tumor necrosis factor-α mRNA transcription in duodenal tissue increased gradually since 1 hpi, peaked at 4 hpi, and then reduced remarkably, indicating that 4 hpi of LPS was the early stage of intestinal inflammation. Meanwhile, the MUC2 expression in duodenal tissue was dramatically reduced since 1 hpi of LPS. The ISC marker, Lgr5 and Bmi1, in the duodenal crypt were reduced from 1 hpi to 4 hpi and elevated later. Accordingly, the hydroethidine staining showed that the reactive oxygen species level, which drives the differentiation of ISC, in the duodenal crypt reduced obviously at 1 hpi and recovered gradually since 4 hpi. The analysis of Paneth cells showed that many swollen mitochondria appeared in Paneth cells at 4 hpi of LPS. Meanwhile, the Lysozyme transcription in the duodenal crypt was substantially decreased since 1 hpi of LPS. However, the Wnt3a and Dll1 in duodenal crypt decreased at 1 hpi of LPS, then increased gradually. In conclusion, Paneth cells were impaired at the early stage of intestinal inflammation, then recovered rapidly. Thus, the ISC activity was reduced at first and recovery soon.
Collapse
Affiliation(s)
- Lingzi Yu
- Department of Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Xiaochen Xie
- Department of Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Keyang Jiang
- Department of Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Yi Hong
- Department of Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Zhou Zhou
- Department of Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Yuling Mi
- Department of Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Caiqiao Zhang
- Department of Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, P.R. China
| | - Jian Li
- Department of Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou 310058, P.R. China.
| |
Collapse
|
27
|
Kumar S, Suman S, Fornace AJ, Datta K. Intestinal stem cells acquire premature senescence and senescence associated secretory phenotype concurrent with persistent DNA damage after heavy ion radiation in mice. Aging (Albany NY) 2020; 11:4145-4158. [PMID: 31239406 PMCID: PMC6629005 DOI: 10.18632/aging.102043] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/17/2019] [Indexed: 12/19/2022]
Abstract
Heavy ion radiation, prevalent in outer space and relevant for radiotherapy, is densely ionizing and poses risk to stem cells that are key to intestinal homeostasis. Currently, the molecular spectrum of heavy ion radiation-induced perturbations in intestinal stem cells (ISCs), that could trigger intestinal pathologies, remains largely unexplored. The Lgr5-EGFP-IRES-creERT mice were exposed to 50 cGy of iron radiation. Mice were euthanized 60 d after exposure and ISCs were sorted using fluorescence activated cell sorting. Reactive oxygen species (ROS) and mitochondrial superoxide were measured using fluorescent probes. Since DNA damage is linked to senescence and senescent cells acquire senescence-associated secretory phenotype (SASP), we stained ISCs for both senescence markers p16, p21, and p19 as well as SASP markers IL6, IL8, and VEGF. Due to potential positive effects of SASP on proliferation, we also stained for PCNA. Data show increased ROS and ongoing DNA damage, by staining for γH2AX, and 53BP1, along with accumulation of senescence markers. Results also showed increased SASP markers in senescent cells. Collectively, our data suggest that heavy-ion-induced chronic stress and ongoing DNA damage is promoting SASP in a fraction of the ISCs, which has implications for gastrointestinal function, inflammation, and carcinogenesis in astronauts and patients.
Collapse
Affiliation(s)
- Santosh Kumar
- Department of Biochemistry and Molecular & Cellular Biology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Shubhankar Suman
- Department of Biochemistry and Molecular & Cellular Biology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Albert J Fornace
- Department of Biochemistry and Molecular & Cellular Biology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| | - Kamal Datta
- Department of Biochemistry and Molecular & Cellular Biology and Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA
| |
Collapse
|
28
|
IRF2 is a master regulator of human keratinocyte stem cell fate. Nat Commun 2019; 10:4676. [PMID: 31611556 PMCID: PMC6791852 DOI: 10.1038/s41467-019-12559-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 09/14/2019] [Indexed: 12/25/2022] Open
Abstract
Resident adult epithelial stem cells maintain tissue homeostasis by balancing self-renewal and differentiation. The stem cell potential of human epidermal keratinocytes is retained in vitro but lost over time suggesting extrinsic and intrinsic regulation. Transcription factor-controlled regulatory circuitries govern cell identity, are sufficient to induce pluripotency and transdifferentiate cells. We investigate whether transcriptional circuitry also governs phenotypic changes within a given cell type by comparing human primary keratinocytes with intrinsically high versus low stem cell potential. Using integrated chromatin and transcriptional profiling, we implicate IRF2 as antagonistic to stemness and show that it binds and regulates active cis-regulatory elements at interferon response and antigen presentation genes. CRISPR-KD of IRF2 in keratinocytes with low stem cell potential increases self-renewal, migration and epidermis formation. These data demonstrate that transcription factor regulatory circuitries, in addition to maintaining cell identity, control plasticity within cell types and offer potential for therapeutic modulation of cell function. Epidermal homeostasis requires long term stem cell function. Here, the authors apply transcriptional circuitry analysis based on integrated epigenomic profiling of primary human keratinocytes with high and low stem cell function to identify IRF2 as a negative regulator of stemness.
Collapse
|
29
|
Kopecki Z, Yang G, Treloar S, Mashtoub S, Howarth GS, Cummins AG, Cowin AJ. Flightless I exacerbation of inflammatory responses contributes to increased colonic damage in a mouse model of dextran sulphate sodium-induced ulcerative colitis. Sci Rep 2019; 9:12792. [PMID: 31488864 PMCID: PMC6728368 DOI: 10.1038/s41598-019-49129-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/15/2019] [Indexed: 12/13/2022] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by cytokine driven inflammation that disrupts the mucosa and impedes intestinal structure and functions. Flightless I (Flii) is an immuno-modulatory protein is a member of the gelsolin family of actin-remodelling proteins that regulates cellular and inflammatory processes critical in tissue repair. Here we investigated its involvement in UC and show that Flii is significantly elevated in colonic tissues of patients with inflammatory bowel disease. Using an acute murine model of colitis, we characterised the contribution of Flii to UC using mice with low (Flii+/-), normal (Flii+/+) and high Flii (FliiTg/Tg). High levels of Flii resulted in significantly elevated disease severity index scores, increased rectal bleeding and degree of colon shortening whereas, low Flii expression decreased disease severity, reduced tissue inflammation and improved clinical indicators of UC. Mice with high levels of Flii had significantly increased histological disease severity and elevated mucosal damage with significantly increased inflammatory cell infiltrate and significantly higher levels of TNF-α, IFN-γ, IL-5 and IL-13 pro-inflammatory cytokines. Additionally, Flii overexpression resulted in decreased β-catenin levels, inhibited Wnt/β-catenin signalling and impaired regeneration of colonic crypts. These studies suggest that high levels of Flii, as is observed in patients with UC, may adversely affect mucosal healing via mechanisms involving Th1 and Th2 mediated tissue inflammation and Wnt/β-catenin signalling pathway.
Collapse
Affiliation(s)
- Z Kopecki
- Regenerative Medicine, Future Industries Institute, University of South Australia, Mawson Lakes, Adelaide, South Australia, Australia.
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia.
| | - G Yang
- Regenerative Medicine, Future Industries Institute, University of South Australia, Mawson Lakes, Adelaide, South Australia, Australia
| | - S Treloar
- School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, Adelaide, South Australia, Australia
| | - S Mashtoub
- Department of Gastroenterology, Women's and Children's Hospital, North Adelaide, South Australia, Australia
- Discipline of Physiology, Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - G S Howarth
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - A G Cummins
- Department of Gastroenterology and Hepatology, The Queen Elizabeth Hospital, Woodville South, Adelaide, South Australia, Australia
| | - A J Cowin
- Regenerative Medicine, Future Industries Institute, University of South Australia, Mawson Lakes, Adelaide, South Australia, Australia
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| |
Collapse
|
30
|
Perner F, Perner C, Ernst T, Heidel FH. Roles of JAK2 in Aging, Inflammation, Hematopoiesis and Malignant Transformation. Cells 2019; 8:cells8080854. [PMID: 31398915 PMCID: PMC6721738 DOI: 10.3390/cells8080854] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 12/22/2022] Open
Abstract
Clonal alterations in hematopoietic cells occur during aging and are often associated with the establishment of a subclinical inflammatory environment. Several age-related conditions and diseases may be initiated or promoted by these alterations. JAK2 mutations are among the most frequently mutated genes in blood cells during aging. The most common mutation within the JAK2 gene is JAK2-V617F that leads to constitutive activation of the kinase and thereby aberrant engagement of downstream signaling pathways. JAK2 mutations can act as central drivers of myeloproliferative neoplasia, a pre-leukemic and age-related malignancy. Likewise, hyperactive JAK-signaling is a hallmark of immune diseases and critically influences inflammation, coagulation and thrombosis. In this review we aim to summarize the current knowledge on JAK2 in clonal hematopoiesis during aging, the role of JAK-signaling in inflammation and lymphocyte biology and JAK2 function in age-related diseases and malignant transformation.
Collapse
Affiliation(s)
- Florian Perner
- Innere Medizin 2, Hämatologie und Onkologie, Universitätsklinikum Jena, 07747 Jena, Germany
- Leibniz-Institute on Aging-Fritz Lipmann Institute (FLI), 07745 Jena, Germany
- Dana-Farber Cancer Institute, Department of Pediatric Oncology, Harvard University, Boston, MA 02467, USA
| | - Caroline Perner
- Center for Immunology & Inflammatory Diseases, Massachusetts General Hospital, and Harvard Medical School, Boston, 02129 MA, USA
| | - Thomas Ernst
- Innere Medizin 2, Hämatologie und Onkologie, Universitätsklinikum Jena, 07747 Jena, Germany
| | - Florian H Heidel
- Innere Medizin 2, Hämatologie und Onkologie, Universitätsklinikum Jena, 07747 Jena, Germany.
- Leibniz-Institute on Aging-Fritz Lipmann Institute (FLI), 07745 Jena, Germany.
| |
Collapse
|
31
|
Wnt signaling in intestinal inflammation. Differentiation 2019; 108:24-32. [DOI: 10.1016/j.diff.2019.01.002] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/17/2018] [Accepted: 01/18/2019] [Indexed: 12/12/2022]
|
32
|
Gonzalez LM, Stewart AS, Freund J, Kucera CR, Dekaney CM, Magness ST, Blikslager AT. Preservation of reserve intestinal epithelial stem cells following severe ischemic injury. Am J Physiol Gastrointest Liver Physiol 2019; 316:G482-G494. [PMID: 30714814 PMCID: PMC6483022 DOI: 10.1152/ajpgi.00262.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Intestinal ischemia is an abdominal emergency with a mortality rate >50%, leading to epithelial barrier loss and subsequent sepsis. Epithelial renewal and repair after injury depend on intestinal epithelial stem cells (ISC) that reside within the crypts of Lieberkühn. Two ISC populations critical to epithelial repair have been described: 1) active ISC (aISC; highly proliferative; leucine-rich-repeat-containing G protein-coupled receptor 5 positive, sex determining region Y-box 9 positive) and 2) reserve ISC [rISC; less proliferative; homeodomain only protein X (Hopx)+]. Yorkshire crossbred pigs (8-10 wk old) were subjected to 1-4 h of ischemia and 1 h of reperfusion or recovery by reversible mesenteric vascular occlusion. This study was designed to evaluate whether ISC-expressing biomarkers of aISCs or rISCs show differential resistance to ischemic injury and different contributions to the subsequent repair and regenerative responses. Our data demonstrate that, following 3-4 h ischemic injury, aISC undergo apoptosis, whereas rISC are preserved. Furthermore, these rISC are retained ex vivo in spheroids in which cell populations are enriched in the rISC biomarker Hopx. These cells appear to go on to provide a proliferative pool of cells during the recovery period. Taken together, these data indicate that Hopx+ cells are resistant to injury and are the likely source of epithelial renewal following prolonged ischemic injury. It is therefore possible that targeting reserve stem cells will lead to new therapies for patients with severe intestinal injury. NEW & NOTEWORTHY The population of reserve less-proliferative intestinal epithelial stem cells appears resistant to injury despite severe epithelial cell loss, including that of the active stem cell population, which results from prolonged mesenteric ischemia. These cells can change to an activated state and are likely indispensable to regenerative processes. Reserve stem cell targeted therapies may improve treatment and outcome of patients with ischemic disease.
Collapse
Affiliation(s)
- Liara M. Gonzalez
- 1Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina,4Center for Gastrointestinal Biologyand Disease, Joint Center at University of North Carolina Chapel Hill and North Carolina State University, Raleigh,North Carolina
| | - Amy Stieler Stewart
- 1Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - John Freund
- 1Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Cecilia Renee Kucera
- 1Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina
| | - Christopher M. Dekaney
- 2Department of Molecular and Biological Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina,4Center for Gastrointestinal Biologyand Disease, Joint Center at University of North Carolina Chapel Hill and North Carolina State University, Raleigh,North Carolina
| | - Scott T. Magness
- 3University of North Carolina, Chapel Hill, North Carolina,4Center for Gastrointestinal Biologyand Disease, Joint Center at University of North Carolina Chapel Hill and North Carolina State University, Raleigh,North Carolina
| | - Anthony T. Blikslager
- 1Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina,4Center for Gastrointestinal Biologyand Disease, Joint Center at University of North Carolina Chapel Hill and North Carolina State University, Raleigh,North Carolina
| |
Collapse
|
33
|
Parham LR, Williams PA, Chatterji P, Whelan KA, Hamilton KE. RNA regulons are essential in intestinal homeostasis. Am J Physiol Gastrointest Liver Physiol 2019; 316:G197-G204. [PMID: 30520692 PMCID: PMC6383383 DOI: 10.1152/ajpgi.00403.2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Intestinal epithelial cells are among the most rapidly proliferating cell types in the human body. There are several different subtypes of epithelial cells, each with unique functional roles in responding to the ever-changing environment. The epithelium's ability for rapid and customized responses to environmental changes requires multitiered levels of gene regulation. An emerging paradigm in gastrointestinal epithelial cells is the regulation of functionally related mRNA families, or regulons, via RNA-binding proteins (RBPs). RBPs represent a rapid and efficient mechanism to regulate gene expression and cell function. In this review, we will provide an overview of intestinal epithelial RBPs and how they contribute specifically to intestinal epithelial stem cell dynamics. In addition, we will highlight key gaps in knowledge in the global understanding of RBPs in gastrointestinal physiology as an opportunity for future studies.
Collapse
Affiliation(s)
- Louis R. Parham
- 1Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Patrick A. Williams
- 1Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Priya Chatterji
- 2Division of Gastroenterology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kelly A. Whelan
- 3Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania,4Fels Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Kathryn E. Hamilton
- 1Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
34
|
May S, Owen H, Phesse TJ, Greenow KR, Jones G, Blackwood A, Cook PC, Towers C, Gallimore AM, Williams GT, Stürzl M, Britzen‐Laurent N, Sansom OJ, MacDonald AS, Bird AP, Clarke AR, Parry L. Mbd2 enables tumourigenesis within the intestine while preventing tumour-promoting inflammation. J Pathol 2018; 245:270-282. [PMID: 29603746 PMCID: PMC6032908 DOI: 10.1002/path.5074] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 02/22/2018] [Accepted: 03/08/2018] [Indexed: 01/14/2023]
Abstract
Epigenetic regulation plays a key role in the link between inflammation and cancer. Here we examine Mbd2, which mediates epigenetic transcriptional silencing by binding to methylated DNA. In separate studies the Mbd2-/- mouse has been shown (1) to be resistant to intestinal tumourigenesis and (2) to have an enhanced inflammatory/immune response, observations that are inconsistent with the links between inflammation and cancer. To clarify its role in tumourigenesis and inflammation, we used constitutive and conditional models of Mbd2 deletion to explore its epithelial and non-epithelial roles in the intestine. Using a conditional model, we found that suppression of intestinal tumourigenesis is due primarily to the absence of Mbd2 within the epithelia. Next, we demonstrated, using the DSS colitis model, that non-epithelial roles of Mbd2 are key in preventing the transition from acute to tumour-promoting chronic inflammation. Combining models revealed that prior to inflammation the altered Mbd2-/- immune response plays a role in intestinal tumour suppression. However, following inflammation the intestine converts from tumour suppressive to tumour promoting. To summarise, in the intestine the normal function of Mbd2 is exploited by cancer cells to enable tumourigenesis, while in the immune system it plays a key role in preventing tumour-enabling inflammation. Which role is dominant depends on the inflammation status of the intestine. As environmental interactions within the intestine can alter DNA methylation patterns, we propose that Mbd2 plays a key role in determining whether these interactions are anti- or pro-tumourigenic and this makes it a useful new epigenetic model for inflammation-associated carcinogenesis. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Stephanie May
- European Cancer Stem Cell Research InstituteCardiff University, School of BiosciencesCardiffUK
| | - Heather Owen
- Wellcome Trust Centre for Cell BiologyUniversity of Edinburgh, Michael Swann BuildingEdinburghUK
| | - Toby J Phesse
- European Cancer Stem Cell Research InstituteCardiff University, School of BiosciencesCardiffUK
| | - Kirsty R Greenow
- European Cancer Stem Cell Research InstituteCardiff University, School of BiosciencesCardiffUK
| | - Gareth‐Rhys Jones
- Manchester Collaborative Centre for Inflammation ResearchManchesterUK
| | - Adam Blackwood
- European Cancer Stem Cell Research InstituteCardiff University, School of BiosciencesCardiffUK
| | - Peter C Cook
- Manchester Collaborative Centre for Inflammation ResearchManchesterUK
| | - Christopher Towers
- European Cancer Stem Cell Research InstituteCardiff University, School of BiosciencesCardiffUK
| | - Awen M Gallimore
- Cardiff Institute of Infection and Immunity, Henry Wellcome BuildingCardiffUK
| | - Geraint T Williams
- Institute of Cancer and GeneticsCardiff University School of MedicineCardiffUK
| | - Michael Stürzl
- Division of Molecular and Experimental Surgery, Department of SurgeryFriedrich‐Alexander‐Universität (FAU) Erlangen‐Nürnberg and Universitätsklinikum ErlangenErlangenGermany
| | - Nathalie Britzen‐Laurent
- Division of Molecular and Experimental Surgery, Department of SurgeryFriedrich‐Alexander‐Universität (FAU) Erlangen‐Nürnberg and Universitätsklinikum ErlangenErlangenGermany
| | | | | | - Adrian P Bird
- Wellcome Trust Centre for Cell BiologyUniversity of Edinburgh, Michael Swann BuildingEdinburghUK
| | - Alan R Clarke
- European Cancer Stem Cell Research InstituteCardiff University, School of BiosciencesCardiffUK
| | - Lee Parry
- European Cancer Stem Cell Research InstituteCardiff University, School of BiosciencesCardiffUK
| |
Collapse
|