51
|
Fernandes MA, Usatin D, Allen IE, Rhee S, Vu L. Improved enteral tolerance following step procedure: systematic literature review and meta-analysis. Pediatr Surg Int 2016; 32:921-6. [PMID: 27461428 DOI: 10.1007/s00383-016-3927-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/20/2016] [Indexed: 10/21/2022]
Abstract
Surgical management of children with short bowel syndrome (SBS) changed with the introduction of the serial transverse enteroplasty procedure (STEP). We conducted a systematic review and meta-analysis using MEDLINE and SCOPUS to determine if children with SBS had improved enteral tolerance following STEP. Studies were included if information about a child's pre- and post-STEP enteral tolerance was provided. A random effects meta-analysis provided a summary estimate of the proportion of children with enteral tolerance increase following STEP. From 766 abstracts, seven case series involving 86 children were included. Mean percent tolerance of enteral nutrition improved from 35.1 to 69.5. Sixteen children had no enteral improvement following STEP. A summary estimate showed that 87 % (95 % CI 77-95 %) of children who underwent STEP had an increase in enteral tolerance. Compilation of the literature supports the belief that SBS subjects' enteral tolerance improves following STEP. Enteral nutritional tolerance is a measure of efficacy of STEP and should be presented as a primary or secondary outcome. By standardizing data collection on children undergoing STEP procedure, better determination of nutritional benefit from STEP can be ascertained.
Collapse
Affiliation(s)
- Melissa A Fernandes
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, San Francisco, USA
| | - Danielle Usatin
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, San Francisco, USA
| | - Isabel E Allen
- Department of Epidemiology and Biostatistics, School of Medicine, University of California, San Francisco, San Francisco, USA
| | - Sue Rhee
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Benioff Children's Hospital, University of California, San Francisco, San Francisco, USA
| | - Lan Vu
- Division of Pediatric Surgery, Department of Surgery, Benioff Children's Hospital, University of California, San Francisco, 550 16th Street 5th Floor, San Francisco, CA, 94143, USA.
| |
Collapse
|
52
|
Enteric nervous system assembly: Functional integration within the developing gut. Dev Biol 2016; 417:168-81. [PMID: 27235816 DOI: 10.1016/j.ydbio.2016.05.030] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 04/26/2016] [Accepted: 05/24/2016] [Indexed: 02/08/2023]
Abstract
Co-ordinated gastrointestinal function is the result of integrated communication between the enteric nervous system (ENS) and "effector" cells in the gastrointestinal tract. Unlike smooth muscle cells, interstitial cells, and the vast majority of cell types residing in the mucosa, enteric neurons and glia are not generated within the gut. Instead, they arise from neural crest cells that migrate into and colonise the developing gastrointestinal tract. Although they are "later" arrivals into the developing gut, enteric neural crest-derived cells (ENCCs) respond to many of the same secreted signalling molecules as the "resident" epithelial and mesenchymal cells, and several factors that control the development of smooth muscle cells, interstitial cells and epithelial cells also regulate ENCCs. Much progress has been made towards understanding the migration of ENCCs along the gastrointestinal tract and their differentiation into neurons and glia. However, our understanding of how enteric neurons begin to communicate with each other and extend their neurites out of the developing plexus layers to innervate the various cell types lining the concentric layers of the gastrointestinal tract is only beginning. It is critical for postpartum survival that the gastrointestinal tract and its enteric circuitry are sufficiently mature to cope with the influx of nutrients and their absorption that occurs shortly after birth. Subsequently, colonisation of the gut by immune cells and microbiota during postnatal development has an important impact that determines the ultimate outline of the intrinsic neural networks of the gut. In this review, we describe the integrated development of the ENS and its target cells.
Collapse
|
53
|
Abstract
Antibiotics are by far the most common medications prescribed for children. Recent epidemiological data suggests an association between early antibiotic use and disease phenotypes in adulthood. Antibiotic use during infancy induces imbalances in gut microbiota, called dysbiosis. The gut microbiome's responses to antibiotics and its potential link to disease development are especially complex to study in the changing infant gut. Here, we synthesize current knowledge linking antibiotics, dysbiosis, and disease and propose a framework for studying antibiotic-related dysbiosis in children. We recommend future studies into the microbiome-mediated effects of antibiotics focused on four types of dysbiosis: loss of keystone taxa, loss of diversity, shifts in metabolic capacity, and blooms of pathogens. Establishment of a large and diverse baseline cohort to define healthy infant microbiome development is essential to advancing diagnosis, interpretation, and eventual treatment of pediatric dysbiosis. This approach will also help provide evidence-based recommendations for antibiotic usage in infancy.
Collapse
Affiliation(s)
- Pajau Vangay
- Biomedical Informatics and Computational Biology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Tonya Ward
- Biotechnology Institute, University of Minnesota, Saint Paul, MN 55108, USA
| | - Jeffrey S Gerber
- Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Dan Knights
- Biotechnology Institute, University of Minnesota, Saint Paul, MN 55108, USA; Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
54
|
O'Hara K, Wright IMR, Schneider JJ, Jones AL, Martin JH. Pharmacokinetics in neonatal prescribing: evidence base, paradigms and the future. Br J Clin Pharmacol 2015; 80:1281-8. [PMID: 26256466 PMCID: PMC4693494 DOI: 10.1111/bcp.12741] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 07/15/2015] [Accepted: 08/03/2015] [Indexed: 01/21/2023] Open
Abstract
Paediatric patients, particularly preterm neonates, present many pharmacological challenges. Due to the difficulty in conducting clinical trials in these populations dosing information is often extrapolated from adult populations. As the processes of absorption, distribution, metabolism and excretion of drugs change throughout growth and development extrapolation presents risk of over or underestimating the doses required. Information about the development these processes, particularly drug metabolism pathways, is still limited with weight based dose adjustment presenting the best method of estimating pharmacokinetic changes due to growth and development. New innovations in pharmacokinetic research, such as population pharmacokinetic modelling, present unique opportunities to conduct clinical trials in these populations improving the safety and effectiveness of the drugs used. More research is required into this area to ensure the best outcomes for our most vulnerable patients.
Collapse
Affiliation(s)
- Kate O'Hara
- School of Medicine and Public HealthUniversity of NewcastleNewcastle
| | - Ian M. R. Wright
- University of Wollongong and Illawarra Health & Medical Research InstituteWollongong
- University of Newcastle and Hunter Medical Research InstituteNewcastle
- Clinical Lead, Newborn Services, Division of PaediatricsIllawarra Shoalhaven Local Health DistrictNSW
| | | | - Alison L. Jones
- School of Medicine and Public HealthUniversity of NewcastleNewcastle
- Executive Dean Faculty Science, Medicine and HealthUniversity of Wollongong, Wollongong
- Deputy DirectorIllawarra Health and Medical Research InstituteWollongongAustralia
| | | |
Collapse
|
55
|
More similar than you think: Frog metamorphosis as a model of human perinatal endocrinology. Dev Biol 2015; 408:188-95. [DOI: 10.1016/j.ydbio.2015.02.018] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 02/05/2015] [Accepted: 02/20/2015] [Indexed: 11/21/2022]
|
56
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015; 9:392. [PMID: 26528128 PMCID: PMC4604320 DOI: 10.3389/fncel.2015.00392] [Citation(s) in RCA: 649] [Impact Index Per Article: 72.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 09/21/2015] [Indexed: 12/12/2022] Open
Abstract
The emerging links between our gut microbiome and the central nervous system (CNS) are regarded as a paradigm shift in neuroscience with possible implications for not only understanding the pathophysiology of stress-related psychiatric disorders, but also their treatment. Thus the gut microbiome and its influence on host barrier function is positioned to be a critical node within the brain-gut axis. Mounting preclinical evidence broadly suggests that the gut microbiota can modulate brain development, function and behavior by immune, endocrine and neural pathways of the brain-gut-microbiota axis. Detailed mechanistic insights explaining these specific interactions are currently underdeveloped. However, the concept that a "leaky gut" may facilitate communication between the microbiota and these key signaling pathways has gained traction. Deficits in intestinal permeability may underpin the chronic low-grade inflammation observed in disorders such as depression and the gut microbiome plays a critical role in regulating intestinal permeability. In this review we will discuss the possible role played by the gut microbiota in maintaining intestinal barrier function and the CNS consequences when it becomes disrupted. We will draw on both clinical and preclinical evidence to support this concept as well as the key features of the gut microbiota which are necessary for normal intestinal barrier function.
Collapse
Affiliation(s)
- John R Kelly
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork Cork, Ireland ; Department of Psychiatry and Neurobehavioural Science, University College Cork Cork, Ireland
| | - Paul J Kennedy
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork Cork, Ireland
| | - John F Cryan
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork Cork, Ireland ; Department of Anatomy and Neuroscience, University College Cork Cork, Ireland
| | - Timothy G Dinan
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork Cork, Ireland ; Department of Psychiatry and Neurobehavioural Science, University College Cork Cork, Ireland
| | - Gerard Clarke
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork Cork, Ireland ; Department of Psychiatry and Neurobehavioural Science, University College Cork Cork, Ireland
| | - Niall P Hyland
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork Cork, Ireland ; Department of Pharmacology and Therapeutics, University College Cork Cork, Ireland
| |
Collapse
|
57
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 order by 1-- -] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
58
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 order by 8029-- -] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
59
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 order by 8029-- #] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
60
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 order by 1-- gadu] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
61
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 order by 8029-- awyx] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
62
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 order by 1-- #] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
63
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 and 1880=1880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
64
|
Gomes AS, Alves RN, Rønnestad I, Power DM. Orchestrating change: The thyroid hormones and GI-tract development in flatfish metamorphosis. Gen Comp Endocrinol 2015; 220:2-12. [PMID: 24975541 DOI: 10.1016/j.ygcen.2014.06.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 06/06/2014] [Accepted: 06/10/2014] [Indexed: 10/25/2022]
Abstract
Metamorphosis in flatfish (Pleuronectiformes) is a late post-embryonic developmental event that prepares the organism for the larval-to-juvenile transition. Thyroid hormones (THs) play a central role in flatfish metamorphosis and the basic elements that constitute the thyroid axis in vertebrates are all present at this stage. The advantage of using flatfish to study the larval-to-juvenile transition is the profound change in external morphology that accompanies metamorphosis making it easy to track progression to climax. This important lifecycle transition is underpinned by molecular, cellular, structural and functional modifications of organs and tissues that prepare larvae for a successful transition to the adult habitat and lifestyle. Understanding the role of THs in the maturation of organs and tissues with diverse functions during metamorphosis is a major challenge. The change in diet that accompanies the transition from a pelagic larvae to a benthic juvenile in flatfish is associated with structural and functional modifications in the gastrointestinal tract (GI-tract). The present review will focus on the maturation of the GI-tract during metamorphosis giving particular attention to organogenesis of the stomach a TH triggered event. Gene transcripts and biological processes that are associated with GI-tract maturation during Atlantic halibut metamorphosis are identified. Gene ontology analysis reveals core biological functions and putative TH-responsive genes that underpin TH-driven metamorphosis of the GI-tract in Atlantic halibut. Deciphering the specific role remains a challenge. Recent advances in characterizing the molecular, structural and functional modifications that accompany the appearance of a functional stomach in Atlantic halibut are considered and future research challenges identified.
Collapse
Affiliation(s)
- A S Gomes
- Department of Biology, University of Bergen, 5020 Bergen, Norway
| | - R N Alves
- Centre for Marine Sciences (CCMAR), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - I Rønnestad
- Department of Biology, University of Bergen, 5020 Bergen, Norway
| | - D M Power
- Centre for Marine Sciences (CCMAR), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal.
| |
Collapse
|
65
|
An N, Shi X, Zhang Y, Lv N, Feng L, Di X, Han N, Wang G, Cheng S, Zhang K. Discovery of a Novel Immune Gene Signature with Profound Prognostic Value in Colorectal Cancer: A Model of Cooperativity Disorientation Created in the Process from Development to Cancer. PLoS One 2015; 10:e0137171. [PMID: 26325386 PMCID: PMC4556644 DOI: 10.1371/journal.pone.0137171] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 08/13/2015] [Indexed: 02/06/2023] Open
Abstract
Immune response-related genes play a major role in colorectal carcinogenesis by mediating inflammation or immune-surveillance evasion. Although remarkable progress has been made to investigate the underlying mechanism, the understanding of the complicated carcinogenesis process was enormously hindered by large-scale tumor heterogeneity. Development and carcinogenesis share striking similarities in their cellular behavior and underlying molecular mechanisms. The association between embryonic development and carcinogenesis makes embryonic development a viable reference model for studying cancer thereby circumventing the potentially misleading complexity of tumor heterogeneity. Here we proposed that the immune genes, responsible for intra-immune cooperativity disorientation (defined in this study as disruption of developmental expression correlation patterns during carcinogenesis), probably contain untapped prognostic resource of colorectal cancer. In this study, we determined the mRNA expression profile of 137 human biopsy samples, including samples from different stages of human colonic development, colorectal precancerous progression and colorectal cancer samples, among which 60 were also used to generate miRNA expression profile. We originally established Spearman correlation transition model to quantify the cooperativity disorientation associated with the transition from normal to precancerous to cancer tissue, in conjunction with miRNA-mRNA regulatory network and machine learning algorithm to identify genes with prognostic value. Finally, a 12-gene signature was extracted, whose prognostic value was evaluated using Kaplan–Meier survival analysis in five independent datasets. Using the log-rank test, the 12-gene signature was closely related to overall survival in four datasets (GSE17536, n = 177, p = 0.0054; GSE17537, n = 55, p = 0.0039; GSE39582, n = 562, p = 0.13; GSE39084, n = 70, p = 0.11), and significantly associated with disease-free survival in four datasets (GSE17536, n = 177, p = 0.0018; GSE17537, n = 55, p = 0.016; GSE39582, n = 557, p = 4.4e-05; GSE14333, n = 226, p = 0.032). Cox regression analysis confirmed that the 12-gene signature was an independent factor in predicting colorectal cancer patient’s overall survival (hazard ratio: 1.759; 95% confidence interval: 1.126–2.746; p = 0.013], as well as disease-free survival (hazard ratio: 2.116; 95% confidence interval: 1.324–3.380; p = 0.002).
Collapse
Affiliation(s)
- Ning An
- State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaoyu Shi
- State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yueming Zhang
- Department of Endoscopy, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Ning Lv
- Department of Pathology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Lin Feng
- State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xuebing Di
- State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Naijun Han
- State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Guiqi Wang
- Department of Endoscopy, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Shujun Cheng
- State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- * E-mail: (SC); (KZ)
| | - Kaitai Zhang
- State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- * E-mail: (SC); (KZ)
| |
Collapse
|
66
|
Abstract
Necrotizing enterocolitis (NEC) is a leading cause of mortality in preterm infants. This article reviews the immunologic and hematological abnormalities typically seen in infants with NEC, such as elevated plasma cytokine levels, thrombocytopenia, increased or decreased neutrophil counts, low monocyte counts, and anemia. Some of these findings may provide important diagnostic and prognostic information.
Collapse
Affiliation(s)
- Akhil Maheshwari
- Department of Pediatrics, Morsani College of Medicine, Tampa, FL 33606, USA; Department of Molecular Medicine, Morsani College of Medicine, Tampa, FL 33612, USA; Department of Community and Family Health, College of Public Health, University of South Florida, Tampa, FL 33612, USA.
| |
Collapse
|
67
|
Mould AW, Morgan MAJ, Nelson AC, Bikoff EK, Robertson EJ. Blimp1/Prdm1 Functions in Opposition to Irf1 to Maintain Neonatal Tolerance during Postnatal Intestinal Maturation. PLoS Genet 2015; 11:e1005375. [PMID: 26158850 PMCID: PMC4497732 DOI: 10.1371/journal.pgen.1005375] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 06/19/2015] [Indexed: 11/18/2022] Open
Abstract
The neonatal intestine is a very complex and dynamic organ that must rapidly adapt and remodel in response to a barrage of environmental stimuli during the first few postnatal weeks. Recent studies demonstrate that the zinc finger transcriptional repressor Blimp1/Prdm1 plays an essential role governing postnatal reprogramming of intestinal enterocytes during this period. Functional loss results in global changes in gene expression patterns, particularly in genes associated with metabolic function. Here we engineered a knock-in allele expressing an eGFP-tagged fusion protein under control of the endogenous regulatory elements and performed genome wide ChIP-seq analysis to identify direct Blimp1 targets and further elucidate the function of Blimp1 in intestinal development. Comparison with published human and mouse datasets revealed a highly conserved core set of genes including interferon-inducible promoters. Here we show that the interferon-inducible transcriptional activator Irf1 is constitutively expressed throughout fetal and postnatal intestinal epithelium development. ChIP-seq demonstrates closely overlapping Blimp1 and Irf1 peaks at key components of the MHC class I pathway in fetal enterocytes. The onset of MHC class I expression coincides with down-regulated Blimp1 expression during the suckling to weaning transition. Collectively, these experiments strongly suggest that in addition to regulating the enterocyte metabolic switch, Blimp1 functions as a gatekeeper in opposition to Irf1 to prevent premature activation of the MHC class I pathway in villus epithelium to maintain tolerance in the neonatal intestine. The transcriptional repressor Blimp1/Prdm1 plays a pivotal role in the metabolic switch that occurs in the small intestine during the suckling to weaning transition. Notably, expression profiling of perinatal Blimp1-deficient small intestine revealed premature activation of metabolic genes normally restricted to post-weaning enterocytes. To further elucidate the function of Blimp1 in intestinal development, we engineered a novel Blimp1-eGFP-fusion knock-in mouse strain to perform ChIP-seq analysis. In addition to identifying which metabolic genes are direct Blimp1 targets, ChIP-seq analysis revealed a highly conserved Blimp1/Irf-1 overlapping sites that function to control MHC class I antigen processing during acquisition of neonatal tolerance in the first weeks after birth during early colonization of the intestinal tract by commensal microorganisms. Moreover, immunohistochemical analysis of human fetal intestine suggests that a BLIMP1/IRF-1 axis may also function in human intestinal epithelium development.
Collapse
Affiliation(s)
- Arne W. Mould
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Marc A. J. Morgan
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Andrew C. Nelson
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Elizabeth K. Bikoff
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
- * E-mail: (EKB); (EJR)
| | - Elizabeth J. Robertson
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
- * E-mail: (EKB); (EJR)
| |
Collapse
|
68
|
Porcine models of digestive disease: the future of large animal translational research. Transl Res 2015; 166:12-27. [PMID: 25655839 PMCID: PMC4458388 DOI: 10.1016/j.trsl.2015.01.004] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 01/03/2015] [Accepted: 01/07/2015] [Indexed: 12/14/2022]
Abstract
There is increasing interest in nonrodent translational models for the study of human disease. The pig, in particular, serves as a useful animal model for the study of pathophysiological conditions relevant to the human intestine. This review assesses currently used porcine models of gastrointestinal physiology and disease and provides a rationale for the use of these models for future translational studies. The pig has proven its utility for the study of fundamental disease conditions such as ischemia-reperfusion injury, stress-induced intestinal dysfunction, and short bowel syndrome. Pigs have also shown great promise for the study of intestinal barrier function, surgical tissue manipulation and intervention, as well as biomaterial implantation and tissue transplantation. Advantages of pig models highlighted by these studies include the physiological similarity to human intestine and mechanisms of human disease. Emerging future directions for porcine models of human disease include the fields of transgenics and stem cell biology, with exciting implications for regenerative medicine.
Collapse
|
69
|
An efficient nano-based theranostic system for multi-modal imaging-guided photothermal sterilization in gastrointestinal tract. Biomaterials 2015; 56:206-18. [PMID: 25934293 DOI: 10.1016/j.biomaterials.2015.04.005] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 03/31/2015] [Accepted: 04/02/2015] [Indexed: 12/23/2022]
Abstract
Since understanding the healthy status of gastrointestinal tract (GI tract) is of vital importance, clinical implementation for GI tract-related disease have attracted much more attention along with the rapid development of modern medicine. Here, a multifunctional theranostic system combining X-rays/CT/photothermal/photoacoustic mapping of GI tract and imaging-guided photothermal anti-bacterial treatment is designed and constructed. PEGylated W18O49 nanosheets (PEG-W18O49) are created via a facile solvothermal method and an in situ probe-sonication approach. In terms of excellent colloidal stability, low cytotoxicity, and neglectable hemolysis of PEG-W18O49, we demonstrate the first example of high-performance four-modal imaging of GI tract by using these nanosheets as contrast agents. More importantly, due to their intrinsic absorption of NIR light, glutaraldehyde-modified PEG-W18O49 are successfully applied as fault-free targeted photothermal agents for imaging-guided killing of bacteria on a mouse infection model. Critical to pre-clinical and clinical prospects, long-term toxicity is further investigated after oral administration of these theranostic agents. These kinds of tungsten-based nanomaterials exhibit great potential as multi-modal contrast agents for directed visualization of GI tract and anti-bacterial agents for phothothermal sterilization.
Collapse
|
70
|
Functional significance of single nucleotide polymorphisms in the lactase gene in diverse US patients and evidence for a novel lactase persistence allele at -13909 in those of European ancestry. J Pediatr Gastroenterol Nutr 2015; 60:182-91. [PMID: 25625576 PMCID: PMC4308731 DOI: 10.1097/mpg.0000000000000595] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES Recent data from mainly homogeneous European and African populations implicate a 140-bp region 5' to the transcriptional start site of LCT (the lactase gene) as a regulatory site for lactase persistence and nonpersistence. Because there are no studies of US nonhomogeneous populations, we performed genotype/phenotype analysis of the -13910 and -22018 LCT single nucleotide polymorphisms (SNPs) in New England children, mostly of European ancestry. METHODS Duodenal biopsies were processed for disaccharidase activities, RNA quantification by reverse transcription polymerase chain reaction (RT-PCR), allelic expression ratios by PCR, and genotyping and SNP analysis. Results were compared with clinical information. RESULTS Lactase activity and mRNA levels, and sucrase-to-lactase ratios of enzyme activity and mRNA, showed robust correlations with genotype. None of the other LCT SNPs showed as strong a correlation with enzyme or mRNA levels as did -13910. Data were consistent, with the -13910 being the causal sequence variant instead of -22018. Four individuals heterozygous for -13910T/C had allelic expression patterns similar to individuals with -13910C/C genotypes; of these, 2 showed equal LCT expression from the 2 alleles and a novel variant (-13909C>A) associated with lactase persistence. CONCLUSIONS The identification of -13910C/C genotype is likely to predict lactase nonpersistence, consistent with prior published studies. A -13910T/T genotype will frequently, but not perfectly, predict lactase persistence in this mixed European-ancestry population; a -13910T/C genotype will not predict the phenotype. A long, rare haplotype in 2 individuals with -13910T/C genotype but equal allele-specific expression contains a novel lactase persistence allele present at -13909.
Collapse
|
71
|
The transcription factor FOXA2 suppresses gastric tumorigenesis in vitro and in vivo. Dig Dis Sci 2015; 60:109-17. [PMID: 25129104 DOI: 10.1007/s10620-014-3290-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 07/11/2014] [Indexed: 01/04/2023]
Abstract
BACKGROUND AND AIMS The transcription factor forkhead box A2 (FOXA2) plays a central role in the development of endoderm-derived organs. It has been reported that FOXA2 acts as a suppressor in many kinds of tumor. However, little is known about the role of FOXA2 in gastric cancer. METHODS The expression of FOXA2 in gastric cancer tissue samples from 89 patients was assessed by immunohistochemistry, and the clinicopathological characteristics of the samples were analyzed. The human gastric cancer cell line, BGC-823, was used to investigate the effects of FOXA2 in gastric cancer in vitro and in vivo and the potential mechanism involved was explored. RESULTS FOXA2 expression in human gastric cancer cell lines and human gastric cancer tissues was lower compared with the normal gastric epithelium cell line GES1 and normal adult gastric tissues, respectively. Patients with high FOXA2 expression level had longer 5-year overall survival than those with low FOXA2 expression level. FOXA2 markedly inhibited growth of BGC-823 cells accompanied with the cell cycle arrest and apoptosis. Infection of BGC-823 cells by FOXA2 lentivirus resulted in reduced cell tumorigenesis in vitro and in vivo. Moreover, expression of Mucin 5AC was up-regulated along with increased expression of exogenous FOXA2 in BGC-823 cells; in contrast, dedifferentiation markers, BMI, CD54 and CD24, were down-regulated. CONCLUSIONS These results suggest that FOXA2 induces the differentiation of gastric cancer and highlight FOXA2 as a novel therapeutic target and prognostic marker for human gastric cancer.
Collapse
|
72
|
Takahashi T, Ohnishi H, Sugiura Y, Honda K, Suematsu M, Kawasaki T, Deguchi T, Fujii T, Orihashi K, Hippo Y, Watanabe T, Yamagaki T, Yuba S. Non‐neuronal acetylcholine as an endogenous regulator of proliferation and differentiation of Lgr5‐positive stem cells in mice. FEBS J 2014; 281:4672-90. [DOI: 10.1111/febs.12974] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 07/22/2014] [Accepted: 08/14/2014] [Indexed: 12/13/2022]
Affiliation(s)
- Toshio Takahashi
- Suntory Foundation for Life Sciences Bioorganic Research Institute Osaka Japan
| | - Hiroe Ohnishi
- National Institute of Advanced Industrial Science and Technology Hyogo Japan
| | - Yuki Sugiura
- Department of Biochemistry School of Medicine Keio University Tokyo Japan
- Precursory Research for Embryonic Science and Technology Tokyo Japan
| | - Kurara Honda
- Department of Biochemistry School of Medicine Keio University Tokyo Japan
- Precursory Research for Embryonic Science and Technology Tokyo Japan
| | - Makoto Suematsu
- Department of Biochemistry School of Medicine Keio University Tokyo Japan
- Japan Science Technology Agency Exploratory Research for Advanced Technology Suematsu Gas Biology Project Tokyo Japan
| | - Takashi Kawasaki
- National Institute of Advanced Industrial Science and Technology Hyogo Japan
| | - Tomonori Deguchi
- National Institute of Advanced Industrial Science and Technology Hyogo Japan
| | - Takeshi Fujii
- Department of Pharmacology Faculty of Pharmaceutical Sciences Doshisha Women's College of Liberal Arts Kyoto Japan
| | - Kaoru Orihashi
- Division of Cancer Development System National Cancer Research Institute Tokyo Japan
| | - Yoshitaka Hippo
- Division of Cancer Development System National Cancer Research Institute Tokyo Japan
| | - Takehiro Watanabe
- Suntory Foundation for Life Sciences Bioorganic Research Institute Osaka Japan
| | - Tohru Yamagaki
- Suntory Foundation for Life Sciences Bioorganic Research Institute Osaka Japan
| | - Shunsuke Yuba
- National Institute of Advanced Industrial Science and Technology Hyogo Japan
| |
Collapse
|
73
|
Rodrigues MN, Carvalho RC, Franciolli ALR, Rodrigues RF, Rigoglio NN, Jacob JCF, Gastal EL, Miglino MA. Prenatal Development of the Digestive System in the Horse. Anat Rec (Hoboken) 2014; 297:1218-27. [DOI: 10.1002/ar.22929] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 02/24/2014] [Accepted: 03/11/2014] [Indexed: 11/10/2022]
Affiliation(s)
- Marcio N. Rodrigues
- Department of Surgery; School of Veterinary Medicine and Animal Science, University of Sao Paulo; Butanta 05508-270 Sao Paulo Brazil
| | - Rafael C. Carvalho
- Department of Surgery; School of Veterinary Medicine and Animal Science, University of Sao Paulo; Butanta 05508-270 Sao Paulo Brazil
| | - Andre L. R. Franciolli
- Department of Surgery; School of Veterinary Medicine and Animal Science, University of Sao Paulo; Butanta 05508-270 Sao Paulo Brazil
| | - Rosângela F. Rodrigues
- Department of Surgery; School of Veterinary Medicine and Animal Science, University of Sao Paulo; Butanta 05508-270 Sao Paulo Brazil
| | - Nathia N. Rigoglio
- Department of Surgery; School of Veterinary Medicine and Animal Science, University of Sao Paulo; Butanta 05508-270 Sao Paulo Brazil
| | - Julio C. F. Jacob
- Department of Reproduction and Animal Evaluation; Federal Rural University of Rio de Janeiro; 23890-000 Seropedica Rio de Janeiro Brazil
| | - Eduardo L. Gastal
- Department of Animal Science; Food and Nutrition, Southern Illinois University; 62901 Carbondale Illinois
| | - Maria A. Miglino
- Department of Surgery; School of Veterinary Medicine and Animal Science, University of Sao Paulo; Butanta 05508-270 Sao Paulo Brazil
| |
Collapse
|
74
|
Aronson BE, Stapleton KA, Krasinski SD. Role of GATA factors in development, differentiation, and homeostasis of the small intestinal epithelium. Am J Physiol Gastrointest Liver Physiol 2014; 306:G474-90. [PMID: 24436352 PMCID: PMC3949026 DOI: 10.1152/ajpgi.00119.2013] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The small intestinal epithelium develops from embryonic endoderm into a highly specialized layer of cells perfectly suited for the digestion and absorption of nutrients. The development, differentiation, and regeneration of the small intestinal epithelium require complex gene regulatory networks involving multiple context-specific transcription factors. The evolutionarily conserved GATA family of transcription factors, well known for its role in hematopoiesis, is essential for the development of endoderm during embryogenesis and the renewal of the differentiated epithelium in the mature gut. We review the role of GATA factors in the evolution and development of endoderm and summarize our current understanding of the function of GATA factors in the mature small intestine. We offer perspective on the application of epigenetics approaches to define the mechanisms underlying context-specific GATA gene regulation during intestinal development.
Collapse
Affiliation(s)
- Boaz E. Aronson
- 1Division of Gastroenterology and Nutrition, Department of Medicine, Children's Hospital Boston, and Harvard Medical School, Boston, Massachusetts; ,2Department of Pediatrics, Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; and
| | - Kelly A. Stapleton
- 1Division of Gastroenterology and Nutrition, Department of Medicine, Children's Hospital Boston, and Harvard Medical School, Boston, Massachusetts;
| | - Stephen D. Krasinski
- 1Division of Gastroenterology and Nutrition, Department of Medicine, Children's Hospital Boston, and Harvard Medical School, Boston, Massachusetts; ,3Gerald J. and Dorothy R. Friedman School of Nutrition Science and Policy, Tufts University, Boston, Massachusetts
| |
Collapse
|
75
|
Guezguez A, Paré F, Benoit YD, Basora N, Beaulieu JF. Modulation of stemness in a human normal intestinal epithelial crypt cell line by activation of the WNT signaling pathway. Exp Cell Res 2014; 322:355-64. [PMID: 24534551 DOI: 10.1016/j.yexcr.2014.02.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 02/04/2014] [Accepted: 02/07/2014] [Indexed: 12/21/2022]
Abstract
The small intestine consists of two histological compartments composed of the crypts and the villi. The function of the adult small intestinal epithelium is mediated by four different types of mature cells: enterocytes, goblet, enteroendocrine and Paneth. Undifferentiated cells reside in the crypts and produce these four types of mature cells. The niche-related Wnt and Bmp signaling pathways have been suggested to be involved in the regulation and maintenance of the stem cell microenvironment. In our laboratory, we isolated the first normal human intestinal epithelial crypt (HIEC) cell model from the human fetal intestine and in this study we investigated the expression of a panel of intestinal stem cell markers in HIEC cells under normal culture parameters as well as under conditions that mimic the stem cell microenvironment. The results showed that short term stimulation of HIEC cells with R-spondin 1 and Wnt-3a±SB-216763, a glycogen synthase kinase 3β (GSK3β) inhibitor, induced β-catenin/TCF activity and expression of the WNT target genes, cyclin D2 and LGR5. Treatment of HIEC cells with noggin, an antagonist of BMP signaling, abolished SMAD2/5/8 phosphorylation. Inducing a switch from inactive WNT/active BMP toward active WNT/inactive BMP pathways was sufficient to trigger a robust intestinal primordial stem-like cell signature with predominant LGR5, PHLDA1, PROM1, SMOC2 and OLFM4 expression. These findings demonstrate that even fully established cultures of intestinal cells can be prompted toward a CBC stem cell-like phenotype. This model should be useful for studying the regulation of human intestinal stem cell self-renewal and differentiation.
Collapse
Affiliation(s)
- Amel Guezguez
- Laboratory of Intestinal Physiopathology, Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada J1H 5N4.
| | - Fréderic Paré
- Laboratory of Intestinal Physiopathology, Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada J1H 5N4.
| | - Yannick D Benoit
- Laboratory of Intestinal Physiopathology, Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada J1H 5N4.
| | - Nuria Basora
- Laboratory of Intestinal Physiopathology, Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada J1H 5N4.
| | - Jean-François Beaulieu
- Laboratory of Intestinal Physiopathology, Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada J1H 5N4.
| |
Collapse
|
76
|
Cizkova K, Konieczna A, Erdosova B, Ehrmann J. Time-dependent expression of cytochrome p450 epoxygenases during human prenatal development. Organogenesis 2014; 10:53-61. [PMID: 24492490 PMCID: PMC4049895 DOI: 10.4161/org.27911] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 01/08/2014] [Accepted: 01/19/2014] [Indexed: 11/19/2022] Open
Abstract
There is growing evidence that some members of cytochrome P450 enzymes contribute to regulation of normal prenatal development. CYP epoxygenases (CYP2C and CYP2J subfamilies) convert arachidonic acid into four regioisomeric epoxyeicosatrienoic acids (EETs), biologically active molecules involved in mitogenesis and cell signaling. Almost nothing is known about localization of their expression in tissues during human prenatal development. The spatio-temporal expression pattern of CYP2C8, CYP2C9, CYP2C19 and CYP2J2 in human embryonic/fetal intestines, liver, and kidney was investigated by immunohistochemical method. CYP epoxygenases are expressed already in early stages of development in these embryonic/fetal tissues (as early as 7th week of IUD in the intestines, 5th week of IUD in the liver, and 6th week of IUD in the kidney). In kidney, CYP epoxygenases are expressed in the metanephrogenic blastema (but not in the uninduced mesenchyme) and in the tubular system. In the intestines, diverse CYP epoxygenases distribution along crypt-villus axis could suggest role in cell differentiation. Moreover, we detected higher CYP2J2 level in these organs than in adult tissue samples.
Collapse
Affiliation(s)
- Katerina Cizkova
- Department of Histology and Embryology; Palacky University; Olomouc, Czech Republic
| | - Anna Konieczna
- Department of Histology and Embryology; Palacky University; Olomouc, Czech Republic
| | - Bela Erdosova
- Department of Histology and Embryology; Palacky University; Olomouc, Czech Republic
| | - Jiri Ehrmann
- Department of Histology and Embryology; Palacky University; Olomouc, Czech Republic
- Department of Clinical and Molecular Pathology & Laboratory of Molecular Pathology; Palacky University; Olomouc, Czech Republic
| |
Collapse
|
77
|
Fordham R, Yui S, Hannan N, Soendergaard C, Madgwick A, Schweiger P, Nielsen O, Vallier L, Pedersen R, Nakamura T, Watanabe M, Jensen K. Transplantation of expanded fetal intestinal progenitors contributes to colon regeneration after injury. Cell Stem Cell 2013; 13:734-44. [PMID: 24139758 PMCID: PMC3858813 DOI: 10.1016/j.stem.2013.09.015] [Citation(s) in RCA: 293] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 09/04/2013] [Accepted: 09/27/2013] [Indexed: 12/12/2022]
Abstract
Regeneration and homeostasis in the adult intestinal epithelium is driven by proliferative resident stem cells, whose functional properties during organismal development are largely unknown. Here, we show that human and mouse fetal intestine contains proliferative, immature progenitors, which can be expanded in vitro as Fetal Enterospheres (FEnS). A highly similar progenitor population can be established during intestinal differentiation of human induced pluripotent stem cells. Established cultures of mouse fetal intestinal progenitors express lower levels of Lgr5 than mature progenitors and propagate in the presence of the Wnt antagonist Dkk1, and new cultures can be induced to form mature intestinal organoids by exposure to Wnt3a. Following transplantation in a colonic injury model, FEnS contribute to regeneration of colonic epithelium by forming epithelial crypt-like structures expressing region-specific differentiation markers. This work provides insight into mechanisms underlying development of the mammalian intestine and points to future opportunities for patient-specific regeneration of the digestive tract.
Collapse
Affiliation(s)
- Robert P. Fordham
- Wellcome Trust & Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 1QR, UK
- Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Cambridge, CB2 0SZ, UK
| | - Shiro Yui
- BRIC: Biotech Research and Innovation Centre, University of Copenhagen, DK-2200 Copenhagen N, Denmark
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Nicholas R.F. Hannan
- Wellcome Trust & Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 1QR, UK
- Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Cambridge, CB2 0SZ, UK
| | - Christoffer Soendergaard
- Department of Gastroenterology, Medical Section, Herlev Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2730 Herlev, Denmark
| | - Alison Madgwick
- Wellcome Trust & Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 1QR, UK
| | - Pawel J. Schweiger
- BRIC: Biotech Research and Innovation Centre, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Ole H. Nielsen
- Department of Gastroenterology, Medical Section, Herlev Hospital, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2730 Herlev, Denmark
| | - Ludovic Vallier
- Wellcome Trust & Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 1QR, UK
- Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Cambridge, CB2 0SZ, UK
| | - Roger A. Pedersen
- Wellcome Trust & Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 1QR, UK
- Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Cambridge, CB2 0SZ, UK
| | - Tetsuya Nakamura
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Mamoru Watanabe
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Kim B. Jensen
- Wellcome Trust & Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, CB2 1QR, UK
- BRIC: Biotech Research and Innovation Centre, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| |
Collapse
|
78
|
Zhu MJ, Du M, Ford SP. CELL BIOLOGY SYMPOSIUM: Impacts of maternal obesity on placental and gut inflammation and health. J Anim Sci 2013; 92:1840-9. [PMID: 24243902 DOI: 10.2527/jas.2013-7106] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Obesity in pregnant women is a growing public health concern that negatively affects fetal development and has long-term impacts on offspring health. The placenta plays an essential role in nutrient transport to the fetus and supports fetal growth and development. Maternal obesity (MO) induces an exacerbated proinflammatory milieu in the placenta providing an inflammatory environment for fetuses. The gut is one of the largest immune organs and mainly develops during the fetal stage. Maternal obesity and the corresponding inflammatory uteroplacental environment affect gut development, incurring inflammatory responses in the fetal intestine that further prime or program the offspring gut to enhance inflammation and impair intestinal barrier integrity. This review summarizes the impact of MO on inflammatory responses in placenta and fetal intestine and the long-term effects on offspring intestinal health. Because "leaky gut" is one of the main etiological factors for a number of common diseases, including inflammatory bowel diseases, type I diabetes, and related autoimmune diseases, the adverse effect of MO on the overall health of progeny is further discussed.
Collapse
Affiliation(s)
- M J Zhu
- School of Food Science, Washington State University, Pullman 99164
| | | | | |
Collapse
|
79
|
Carlson SJ, Chang MI, Nandivada P, Cowan E, Puder M. Neonatal intestinal physiology and failure. Semin Pediatr Surg 2013; 22:190-4. [PMID: 24331093 DOI: 10.1053/j.sempedsurg.2013.10.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The neonatal intestine is a complex organ that regulates the absorption of nutrients essential for growth and development. Intestinal failure results from insufficient or functionally inadequate bowel and can lead to failure of neonatal growth and development. Current literature on neonatal intestinal physiology and failure was reviewed and summarized. A homeostatic interplay of electrolytes, enzymes, and hormonal regulators is essential to achieve the physiologic balance needed for adequate intestinal performance. Physiologic consequences of intestinal failure are dependent on the length and anatomic location of the diseased or surgically resected bowel. Intestinal failure leads to disruption of normal intestinal physiology and may have long-term consequences for growth and development if inadequately treated. Parenteral nutrition remains the mainstay of treatment for neonatal intestinal failure.
Collapse
Affiliation(s)
- Sarah J Carlson
- Department of Surgery and the Vascular Biology Program, Boston Children's Hospital, 300 Longwood Ave, Fegan 3, Boston, Massachusetts 02115
| | - Melissa I Chang
- Department of Surgery and the Vascular Biology Program, Boston Children's Hospital, 300 Longwood Ave, Fegan 3, Boston, Massachusetts 02115
| | - Prathima Nandivada
- Department of Surgery and the Vascular Biology Program, Boston Children's Hospital, 300 Longwood Ave, Fegan 3, Boston, Massachusetts 02115
| | - Eileen Cowan
- Department of Surgery and the Vascular Biology Program, Boston Children's Hospital, 300 Longwood Ave, Fegan 3, Boston, Massachusetts 02115
| | - Mark Puder
- Department of Surgery and the Vascular Biology Program, Boston Children's Hospital, 300 Longwood Ave, Fegan 3, Boston, Massachusetts 02115.
| |
Collapse
|
80
|
Nanthakumar NN, Meng D, Newburg DS. Glucocorticoids and microbiota regulate ontogeny of intestinal fucosyltransferase 2 requisite for gut homeostasis. Glycobiology 2013; 23:1131-41. [PMID: 23887940 PMCID: PMC3766278 DOI: 10.1093/glycob/cwt050] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 06/21/2013] [Accepted: 06/26/2013] [Indexed: 12/28/2022] Open
Abstract
At weaning, the intestinal mucosa surface glycans change from predominantly sialylated to fucosylated. Intestinal adaptation from milk to solid food is regulated by intrinsic and extrinsic factors. The contribution by glucocorticoid, an intrinsic factor, and colonization by microbiota, an extrinsic factor, was measured as the induction of α1,2/3-fucosyltransferase and sucrase-isomaltase (SI) activity and gene expression in conventionally raised, germ-free, and bacteria-depleted mice. In conventionally raised mice, cortisone acetate (CA) precociously accelerated SI gene expression up to 3 weeks and fut2 to 4 weeks of age. In germ-free mice, CA treatment induces only SI expression but not fucosyltransferase. In post-weaning bacteria-deficient (germ-free and bacteria-depleted) mice, fut2 expression remains at low suckling levels. In microbiota deficient mice, intestinal fut2 (but not fut1, fut4 or fut7) was induced only by adult microbiota, but not immature microbiota or CA. Fut2 induction could also be restored by colonization by Bacteroides fragilis, but not by a B. fragilis mutant unable to utilize fucose. Restoration of fut2 expression (by either microbiota or B. fragilis) in bacteria-depleted mice is necessary for recovery from dextran sulfate sodium-induced mucosal injury. Thus, glucocorticoids and microbes regulate distinct aspects of gut ontogeny: CA precociously accelerates SI expression and, only in colonized mice, fut2 early expression. The adult microbiota is required for the fut2 induction responsible for the highly fucosylated adult gut phenotype and is necessary for recovery from intestinal injury. Fut2-dependent recovery from inflammation may explain the high incidence of inflammatory disease (Crohn's and necrotizing enterocolitis) in populations with mutant FUT2 polymorphic alleles.
Collapse
Affiliation(s)
- N Nanda Nanthakumar
- Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, VA 24060, USA
| | - Di Meng
- Pediatrics, Harvard Medical School, Boston, MA, USA
| | - David S Newburg
- Program in Glycobiology, Department of Biology, Boston College, Chestnut Hill, MA 02467, USA
| |
Collapse
|
81
|
Sangild PT, Thymann T, Schmidt M, Stoll B, Burrin DG, Buddington RK. Invited review: the preterm pig as a model in pediatric gastroenterology. J Anim Sci 2013; 91:4713-29. [PMID: 23942716 DOI: 10.2527/jas.2013-6359] [Citation(s) in RCA: 196] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
At birth, the newborn mammal undergoes a transition from a sterile uterine environment with a constant nutrient supply, to a microbe-rich environment with intermittent oral intake of complex milk nutrients via the gastrointestinal tract (GIT). These functional challenges partly explain the relatively high morbidity and mortality of neonates. Preterm birth interrupts prenatal organ maturation, including that of the GIT, and increases disease risk. Exemplary is necrotizing enterocolitis (NEC), which is associated closely with GIT immaturity, enteral feeding, and bacterial colonization. Infants with NEC may require resection of the necrotic parts of the intestine, leading to short bowel syndrome (SBS), characterized by reduced digestive capacity, fluid loss, and dependency on parenteral nutrition. This review presents the preterm pig as a translational model in pediatric gastroenterology that has provided new insights into important pediatric diseases such as NEC and SBS. We describe protocols for delivery, care, and handling of preterm pigs, and show how the immature GIT responds to delivery method and different nutritional and therapeutic interventions. The preterm pig may also provide a sensitive model for postnatal adaptation of weak term piglets showing high mortality. Attributes of the preterm pig model include close similarities with preterm infants in body size, organ development, and many clinical features, thereby providing a translational advantage relative to rodent models of GIT immaturity. On the other hand, the need for a sow surgical facility, a piglet intensive care unit, and clinically trained personnel may limit widespread use of preterm pigs. Studies on organ adaptation in preterm pigs help to identify the physiological basis of neonatal survival for hypersensitive newborns and aid in defining the optimal diet and rearing conditions during the critical neonatal period.
Collapse
Affiliation(s)
- P T Sangild
- Department of Nutrition, Exercise, and Sports
| | | | | | | | | | | |
Collapse
|
82
|
Estores D, Velanovich V. Barrett esophagus: epidemiology, pathogenesis, diagnosis, and management. Curr Probl Surg 2013; 50:192-226. [PMID: 23601575 DOI: 10.1067/j.cpsurg.2013.01.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
83
|
Hunter AC, Elsom J, Wibroe PP, Moghimi SM. Polymeric particulate technologies for oral drug delivery and targeting: A pathophysiological perspective. Maturitas 2012; 73:5-18. [DOI: 10.1016/j.maturitas.2012.05.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 05/25/2012] [Indexed: 11/25/2022]
|
84
|
GATA-4/-6 and HNF-1/-4 families of transcription factors control the transcriptional regulation of the murine Muc5ac mucin during stomach development and in epithelial cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2012; 1819:869-76. [DOI: 10.1016/j.bbagrm.2012.04.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 04/13/2012] [Accepted: 04/17/2012] [Indexed: 02/07/2023]
|
85
|
Hunter AC, Elsom J, Wibroe PP, Moghimi SM. Polymeric particulate technologies for oral drug delivery and targeting: a pathophysiological perspective. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2012; 8 Suppl 1:S5-20. [PMID: 22846372 DOI: 10.1016/j.nano.2012.07.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 05/25/2012] [Indexed: 01/01/2023]
Abstract
The oral route for delivery of pharmaceuticals is the most widely used and accepted. Nanoparticles and microparticles are increasingly being applied within this arena to optimize drug targeting and bioavailability. Frequently the carrier systems used are either constructed from or contain polymeric materials. Examples of these nanocarriers include polymeric nanoparticles, solid lipid nanocarriers, self-nanoemulsifying drug delivery systems and nanocrystals. It is the purpose of this review to describe these cutting edge technologies and specifically focus on the interaction and fate of these polymers within the gastrointestinal system.
Collapse
Affiliation(s)
- A Christy Hunter
- University of Manchester, Department of Pharmacy and Pharmaceutical Sciences, Stopford Building, Oxford Road, Manchester, M13 9PT, United Kingdom.
| | | | | | | |
Collapse
|
86
|
Wnt blockade with dickkopf reduces intestinal crypt fission and intestinal growth in infant rats. J Pediatr Gastroenterol Nutr 2012; 55:26-31. [PMID: 22193181 DOI: 10.1097/mpg.0b013e318246b42d] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Intestinal crypt fission peaks during infancy. In human and experimental familial polyposis coli, increased crypt fission is due to activation of Wnt/β-catenin signalling, but the molecular basis of crypt fission during intestinal growth has not been examined. The aim of this project was to investigate whether crypt fission and intestinal growth are affected by experimental blockade of the Wnt/β-catenin signalling pathway. METHODS Hooded Wistar rats were given either the Wnt inhibitor, dickkopf (30 and 100 ng), daily or vehicle control intraperitoneally from days 11 to 15 and were killed at day 16. Intestinal morphometry was used to measure villous area, crypt area, percentage of crypt fission, and crypt mitotic count. Intestinal stem cells were assessed by expression of real time-polymerase chain reaction for Lgr5 (a stem cell marker), and the number of β-catenin-expressing crypts by immunostaining was determined after 100-ng dickkopf treatment. RESULTS Dickkopf at 30 and 100 ng/day reduced villous area to 71% (P = 0.013) and 29% (P < 0.0001), crypt area to 42% (P = 0.0026) and 30% (P = 0.0067), and crypt fission to 51% (P = 0.006) and 29% (P < 0.0001), respectively, of control values. Mitotic count per crypt did not change. Lgr5 RNA expression and the number of β-catenin-expressing crypts decreased in dickkopf-treated animals. CONCLUSIONS We conclude that intestinal crypt fission during infancy is mediated by Wnt signalling. It is possible that local treatment with Wnt agonists could be used to increase intestinal growth.
Collapse
|
87
|
Chailler P, Beaulieu JF, Ménard D. Isolation and functional studies of human fetal gastric epithelium in primary culture. Methods Mol Biol 2012; 806:137-55. [PMID: 22057450 DOI: 10.1007/978-1-61779-367-7_10] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Our understanding of gastric epithelial physiology in man is limited by the absence of normal or appropriate cancer cell lines that could serve as an in vitro model. Research mostly relied on primary culture of gastric epithelial cells of animal species, enriched with surface mucous cells, and devoid of glandular zymogenic chief cells. We successfully applied a new nonenzymatic procedure using Matrisperse Cell Recovery Solution to dissociate the entire epithelium from human fetal stomach. Cultures were generated by seeding multicellular aggregates prepared by mechanical fragmentation. We further demonstrate that this simple and convenient technique allows for the maintenance of heterogenous gastric epithelial primary cultures on plastic without a biological matrix as well as the persistence of viable chief cells able to synthesize and secrete gastric digestive enzymes, i.e., pepsinogen and gastric lipase. In wounding experiments, epithelial restitution occurred in serum-reduced conditions and was modulated by exogenous agents. This culture system is thus representative of the foveolus-gland axis and offers new perspectives to establish the influence of individual growth factors and extracellular matrix components as well as their combinatory effects on gastric epithelium homeostasis.
Collapse
Affiliation(s)
- Pierre Chailler
- CIHR Team on Digestive Epithelium, Département d'anatomie et de biologie cellulaire, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | | |
Collapse
|
88
|
Zhang C, Sherman MP, Prince LS, Bader D, Weitkamp JH, Slaughter JC, McElroy SJ. Paneth cell ablation in the presence of Klebsiella pneumoniae induces necrotizing enterocolitis (NEC)-like injury in the small intestine of immature mice. Dis Model Mech 2012; 5:522-32. [PMID: 22328592 PMCID: PMC3380715 DOI: 10.1242/dmm.009001] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a leading cause of morbidity and mortality in premature infants. During NEC pathogenesis, bacteria are able to penetrate innate immune defenses and invade the intestinal epithelial layer, causing subsequent inflammation and tissue necrosis. Normally, Paneth cells appear in the intestinal crypts during the first trimester of human pregnancy. Paneth cells constitute a major component of the innate immune system by producing multiple antimicrobial peptides and proinflammatory mediators. To better understand the possible role of Paneth cell disruption in NEC, we quantified the number of Paneth cells present in infants with NEC and found that they were significantly decreased compared with age-matched controls. We were able to model this loss in the intestine of postnatal day (P)14-P16 (immature) mice by treating them with the zinc chelator dithizone. Intestines from dithizone-treated animals retained approximately half the number of Paneth cells compared with controls. Furthermore, by combining dithizone treatment with exposure to Klebsiella pneumoniae, we were able to induce intestinal injury and inflammatory induction that resembles human NEC. Additionally, this novel Paneth cell ablation model produces NEC-like pathology that is consistent with other currently used animal models, but this technique is simpler to use, can be used in older animals that have been dam fed, and represents a novel line of investigation to study NEC pathogenesis and treatment.
Collapse
Affiliation(s)
- Chunxian Zhang
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | | | | | | | | | | |
Collapse
|
89
|
Ménard D, Tremblay E, Ferretti E, Babakissa C, Perron N, Seidman EG, Levy E, Beaulieu JF. Anti-inflammatory effects of epidermal growth factor on the immature human intestine. Physiol Genomics 2012; 44:268-80. [PMID: 22214601 DOI: 10.1152/physiolgenomics.00101.2011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The inflammatory response of the preterm infants' intestine underlines its inability to respond to hemodynamic stress, microbes, and nutrients. Recent evidence suggests that exogenous epidermal growth factor (EGF) exerts a therapeutic influence on neonatal enteropathies. However, the molecular mechanisms underlying the beneficial effects of EGF remain to be clarified. The purpose of this study was to evaluate the impact of EGF on the gene expression profiles of the developing human small and large intestine at midgestation in serum-free organ cultures using microarrays. The gene expression profiles of cultured human fetal ileal and colonic explants were investigated in the absence or presence of a physiological concentration of 50 ng/ml EGF for 48 h. Data were analyzed with the Ingenuity Pathway Analysis (IPA) software and confirmed by qPCR. We found a total of 6,474 differentially expressed genes in the two segments in response to EGF. IPA functional analysis revealed that in addition to differentially modulating distinct cellular, molecular, and physiological functions in the small and large intestine, EGF regulated the inflammatory response in both intestinal segments in a distinct manner. For instance, several intestinal-derived chemokines such as CCL2, CCL25, CXCL5, and CXCL10 were found to be differentially regulated by EGF in the immature ileum and colon. The findings showing the anti-inflammatory influence of exogenous EGF suggests a mechanistic basis for the beneficial effects of EGF on neonatal enteropathies. These results reinforce growing evidence that by midgestation, the human small intestine and colon rely on specific and distinct regulatory pathways.
Collapse
Affiliation(s)
- Daniel Ménard
- Canadian Institutes of Health Research Team on the Digestive Epithelium, Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | | | | | | | | | | | | | | |
Collapse
|
90
|
|
91
|
Beaulieu JF, Ménard D. Isolation, characterization, and culture of normal human intestinal crypt and villus cells. Methods Mol Biol 2012; 806:157-173. [PMID: 22057451 DOI: 10.1007/978-1-61779-367-7_11] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
The intestinal epithelium is a highly dynamic tissue undergoing constant and rapid renewal. It consists of a functional villus compartment responsible for terminal digestion and nutrient absorption and a progenitor cell compartment located in the crypts that produce new cells. The mechanisms regulating cell proliferation in the crypt, their migration, and differentiation are still incompletely understood. Until recently, normal human intestinal cell models allowing the study of these mechanisms have been lacking. In our laboratory, using fetal human intestines obtained at mid-gestation, we have generated the first normal human intestinal epithelial crypt-like (HIEC) cell line and villus-like primary cultures of differentiated enterocytes (PCDE). In this chapter, we provide a detailed description of the methodologies used to generate and characterize these normal intestinal crypt and villus cell models.
Collapse
Affiliation(s)
- Jean-François Beaulieu
- CIHR Team on Digestive Epithelium, Département d'anatomie et de biologie cellulaire, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
| | | |
Collapse
|
92
|
Tremblay E, Ferretti E, Babakissa C, Seidman EG, Levy E, Ménard D, Beaulieu JF. Gene-expression profile analysis in the mid-gestation human intestine discloses greater functional immaturity of the colon as compared with the ileum. J Pediatr Gastroenterol Nutr 2011; 52:670-678. [PMID: 21478755 DOI: 10.1097/mpg.0b013e3182078370] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
BACKGROUND AND OBJECTIVES The occurrence of many neonatal inflammatory intestinal diseases in preterm infants highlights the susceptibility of the immature intestine to responding inadequately to nutrients and microbes. A better understanding of functional intestinal development is essential for the design of optimal treatments ensuring survival and growth of premature infants. The purpose of this study was to evaluate the gene expression profiles of the human ileum and colon at mid-gestation because these 2 segments are considered to be similar at this stage and are the sites of the most frequent pathologies in preterm infants. SUBJECTS AND METHODS We compared the gene-expression profiles of human fetal small and large intestines using a cDNA microarray and analyzed the data with Ingenuity Pathway Analysis software. RESULTS We found that a significant proportion of the genes was differentially expressed in the 2 segments. Gene cluster analysis revealed an even higher level of transcriptional dissimilarity at the functional level. For instance, segment-specific/overexpressed gene clusters in the ileum included genes involved with amino acid, vitamin, and mineral metabolism, reflecting the higher level of maturity of the small intestine as compared with the colon in which genes involved with cell cycle, cell death, and cell signaling were the predominant clusters of genes expressed. CONCLUSIONS Functional clustering analysis of the differentially expressed genes revealed important functional differences between the 2 segments and a relative immaturity of the colon, suggesting that already at mid-gestation, the 2 intestinal segments should be considered as 2 distinct organs.
Collapse
Affiliation(s)
- Eric Tremblay
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | | | | | | | | | | | | |
Collapse
|
93
|
Affiliation(s)
- Rao N. Jaladanki
- University of Maryland School of Medicine and Baltimore Veterans Affairs Medical Center
| | - Jian-Ying Wang
- University of Maryland School of Medicine and Baltimore Veterans Affairs Medical Center
| |
Collapse
|
94
|
The pathogenesis of Barrett's metaplasia and the progression to esophageal adenocarcinoma. Recent Results Cancer Res 2010; 182:39-63. [PMID: 20676870 DOI: 10.1007/978-3-540-70579-6_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The most important risk factor for the development of Barrett's esophagus is the reflux of both gastric and duodenal contents into the esophagus. The reason why Barrett's metaplasia develops only in a minority of patients suffering from gastroesophageal reflux disease remains unknown.The exact mechanism behind the transition of normal squamous epithelium into specialized columnar epithelium is also unclear. It is likely that stem cells are involved in this metaplastic change, as they are the only permanent residents of the epithelium. Several tumorigenic steps that lead to the underlying genetic instability, which is indispensable in the progression from columnar metaplasia to esophageal adenocarcinoma have been described. This review outlines the process of pathogenesis of Barrett's metaplasia and its progression to esophageal adenocarcinoma.
Collapse
|
95
|
Li P, Waldman SA. Corruption of homeostatic mechanisms in the guanylyl cyclase C signaling pathway underlying colorectal tumorigenesis. Cancer Biol Ther 2010; 10:211-8. [PMID: 20592492 DOI: 10.4161/cbt.10.3.12539] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Colon cancer, the second leading cause of cancer-related mortality worldwide, originates from the malignant transformation of intestinal epithelial cells. The intestinal epithelium undergoes a highly organized process of rapid regeneration along the crypt-villus axis, characterized by proliferation, migration, differentiation and apoptosis, whose coordination is essential to maintaining the mucosal barrier. Disruption of these homeostatic processes predisposes cells to mutations in tumor suppressors or oncogenes, whose dysfunction provides transformed cells an evolutionary growth advantage. While sequences of genetic mutations at different stages along the neoplastic continuum have been established, little is known of the events initiating tumorigenesis prior to adenomatous polyposis coli (APC) mutations. Here, we examine a role for the corruption of homeostasis induced by silencing novel tumor suppressors, including the intestine-specific transcription factor CDX2 and its gene target guanylyl cyclase C (GCC), as early events predisposing cells to mutations in APC and other sequential genes that initiate colorectal cancer. CDX2 and GCC maintain homeostatic regeneration in the intestine by restricting cell proliferation, promoting cell maturation and adhesion, regulating cell migration and defending the intestinal barrier and genomic integrity. Elimination of CDX2 or GCC promotes intestinal tumor initiation and growth in aged mice, mice carrying APC mutations or mice exposed to carcinogens. The roles of CDX2 and GCC in suppressing intestinal tumorigenesis, universal disruption in their signaling through silencing of hormones driving GCC, and the uniform overexpression of GCC by tumors underscore the potential value of oral replacement with GCC ligands as targeted prevention and therapy for colorectal cancer.
Collapse
Affiliation(s)
- Peng Li
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA, USA.
| | | |
Collapse
|
96
|
Colorectal cancer is a paracrine deficiency syndrome amenable to oral hormone replacement therapy. Clin Transl Sci 2010; 1:163-7. [PMID: 19727435 DOI: 10.1111/j.1752-8062.2008.00040.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The most commonly lost gene products in colorectal carcinogenesis include the paracrine hormones guanylin and uroguanylin, the endogenous ligands for guanylyl cyclase C (GCC), the intestinal receptor for diarrheagenic bacterial enterotoxins. Recently, GCC-cGMP signaling has emerged as a principal regulator of proliferation, genetic integrity and metabolic programming in normal human enterocytes and colon cancer cells. Elimination of GCC in mice produced hyperplasia of the proliferating compartment associated with increases in rapidly cycling progenitor cells, and reprogrammed enterocyte metabolism, with a shift from oxidative phosphorylation to glycolysis. In addition, in colons of mice carrying mutations in Apc (Apc(Min) (/+)) or exposed to the carcinogen azoxymethane, elimination of GCC increased tumor initiation and promotion by disrupting genomic integrity and releasing cell cycle restriction. These previously unrecognized roles for GCC as a fundamental regulator of intestinal homeostasis and as an intestinal tumor suppressor suggest that receptor dysregulation reflecting paracrine hormone insufficiency is a key event during the initial stages of colorectal tumorigenesis. Together with the uniform over-expression of GCC in human tumors, these novel roles for GCC underscore the potential of oral replacement with GCC ligands for targeted prevention and therapy of colorectal cancer.
Collapse
|
97
|
Beaulieu JF. Integrin α6β4 in colorectal cancer. World J Gastrointest Pathophysiol 2010; 1:3-11. [PMID: 21607137 PMCID: PMC3097941 DOI: 10.4291/wjgp.v1.i1.3] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Revised: 03/18/2010] [Accepted: 03/25/2010] [Indexed: 02/06/2023] Open
Abstract
The ability of cells to interact with extracellular matrix macromolecules is at the forefront of the regulation of cell phenotype and organization. Indeed most if not all cells bear specific cell surface receptors for these molecules, namely the integrins, which are specific for the ligation of various macromolecules such as the laminins, fibronectins and tenascins. It is now well established that integrins can regulate a variety of biological activities, most notably cell cycle and tissue-specific gene expression. In the intestine, several observations suggest functional roles for cell-matrix interactions in the regulation of epithelial cell functions. This article focuses on integrin α6β4 as a paradigm to illustrate the importance as well as the complexity of integrins in the mediation of cell-matrix interactions. Indeed, α6β4 has been well-characterized for its involvement as a link between the cytoskeleton and extracellular matrix molecules as well as in the activation of a variety of intracellular signalization processes in cooperation with growth factor receptors. Furthermore, recent studies show that distinct forms of α6 and β4 subunits are expressed in the human intestine and, more importantly, recent work provides experimental evidence that various forms of α6β4 can differentially regulate intestinal epithelial cell functions under both normal and pathological conditions. For instance, it has been discovered that colorectal cancer cells express a hybrid form of α6β4 that is never seen in normal cells. Although further work is needed, integrin α6β4 is emerging as a key regulator of intestinal functions in both intestinal health and disease.
Collapse
|
98
|
Drozdowski LA, Clandinin T, Thomson ABR. Ontogeny, growth and development of the small intestine: Understanding pediatric gastroenterology. World J Gastroenterol 2010; 16:787-99. [PMID: 20143457 PMCID: PMC2825325 DOI: 10.3748/wjg.v16.i7.787] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Throughout our lifetime, the intestine changes. Some alterations in its form and function may be genetically determined, and some are the result of adaptation to diet, temperature, or stress. The critical period programming of the intestine can be modified, such as from subtle differences in the types and ratios of n3:m6 fatty acids in the diet of the pregnant mother, or in the diet of the weanlings. This early forced adaptation may persist in later life, such as the unwanted increased intestinal absorption of sugars, fatty acids and cholesterol. Thus, the ontogeny, early growth and development of the intestine is important for the adult gastroenterologist to appreciate, because of the potential for these early life events to affect the responsiveness of the intestine to physiological or pathological challenges in later life.
Collapse
|
99
|
Kwek J, De Iongh R, Nicholas K, Familari M. Molecular insights into evolution of the vertebrate gut: focus on stomach and parietal cells in the marsupial,Macropus eugenii. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2009; 312:613-24. [DOI: 10.1002/jez.b.21227] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
100
|
Sri Paran T, Rolle U, Puri P. Age-related changes in the myenteric plexus of the porcine bowel. J Pediatr Surg 2009; 44:1771-7. [PMID: 19735824 DOI: 10.1016/j.jpedsurg.2008.12.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2008] [Revised: 12/10/2008] [Accepted: 12/11/2008] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Myenteric plexus (MP) is well recognized as an important regulator of peristaltic activity. Knowledge regarding prenatal and postnatal normal morphological changes is important when interpreting histopathologic findings in motility disorders of childhood. The aim of this study was to determine the neuronal density and morphology of the myenteric plexus (MP) of the porcine bowel from fetal life to adulthood. METHOD Small and large bowel whole-mount preparations of the MP were stained using NADPH diaphorase histochemistry in animals from 6 different age groups (60 and 90 days of gestation, newborn, 4-week and 12-week-old, and adult pigs). Using light microscopy, above parameters was quantified, and cell/nucleus sizes were measured. Results were analyzed using 1-way analysis of variance test. RESULTS There were significant regional and age-related differences in cell numbers per ganglia noted in MP throughout the lifetime of a pig. There was an abrupt increase in cell numbers per ganglia from the newborn to 4-week-old animals, which then stabilized in most parts of the bowel, except in the distal large bowel, where it continued to increase. Ganglion density and ganglia cell density both decreased steadily with advancing age. Cell size increased with age, mostly secondary to increase in the cytoplasm. CONCLUSION Our results show that significant changes occur in the MP in relation to age and the region of the bowel. These changes are most evident in the immediate period after birth but continue throughout life. Such age-related changes must be taken into account during morphological evaluation of biopsy specimens taken from infants who had constipation.
Collapse
|