1
|
Ardicli S, Ardicli O, Yazici D, Pat Y, Babayev H, Xiong P, Zeyneloglu C, Garcia-Sanchez A, Shi LL, Viscardi OG, Skolnick S, Ogulur I, Dhir R, Jutel M, Agache I, Janda J, Pali-Schöll I, Nadeau KC, Akdis M, Akdis CA. Epithelial barrier dysfunction and associated diseases in companion animals: Differences and similarities between humans and animals and research needs. Allergy 2024. [PMID: 39417247 DOI: 10.1111/all.16343] [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: 06/30/2024] [Revised: 09/04/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024]
Abstract
Since the 1960s, more than 350,000 new chemicals have been introduced into the lives of humans and domestic animals. Many of them have become part of modern life and some are affecting nature as pollutants. Yet, our comprehension of their potential health risks for both humans and animals remains partial. The "epithelial barrier theory" suggests that genetic predisposition and exposure to diverse factors damaging the epithelial barriers contribute to the emergence of allergic and autoimmune conditions. Impaired epithelial barriers, microbial dysbiosis, and tissue inflammation have been observed in a high number of mucosal inflammatory, autoimmune and neuropsychiatric diseases, many of which showed increased prevalence in the last decades. Pets, especially cats and dogs, share living spaces with humans and are exposed to household cleaners, personal care products, air pollutants, and microplastics. The utilisation of cosmetic products and food additives for pets is on the rise, unfortunately, accompanied by less rigorous safety regulations than those governing human products. In this review, we explore the implications of disruptions in epithelial barriers on the well-being of companion animals, drawing comparisons with humans, and endeavour to elucidate the spectrum of diseases that afflict them. In addition, future research areas with the interconnectedness of human, animal, and environmental well-being are highlighted in line with the "One Health" concept.
Collapse
Affiliation(s)
- Sena Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Genetics, Faculty of Veterinary Medicine, Bursa Uludag University, Bursa, Türkiye
| | - Ozge Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Division of Food Processing, Milk and Dairy Products Technology Program, Karacabey Vocational School, Bursa Uludag University, Bursa, Türkiye
| | - Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Huseyn Babayev
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Peng Xiong
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Can Zeyneloglu
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Asuncion Garcia-Sanchez
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Biomedical & Diagnostic Sciences, Faculty of Medicine, University of Salamanca, Salamanca, Spain
| | - Li-Li Shi
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | | | - Stephen Skolnick
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- SEED Inc. Co., Los Angeles, California, USA
| | - Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Raja Dhir
- SEED Inc. Co., Los Angeles, California, USA
| | - Marek Jutel
- Department of Clinical Immunology, Wrocław Medical University, Wroclaw, Poland
- ALL-MED Medical Research Institute, Wrocław, Poland
| | - Ioana Agache
- Faculty of Medicine, Department of Allergy and Clinical Immunology, Transylvania University, Brasov, Romania
| | - Jozef Janda
- Faculty of Science, Charles University, Prague, Czech Republic
| | - Isabella Pali-Schöll
- The Interuniversity Messerli Research Institute of the University of Veterinary Medicine and Medical University Vienna, Vienna, Austria
- Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| |
Collapse
|
2
|
Anthney A, Do ADT, Alrubaye AAK. Bacterial chondronecrosis with osteomyelitis lameness in broiler chickens and its implications for welfare, meat safety, and quality: a review. Front Physiol 2024; 15:1452318. [PMID: 39268189 PMCID: PMC11390708 DOI: 10.3389/fphys.2024.1452318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/06/2024] [Indexed: 09/15/2024] Open
Abstract
The exponential increase in global population continues to present an ongoing challenge for livestock producers worldwide to consistently provide a safe, high-quality, and affordable source of protein for consumers. In the last 50 years, the poultry industry has spearheaded this effort thanks to focused genetic and genomic selection for feed-efficient, high-yielding broilers. However, such intense selection for productive traits, along with conventional industry farming practices, has also presented the industry with a myriad of serious issues that negatively impacted animal health, welfare, and productivity-such as woody breast and virulent diseases commonly associated with poultry farming. Bacterial chondronecrosis with osteomyelitis (BCO) lameness is one such issue, having rapidly become a key issue affecting the poultry industry with serious impacts on broiler welfare, meat quality, production, food safety, and economic losses since its discovery in 1972. This review focuses on hallmark clinical symptoms, diagnosis, etiology, and impact of BCO lameness on key issues facing the poultry industry.
Collapse
Affiliation(s)
- Amanda Anthney
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
| | - Anh Dang Trieu Do
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR, United States
| | - Adnan A K Alrubaye
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR, United States
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR, United States
| |
Collapse
|
3
|
Giugliano FP, Navis M, Ouahoud S, Garcia TM, Kreulen IA, Ferrantelli E, Meisner S, Vermeulen JL, van Roest M, Billaud JN, Koster J, Dawood Y, de Bakker BS, Picavet-Havik DI, Schimmel IM, van der Wel NN, Koelink PJ, Wildenberg ME, Derikx JP, de Jonge WJ, Renes IB, van Elburg RM, Muncan V. Pro-inflammatory T cells-derived cytokines enhance the maturation of the human fetal intestinal epithelial barrier. iScience 2024; 27:109909. [PMID: 38812539 PMCID: PMC11134877 DOI: 10.1016/j.isci.2024.109909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/31/2024] [Accepted: 05/03/2024] [Indexed: 05/31/2024] Open
Abstract
Small intestine (SI) maturation during early life is pivotal in preventing the onset of gut diseases. In this study we interrogated the milestones of SI development by gene expression profiling and ingenuity pathway analyses. We identified a set of cytokines as main regulators of changes observed across different developmental stages. Upon cytokines stimulation, with IFNγ as the most contributing factor, human fetal organoids (HFOs) increase brush border gene expression and enzyme activity as well as trans-epithelial electrical resistance. Electron microscopy revealed developed brush border and loss of fetal cell characteristics in HFOs upon cytokine stimulation. We identified T cells as major source of IFNγ production in the fetal SI lamina propria. Co-culture of HFOs with T cells recapitulated the major effects of cytokine stimulation. Our findings underline pro-inflammatory cytokines derived from T cells as pivotal factors inducing functional SI maturation in vivo and capable of modulating the barrier maturation of HFOs in vitro.
Collapse
Affiliation(s)
- Francesca P. Giugliano
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Marit Navis
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Sarah Ouahoud
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Tânia Martins Garcia
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Irini A.M. Kreulen
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Evelina Ferrantelli
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Sander Meisner
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Jacqueline L.M. Vermeulen
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Manon van Roest
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Jean-Noël Billaud
- QIAGEN Digital Insights, 1001 Marshall Street, Redwood City, CA, USA
- DNAnexus, 204 El Camino Real, Mountain View, CA, USA
| | - Jan Koster
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Yousif Dawood
- Department of Obstetrics and Gynecology, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Reproduction and Development research institute, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Bernadette S. de Bakker
- Department of Obstetrics and Gynecology, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Reproduction and Development research institute, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Daisy I. Picavet-Havik
- Department of Medical Biology, Electron Microscopy Center Amsterdam, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Irene M. Schimmel
- Department of Medical Biology, Electron Microscopy Center Amsterdam, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Nicole N. van der Wel
- Department of Medical Biology, Electron Microscopy Center Amsterdam, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Pim J. Koelink
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Manon E. Wildenberg
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Joep P.M. Derikx
- Department of Pediatric Surgery, Pediatric Surgery Center of Amsterdam, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Wouter J. de Jonge
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| | - Ingrid B. Renes
- Department of Pediatrics, Amsterdam University Medical Center (AUMC), Emma Children’s Hospital, Amsterdam, the Netherlands
- Danone Nutricia Research, Utrecht, the Netherlands
| | - Ruurd M. van Elburg
- Department of Pediatrics, Amsterdam University Medical Center (AUMC), Emma Children’s Hospital, Amsterdam, the Netherlands
| | - Vanesa Muncan
- Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center (AUMC), University of Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
4
|
Fleming SA, Reyes SM, Donovan SM, Hernell O, Jiang R, Lönnerdal B, Neu J, Steinman L, Sørensen ES, West CE, Kleinman R, Wallingford JC. An expert panel on the adequacy of safety data and physiological roles of dietary bovine osteopontin in infancy. Front Nutr 2024; 11:1404303. [PMID: 38919388 PMCID: PMC11197938 DOI: 10.3389/fnut.2024.1404303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 04/29/2024] [Indexed: 06/27/2024] Open
Abstract
Human milk, due to its unique composition, is the optimal standard for infant nutrition. Osteopontin (OPN) is abundant in human milk but not bovine milk. The addition of bovine milk osteopontin (bmOPN) to formula may replicate OPN's concentration and function in human milk. To address safety concerns, we convened an expert panel to assess the adequacy of safety data and physiological roles of dietary bmOPN in infancy. The exposure of breastfed infants to human milk OPN (hmOPN) has been well-characterized and decreases markedly over the first 6 months of lactation. Dietary bmOPN is resistant to gastric and intestinal digestion, absorbed and cleared from circulation within 8-24 h, and represents a small portion (<5%) of total plasma OPN. Label studies on hmOPN suggest that after 3 h, intact or digested OPN is absorbed into carcass (62%), small intestine (23%), stomach (5%), and small intestinal perfusate (4%), with <2% each found in the cecum, liver, brain, heart, and spleen. Although the results are heterogenous with respect to bmOPN's physiologic impact, no adverse impacts have been reported across growth, gastrointestinal, immune, or brain-related outcomes. Recombinant bovine and human forms demonstrate similar absorption in plasma as bmOPN, as well as effects on cognition and immunity. The panel recommended prioritization of trials measuring a comprehensive set of clinically relevant outcomes on immunity and cognition to confirm the safety of bmOPN over that of further research on its absorption, distribution, metabolism, and excretion. This review offers expert consensus on the adequacy of data available to assess the safety of bmOPN for use in infant formula, aiding evidence-based decisions on the formulation of infant formula.
Collapse
Affiliation(s)
| | | | - Sharon M. Donovan
- Department of Food Science and Human Nutrition, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Olle Hernell
- Department of Clinical Sciences and Pediatrics, Umeå University, Umeå, Sweden
| | - Rulan Jiang
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Bo Lönnerdal
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Josef Neu
- Department of Pediatrics, Division of Neonatology, University of Florida, Gainesville, FL, United States
| | - Lawrence Steinman
- Departments of Pediatrics and of Neurology and Neurological Sciences, Interdepartmental Program in Immunology, Beckman Center for Molecular Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Esben S. Sørensen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Christina E. West
- Department of Clinical Sciences and Pediatrics, Umeå University, Umeå, Sweden
| | - Ronald Kleinman
- Harvard Medical School, Boston, MA, United States
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA, United States
| | | |
Collapse
|
5
|
Hopton RE, Jahahn NJ, Zemper AE. Lrig1 drives cryptogenesis and restrains proliferation during colon development. Am J Physiol Gastrointest Liver Physiol 2023; 325:G570-G581. [PMID: 37873577 PMCID: PMC11192189 DOI: 10.1152/ajpgi.00094.2023] [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: 05/03/2023] [Revised: 10/04/2023] [Accepted: 10/13/2023] [Indexed: 10/25/2023]
Abstract
Growth and specification of the mouse intestine occurs in utero and concludes after birth. Although numerous studies have examined this developmental process in the small intestine, far less is known about the cellular and molecular cues required for colon development. In this study, we examine the morphological events leading to crypt formation, epithelial cell differentiation, proliferation, and the emergence and expression of a stem and progenitor cell marker Lrig1. Through multicolor lineage tracing, we show Lrig1-expressing cells are present at birth and behave as stem cells to establish clonal crypts within 3 wk of life. In addition, we use an inducible knockout mouse to eliminate Lrig1 and show Lrig1 restrains proliferation within a critical developmental time window, without impacting colonic epithelial cell differentiation. Our study illustrates morphological changes during crypt development and the importance of Lrig1 in the developing colon.NEW & NOTEWORTHY Our studies define the importance of studying Lrig1 in colon development. We address a critical gap in the intestinal development literature and provide new information about the molecular cues that guide colon development. Using a novel, inducible knockout of Lrig1, we show Lrig1 is required for appropriate colon epithelial growth and illustrate the importance of Lrig1-expressing cells in the establishment of colonic crypts.
Collapse
Affiliation(s)
- Rachel E Hopton
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon, United States
- Department of Biology, University of Oregon, Eugene, Oregon, United States
| | - Nicholas J Jahahn
- Institute of Molecular Biology, University of Oregon, Eugene, Oregon, United States
- Department of Biology, University of Oregon, Eugene, Oregon, United States
| | - Anne E Zemper
- Department of Biology, University of Oregon, Eugene, Oregon, United States
| |
Collapse
|
6
|
Che X, Yang C, Pan L, Gu D, Dai G, Shu J, Yang L. Achieving safe and high-performance gastrointestinal tract spectral CT imaging with small-molecule lanthanide complex. Biomater Res 2023; 27:119. [PMID: 37990349 PMCID: PMC10664581 DOI: 10.1186/s40824-023-00463-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 11/13/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND Non-intrusive imaging of gastrointestinal (GI) tract using computed tomography (CT) contrast agents is of the most significant issues in the diagnosis and treatment of GI diseases. Moreover, spectral CT, which can generate monochromatic images to display the X-ray attenuation characteristics of contrast agents, provides a better imaging sensitivity for diagnose inflammatory bowel disease (IBD) than convention CT imaging. METHODS Herein, a convenient and one-pot synthesis method is provided for the fabrication of small-molecule lanthanide complex Holmium-tetraazacyclododecane-1, 4, 7, 10-tetraacetic acid (Ho-DOTA) as a biosafe and high-performance spectral CT contrast agent for GI imaging with IBD. In vivo CT imaging was administered with both healthy mice and colitis mice induced by dextran sodium sulfate. RESULTS We found that Ho-DOTA accumulated in inflammation sites of large intestines and produced high CT contrast compared with healthy mice. Both in vitro and in vivo experimental results also showed that Ho-DOTA provided much more diagnostic sensitivity and accuracy due to the excellent X-ray attenuation characteristics of Ho-DOTA compared with clinical iodinate agent. Furthermore, the proposed contrast media could be timely excreted from the body via the urinary and digestive system, keeping away from the potential side effects due to long-term retention in vivo. CONCLUSION Accordingly, Ho-DOTA with excellent biocompatibility can be useful as a potential high-performance spectral CT contrast agent for further clinical imaging of gastrointestinal tract and diagnosis of intestinal system diseases.
Collapse
Affiliation(s)
- Xiaoling Che
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Chunmei Yang
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Liping Pan
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Didi Gu
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Guidong Dai
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China
| | - Jian Shu
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China.
| | - Lu Yang
- Department of Radiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, People's Republic of China.
| |
Collapse
|
7
|
Kable ME, Chin EL, Huang L, Stephensen CB, Lemay DG. Association of Estimated Daily Lactose Consumption, Lactase Persistence Genotype (rs4988235), and Gut Microbiota in Healthy Adults in the United States. J Nutr 2023; 153:2163-2173. [PMID: 37354976 DOI: 10.1016/j.tjnut.2023.06.025] [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: 03/01/2023] [Revised: 05/26/2023] [Accepted: 06/21/2023] [Indexed: 06/26/2023] Open
Abstract
BACKGROUND Lactase persistence (LP) is a heritable trait in which lactose can be digested throughout adulthood. Lactase nonpersistent (LNP) individuals who consume lactose may experience microbial adaptations in response to undigested lactose. OBJECTIVES The objective of the study was to estimate lactose from foods reported in the Automated Self-Administered 24-Hour Dietary Assessment Tool (ASA24) and determine the interaction between lactose consumption, LP genotype, and gut microbiome in an observational cross-sectional study of healthy adults in the United States (US). METHODS Average daily lactose consumption was estimated for 279 healthy US adults, genotyped for the lactase gene -13910G>A polymorphism (rs4988235) by matching ASA24-reported foods to foods in the Nutrition Coordinating Center Food and Nutrient Database. Analysis of covariance was used to identify whether the A genotype (LP) influenced lactose and total dairy consumption, with total energy intake and weight as covariates. The 16S rRNA V4/V5 region, amplified from bacterial DNA extracted from each frozen stool sample, was sequenced using Illumina MiSeq (300 bp paired-end) and analyzed using Quantitative Insights Into Microbial Ecology (QIIME)2 (version 2019.10). Differential abundances of bacterial taxa were analyzed using DESeq2 likelihood ratio tests. RESULTS Across a diverse set of ethnicities, LP subjects consumed more lactose than LNP subjects. Lactobacillaceae abundance was highest in LNP subjects who consumed more than 12.46 g/d (upper tercile). Within Caucasians and Hispanics, family Lachnospiraceae was significantly enriched in the gut microbiota of LNP individuals consuming the upper tercile of lactose across both sexes. CONCLUSIONS Elevated lactose consumption in individuals with the LNP genotype is associated with increased abundance of family Lactobacillaceae and Lachnospriaceae, taxa that contain multiple genera capable of utilizing lactose. This trial was registered on clinicaltrials.gov as NCT02367287.
Collapse
Affiliation(s)
- Mary E Kable
- United States Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, USA; Department of Nutrition, University of California-Davis, Davis, CA, USA.
| | - Elizabeth L Chin
- United States Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, USA
| | - Liping Huang
- United States Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, USA; Department of Nutrition, University of California-Davis, Davis, CA, USA
| | - Charles B Stephensen
- United States Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, USA; Department of Nutrition, University of California-Davis, Davis, CA, USA
| | - Danielle G Lemay
- United States Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, Davis, CA, USA; Department of Nutrition, University of California-Davis, Davis, CA, USA.
| |
Collapse
|
8
|
Hopton RE, Jahahn NJ, Zemper AE. The Role of Lrig1 in the Development of the Colonic Epithelium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.02.539114. [PMID: 37205411 PMCID: PMC10187246 DOI: 10.1101/2023.05.02.539114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Growth and specification of the mouse intestine occurs in utero and concludes after birth. While numerous studies have examined this developmental process in the small intestine, far less is known about the cellular and molecular cues required for colon development. In this study, we examine the morphological events leading to crypt formation, epithelial cell differentiation, areas of proliferation, and the emergence and expression of a stem and progenitor cell marker Lrig1. Through multicolor lineage tracing, we show Lrig1 expressing cells are present at birth and behave as stem cells to establish clonal crypts within three weeks after birth. In addition, we use an inducible knockout mouse to eliminate Lrig1 during colon development and show loss of Lrig1 restrains proliferation within a critical developmental time window, without impacting colonic epithelial cell differentiation. Our study illustrates the morphological changes that occur during crypt development and the importance of Lrig1 in the developing colon.
Collapse
|
9
|
Singh A, Poling HM, Chaturvedi P, Thorner K, Sundaram N, Kechele DO, Childs CJ, McCauley HA, Fisher GW, Brown NE, Spence JR, Wells JM, Helmrath MA. Transplanted human intestinal organoids: a resource for modeling human intestinal development. Development 2023; 150:dev201416. [PMID: 37070767 PMCID: PMC10259511 DOI: 10.1242/dev.201416] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/28/2023] [Indexed: 04/19/2023]
Abstract
The in vitro differentiation of pluripotent stem cells into human intestinal organoids (HIOs) has served as a powerful means for creating complex three-dimensional intestinal structures. Owing to their diverse cell populations, transplantation into an animal host is supported with this system and allows the temporal formation of fully laminated structures, including crypt-villus architecture and smooth muscle layers that resemble native human intestine. Although the endpoint of HIO engraftment has been well described, here we aim to elucidate the developmental stages of HIO engraftment and establish whether it parallels fetal human intestinal development. We analyzed a time course of transplanted HIOs histologically at 2, 4, 6 and 8 weeks post-transplantation, and demonstrated that HIO maturation closely resembles key stages of fetal human intestinal development. We also utilized single-nuclear RNA sequencing to determine and track the emergence of distinct cell populations over time, and validated our transcriptomic data through in situ protein expression. These observations suggest that transplanted HIOs do indeed recapitulate early intestinal development, solidifying their value as a human intestinal model system.
Collapse
Affiliation(s)
- Akaljot Singh
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Holly M. Poling
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Praneet Chaturvedi
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Konrad Thorner
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Nambirajan Sundaram
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Daniel O. Kechele
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Charlie J. Childs
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Heather A. McCauley
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Garrett W. Fisher
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Nicole E. Brown
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jason R. Spence
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - James M. Wells
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Michael A. Helmrath
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
10
|
McCarthy N, Tie G, Madha S, He R, Kraiczy J, Maglieri A, Shivdasani RA. Smooth muscle contributes to the development and function of a layered intestinal stem cell niche. Dev Cell 2023; 58:550-564.e6. [PMID: 36924771 PMCID: PMC10089980 DOI: 10.1016/j.devcel.2023.02.012] [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: 06/17/2022] [Revised: 12/05/2022] [Accepted: 02/20/2023] [Indexed: 03/17/2023]
Abstract
Wnt and Rspondin (RSPO) signaling drives proliferation, and bone morphogenetic protein inhibitors (BMPi) impede differentiation, of intestinal stem cells (ISCs). Here, we identify the mouse ISC niche as a complex, multi-layered structure that encompasses distinct mesenchymal and smooth muscle populations. In young and adult mice, diverse sub-cryptal cells provide redundant ISC-supportive factors; few of these are restricted to single cell types. Niche functions refine during postnatal crypt morphogenesis, in part to oppose the dense aggregation of differentiation-promoting BMP+ sub-epithelial myofibroblasts at crypt-villus junctions. Muscularis mucosae, a specialized muscle layer, first appears during this period and supplements neighboring RSPO and BMPi sources. Components of this developing niche are conserved in human fetuses. The in vivo ablation of mouse postnatal smooth muscle increases BMP signaling activity, potently limiting a pre-weaning burst of crypt fission. Thus, distinct and progressively specialized mesenchymal cells together create the milieu that is required to propagate crypts during rapid organ growth and to sustain adult ISCs.
Collapse
Affiliation(s)
- Neil McCarthy
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA.
| | - Guodong Tie
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Shariq Madha
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Ruiyang He
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Judith Kraiczy
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Adrianna Maglieri
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Ramesh A Shivdasani
- Department of Medical Oncology and Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02139, USA.
| |
Collapse
|
11
|
Gonçalves EDS, Ribeiro JD, Marson FAL, Montes CG, Ribeiro AF, Mainz JG, Toro AADC, Zamariola JH, Borgli DSP, Lomazi EA, Servidoni MDFCP. Combined multi-channel intraluminal impedance measurement and pHmetry in the detection of gastroesophageal reflux disease in children with cystic fibrosis. J Pediatr (Rio J) 2022; 99:269-277. [PMID: 36564006 DOI: 10.1016/j.jped.2022.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 11/05/2022] [Accepted: 11/07/2022] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE To assess the prevalence of GERD exclusively by means of multichannel intraluminal impedanciometry associated with pH monitoring (MIIpH) and compare it with respiratory symptoms in children with CF. To compare MIIpH with pHmetry alone to perform GERD diagnosis. METHODS An analytical cross-sectional study was conducted with children diagnosed with CF who underwent MIIpH. Clinical and laboratory markers, including respiratory and digestive symptoms, were used for comparative analyses. High-resolution chest computed tomography was performed on patients with symptoms of chronic lung disease. Severity was classified according to the Bhalla score. RESULTS A total of 29 children < 10 yo (18 girls) were evaluated; 19 of whom with physiological GER and 10 with GERD. Of the children with GERD, seven had predominantly acid GER, two acid+non-acid GER, and one non-acid GER. Three patients had GERD diagnosed only by MIIpH. Bhalla scores ranged from seven to 17.75 with no significant relationship with GERD. The number of pulmonary exacerbations was associated with a decrease in esophageal clearance regardless of the position in pHmetry and MIIpH. CONCLUSIONS The prevalence of GERD was 34% in children with CF. There was no association between respiratory disease severity and GER types. MIIpH detected 30% more patients with GERD than pHmetry.
Collapse
Affiliation(s)
- Emília da Silva Gonçalves
- Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Pediatria, Centro de Pesquisa Pediátrica, Laboratório de Fibrose Cística e Laboratório de Fisiologia Pulmonar, Campinas, SP, Brazil.
| | - José Dirceu Ribeiro
- Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Pediatria, Centro de Pesquisa Pediátrica, Laboratório de Fibrose Cística e Laboratório de Fisiologia Pulmonar, Campinas, SP, Brazil.
| | - Fernando Augusto Lima Marson
- Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Pediatria, Centro de Pesquisa Pediátrica, Laboratório de Fibrose Cística e Laboratório de Fisiologia Pulmonar, Campinas, SP, Brazil; Universidade São Francisco, Laboratório de Genética Médica e Medicina Genômica, Pós-Graduação em Ciências da Saúde, Bragança Paulista, SP, Brazil; Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Centro de Diagnóstico de Doenças do Aparelho Digestivo (Gastrocentro), Campinas, SP, Brazil
| | - Ciro Garcia Montes
- Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Centro de Diagnóstico de Doenças do Aparelho Digestivo (Gastrocentro), Campinas, SP, Brazil
| | - Antônio Fernando Ribeiro
- Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Pediatria, Centro de Pesquisa Pediátrica, Laboratório de Fibrose Cística e Laboratório de Fisiologia Pulmonar, Campinas, SP, Brazil
| | - Jochen Georg Mainz
- University Klinikum Westbrandenburg Klinik für Kinder- und Jugendmedizin, Brandenburg Medical School (MHB), Head of Pediatric Pulmonology and Cystic Fibrosis, Hochstraße, Germany
| | - Adyléia Aparecida Dalbo Contrera Toro
- Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Pediatria, Centro de Pesquisa Pediátrica, Laboratório de Fibrose Cística e Laboratório de Fisiologia Pulmonar, Campinas, SP, Brazil
| | - Juliana Helena Zamariola
- Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Pediatria, Centro de Pesquisa Pediátrica, Laboratório de Fibrose Cística e Laboratório de Fisiologia Pulmonar, Campinas, SP, Brazil
| | - Daniela Souza Paiva Borgli
- Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Pediatria, Centro de Pesquisa Pediátrica, Laboratório de Fibrose Cística e Laboratório de Fisiologia Pulmonar, Campinas, SP, Brazil
| | - Elizete Aparecida Lomazi
- Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Pediatria, Centro de Pesquisa Pediátrica, Laboratório de Fibrose Cística e Laboratório de Fisiologia Pulmonar, Campinas, SP, Brazil
| | - Maria de Fátima Corrêa Pimenta Servidoni
- Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Departamento de Pediatria, Centro de Pesquisa Pediátrica, Laboratório de Fibrose Cística e Laboratório de Fisiologia Pulmonar, Campinas, SP, Brazil; Universidade Estadual de Campinas, Faculdade de Ciências Médicas, Centro de Diagnóstico de Doenças do Aparelho Digestivo (Gastrocentro), Campinas, SP, Brazil
| |
Collapse
|
12
|
Gustafsson JK, Johansson MEV. The role of goblet cells and mucus in intestinal homeostasis. Nat Rev Gastroenterol Hepatol 2022; 19:785-803. [PMID: 36097076 DOI: 10.1038/s41575-022-00675-x] [Citation(s) in RCA: 175] [Impact Index Per Article: 87.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/04/2022] [Indexed: 12/08/2022]
Abstract
The intestinal tract faces numerous challenges that require several layers of defence. The tight epithelium forms a physical barrier that is further protected by a mucus layer, which provides various site-specific protective functions. Mucus is produced by goblet cells, and as a result of single-cell RNA sequencing identifying novel goblet cell subpopulations, our understanding of their various contributions to intestinal homeostasis has improved. Goblet cells not only produce mucus but also are intimately linked to the immune system. Mucus and goblet cell development is tightly regulated during early life and synchronized with microbial colonization. Dysregulation of the developing mucus systems and goblet cells has been associated with infectious and inflammatory conditions and predisposition to chronic disease later in life. Dysfunctional mucus and altered goblet cell profiles are associated with inflammatory conditions in which some mucus system impairments precede inflammation, indicating a role in pathogenesis. In this Review, we present an overview of the current understanding of the role of goblet cells and the mucus layer in maintaining intestinal health during steady-state and how alterations to these systems contribute to inflammatory and infectious disease.
Collapse
Affiliation(s)
- Jenny K Gustafsson
- Department of Physiology, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Malin E V Johansson
- Department of Medical Biochemisty and Cell biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
13
|
Campion S, Inselman A, Hayes B, Casiraghi C, Joseph D, Facchinetti F, Salomone F, Schmitt G, Hui J, Davis-Bruno K, Van Malderen K, Morford L, De Schaepdrijver L, Wiesner L, Kourula S, Seo S, Laffan S, Urmaliya V, Chen C. The benefits, limitations and opportunities of preclinical models for neonatal drug development. Dis Model Mech 2022; 15:dmm049065. [PMID: 35466995 PMCID: PMC9066504 DOI: 10.1242/dmm.049065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Increased research to improve preclinical models to inform the development of therapeutics for neonatal diseases is an area of great need. This article reviews five common neonatal diseases - bronchopulmonary dysplasia, retinopathy of prematurity, necrotizing enterocolitis, perinatal hypoxic-ischemic encephalopathy and neonatal sepsis - and the available in vivo, in vitro and in silico preclinical models for studying these diseases. Better understanding of the strengths and weaknesses of specialized neonatal disease models will help to improve their utility, may add to the understanding of the mode of action and efficacy of a therapeutic, and/or may improve the understanding of the disease pathology to aid in identification of new therapeutic targets. Although the diseases covered in this article are diverse and require specific approaches, several high-level, overarching key lessons can be learned by evaluating the strengths, weaknesses and gaps in the available models. This Review is intended to help guide current and future researchers toward successful development of therapeutics in these areas of high unmet medical need.
Collapse
Affiliation(s)
- Sarah Campion
- Pfizer Worldwide Research, Development, and Medical, Groton, CT 06340, USA
| | - Amy Inselman
- U.S. Food and Drug Administration, National Center for Toxicological Research, Division of Systems Biology, Jefferson, AR 72079, USA
| | - Belinda Hayes
- U.S. Food and Drug Administration, Center for Drug Evaluation and Research, Office of New Drugs, Silver Spring, MD 20993, USA
| | - Costanza Casiraghi
- Department of Experimental Pharmacology and Translational Science, Chiesi Farmaceutici S.p.A., 43122 Parma, Italy
| | - David Joseph
- U.S. Food and Drug Administration, Center for Drug Evaluation and Research, Office of New Drugs, Silver Spring, MD 20993, USA
| | - Fabrizio Facchinetti
- Department of Experimental Pharmacology and Translational Science, Chiesi Farmaceutici S.p.A., 43122 Parma, Italy
| | - Fabrizio Salomone
- Department of Experimental Pharmacology and Translational Science, Chiesi Farmaceutici S.p.A., 43122 Parma, Italy
| | - Georg Schmitt
- Pharma Research and Early Development, Roche Innovation Center Basel, Pharmaceutical Sciences, F. Hoffmann-La Roche, 4070 Basel, Switzerland
| | - Julia Hui
- Bristol Myers Squibb, Nonclinical Research and Development, Summit, NJ 07901, USA
| | - Karen Davis-Bruno
- U.S. Food and Drug Administration, Center for Drug Evaluation and Research, Office of New Drugs, Silver Spring, MD 20993, USA
| | - Karen Van Malderen
- Federal Agency for Medicines and Health Products (FAMHP), Department DG PRE authorization, 1210 Brussels, Belgium
| | - LaRonda Morford
- Eli Lilly, Global Regulatory Affairs, Indianapolis, IN 46285, USA
| | | | - Lutz Wiesner
- Federal Institute for Drugs and Medical Devices, Clinical Trials, 53175 Bonn, Germany
| | - Stephanie Kourula
- Janssen R&D, Drug Metabolism & Pharmacokinetics, 2340 Beerse, Belgium
| | - Suna Seo
- U.S. Food and Drug Administration, Center for Drug Evaluation and Research, Office of New Drugs, Silver Spring, MD 20993, USA
| | - Susan Laffan
- GlaxoSmithKline, Non-Clinical Safety, Collegeville, PA 19406, USA
| | | | - Connie Chen
- Health and Environmental Sciences Institute, Washington, DC 20005, USA
| |
Collapse
|
14
|
Garg PM, Paschal JL, Lett K, Middleton C, Varshney N, Maheshwari A. Intestinal Resection is More Likely to be Effective in Necrotizing Enterocolitis Extending to Colon than in Disease Limited to the Small Intestine. NEWBORN 2022; 1:14-26. [PMID: 35846895 PMCID: PMC9282977 DOI: 10.5005/jp-journals-11002-0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Background: The prognosis in surgical necrotizing enterocolitis (NEC) has focused on the total length of the resected bowel; the relative impact of small intestinal vs colonic resection is not well studied. Objective: We hypothesized that intestinal resections may reduce mortality and length of hospital stay (LOS) more likely in infants who have NEC extending into the colon than in those with disease limited to the small intestine. We also investigated the relationship between gestational maturation and NEC-related mortality. Methods: A retrospective study of 153 patients compared demographic, clinical, and histopathological information in infants who had NEC limited to the small intestine vs disease with colonic involvement. Results: Our 153 infants had a mean (±standard deviation) gestational age of 27.4 ± 3.4 weeks and a birth weight of 987 ± 505 g. NEC was limited to the small intestine in 103 (67.3%) infants and extended into the colon in 50 (32.7%). Infants with small intestinal NEC needed shorter bowel resections of 28 ± 31.9 cm than 42.2 ± 40.7 cm in those with colonic involvement (p = 0.02). The LOS was longer in NEC limited to the small intestine than in disease with colonic lesions (96 ± 88.1 vs 69.7 ± 19.1 days; p <0.05). In small intestinal NEC, mortality decreased to <50% beyond a gestational age (GA) >37 weeks. In contrast, infants with NEC that involved the colon had mortality <50% mortality beyond 27.3 weeks’ GA (p = 0.008). Conclusions: Bowel resections may be more likely associated with shorter LOS in surgical NEC that involves both the small bowel and colon, even when longer segments of the gastrointestinal tract are removed, than in disease limited to the small intestine.
Collapse
Affiliation(s)
- Parvesh M Garg
- Department of Pediatrics/Neonatology, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Jaslyn L Paschal
- Department of Pediatrics/Neonatology, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Katherine Lett
- Department of Pediatrics/Neonatology, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Charles Middleton
- Department of Pathology, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Neha Varshney
- Department of Pathology, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Akhil Maheshwari
- Global Newborn Society, Clarksville, Maryland, United States of America
| |
Collapse
|
15
|
Development of the Gastrointestinal Tract in Newborns as a Challenge for an Appropriate Nutrition: A Narrative Review. Nutrients 2022; 14:nu14071405. [PMID: 35406018 PMCID: PMC9002905 DOI: 10.3390/nu14071405] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/23/2022] [Accepted: 03/23/2022] [Indexed: 02/07/2023] Open
Abstract
The second and third trimesters of pregnancy are crucial for the anatomical and functional development of the gastrointestinal (GI) tract. If premature birth occurs, the immaturity of the digestive and absorptive processes and of GI motility represent a critical challenge to meet adequate nutritional needs, leading to poor extrauterine growth and to other critical complications. Knowledge of the main developmental stages of the processes involved in the digestion and absorption of proteins, carbohydrates, and lipids, as well as of the maturational phases underlying the development of GI motility, may aid clinicians to optimize the nutritional management of preterm infants. The immaturity of these GI systems and functions may negatively influence the patterns of gut colonization, predisposing to an abnormal microbiome. This, in turn, further contributes to alter the functional, immune, and neural development of the GI tract and, especially in preterm infants, has been associated with an increased risk of severe GI complications, such as necrotizing enterocolitis. Deeper understanding of the physiological colonization patterns in term and preterm infants may support the promotion of these patterns and the avoidance of microbial perturbations associated with the development of several diseases throughout life. This review aims to provide a global overview on the maturational features of the main GI functions and on their implications following preterm birth. We will particularly focus on the developmental differences in intestinal digestion and absorption functionality, motility, gut–brain axis interaction, and microbiomes.
Collapse
|
16
|
Challenges to, and prospects for, reverse engineering the gastrointestinal tract using organoids. Trends Biotechnol 2022; 40:932-944. [DOI: 10.1016/j.tibtech.2022.01.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/09/2022] [Accepted: 01/10/2022] [Indexed: 12/29/2022]
|
17
|
Barreto E Barreto L, Rattes IC, da Costa AV, Gama P. Paneth cells and their multiple functions. Cell Biol Int 2022; 46:701-710. [PMID: 35032139 DOI: 10.1002/cbin.11764] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/30/2021] [Accepted: 01/08/2022] [Indexed: 01/08/2023]
Abstract
The small intestine mucosa is lined by specialized cells that form the crypt-villus axis, which expands its surface. Among the six intestinal epithelial cell types, the Paneth cell is located at the base of the crypt, and it contains numerous granules in its cytoplasm, composed of antimicrobial peptides, such as defensins and lysozyme, and growth factors, such as EGF, TGF-alpha, and Wnt ligands. Together, these elements act in the defense against microorganisms, regulation of intestinal microbiota, maintenance, and regulation of stem cell identity. Pathologies that target Paneth cells can disturb such defense activity, but they also affect the maintenance of stem cell niche. In that way, Crohn's disease, necrotizing enterocolitis, and graft-versus-host disease promote a reduction of Paneth cell population, and consequently of secretion of their products into the lumen of the crypts, making the affected organism predisposed to infections and dysbiosis. Additionally, the emergence of new intestinal cells is also decreased. This review aims to address the main characteristics of Paneth cells, highlighting their multiple functions and the importance of their preservation to ensure bowel homeostasis. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Laylla Barreto E Barreto
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Isadora Campos Rattes
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Aline Vasques da Costa
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Patrícia Gama
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, SP, Brazil
| |
Collapse
|
18
|
Ye L, Rawls JF. Microbial influences on gut development and gut-brain communication. Development 2021; 148:dev194936. [PMID: 34758081 PMCID: PMC8627602 DOI: 10.1242/dev.194936] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 10/07/2021] [Indexed: 12/15/2022]
Abstract
The developmental programs that build and sustain animal forms also encode the capacity to sense and adapt to the microbial world within which they evolved. This is abundantly apparent in the development of the digestive tract, which typically harbors the densest microbial communities of the body. Here, we review studies in human, mouse, zebrafish and Drosophila that are revealing how the microbiota impacts the development of the gut and its communication with the nervous system, highlighting important implications for human and animal health.
Collapse
|
19
|
Zhuang P, Xiang K, Meng X, Wang G, Li Z, Lu Y, Kan D, Zhang X, Sun SK. Gram-scale synthesis of a neodymium chelate as a spectral CT and second near-infrared window imaging agent for visualizing the gastrointestinal tract in vivo. J Mater Chem B 2021; 9:2285-2294. [PMID: 33616148 DOI: 10.1039/d0tb02276d] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The diagnosis of gastrointestinal (GI) tract diseases is frequently performed in the clinic, so it is crucial to develop high-performance contrast agents for real-time and non-invasive imaging examination of the GI tract. Herein, we show a novel method to synthesize a neodymium (Nd) chelate, Nd-diethylenetriaminepentaacetic acid (Nd-DTPA), on a large scale without byproducts for spectral computed tomography (CT) and second near-infrared window imaging of the GI tract in vivo. The Nd-DTPA was simply generated by heating the mixture of Nd2O3 and DTPA in water at 85 °C for 2 h. This dual-modal imaging agent has the advantages of a simple and green synthesis route, no need of purification process, high yield (86.24%), large-scale production capability (>10 g in lab synthesis), good chemical stability and excellent water solubility (≈2 g mL-1). Moreover, the Nd-DTPA emitted strong near-infrared fluorescence at 1308 nm, and exhibited superior X-ray attenuation ability compared to clinical iohexol. The proposed Nd-DTPA can integrate the complementary merits of dual-modal imaging to realize spatial-temporal and highly sensitive imaging of the GI tract in vivo, and accurate diagnosis of the location of intestinal obstruction and monitor its recovery after surgery. The developed highly efficient method for the gram-scale synthesis of Nd-DTPA and the proposed spectral CT and second near-infrared window dual-modal imaging strategy provide a promising route for accurate visualization of the GI tract in vivo.
Collapse
Affiliation(s)
- Pengrui Zhuang
- Department of Medical imaging, Tianjin Medical University, Tianjin 300203, China.
| | - Ke Xiang
- Department of Medical imaging, Tianjin Medical University, Tianjin 300203, China.
| | - Xiangxi Meng
- Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Guohe Wang
- Department of Medical imaging, Tianjin Medical University, Tianjin 300203, China.
| | - Ziyuan Li
- Department of Biomedical Engineering, Peking University, Beijing 100871, China
| | - Yanye Lu
- Department of Biomedical Engineering, Peking University, Beijing 100871, China
| | - Di Kan
- Department of Medical imaging, Tianjin Medical University, Tianjin 300203, China.
| | - Xuejun Zhang
- Department of Medical imaging, Tianjin Medical University, Tianjin 300203, China.
| | - Shao-Kai Sun
- Department of Medical imaging, Tianjin Medical University, Tianjin 300203, China.
| |
Collapse
|
20
|
Fournier E, Roussel C, Dominicis A, Ley D, Peyron MA, Collado V, Mercier-Bonin M, Lacroix C, Alric M, Van de Wiele T, Chassard C, Etienne-Mesmin L, Blanquet-Diot S. In vitro models of gut digestion across childhood: current developments, challenges and future trends. Biotechnol Adv 2021; 54:107796. [PMID: 34252564 DOI: 10.1016/j.biotechadv.2021.107796] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 02/08/2023]
Abstract
The human digestion is a multi-step and multi-compartment process essential for human health, at the heart of many issues raised by academics, the medical world and industrials from the food, nutrition and pharma fields. In the first years of life, major dietary changes occur and are concomitant with an evolution of the whole child digestive tract anatomy and physiology, including colonization of gut microbiota. All these phenomena are influenced by child exposure to environmental compounds, such as drugs (especially antibiotics) and food pollutants, but also childhood infections. Due to obvious ethical, regulatory and technical limitations, in vivo approaches in animal and human are more and more restricted to favor complementary in vitro approaches. This review summarizes current knowledge on the evolution of child gut physiology from birth to 3 years old regarding physicochemical, mechanical and microbial parameters. Then, all the available in vitro models of the child digestive tract are described, ranging from the simplest static mono-compartmental systems to the most sophisticated dynamic and multi-compartmental models, and mimicking from the oral phase to the colon compartment. Lastly, we detail the main applications of child gut models in nutritional, pharmaceutical and microbiological studies and discuss the limitations and challenges facing this field of research.
Collapse
Affiliation(s)
- Elora Fournier
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, CRNH Auvergne, 63000 Clermont-Ferrand, France; Toxalim, Research Centre in Food Toxicology, INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, 31000 Toulouse, France
| | - Charlène Roussel
- Laval University, INAF Institute of Nutrition and Functional Foods, G1V 0A6 Quebec, Canada
| | - Alessandra Dominicis
- European Reference Laboratory for E. coli, Istituto Superiore di Sanità, Rome, Italy
| | - Delphine Ley
- Université Lille 2, Faculté de Médecine, Inserm U995 Nutritional Modulation of Infection and Inflammation, 59045 Lille, France
| | - Marie-Agnès Peyron
- Université Clermont Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH Auvergne, 63000 Clermont-Ferrand, France
| | - Valérie Collado
- Université Clermont Auvergne, EA 4847, CROC, Centre de Recherche en Odontologie Clinique, 63000 Clermont-Ferrand, France
| | - Muriel Mercier-Bonin
- Toxalim, Research Centre in Food Toxicology, INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, 31000 Toulouse, France
| | - Christophe Lacroix
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, 8092 Zürich, Switzerland
| | - Monique Alric
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, CRNH Auvergne, 63000 Clermont-Ferrand, France
| | - Tom Van de Wiele
- Ghent University, Center for Microbial Ecology and Technology (CMET), Coupure Links 653, 9000 Ghent, Belgium
| | - Christophe Chassard
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF, 15000 Aurillac, France
| | - Lucie Etienne-Mesmin
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, CRNH Auvergne, 63000 Clermont-Ferrand, France
| | - Stéphanie Blanquet-Diot
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, CRNH Auvergne, 63000 Clermont-Ferrand, France.
| |
Collapse
|
21
|
Takahashi T, Shiraishi A, Murata J, Matsubara S, Nakaoka S, Kirimoto S, Osawa M. Muscarinic receptor M3 contributes to intestinal stem cell maintenance via EphB/ephrin-B signaling. Life Sci Alliance 2021; 4:4/9/e202000962. [PMID: 34244422 PMCID: PMC8321669 DOI: 10.26508/lsa.202000962] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 12/31/2022] Open
Abstract
Acetylcholine (ACh) signaling through activation of nicotinic and muscarinic ACh receptors regulates expression of specific genes that mediate and sustain proliferation, differentiation, and homeostasis in the intestinal crypts. This signaling plays a pivotal role in the regulation of intestinal stem cell function, but the details have not been clarified. Here, we performed experiments using type 3 muscarinic acetylcholine receptor (M3) knockout mice and their intestinal organoids and report that endogenous ACh affects the size of the intestinal stem niche via M3 signaling. RNA sequencing of crypts identified up-regulation of the EphB/ephrin-B signaling pathway. Furthermore, using an MEK inhibitor (U0126), we found that mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signaling, which is downstream of EphB/ephrin-B signaling, is activated in M3-deficient crypts. Collectively, M3, EphB/ephrin-B, and the MAPK/ERK signaling cascade work together to maintain the homeostasis of intestinal epithelial cell growth and differentiation following modifications of the cholinergic intestinal niche.
Collapse
Affiliation(s)
- Toshio Takahashi
- Suntory Foundation for Life Sciences, Bioorganic Research Institute, Kyoto, Japan
| | - Akira Shiraishi
- Suntory Foundation for Life Sciences, Bioorganic Research Institute, Kyoto, Japan
| | - Jun Murata
- Suntory Foundation for Life Sciences, Bioorganic Research Institute, Kyoto, Japan
| | - Shin Matsubara
- Suntory Foundation for Life Sciences, Bioorganic Research Institute, Kyoto, Japan
| | | | | | - Masatake Osawa
- Department of Regenerative Medicine and Applied Biomedical Sciences, Graduate School of Medicine, Gifu University, Gifu, Japan.,Center for Highly Advanced Integration of Nano and Life Sciences, Gifu University (G-CHAIN), Gifu, Japan
| |
Collapse
|
22
|
Maternal effects in mammals: Broadening our understanding of offspring programming. Front Neuroendocrinol 2021; 62:100924. [PMID: 33992652 DOI: 10.1016/j.yfrne.2021.100924] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/18/2021] [Accepted: 05/11/2021] [Indexed: 12/17/2022]
Abstract
The perinatal period is a sensitive time in mammalian development that can have long-lasting consequences on offspring phenotype via maternal effects. Maternal effects have been most intensively studied with respect to two major conditions: maternal diet and maternal stress. In this review, we shift the focus by discussing five major additional maternal cues and their influence on offspring phenotype: maternal androgen levels, photoperiod (melatonin), microbiome, immune regulation, and milk composition. We present the key findings for each of these topics in mammals, their mechanisms of action, and how they interact with each other and with the maternal influences of diet and stress. We explore their impacts in the contexts of both predictive adaptive responses and the developmental origins of disease, identify knowledge gaps and research opportunities in the field, and place a particular emphasis on the application and consideration of these effects in non-model species and natural ecological systems.
Collapse
|
23
|
Luo S, Terciolo C, Neves M, Puel S, Naylies C, Lippi Y, Pinton P, Oswald IP. Comparative sensitivity of proliferative and differentiated intestinal epithelial cells to the food contaminant, deoxynivalenol. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 277:116818. [PMID: 33752036 DOI: 10.1016/j.envpol.2021.116818] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 02/11/2021] [Accepted: 02/19/2021] [Indexed: 06/12/2023]
Abstract
The intestinal epithelium is a functional and physical barrier formed by a cell monolayer that constantly differentiates from a stem cell in the crypt. This is the first target for food contaminants, especially mycotoxins. Deoxynivalenol (DON) is one of the most prevalent mycotoxins. This study compared the effects of DON (0-100 μM) on proliferative and differentiated intestinal epithelial cells. Three cell viability assays (LDH release, ATP content and neutral red uptake) indicated that proliferative Caco-2 cells are more sensitive to DON than differentiated ones. The establishment of transepithelial electrical resistance (TEER), as a read out of the differentiation process, was delayed in proliferative cells after exposure to 1 μM DON. Transcriptome analysis of proliferative and differentiated exposure to 0-3 μM DON for 24 h revealed 4862 differentially expressed genes (DEG) and indicated an effect of both the differentiation status and the DON treatment. KEGG enrichment analysis indicated involvement of metabolism, ECM receptors and tight junctions in the differentiation process, while ribosome biogenesis, mRNA surveillance, and the MAPK pathway were involved in the response to DON. The number of differentially expressed genes and the amplitude of the effect were higher in proliferative cells exposed to DON than that in differentiated cells. In conclusion, our study shows that proliferative cells are more susceptible than differentiated ones to DON and that the mycotoxin delays the differentiation process.
Collapse
Affiliation(s)
- Su Luo
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Chloe Terciolo
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Manon Neves
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Sylvie Puel
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Claire Naylies
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Yannick Lippi
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Philippe Pinton
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Isabelle P Oswald
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France.
| |
Collapse
|
24
|
Zgheib H, Belguesmia Y, Boukherroub R, Drider D. Alginate Nanoparticles Enhance Anti-Clostridium perfringens Activity of the Leaderless Two-Peptide Enterocin DD14 and Affect Expression of Some Virulence Factors. Probiotics Antimicrob Proteins 2021; 13:1213-1227. [PMID: 33481224 DOI: 10.1007/s12602-020-09730-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2020] [Indexed: 02/06/2023]
Abstract
Here, we report a novel approach to improve the anti-Clostridium perfringens activity of the leaderless two-peptide enterocin 14 (EntDD14), produced by Enterococcus faecalis 14. This strategy consists of loading EntDD14 onto alginate nanoparticles (Alg NPs), which are made of a safe polymer. The resulting formulation (EntDD14/Alg NPs) was able to reduce up to four times the minimum inhibitory concentration (MIC) of EntDD14 against C. perfringens pathogenic strains isolated from a chicken affected by necrotic enteritis (NE). Interestingly, this formulation remained active under conditions mimicking the human and chicken gastric tract. Assays conducted to establish the impact of this formulation on the intestinal epithelial cell line Caco-2 and the human colorectal adenocarcinoma cell line HT29 revealed the absence of cytotoxicity of both free-EntDD14 and EntDD14 loaded onto the alginate nanoparticles (EntDD14/Alg NPs) against the aforementioned eukaryotic cells, after 24 h of contact. Notably, EntDD14 and EntDD14/Alg NPs, both at a sub-inhibitory concentration, affected the expression of genes coding for clostridial toxins such as toxin α, enteritis B-like toxin, collagen adhesion protein and thiol-activated cytolysin. Further, expression of these genes was significantly down-regulated following the addition of EntDD14/Alg NPs, but not affected upon addition of EntDD14 alone. This study revealed that adsorption of EntDD14 onto Alg NPs leads to a safe and active formulation (EntDD14/Alg NPs) capable of affecting the pathogenicity of C. perfringens. This formulation could therefore be used in the poultry industry as a novel approach to tackle NE.
Collapse
Affiliation(s)
- Hassan Zgheib
- UMR Transfrontalière BioEcoAgro1158, Univ. Lille, INRAE, Univ. Liège, UPJV, YNCREA, Univ. Artois, Univ. Littoral Côte D'Opale, ICV - Institut Charles Viollette, 59000, Lille, France
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France. UMR, 8520 - IEMN, 59000, Lille, France
| | - Yanath Belguesmia
- UMR Transfrontalière BioEcoAgro1158, Univ. Lille, INRAE, Univ. Liège, UPJV, YNCREA, Univ. Artois, Univ. Littoral Côte D'Opale, ICV - Institut Charles Viollette, 59000, Lille, France.
| | - Rabah Boukherroub
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France. UMR, 8520 - IEMN, 59000, Lille, France
| | - Djamel Drider
- UMR Transfrontalière BioEcoAgro1158, Univ. Lille, INRAE, Univ. Liège, UPJV, YNCREA, Univ. Artois, Univ. Littoral Côte D'Opale, ICV - Institut Charles Viollette, 59000, Lille, France
| |
Collapse
|
25
|
Exploring the impact of intestinal ion transport on the gut microbiota. Comput Struct Biotechnol J 2020; 19:134-144. [PMID: 33425246 PMCID: PMC7773683 DOI: 10.1016/j.csbj.2020.12.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota and the host are intimately connected. The host physiology dictates the intestinal environment through regulation of pH, ion concentration, mucus production, etc., all of which exerts a selective pressure on the gut microbiota. Since different regions of the gastrointestinal tract are characterized by their own physicochemical conditions, distinct microbial communities are present in these locations. While it is widely accepted that the intestinal microbiome influences the host (tight junctions, cytokine/immune responses, diarrhea, etc.), the reciprocal interaction of the host on the microbiome is under-explored. This review aims to address these gaps in knowledge by focusing on how the host intestinal ion transport influences the luminal environment and thereby modulates the gut microbiota composition.
Collapse
Key Words
- CFTR
- CFTR, cystic fibrosis transmembrane regulator
- ClC, chloride channel
- DRA
- DRA, down-regulated in adenoma
- ENaC, epithelial Na+ channel
- GI, gastrointestinal
- GLUT2
- GLUT2, glucose transporter 2
- Gastrointestinal
- Ion transport
- Microbiome
- Microbiota
- NHE2
- NHE2, sodium-hydrogen exchanger isoform 2
- NHE3
- NHE3, sodium-hydrogen exchanger isoform 3
- NKCC1, Na+-K+-2Cl− co-transporter
- OTUs, operational taxonomic units
- SGLT1, sodium glucose co-transporter 1
Collapse
|
26
|
Turner JM, George P, Lansing M, Slim G, Wizzard PR, Nation P, Brubaker PL, Wales PW. In the Short-term, Milk Fat Globule Epidermal Growth Factor-8 Causes Site-specific Intestinal Growth in Resected Piglets. J Pediatr Gastroenterol Nutr 2020; 71:543-549. [PMID: 32910624 DOI: 10.1097/mpg.0000000000002818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Short bowel syndrome (SBS) remains the leading cause of neonatal intestinal failure. Milk fat globule epidermal growth factor-8 (MFG-E8), present in human milk, has homology with epidermal growth factor (EGF), known to enhance adaptation in SBS. In this pilot study, the role of oral MFG-E8 treatment in SBS was explored in neonatal piglets. METHODS Neonatal piglets underwent 75% intestinal resection, either distal (jejunal-colonic [JC] anastomosis) or mid-intestinal (jejunal-ileal [JI] anastomosis). Piglets were randomized to intragastric treatment with MFG-E8 (5 mg/kg per day) or saline and were maintained on parenteral nutrition and enteral nutrition for 7 days. Adaptation was assessed by intestinal length and weight, histopathology, fecal fat analysis and RT-qPCR analysis of mucosal transcripts, including growth factors. RESULTS JI piglets demonstrated intestinal lengthening (P < 0.001), 2-fold greater in ileum than jejunum (P = 0.02), where lengthening was increased by MFG-E8 treatment (P = 0.02). JC piglets did not exhibit jejunal lengthening, regardless of treatment. Fat absorption was greater for JI piglets (P = 0.02), unaffected by treatment. In JI piglets, expression of Egf was increased in the ileum (P < 0.01) and MFG-E8 treatment increased Egfr (receptor) expression (P = 0.02). CONCLUSIONS MF-EG8 demonstrated site-specific trophic effects, only with JI anatomy. This may limit the utility of this treatment for SBS, except for rare patients with retained ileum. The mechanisms of these site-specific effects, however, and the role of MFG-E8 in neonatal gut growth and in diseases, such as necrotizing enterocolitis that commonly target ileum, warrant further exploration.
Collapse
Affiliation(s)
| | | | | | | | | | - Patrick Nation
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta
| | | | - Paul W Wales
- Department of Pediatrics.,Department of Surgery, University of Toronto.,Group for the Improvement of Intestinal Function and Treatment (GIFT), Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
27
|
Anguita-Ruiz A, Aguilera CM, Gil Á. Genetics of Lactose Intolerance: An Updated Review and Online Interactive World Maps of Phenotype and Genotype Frequencies. Nutrients 2020; 12:nu12092689. [PMID: 32899182 PMCID: PMC7551416 DOI: 10.3390/nu12092689] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 08/28/2020] [Accepted: 09/01/2020] [Indexed: 01/15/2023] Open
Abstract
In humans the ability to digest milk lactose is conferred by a β-galactosidase enzyme called lactase-phlorizin hydrolase (LPH). While in some humans (approximately two-thirds of humankind) the levels of this enzyme decline drastically after the weaning phase (a trait known as lactase non-persistence (LNP)), some other individuals are capable of maintaining high levels of LPH lifelong (lactase persistence (LP)), thus being able to digest milk during adulthood. Both lactase phenotypes in humans present a complex genetic basis and have been widely investigated during the last decades. The distribution of lactase phenotypes and their associated single nucleotide polymorphisms (SNPs) across human populations has also been extensively studied, though not recently reviewed. All available information has always been presented in the form of static world maps or large dimension tables, so that it would benefit from the newly available visualization tools, such as interactive world maps. Taking all this into consideration, the aims of the present review were: (1) to gather and summarize all available information on LNP and LP genetic mechanisms and evolutionary adaptation theories, and (2) to create online interactive world maps, including all LP phenotype and genotype frequency data reported to date. As a result, we have created two online interactive resources, which constitute an upgrade over previously published static world maps, and allow users a personalized data exploration, while at the same time accessing complete reports by population or ethnicity.
Collapse
Affiliation(s)
- Augusto Anguita-Ruiz
- Department of Biochemistry and Molecular Biology II, Institute of Nutrition and Food Technology “José Mataix”, Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n. Armilla, 18016 Granada, Spain; (A.A.-R.); (C.M.A.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18014 Granada, Spain
- CIBEROBN (Physiopathology of Obesity and Nutrition Network CB12/03/30038), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Concepción M. Aguilera
- Department of Biochemistry and Molecular Biology II, Institute of Nutrition and Food Technology “José Mataix”, Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n. Armilla, 18016 Granada, Spain; (A.A.-R.); (C.M.A.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18014 Granada, Spain
- CIBEROBN (Physiopathology of Obesity and Nutrition Network CB12/03/30038), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
| | - Ángel Gil
- Department of Biochemistry and Molecular Biology II, Institute of Nutrition and Food Technology “José Mataix”, Center of Biomedical Research, University of Granada, Avda. del Conocimiento s/n. Armilla, 18016 Granada, Spain; (A.A.-R.); (C.M.A.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18014 Granada, Spain
- CIBEROBN (Physiopathology of Obesity and Nutrition Network CB12/03/30038), Institute of Health Carlos III (ISCIII), 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-958241000 (ext. 20307)
| |
Collapse
|
28
|
Jena A, Montoya CA, Mullaney JA, Dilger RN, Young W, McNabb WC, Roy NC. Gut-Brain Axis in the Early Postnatal Years of Life: A Developmental Perspective. Front Integr Neurosci 2020; 14:44. [PMID: 32848651 PMCID: PMC7419604 DOI: 10.3389/fnint.2020.00044] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/20/2020] [Indexed: 12/11/2022] Open
Abstract
Emerging evidence suggests that alterations in the development of the gastrointestinal (GI) tract during the early postnatal period can influence brain development and vice-versa. It is increasingly recognized that communication between the GI tract and brain is mainly driven by neural, endocrine, immune, and metabolic mediators, collectively called the gut-brain axis (GBA). Changes in the GBA mediators occur in response to the developmental changes in the body during this period. This review provides an overview of major developmental events in the GI tract and brain in the early postnatal period and their parallel developmental trajectories under physiological conditions. Current knowledge of GBA mediators in context to brain function and behavioral outcomes and their synthesis and metabolism (site, timing, etc.) is discussed. This review also presents hypotheses on the role of the GBA mediators in response to the parallel development of the GI tract and brain in infants.
Collapse
Affiliation(s)
- Ankita Jena
- School of Food & Advanced Technology, College of Sciences, Massey University, Palmerston North, New Zealand.,The Riddet Institute, Massey University, Palmerston North, New Zealand.,Food Nutrition & Health, Grasslands Research Centre, AgResearch, Palmerston North, New Zealand
| | - Carlos A Montoya
- The Riddet Institute, Massey University, Palmerston North, New Zealand.,Food Nutrition & Health, Grasslands Research Centre, AgResearch, Palmerston North, New Zealand
| | - Jane A Mullaney
- The Riddet Institute, Massey University, Palmerston North, New Zealand.,Food Nutrition & Health, Grasslands Research Centre, AgResearch, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Ryan N Dilger
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Wayne Young
- The Riddet Institute, Massey University, Palmerston North, New Zealand.,Food Nutrition & Health, Grasslands Research Centre, AgResearch, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Warren C McNabb
- The Riddet Institute, Massey University, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Nicole C Roy
- The Riddet Institute, Massey University, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand.,Liggins Institute, The University of Auckland, Auckland, New Zealand.,Department of Human Nutrition, University of Otago, Dunedin, New Zealand
| |
Collapse
|
29
|
Burrin D, Sangild PT, Stoll B, Thymann T, Buddington R, Marini J, Olutoye O, Shulman RJ. Translational Advances in Pediatric Nutrition and Gastroenterology: New Insights from Pig Models. Annu Rev Anim Biosci 2020; 8:321-354. [PMID: 32069436 DOI: 10.1146/annurev-animal-020518-115142] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Pigs are increasingly important animals for modeling human pediatric nutrition and gastroenterology and complementing mechanistic studies in rodents. The comparative advantages in size and physiology of the neonatal pig have led to new translational and clinically relevant models of important diseases of the gastrointestinal tract and liver in premature infants. Studies in pigs have established the essential roles of prematurity, microbial colonization, and enteral nutrition in the pathogenesis of necrotizing enterocolitis. Studies in neonatal pigs have demonstrated the intestinal trophic effects of akey gut hormone, glucagon-like peptide 2 (GLP-2), and its role in the intestinal adaptation process and efficacy in the treatment of short bowel syndrome. Further, pigs have been instrumental in elucidating the physiology of parenteral nutrition-associated liver disease and the means by which phytosterols, fibroblast growth factor 19, and a new generation of lipid emulsions may modify disease. The premature pig will continue to be a valuable model in the development of optimal infant diets (donor human milk, colostrum), specific milk bioactives (arginine, growth factors), gut microbiota modifiers (pre-, pro-, and antibiotics), pharmaceutical drugs (GLP-2 analogs, FXR agonists), and novel diagnostic tools (near-infrared spectroscopy) to prevent and treat these pediatric diseases.
Collapse
Affiliation(s)
- Douglas Burrin
- USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Baylor College of Medicine, Houston, Texas 77030, USA;
| | - Per Torp Sangild
- Comparative Pediatrics and Nutrition, University of Copenhagen, DK-1870 Frederiksberg C., Copenhagen, Denmark
| | - Barbara Stoll
- USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Baylor College of Medicine, Houston, Texas 77030, USA;
| | - Thomas Thymann
- Comparative Pediatrics and Nutrition, University of Copenhagen, DK-1870 Frederiksberg C., Copenhagen, Denmark
| | - Randal Buddington
- College of Nursing, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Juan Marini
- USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Baylor College of Medicine, Houston, Texas 77030, USA; .,Department of Pediatrics, Section of Critical Care Medicine, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Oluyinka Olutoye
- Division of Pediatric Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Robert J Shulman
- USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Baylor College of Medicine, Houston, Texas 77030, USA;
| |
Collapse
|
30
|
Khanal P, D. Axel AM, Safayi S, Elbrønd VS, Nielsen MO. Prenatal over- and undernutrition differentially program small intestinal growth, angiogenesis, absorptive capacity, and endocrine function in sheep. Physiol Rep 2020; 8:e14498. [PMID: 32597039 PMCID: PMC7322502 DOI: 10.14814/phy2.14498] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/27/2020] [Accepted: 06/02/2020] [Indexed: 12/20/2022] Open
Abstract
The aim was to test the hypothesis that prenatal under- and overnutrition in late gestation can program small intestinal (SI) growth, angiogenesis, and endocrine function to predispose for a hyperabsorptive state, thereby increasing the susceptibility to the adverse effects of an early postnatal obesogenic diet. Twin-pregnant ewes were exposed to adequate (NORM), LOW (50% of NORM), or HIGH (150% energy and 110% protein of NORM) diets through the last trimester (term ~147 days). From 3 days to 6 months of age, their lambs were fed either a moderate (CONV) or a high-carbohydrate high-fat (HCHF) diet. At 6 months of age, responses in plasma metabolites and insulin to refeeding after fasting were determined and then different segments of the SI were sampled at autopsy. Prenatal overnutrition impacts were most abundant in the duodenum where HIGH had increased villus amplification factor and lowered villi thickness with increased IRS-1 and reduced GH-R expressions. In jejunum, HIGH lambs had an increased expression of Lactate gene and amplified when exposed to HCHF postnatally. Specifically, in LOW, sensitivity to HCHF was affected in ileum. Thus, the mismatching LOW-HCHF nutrition increased expressions of angiogenic genes (VEGF, VEGF-R1, ANGPT1, RTK) and increased mucosa layer (tunica mucosa) thickness but reduced muscle layer (Tunica muscularis) thickness. The SI is a target of prenatal nutritional programming, where late gestation overnutrition increased and shifted digestive capacity for carbohydrates toward the jejunum, whereas late gestation undernutrition predisposed for ileal angiogenesis and carbohydrate and fat hyperabsorptive capacity upon subsequent exposure to postnatal obesogenic diet.
Collapse
Affiliation(s)
- Prabhat Khanal
- Faculty of Biosciences and AquacultureAnimal Science, Production and Welfare DivisionNord UniversitySteinkjerNorway
| | - Anne Marie D. Axel
- Department of Veterinary and Animal SciencesFaculty of Health and Medical SciencesUniversity of CopenhagenFrederiksbergDenmark
| | | | - Vibeke S. Elbrønd
- Department of Veterinary and Animal SciencesFaculty of Health and Medical SciencesUniversity of CopenhagenFrederiksbergDenmark
| | - Mette O. Nielsen
- Department of Animal ScienceFaculty of Technical SciencesAarhus UniversityTjeleDenmark
| |
Collapse
|
31
|
Coon JI, Jain S, Sepuru KM, Chung Y, Mohankumar K, Rajarathnam K, Jain SK. Lyophilization of human amniotic fluid is feasible without affecting biological activity. Pediatr Res 2020; 87:847-852. [PMID: 31756731 DOI: 10.1038/s41390-019-0632-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 09/20/2019] [Accepted: 10/02/2019] [Indexed: 11/09/2022]
Abstract
BACKGROUND Fetal swallowing of human amniotic fluid (hAF) containing trophic factors (TFs) promotes gastrointestinal tract (GIT) development. Preterm birth interrupts hAF swallowing, which may increase the risk of necrotizing enterocolitis (NEC). Postnatally, it is difficult to replicate fetal swallowing of hAF due to volume. We aimed to evaluate whether hAF lyophilization is feasible and its effect on hAF-borne TFs. METHODS We collected hAF (n = 16) from uncomplicated pregnancies. hAF was divided into three groups: unprocessed control (C), concentration by microfiltration (F), and by dialysis and lyophilization (L). EGF, HGF, GM-CSF, and TGF-α were measured in each group by multiplex assay. Bioavailability of TFs was measured by proliferation and LPS-induced IL-8 production by intestinal epithelial cells FHs74. RESULTS After dialysis/lyophilization, GM-CSF and TGF-α were preserved with partial loss of EGF and HGF. hAF increased cell proliferation and reduced LPS-induced IL-8 production compared to medium alone. Compared to control, dialysis/lyophilization and filtration of hAF increased FHs74 cell proliferation (p < 0.001) and decreased LPS-induced IL-8 production (p < 0.01). CONCLUSIONS Lyophilization and filtration of hAF is feasible with partial loss of TFs but maintains and even improves bioavailability of TFs measured by proliferation and LPS-induced IL-8 production by FHs74.
Collapse
Affiliation(s)
- John I Coon
- Department of Pediatrics, Division of Neonatology, University of Texas Medical Branch, Galveston, TX, USA
| | - Sangeeta Jain
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Krishna M Sepuru
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Yerin Chung
- Department of Pediatrics, Division of Neonatology, John Hopkins University, Baltimore, MD, USA
| | - Krishnan Mohankumar
- Department of Pediatrics, Division of Neonatology, John Hopkins University, Baltimore, MD, USA
| | - Krishna Rajarathnam
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Sunil K Jain
- Department of Pediatrics, Division of Neonatology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
32
|
Somaratne G, Ferrua MJ, Ye A, Nau F, Floury J, Dupont D, Singh J. Food material properties as determining factors in nutrient release during human gastric digestion: a review. Crit Rev Food Sci Nutr 2020; 60:3753-3769. [PMID: 31957483 DOI: 10.1080/10408398.2019.1707770] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The fundamental mechanisms of nutrient release from solid foods during gastric digestion consists of multiple elementary processes. These include the diffusion of gastric juice into the food matrix and its simultaneous enzymatic degradation and mechanical breakdown by the peristaltic activity of the stomach. Understanding the relative role of these key processes, in association with the composition and structure of foods, is of paramount importance for the design and manufacture of novel foods possessing specific target behavior within the body. This review covers the past and current literature with respect to the in-stomach processes leading to physical and biochemical disintegration of solid foods and release of nutrients. The review outlines recent progress in experimental and modeling methods used for studying food disintegration mechanisms and concludes with a discussion on potential future research directions in this field. Information from pharmaceutical science-based modeling approaches describing nutrient release kinetics as a result of food disintegration in the gastric environment is also reviewed. Future research aimed at understanding gastric digestion is important not only for setting design principles for novel food design but also for understanding mechanisms underpinning dietary guidelines to consume wholesome foods.
Collapse
Affiliation(s)
- Geeshani Somaratne
- Riddet Institute, Massey University, Palmerston North, New Zealand.,School of food and Advanced Technology, Massey University, Palmerston North, New Zealand
| | - Maria J Ferrua
- Riddet Institute, Massey University, Palmerston North, New Zealand.,Fonterra Research and Development Centre, Palmerston North, New Zealand
| | - Aiqian Ye
- Riddet Institute, Massey University, Palmerston North, New Zealand
| | | | | | | | - Jaspreet Singh
- Riddet Institute, Massey University, Palmerston North, New Zealand.,School of food and Advanced Technology, Massey University, Palmerston North, New Zealand
| |
Collapse
|
33
|
Fundora JB, Guha P, Shores DR, Pammi M, Maheshwari A. Intestinal dysbiosis and necrotizing enterocolitis: assessment for causality using Bradford Hill criteria. Pediatr Res 2020; 87:235-248. [PMID: 31238334 PMCID: PMC7224339 DOI: 10.1038/s41390-019-0482-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/11/2019] [Accepted: 06/18/2019] [Indexed: 02/08/2023]
Abstract
In recent years, several studies have shown that premature infants who develop NEC frequently display enteric dysbiosis with increased Gram-negative bacteria for several days to weeks prior to NEC onset. The importance of these findings, for the possibility of a causal role of these bacteria in NEC pathogenesis, and for potential value of gut dysbiosis as a biomarker of NEC, is well-recognized. In this review, we present current evidence supporting the association between NEC in premature infants and enteric dysbiosis, and its evaluation using the Bradford Hill criteria for causality. To provide an objective appraisal, we developed a novel scoring system for causal inference. Despite important methodological and statistical limitations, there is support for the association from several large studies and a meta-analysis. The association draws strength from strong biological plausibility of a role of Gram-negative bacteria in NEC and from evidence for temporality, that dysbiosis may antedate NEC onset. The weakness of the association is in the low level of consistency across studies, and the lack of specificity of effect. There is a need for an improved definition of dysbiosis, either based on a critical threshold of relative abundances or at higher levels of taxonomic resolution.
Collapse
Affiliation(s)
- Jennifer B Fundora
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Pallabi Guha
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Darla R Shores
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mohan Pammi
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Akhil Maheshwari
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
34
|
Early life nutrition influences susceptibility to chronic inflammatory colitis in later life. Sci Rep 2019; 9:18111. [PMID: 31792267 PMCID: PMC6889478 DOI: 10.1038/s41598-019-54308-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 11/08/2019] [Indexed: 12/21/2022] Open
Abstract
The first thousand days of life are a critical time of development in humans during which the risk profile for diseases in later life can be modified. Nevertheless, long-term consequences of early environment on susceptibility to intestinal diseases have not yet been assessed. Using a mouse model of postnatal growth restriction (PNGR), we showed that early life nutrition influences intestinal maturation and gut health in later life. PNGR induced an alteration of the intestinal barrier in pups at weaning, resulting in increased intestinal permeability, and affected gut bacterial colonization. Specifically, pups with PNGR harbored a decreased bacterial diversity, higher Enterococcus spp., Staphylococcus spp., and Escherichia-Shigella spp., and lower Odoribacter spp. and several members of the Lachnospiraceae family. The lack of an efficient intestinal barrier in early life and the dysbiosis induced by PNGR were associated with a higher susceptibility to chronic colitis in adulthood.
Collapse
|
35
|
Wang J, Lu H, Wang W, Zheng N, Wang Y, Hu Z, Ji G. Hepatocyte Nuclear Factor 3β Plays a Suppressive Role in Colorectal Cancer Progression. Front Oncol 2019; 9:1096. [PMID: 31696055 PMCID: PMC6817462 DOI: 10.3389/fonc.2019.01096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 10/04/2019] [Indexed: 11/17/2022] Open
Abstract
Background and Objective: Hepatocyte nuclear factor 3β (HNF3β) is a key transcription factor in the development of the gastrointestinal tract. However, only few studies have examined its' expression, function and potential clinical significance in colorectal cancer tumorigenesis and progression. Methods: HNF3β expression in colorectal cancer tissue samples of 174 patients was assessed by immunohistochemistry. The results were analyzed with respect to patients' clinicopathological characteristics and survival. Following the in vitro cell transfection, MTT, wound healing, and Transwell assays were used to test cell proliferation, migration, and invasion, respectively. Western blot was used to examine IL6, JAK1, and STAT3 protein expression. The potential for tumor formation was evaluated using a mouse xenograft model. Results: HNF3β expression was lower in colon cancer tissue compared to normal tissue and correlated with UICC clinical stage (P = 0.001), depth of invasion (P = 0.004), regional lymph node metastasis (P = 0.007), distant metastasis (P = 0.048), and poor survival (P < 0.001) in patients with colorectal cancer. Furthermore, HNF3β overexpression impeded proliferation, migration and invasion of SW480 cells via JAK-STAT3 signaling in vitro. Moreso, HNF3β overexpression showed a significant growth inhibition of subcutaneous xenograft tumors in vivo. Conclusions: The results show that HNF3β acts as a suppressor of colorectal cancer progression and decreased HNF3 β expression is closely related to the poor prognosis. Thus, HNF3β may be a potential molecular target for inhibition of colorectal cancer cells and development of new anti-tumor therapies.
Collapse
Affiliation(s)
- Juan Wang
- State Key Laboratory of Cancer Biology, Department of Digestive Surgery, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| | - Hao Lu
- Department of General Surgery, Chang Zheng Hospital, Second Military Medical University, Shanghai, China
| | - Wei Wang
- State Key Laboratory of Cancer Biology, Department of Pharmaceutical and Pharmacy Administration, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Nanxin Zheng
- Department of Colorectal Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yi Wang
- Department of General Surgery, Chang Zheng Hospital, Second Military Medical University, Shanghai, China
| | - Zhiqian Hu
- Department of General Surgery, Chang Zheng Hospital, Second Military Medical University, Shanghai, China
| | - Gang Ji
- State Key Laboratory of Cancer Biology, Department of Digestive Surgery, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
36
|
Gonçalves O, Freitas R, Ferreira P, Araújo M, Zhang G, Mazan S, Cohn MJ, Castro LFC, Wilson JM. Molecular ontogeny of the stomach in the catshark Scyliorhinus canicula. Sci Rep 2019; 9:586. [PMID: 30679499 PMCID: PMC6346038 DOI: 10.1038/s41598-018-36413-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 11/21/2018] [Indexed: 01/27/2023] Open
Abstract
The origin of extracellular digestion in metazoans was accompanied by structural and physiological alterations of the gut. These adaptations culminated in the differentiation of a novel digestive structure in jawed vertebrates, the stomach. Specific endoderm/mesenchyme signalling is required for stomach differentiation, involving the growth and transcription factors: 1) Shh and Bmp4, required for stomach outgrowth; 2) Barx1, Sfrps and Sox2, required for gastric epithelium development and 3) Cdx1 and Cdx2, involved in intestinal versus gastric identity. Thus, modulation of endoderm/mesenchyme signalling emerges as a plausible mechanism linked to the origin of the stomach. In order to gain insight into the ancient mechanisms capable of generating this structure in jawed vertebrates, we characterised the development of the gut in the catshark Scyliorhinus canicula. As chondrichthyans, these animals retained plesiomorphic features of jawed vertebrates, including a well-differentiated stomach. We identified a clear molecular regionalization of their embryonic gut, characterised by the expression of barx1 and sox2 in the prospective stomach region and expression of cdx1 and cdx2 in the prospective intestine. Furthermore, we show that gastric gland development occurs close to hatching, accompanied by the onset of gastric proton pump activity. Our findings favour a scenario in which the developmental mechanisms involved in the origin of the stomach were present in the common ancestor of chondrichthyans and osteichthyans.
Collapse
Affiliation(s)
- Odete Gonçalves
- CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, Univ. Porto, Porto, Portugal.,Institute of Biomedical Sciences Abel Salazar (ICBAS), Univ. Porto, Porto, Portugal
| | - Renata Freitas
- I3S- Institute for Innovation and Health Research, Univ. Porto, Porto, Portugal. .,IBMC- Institute for Molecular and Cell Biology, Univ. Porto, Porto, Portugal.
| | - Patrícia Ferreira
- CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, Univ. Porto, Porto, Portugal.,Institute of Biomedical Sciences Abel Salazar (ICBAS), Univ. Porto, Porto, Portugal
| | - Mafalda Araújo
- I3S- Institute for Innovation and Health Research, Univ. Porto, Porto, Portugal.,IBMC- Institute for Molecular and Cell Biology, Univ. Porto, Porto, Portugal
| | - GuangJun Zhang
- Department of Comparative Pathobiology, Purdue Univ., Lafayette, USA.,Purdue Institute for Integrative Neuroscience, Purdue Univ., Lafayette, USA.,Purdue Univ. Center for Cancer, Purdue Univ., Lafayette, USA.,Purdue Institute for Inflammation, Immunology and Infectious Diseases, Purdue Univ., Lafayette, USA
| | - Sylvie Mazan
- CNRS, Sorbonne Universités, UPMC Univ. Paris, Observatoire Océanologique, Banyuls, France
| | - Martin J Cohn
- Howard Hughes Medical Institute, UF Genetics Institute, Univ. Florida, Florida, USA.,Department of Biology, UF Genetics Institute, Univ. Florida, Florida, USA.,Department of Molecular Genetics and Microbiology, UF Genetics Institute, Univ. Florida, Florida, USA
| | - L Filipe C Castro
- CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, Univ. Porto, Porto, Portugal. .,Department of Biology, Faculty of Sciences, Univ. Porto, Porto, Portugal.
| | - Jonathan M Wilson
- CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, Univ. Porto, Porto, Portugal. .,Department of Biology, Wilfrid Laurier Univ., Waterloo, Canada.
| |
Collapse
|
37
|
Lenfestey MW, Neu J. Gastrointestinal Development: Implications for Management of Preterm and Term Infants. Gastroenterol Clin North Am 2018; 47:773-791. [PMID: 30337032 DOI: 10.1016/j.gtc.2018.07.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The gastrointestinal (GI) system provides digestive, absorptive, neuroendocrine, and immunologic functions to support overall health. If normal development is interrupted, a variety of complications and disease can arise. This article explores normal development of the GI tract and specific clinical challenges pertinent to preterm and term infants. Specific topics include abnormal motility, gastroesophageal reflux, current feeding recommendations for preterm infants, effects of parenteral nutrition, and the relationship between the GI tract and the immune system.
Collapse
Affiliation(s)
- Mary W Lenfestey
- Department of Pediatrics, University of Florida, PO Box 100296, Gainesville, FL 32610, USA
| | - Josef Neu
- Department of Pediatrics, University of Florida, 6516 Southwest 93rd Avenue, Gainesville, FL 32610, USA.
| |
Collapse
|
38
|
McLeod JS, Church JT, Yerramilli P, Coughlin MA, Perkins EM, Rabah R, Bartlett RH, Rojas-Pena A, Greenson JK, Perrone EE, Mychaliska GB. Gastrointestinal mucosal development and injury in premature lambs supported by the artificial placenta. J Pediatr Surg 2018; 53:1240-1245. [PMID: 29605266 PMCID: PMC5994371 DOI: 10.1016/j.jpedsurg.2018.02.092] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 02/27/2018] [Indexed: 01/19/2023]
Abstract
BACKGROUND An Artificial Placenta (AP) utilizing extracorporeal life support (ECLS) could revolutionize care of extremely premature newborns, but its effects on gastrointestinal morphology and injury need investigation. METHODS Lambs (116-121days GA, term=145; n=5) were delivered by C-section, cannulated for ECLS, had total parenteral nutrition (TPN) provided, and were supported for 7days before euthanasia. Early and Late Tissue Controls (ETC, n=5 and LTC, n=5) delivered at 115-121days and 125-131days, respectively, were immediately sacrificed. Standardized jejunal samples were formalin-fixed for histology. Crypt depth (CD), villus height (VH), and VH:CD ratios were measured. Measurements also included enterocyte proliferation (Ki-67), Paneth cell count (Lysozyme), and injury scores (H&E). ANOVA and Chi Square were used with p<0.05 considered significant. RESULTS CD, VH, and VH:CD were similar between groups (p>0.05). AP demonstrated more enterocyte proliferation (95.7±21.8) than ETC (49.4±23.4; p=0.003) and LTC (66.1+11.8; p=0.04), and more Paneth cells (81.7±17.5) than ETC (41.6±7.0; p=0.0005) and LTC (40.7±8.2, p=0.0004). Presence of epithelial injury and congestion in the bowel of all groups were not statistically different. No villus atrophy or inflammation was present in any group. CONCLUSIONS This suggests preserved small bowel mucosal architecture, high cellular turnover, and minimal evidence of injury. STUDY TYPE Research paper/therapeutic potential. LEVEL OF EVIDENCE N/A.
Collapse
Affiliation(s)
- Jennifer S McLeod
- Extracorporeal Life Support Laboratory, Department of Surgery, Michigan Medicine, Ann Arbor, MI.
| | - Joseph T Church
- Extracorporeal Life Support Laboratory, Department of Surgery, Michigan Medicine, Ann Arbor, MI
| | - Prathusha Yerramilli
- Extracorporeal Life Support Laboratory, Department of Surgery, Michigan Medicine, Ann Arbor, MI
| | - Megan A Coughlin
- Extracorporeal Life Support Laboratory, Department of Surgery, Michigan Medicine, Ann Arbor, MI
| | - Elena M Perkins
- Extracorporeal Life Support Laboratory, Department of Surgery, Michigan Medicine, Ann Arbor, MI
| | - Raja Rabah
- Department of Pathology, Michigan Medicine, Ann Arbor, MI
| | - Robert H Bartlett
- Extracorporeal Life Support Laboratory, Department of Surgery, Michigan Medicine, Ann Arbor, MI
| | - Alvaro Rojas-Pena
- Extracorporeal Life Support Laboratory, Department of Surgery, Michigan Medicine, Ann Arbor, MI
| | - Joel K Greenson
- Department of Gastrointestinal Pathology, Michigan Medicine, Ann Arbor, MI
| | - Erin E Perrone
- Extracorporeal Life Support Laboratory, Department of Surgery, Michigan Medicine, Ann Arbor, MI; Fetal Diagnosis and Treatment Center, C.S. Mott Children's Hospital, Michigan Medicine, Ann Arbor, MI
| | - George B Mychaliska
- Extracorporeal Life Support Laboratory, Department of Surgery, Michigan Medicine, Ann Arbor, MI; Fetal Diagnosis and Treatment Center, C.S. Mott Children's Hospital, Michigan Medicine, Ann Arbor, MI
| |
Collapse
|
39
|
Sanctuary MR, Kain JN, Angkustsiri K, German JB. Dietary Considerations in Autism Spectrum Disorders: The Potential Role of Protein Digestion and Microbial Putrefaction in the Gut-Brain Axis. Front Nutr 2018; 5:40. [PMID: 29868601 PMCID: PMC5968124 DOI: 10.3389/fnut.2018.00040] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 04/30/2018] [Indexed: 12/13/2022] Open
Abstract
Children with autism spectrum disorders (ASD), characterized by a range of behavioral abnormalities and social deficits, display high incidence of gastrointestinal (GI) co-morbidities including chronic constipation and diarrhea. Research is now increasingly able to characterize the “fragile gut” in these children and understand the role that impairment of specific GI functions plays in the GI symptoms associated with ASD. This mechanistic understanding is extending to the interactions between diet and ASD, including food structure and protein digestive capacity in exacerbating autistic symptoms. Children with ASD and gut co-morbidities exhibit low digestive enzyme activity, impaired gut barrier integrity and the presence of antibodies specific for dietary proteins in the peripheral circulation. These findings support the hypothesis that entry of dietary peptides from the gut lumen into the vasculature are associated with an aberrant immune response. Furthermore, a subset of children with ASD exhibit high concentrations of metabolites originating from microbial activity on proteinaceous substrates. Taken together, the combination of specific protein intakes poor digestion, gut barrier integrity, microbiota composition and function all on a background of ASD represents a phenotypic pattern. A potential consequence of this pattern of conditions is that the fragile gut of some children with ASD is at risk for GI symptoms that may be amenable to improvement with specific dietary changes. There is growing evidence that shows an association between gut dysfunction and dysbiosis and ASD symptoms. It is therefore urgent to perform more experimental and clinical research on the “fragile gut” in children with ASD in order to move toward advancements in clinical practice. Identifying those factors that are of clinical value will provide an evidence-based path to individual management and targeted solutions; from real time sensing to the design of diets with personalized protein source/processing, all to improve GI function in children with ASD.
Collapse
Affiliation(s)
- Megan R Sanctuary
- Department of Nutrition, University of California, Davis, Davis, CA, United States
| | - Jennifer N Kain
- Department of Neurobiology, Physiology and Behavior Department, University of California, Davis, Davis, CA, United States
| | - Kathleen Angkustsiri
- School of Medicine, Department of Pediatrics, University of California, Davis, Sacramento, CA, United States.,Department of Pediatrics, UC Davis MIND Institute, Sacramento, CA, United States
| | - J Bruce German
- Department of Food Science and Technology, University of California, Davis, Davis, CA, United States.,Foods for Health Institute, University of California, Davis, Davis, CA, United States
| |
Collapse
|
40
|
Özbek M, Ergün E, Beyaz F, Ergün L, Y Ld R M N, Özgenç Ö, Erhan F. Prenatal development and histochemical characteristics of gastrointestinal mucins in sheep fetuses. Microsc Res Tech 2018. [PMID: 29536591 DOI: 10.1002/jemt.23018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The object of this study was to describe the prenatal development and histochemical properties of mucins in the sheep gastrointestinal tract. To determine changes in the mucin profile, the sections were stained with specific histochemical stains for carbohydrates. While neutral and mixed mucins were observed in the superficial epithelial cells of the abomasal pyloric region, acidic mucins were detected in the secretory ducts and corpus of the glands. Acidic mucins consisted predominantly of sialomucins. In the duodenal villi, the number of goblet cells containing neutral mucins increased toward the end of gestation, whereas Brunner's glands contained acidic mucins until the 95th day of gestation and both acidic and neutral mucins thereafter. The jejunal goblet cells contained either acidic, neutral, or mixed mucins. Goblet cells containing acidic mucins, which were mainly localized to the ileal crypts and villi, mostly contained sulfated mucins. While villi were observed in the proximal colon until the 115th day of gestation, later the typical crypt structure emerged. During the period in which the villi were found in the proximal colon, the goblet cells containing sulphomucins were predominant, whereas the goblet cells containing sialomucins were predominant after the typical crypt structure was formed. In conclusion, gastrointestinal mucins may be involved in the formation of meconium during the prenatal period, and acidic mucins may contribute to the strength of the intestinal barrier against pathogens and digestive enzymes, as the barrier is not fully functional after birth.
Collapse
Affiliation(s)
- Mehmet Özbek
- Department of Histology and Embryology, Mehmet Akif Ersoy University, Burdur, Turkey
| | - Emel Ergün
- Department of Histology and Embryology, Ankara University, Ankara, Turkey
| | - Feyzullah Beyaz
- Department of Histology and Embryology, Erciyes University, Kayseri, Turkey
| | - Levent Ergün
- Department of Histology and Embryology, Ankara University, Ankara, Turkey
| | - Nuh Y Ld R M
- Department of Histology and Embryology, Ankara University, Ankara, Turkey
| | - Özge Özgenç
- Department of Histology and Embryology, Ankara University, Ankara, Turkey
| | - Füsun Erhan
- Department of Histology and Embryology, Cumhuriyet University, Sivas, Turkey
| |
Collapse
|
41
|
Perochon J, Carroll LR, Cordero JB. Wnt Signalling in Intestinal Stem Cells: Lessons from Mice and Flies. Genes (Basel) 2018; 9:genes9030138. [PMID: 29498662 PMCID: PMC5867859 DOI: 10.3390/genes9030138] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 02/17/2018] [Accepted: 02/21/2018] [Indexed: 12/12/2022] Open
Abstract
Adult stem cells play critical roles in the basal maintenance of tissue integrity, also known as homeostasis, and in tissue regeneration following damage. The highly conserved Wnt signalling pathway is a key regulator of stem cell fate. In the gastrointestinal tract, Wnt signalling activation drives homeostasis and damage-induced repair. Additionally, deregulated Wnt signalling is a common hallmark of age-associated tissue dysfunction and cancer. Studies using mouse and fruit fly models have greatly improved our understanding of the functional contribution of the Wnt signalling pathway in adult intestinal biology. Here, we summarize the latest knowledge acquired from mouse and Drosophila research regarding canonical Wnt signalling and its key functions during stem cell driven intestinal homeostasis, regeneration, ageing and cancer.
Collapse
Affiliation(s)
- Jessica Perochon
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK.
| | - Lynsey R Carroll
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK.
| | - Julia B Cordero
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK.
- CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK.
| |
Collapse
|
42
|
Skarżyńska E, Kiersztyn B, Wilczyńska P, Jakimiuk A, Lisowska-Myjak B. Total proteolytic activity and concentration of alpha-1 antitrypsin in meconium for assessment of the protease/antiprotease balance. Eur J Obstet Gynecol Reprod Biol 2017; 223:133-138. [PMID: 29029865 DOI: 10.1016/j.ejogrb.2017.10.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 10/02/2017] [Indexed: 01/07/2023]
Abstract
BACKGROUND During intrauterine life, various proteolytic enzymes and their main inhibitor, alpha-1 antitrypsin, accumulate naturally in meconium. A protease/antiprotease balance is required to maintain the biological stability of the environment in which the fetus develops. METHODS The pool of active proteases was determined using the EnzChek Protease Assay Kit. The concentration of alpha-1 antitrypsin in meconium was measured by enzyme-linked immunosorbent assay. Serial portions of meconium (n=80) were collected from healthy full-term neonates (n=19). RESULTS Mean concentrations of active proteases and alpha-1 antitrypsin were 1.55 [standard deviation (SD) 1.3]mgg-1 (range 0.15-6.17) and 3.72 (SD 1.78)mgg-1 (range 0.76-8.55), respectively, with significant correlation (Rs=0.32, p=0.004). A significant increase in the concentration of active proteases was found between the first and last meconium portions (p<0.05). The proteases in the last meconium portions had a higher reaction velocity and affinity for the substrate than the proteases in the first meconium portions. The active protease:alpha-1 antitrypsin ratio was <0.5 in all first meconium portions, but was higher in the last meconium portions. CONCLUSIONS Strong correlation between the concentrations of active proteases and alpha-1 antitrypsin in meconium may indicate their mutual interaction in the intrauterine environment. Alpha-1 antitrypsin maintains the protease/antiprotease balance during fetal development.
Collapse
Affiliation(s)
- Ewa Skarżyńska
- Department of Biochemistry and Clinical Chemistry, Medical University of Warsaw, Warsaw, Poland.
| | - Bartosz Kiersztyn
- Microbial Ecology and Environmental Biotechnology Department, Institute of Botany, Faculty of Biology, University of Warsaw, Biological and Chemical Research Centre, Warsaw, Poland
| | - Paulina Wilczyńska
- Department of Biochemistry and Clinical Chemistry, Medical University of Warsaw, Warsaw, Poland
| | - Artur Jakimiuk
- Institute of Mother and Child Reproductive Health Department, Warsaw, Poland
| | - Barbara Lisowska-Myjak
- Department of Biochemistry and Clinical Chemistry, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
43
|
Ley D, Desseyn JL, Mischke M, Knol J, Turck D, Gottrand F. Early-life origin of intestinal inflammatory disorders. Nutr Rev 2017; 75:175-187. [PMID: 28340001 DOI: 10.1093/nutrit/nuw061] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A growing body of evidence supports the concept of perinatal programming through which the perinatal environment affects the development of the fetus and infant, thereby modifying the risk profile for disease later in life. Increasing attention is focusing on the role of the early environment in the development of chronic intestinal disorders. Epidemiological studies have highlighted the link between perinatal factors, such as breastfeeding, cesarean delivery, and antibiotic use, and an increased risk for inflammatory bowel disease and/or celiac disease. These links are consistent with the concept of perinatal programming of intestinal inflammatory disorders. Animal models have shown that the early-life environment affects the development of the gastrointestinal tract, but further experimental studies are needed to confirm the long-term effects of the perinatal environment on susceptibility to chronic intestinal disorders later in life. Changes in the development and composition of the intestinal microbiota as well as epigenetic changes are emerging as key mechanisms through which the perinatal environment determines susceptibility to intestinal inflammatory disorders.
Collapse
Affiliation(s)
- Delphine Ley
- Lille Inflammation Research International Center (LIRIC) - UMR 995 Inserm, University Lille, CHU Lille, Lille, France
| | - Jean-Luc Desseyn
- Lille Inflammation Research International Center (LIRIC) - UMR 995 Inserm, University Lille, CHU Lille, Lille, France
| | | | - Jan Knol
- Nutricia Research, Utrecht, The Netherlands.,Laboratory of Microbiology, Wageningen University, The Netherlands
| | - Dominique Turck
- Lille Inflammation Research International Center (LIRIC) - UMR 995 Inserm, University Lille, CHU Lille, Lille, France
| | - Frédéric Gottrand
- Lille Inflammation Research International Center (LIRIC) - UMR 995 Inserm, University Lille, CHU Lille, Lille, France
| |
Collapse
|
44
|
The extracellular matrix of the gastrointestinal tract: a regenerative medicine platform. Nat Rev Gastroenterol Hepatol 2017; 14:540-552. [PMID: 28698662 DOI: 10.1038/nrgastro.2017.76] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The synthesis and secretion of components that constitute the extracellular matrix (ECM) by resident cell types occur at the earliest stages of embryonic development, and continue throughout life in both healthy and diseased physiological states. The ECM consists of a complex mixture of insoluble and soluble functional components that are arranged in a tissue-specific 3D ultrastructure, and it regulates numerous biological processes, including angiogenesis, innervation and stem cell differentiation. Owing to its composition and influence on embryonic development, as well as cellular and organ homeostasis, the ECM is an ideal therapeutic substrate for the repair of damaged or diseased tissues. Biologic scaffold materials that are composed of ECM have been used in various surgical and tissue-engineering applications. The gastrointestinal (GI) tract presents distinct challenges, such as diverse pH conditions and the requirement for motility and nutrient absorption. Despite these challenges, the use of homologous and heterologous ECM bioscaffolds for the focal or segmental reconstruction and regeneration of GI tissue has shown promise in early preclinical and clinical studies. This Review discusses the importance of tissue-specific ECM bioscaffolds and highlights the major advances that have been made in regenerative medicine strategies for the reconstruction of functional GI tissues.
Collapse
|
45
|
Yao Y, Voillet V, Jegou M, SanCristobal M, Dou S, Romé V, Lippi Y, Billon Y, Père MC, Boudry G, Gress L, Iannucelli N, Mormède P, Quesnel H, Canario L, Liaubet L, Le Huërou-Luron I. Comparing the intestinal transcriptome of Meishan and Large White piglets during late fetal development reveals genes involved in glucose and lipid metabolism and immunity as valuable clues of intestinal maturity. BMC Genomics 2017; 18:647. [PMID: 28830381 PMCID: PMC5568345 DOI: 10.1186/s12864-017-4001-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Accepted: 08/01/2017] [Indexed: 11/21/2022] Open
Abstract
Background Maturity of intestinal functions is critical for neonatal health and survival, but comprehensive description of mechanisms underlying intestinal maturation that occur during late gestation still remain poorly characterized. The aim of this study was to investigate biological processes specifically involved in intestinal maturation by comparing fetal jejunal transcriptomes of two representative porcine breeds (Large White, LW; Meishan, MS) with contrasting neonatal vitality and maturity, at two key time points during late gestation (gestational days 90 and 110). MS and LW sows inseminated with mixed semen (from breed LW and MS) gave birth to both purebred and crossbred fetuses. We hypothesized that part of the differences in neonatal maturity between the two breeds results from distinct developmental profiles of the fetal intestine during late gestation. Reciprocal crossed fetuses were used to analyze the effect of parental genome. Transcriptomic data and 23 phenotypic variables known to be associated with maturity trait were integrated using multivariate analysis with expectation of identifying relevant genes-phenotypic variable relationships involved in intestinal maturation. Results A moderate maternal genotype effect, but no paternal genotype effect, was observed on offspring intestinal maturation. Four hundred and four differentially expressed probes, corresponding to 274 differentially expressed genes (DEGs), more specifically involved in the maturation process were further studied. In day 110-MS fetuses, Ingenuity® functional enrichment analysis revealed that 46% of DEGs were involved in glucose and lipid metabolism, cell proliferation, vasculogenesis and hormone synthesis compared to day 90-MS fetuses. Expression of genes involved in immune pathways including phagocytosis, inflammation and defense processes was changed in day 110-LW compared to day 90-LW fetuses (corresponding to 13% of DEGs). The transcriptional regulator PPARGC1A was predicted to be an important regulator of differentially expressed genes in MS. Fetal blood fructose level, intestinal lactase activity and villous height were the best predicted phenotypic variables with probes mostly involved in lipid metabolism, carbohydrate metabolism and cellular movement biological pathways. Conclusions Collectively, our findings indicate that the neonatal maturity of pig intestine may rely on functional development of glucose and lipid metabolisms, immune phagocyte differentiation and inflammatory pathways. This process may partially be governed by PPARGC1A. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-4001-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ying Yao
- Nutrition Metabolisms and Cancer (NuMeCan), INRA, INSERM, Université de Rennes 1, UBL, Rennes, Saint-Gilles, France.,Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Valentin Voillet
- GenPhySE, Université de Toulouse, INRA, INPT, ENVT, Castanet Tolosan, France
| | - Maeva Jegou
- Nutrition Metabolisms and Cancer (NuMeCan), INRA, INSERM, Université de Rennes 1, UBL, Rennes, Saint-Gilles, France
| | - Magali SanCristobal
- GenPhySE, Université de Toulouse, INRA, INPT, ENVT, Castanet Tolosan, France
| | - Samir Dou
- PEGASE, INRA, Agrocampus Ouest, Saint-Gilles, France
| | - Véronique Romé
- Nutrition Metabolisms and Cancer (NuMeCan), INRA, INSERM, Université de Rennes 1, UBL, Rennes, Saint-Gilles, France
| | - Yannick Lippi
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRA, ENVT, INP-Purpan, UPS, Toulouse, France
| | | | | | - Gaëlle Boudry
- Nutrition Metabolisms and Cancer (NuMeCan), INRA, INSERM, Université de Rennes 1, UBL, Rennes, Saint-Gilles, France
| | - Laure Gress
- GenPhySE, Université de Toulouse, INRA, INPT, ENVT, Castanet Tolosan, France
| | - Nathalie Iannucelli
- GenPhySE, Université de Toulouse, INRA, INPT, ENVT, Castanet Tolosan, France
| | - Pierre Mormède
- GenPhySE, Université de Toulouse, INRA, INPT, ENVT, Castanet Tolosan, France
| | | | - Laurianne Canario
- GenPhySE, Université de Toulouse, INRA, INPT, ENVT, Castanet Tolosan, France
| | - Laurence Liaubet
- GenPhySE, Université de Toulouse, INRA, INPT, ENVT, Castanet Tolosan, France
| | - Isabelle Le Huërou-Luron
- Nutrition Metabolisms and Cancer (NuMeCan), INRA, INSERM, Université de Rennes 1, UBL, Rennes, Saint-Gilles, France.
| |
Collapse
|
46
|
O'Hara K. Pharmacokinetic changes with growth and development between birth and adulthood. JOURNAL OF PHARMACY PRACTICE AND RESEARCH 2017. [DOI: 10.1002/jppr.1373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Kate O'Hara
- Pharmacy Department; Canberra Hospital and Health Service; Canberra Australia
| |
Collapse
|
47
|
Bile acids and colon cancer: Is FXR the solution of the conundrum? Mol Aspects Med 2017; 56:66-74. [DOI: 10.1016/j.mam.2017.04.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 03/20/2017] [Accepted: 04/07/2017] [Indexed: 02/07/2023]
|
48
|
Moles L, Gómez M, Jiménez E, Bustos G, de Andrés J, Melgar A, Escuder D, Fernández L, Del Campo R, Rodríguez JM. Bacterial Diversity of the Gastric Content of Preterm Infants during Their First Month of Life at the Hospital. Front Nutr 2017; 4:12. [PMID: 28459051 PMCID: PMC5394887 DOI: 10.3389/fnut.2017.00012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 03/29/2017] [Indexed: 12/26/2022] Open
Abstract
Studies focused on the stomach microbiota are relatively scarce, and most of them are focused on the adult population. The aim of this work is to describe the bacterial communities inhabiting the gastric content (GC) of preterm neonates. For that purpose, GC samples were collected weekly from a total of 13 preterm neonates during their first month of life within their hospital stay. Samples were analyzed by using both culture-dependent and -independent techniques. The former allowed the isolation of bacteria belonging mainly to the genera Enterococcus, Staphylococcus, Streptococcus, Serratia, Klebsiella, and Escherichia. The cultured dominant species in the GC samples during all the hospitalization period were Enterococcus faecalis and Staphylococcus epidermidis. Multilocus sequence typing (MLST) analysis revealed the presence of high-risk clonal complexes associated with the hospital environment, which may colonize enteral feeding tubes. Similarly, the 16S rRNA sequencing showed that Streptococcus, Staphylococcus, Lactobacillus, Enterococcus, Corynebacterium, and Propionibacterium were the dominant genera present at 75% of the gastric samples. However, the genera Serratia, Klebsiella, and Streptococcus were the most abundant. Own mother's milk (OMM) and donor milk (DM) were collected after their pass through the external feeding tubes to assess their bacterial content. OMM and DM had a similar bacterial pattern to GC. Based on these data, the GC of preterm neonates is dominated by Proteobacteria and Firmicutes and harbors high-risk bacterial clones, which may colonize enteral feeding tubes, and therefore the feeds that pass through them.
Collapse
Affiliation(s)
- Laura Moles
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Marta Gómez
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Esther Jiménez
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain.,ProbiSearch, S.L., Tres Cantos, Madrid, Spain
| | - Gerardo Bustos
- Servicio de Neonatología, Hospital Universitario 12 de Octubre, Madrid, Spain.,Red de Salud Materno-Infantil y del Desarrollo (SAMID), Barakaldo, Spain
| | - Javier de Andrés
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Ana Melgar
- Servicio de Neonatología, Hospital Universitario 12 de Octubre, Madrid, Spain.,Red de Salud Materno-Infantil y del Desarrollo (SAMID), Barakaldo, Spain
| | - Diana Escuder
- Servicio de Neonatología, Hospital Universitario 12 de Octubre, Madrid, Spain.,Red de Salud Materno-Infantil y del Desarrollo (SAMID), Barakaldo, Spain
| | - Leónides Fernández
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain.,ProbiSearch, S.L., Tres Cantos, Madrid, Spain
| | - Rosa Del Campo
- Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS), Madrid, Spain.,Servicio de Microbiología, Hospital Universitario Ramón y Cajal, Madrid, Spain.,Spanish Network for Research in Infectious Diseases (REIPI), Seville, Spain.,Instituto de Salud Carlos III, Madrid, Spain
| | - Juan Miguel Rodríguez
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain.,ProbiSearch, S.L., Tres Cantos, Madrid, Spain
| |
Collapse
|
49
|
Lima-Ojeda JM, Rupprecht R, Baghai TC. "I Am I and My Bacterial Circumstances": Linking Gut Microbiome, Neurodevelopment, and Depression. Front Psychiatry 2017; 8:153. [PMID: 28878696 PMCID: PMC5572414 DOI: 10.3389/fpsyt.2017.00153] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 08/02/2017] [Indexed: 01/01/2023] Open
Abstract
Recently, there has been renewed interest in the role played by microbiome in both human health and human disease. A correct equilibrium between the human host and their microorganisms is important for an appropriate physiological function. Extensive research has shown that microbes that inhabit the gastrointestinal tract-or gut microbiota-are involved not only in both nutritive and digestive activities but also in immunological processes. Moreover, the gut microbiome influences both central nervous system and energy homeostasis. An altered gut microbiome has been associated with the pathophysiology of different diseases, including neuropsychiatric disorders. Apparently, both environmental-diet, exposition to antibiotics, and infections-and host-genetic factors have a strong influence on gut microbiome, modulating the risk for neuropsychiatric illness. Also, early life disruption of the microbiome-gut-brain (MGB) axis has been associated with an increased risk of developing depression later in life, suggesting a link between gut microbiome, neurodevelopment, and depression. This review aims to contribute to this growing area of research by exploring the role played by the gut microbiome in neurodevelopment and in the etiology of the depressive syndrome, including nutritional, immunological, and energy homeostasis approaches.
Collapse
Affiliation(s)
- Juan M Lima-Ojeda
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Thomas C Baghai
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| |
Collapse
|
50
|
An N, Yang X, Zhang Y, Shi X, Yu X, Cheng S, Zhang K, Wang G. Cell cycle related genes up-regulated in human colorectal development predict the overall survival of late-stage colorectal cancer patients. MOLECULAR BIOSYSTEMS 2016; 12:541-52. [PMID: 26672738 DOI: 10.1039/c5mb00761e] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
A tumor can be perceived as a special "organ" that undergoes aberrant and poorly regulated organogenesis. Embryonic development and carcinogenesis share striking similarities in their cellular behavior and underlying molecular mechanisms. This intimate association makes embryonic development a viable reference model for studying cancer thereby circumventing the potentially misleading complexity of tumor heterogeneity. Therefore, on the basis of global expression profile, the genes simultaneously activated (up-regulated in terms of expression profile) or suppressed (down-regulated) in both the embryonic development and cancer stage, probably contain profound information on the molecular mechanism of cancer. In this study, the Affymetrix expression profile of 1593 colorectal cancer samples was downloaded from Gene Expression Omnibus. The 1396 differentially expressed probes were robustly obtained using 660 colorectal normal and cancer samples, the expression pattern of which was analyzed using our human colorectal developmental data. All of these 1396 probes were classified into 27 distinct patterns based on their expression patterns during the developmental process. By means of gene set enrichment analysis, we collected 393 V probes simultaneously up-regulated in both development and carcinogenesis and 207 A probes down-regulated in both. Functional enrichment analysis indicated that the V probes were significantly related to cell cycle regulation. Notably, 28 cell-cycle related probes within the V probe group were found to be significantly associated with an overall survival of Stage III/IV patients (GSE17536 cross validation, n = 96, p = 5.70 × 10(-3); GSE29621, n = 36, p = 1.70 × 10(-3); GSE39084, n = 38, p = 0.05; GSE39582, n = 264, p = 0.047; GSE17537, n = 36, p = 5.90 × 10(-3)).
Collapse
Affiliation(s)
- Ning An
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, Peking Union Medical College & Cancer Institute (Hospital), Chinese Academy of Medical Sciences, Beijing, China.
| | - Xue Yang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, Peking Union Medical College & Cancer Institute (Hospital), Chinese Academy of Medical Sciences, Beijing, China.
| | - Yueming Zhang
- Department of Endoscopy, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China.
| | - Xiaoyu Shi
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, Peking Union Medical College & Cancer Institute (Hospital), Chinese Academy of Medical Sciences, Beijing, China.
| | - Xuexin Yu
- College of Bioinformatics Science and Technology, Harbin Medical University, China
| | - Shujun Cheng
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, Peking Union Medical College & Cancer Institute (Hospital), Chinese Academy of Medical Sciences, Beijing, China.
| | - Kaitai Zhang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, Peking Union Medical College & Cancer Institute (Hospital), Chinese Academy of Medical Sciences, Beijing, China.
| | - Guiqi Wang
- Department of Endoscopy, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|