1
|
Nucci AM, Bashaw H, Kirpich A, Rudolph J. Retrospective review of growth in pediatric intestinal failure after weaning from parenteral nutrition. Nutr Clin Pract 2025; 40:176-187. [PMID: 39263924 PMCID: PMC11713205 DOI: 10.1002/ncp.11209] [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: 12/26/2023] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND Growth outcomes in children with intestinal failure (IF) after weaning from parenteral nutrition (PN) may be modified by primary diagnosis and interventions aimed at achieving enteral tolerance. We evaluated growth after weaning by diagnosis and intestinal transplant status and during treatment with the glucagon-like peptide-2 analog teduglutide. METHODS A two-center retrospective review was conducted on children diagnosed with IF at age <12 months. The z scores for weight and length/height were examined up to 5 years after PN weaning and in children who received teduglutide for >6 months. Data were reported as median and interquartile range (IQR). RESULTS A total of 362 children (58% male and 72% White) were reviewed; 41% (n = 150) weaned from PN at age 1.5 years (IQR = 0.96-3). Weight and length/height data were available for 144 children; 46 received an intestinal transplant. Median weight and length/height z scores at weaning were -1.15 (IQR = -2.09 to -0.39) and -1.89 (IQR = -2.9 to -1.02), respectively. In those not transplanted, z scores remained stable (± 0.5 change). Children with small bowel atresia experienced accelerated linear growth (> +0.5 change) beginning in year 3. Most children transplanted experienced growth acceleration beginning in year 2. Fourteen children received teduglutide (median = 840 [IQR = 425-1530] days), and growth remained stable throughout treatment. Five were weaned from PN within 1 year. CONCLUSION We observed stable growth with limited catch-up after PN weaning, with minimal variation by diagnosis, and during teduglutide therapy. Children who received an intestinal transplant experienced acceleration in weight and linear growth after weaning.
Collapse
Affiliation(s)
- Anita M. Nucci
- Department of NutritionGeorgia State UniversityAtlantaGeorgiaUSA
| | - Hillary Bashaw
- Division of Gastroenterology, Hepatology, and NutritionEmory University School of MedicineAtlantaGeorgiaUSA
| | - Alexander Kirpich
- Department of Population Health Sciences, School of Public HealthGeorgia State UniversityAtlantaGeorgiaUSA
| | - Jeffrey Rudolph
- Division of Pediatric Gastroenterology, Hepatology, and NutritionUPMC Children's Hospital of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
2
|
Wang SN, Fu YJ, Lu XL, Miao SJ, Zhang P, Wang L, Huang Y, Wang YH. Three patients with new mutations in the EPCAM variant gene for congenital tufting enteropathy and a mutation review in China: a case report. Transl Pediatr 2024; 13:1486-1495. [PMID: 39263299 PMCID: PMC11384436 DOI: 10.21037/tp-24-97] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 08/01/2024] [Indexed: 09/13/2024] Open
Abstract
Background Congenital tufting enteropathy (CTE) is a rare cause of intractable congenital diarrhea in children, always resulting in parenteral nutrition (PN) dependency. We aimed to report novel mutations in Chinese patients and to illustrate the clinical, histopathological, and molecular features of CTE in China. Case Description We report three cases of CTE diagnosed with whole-exome sequencing (WES) and MOC31 [a monoclonal antibody of epithelial cell adhesion molecule (EPCAM)] immunohistochemistry. The main manifestations in the three patients were watery diarrhea and growth retardation. Upper endoscopy in three patients revealed villous atrophy of the duodenal mucosa. Histological examination revealed villus abnormalities and two patients with focal tufting. All of the three patients revealed a complete absence of EPCAM expression through MOC31 immunohistochemistry. Five novel mutations, including c.319delG, c.505_507delGAG, c.491+1G>C, c.60del (p.F20Lfs*17), and c.353G>A, in EPCAM were identified through molecular analysis. In our review, there were 18 different mutations in 11 patients from nine studies, with 12 mutations reported only once. In China, 73% of the patients were compound heterozygotes, and most of the pathogenic variants were in exon 3. All patients presented with congenital diarrhea and needed PN because of growth retardation, even when diarrhea was improved. Of the 11 patients, 3 (27%) died. Conclusions CTE is rare and fatal, and lacks characteristic changes during endoscopy. Patients with CTE require early diagnosis via histological examination and genetic detection to improve survival.
Collapse
Affiliation(s)
- Sheng-Nan Wang
- Department of Gastroenterology, Children's Hospital of Fudan University, Shanghai, China
| | - Yu-Juan Fu
- Department of Pathology, Zhejiang University School of Medicine Sir Run Run Shaw Hospital (Shao Yifu Hospital), Hangzhou, China
| | - Xiao-Lan Lu
- Department of Gastroenterology, Children's Hospital of Fudan University, Shanghai, China
| | - Shi-Jian Miao
- Department of Gastroenterology, Children's Hospital of Fudan University, Shanghai, China
| | - Ping Zhang
- Center for Molecular Medicine, Pediatrics Research Institute, Children's Hospital of Fudan University, Shanghai, China
| | - Lin Wang
- Department of Gastroenterology, Children's Hospital of Fudan University, Shanghai, China
| | - Ying Huang
- Department of Gastroenterology, Children's Hospital of Fudan University, Shanghai, China
| | - Yu-Huan Wang
- Department of Gastroenterology, Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
3
|
Sapmaz T, Kale E, Pence ME, Sevgin K, Tekayev M, Topkaraoglu S, Basol G, Yilmaz MB, Sapmaz E, Irkorucu O. Treatment strategies with vitamin E and C in autologous intraperitoneal ovarian transplantation and its impact on ovarian surface epithelium and follicle reserve. Biochem Biophys Res Commun 2023; 677:81-87. [PMID: 37556954 DOI: 10.1016/j.bbrc.2023.08.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/23/2023] [Accepted: 08/01/2023] [Indexed: 08/11/2023]
Abstract
The objective of this study was to assess the impact of Vitamin E (Vit E) and Vitamin C (Vit C) on markers of the oxidant-antioxidant system, ovarian follicle reserves, and the surface epithelium in autologous intraperitoneal ovarian transplantation conducted in rats. The study aimed to investigate how these antioxidants influence various aspects related to transplantation outcomes, including oxidative stress markers, the preservation of follicle reserves, and the condition of the surface epithelium. A total of 20 adult female Wistar Albino rats were included in the study and randomly assigned to four different groups. Group 1, consisting of 5 rats, served as the control group and underwent a surgical procedure where their abdomens were opened and closed without any further intervention. Group 2, also consisting of 5 rats, underwent ovarian transplantation. In Group 3, comprising 5 rats, an intraperitoneal (IP) administration of 20 mg/kg body weight (b.w.) of Vitamin E (Vit E) was given 15 min prior to ovarian transplantation. Lastly, in Group 4, which included 5 rats, an IP administration of 50 mg/kg body weight (b.w.) of Vitamin C (Vit C) was given 15 min before ovarian transplantation. Vaginal cytology was performed in order to monitor the estrus phase in the rats. Biochemically, tissue and serum malondialdehyde (MDA) levels and erythrocyte superoxide dismutase (SOD) levels were measured. Histopathologically, the number of dysplastic changes in the ovarian surface epithelium and primordial, primary, secondary, Graaffian, and atretic follicles were examined. Dysplastic changes in the surface epithelium of Group 2 were found to be significantly higher than in Group 1 and 4 (p < 0.02). In Group 2, the ovarian follicle reserves (primordial, primary, secondary, and Graaffian follicles) were significantly lower than in other groups (p < 0.02). In addition, a significant decrease in SOD levels was found in Group 2 compared to other groups (p < 0.02). The study showed that Vit E and Vit C in autologous intraperitoneal ovarian transplantation preserved the ovarian follicle reserve. Vit C was found to be more effective than Vit E.
Collapse
Affiliation(s)
- Tansel Sapmaz
- University of Health Sciences, Hamidiye Faculty of Medicine, Department of Histology and Embryology, Istanbul, Turkey.
| | - Ebru Kale
- University of Health Sciences, Hamidiye Faculty of Medicine, Department of Medical Biochemistry, Istanbul, Turkey
| | - Mahmud Esad Pence
- University of Health Sciences, Istanbul Umraniye Training and Research Hospital, Department of Emergency Medicine, Istanbul, Turkey
| | - Kubra Sevgin
- University of Health Sciences, Hamidiye Faculty of Medicine, Department of Histology and Embryology, Istanbul, Turkey
| | - Muhammetnur Tekayev
- University of Health Sciences, Hamidiye Faculty of Medicine, Department of Histology and Embryology, Istanbul, Turkey
| | - Sude Topkaraoglu
- University of Health Sciences, Hamidiye Faculty of Medicine, Department of Histology and Embryology, Istanbul, Turkey
| | - Gulfem Basol
- University of Health Sciences, Lutfi Kirdar City Hospital, Department of Obstetrics and Gynecology, Istanbul, Turkey
| | - Muserref Banu Yilmaz
- University of Health Sciences, Zeynep Kamil Woman's and Children Research and Training Hospital, Istanbul, Turkey
| | - Ekrem Sapmaz
- University of Health Sciences, Adana City Hospital, Department of Obstetrics and Gynecology, Adana, Turkey
| | - Oktay Irkorucu
- University of Sharjah, College of Medicine, Department of Clinical Sciences, Sharjah, United Arab Emirates
| |
Collapse
|
4
|
Babcock SJ, Flores-Marin D, Thiagarajah JR. The genetics of monogenic intestinal epithelial disorders. Hum Genet 2023; 142:613-654. [PMID: 36422736 PMCID: PMC10182130 DOI: 10.1007/s00439-022-02501-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 10/23/2022] [Indexed: 11/27/2022]
Abstract
Monogenic intestinal epithelial disorders, also known as congenital diarrheas and enteropathies (CoDEs), are a group of rare diseases that result from mutations in genes that primarily affect intestinal epithelial cell function. Patients with CoDE disorders generally present with infantile-onset diarrhea and poor growth, and often require intensive fluid and nutritional management. CoDE disorders can be classified into several categories that relate to broad areas of epithelial function, structure, and development. The advent of accessible and low-cost genetic sequencing has accelerated discovery in the field with over 45 different genes now associated with CoDE disorders. Despite this increasing knowledge in the causal genetics of disease, the underlying cellular pathophysiology remains incompletely understood for many disorders. Consequently, clinical management options for CoDE disorders are currently limited and there is an urgent need for new and disorder-specific therapies. In this review, we provide a general overview of CoDE disorders, including a historical perspective of the field and relationship to other monogenic disorders of the intestine. We describe the genetics, clinical presentation, and known pathophysiology for specific disorders. Lastly, we describe the major challenges relating to CoDE disorders, briefly outline key areas that need further study, and provide a perspective on the future genetic and therapeutic landscape.
Collapse
Affiliation(s)
- Stephen J Babcock
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Enders Rm 605, 300 Longwood Ave, Boston, MA, 02115, USA
| | - David Flores-Marin
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Enders Rm 605, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Jay R Thiagarajah
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Harvard Medical School, Enders Rm 605, 300 Longwood Ave, Boston, MA, 02115, USA.
| |
Collapse
|
5
|
Alkhalifa S, Darwish A, Awadh M, Alkhalifa SM, Darwish A. Congenital Tufting Enteropathy, a Rare Cause of Diarrhea and Malnourishment in Arab Child with Genetic and Histopathology Investigations. Case Rep Pediatr 2023; 2023:6301065. [PMID: 36743443 PMCID: PMC9891835 DOI: 10.1155/2023/6301065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 10/03/2022] [Accepted: 01/13/2023] [Indexed: 01/27/2023] Open
Abstract
Congenital tufting enteropathy (CTE), also known as intestinal epithelial dysplasia (IED), is a rare autosomal recessive disorder due to EPCAM gene mutation. It is a rare congenital enteropathy that presents in early infancy as an intractable diarrhea that is independent of breast formula feeding that requires life-long total parental nutrition (TPN) to acquire adequate calories and fluid intake or small bowel transplantation in severe cases. Here, we report a case of intestinal failure due to congenital tufting enteropathy in a 3-year-old girl who presented with loose stools and failure to thrive. This study aims to review the literature about CTE and discuss the clinicopathological aspects and to be able to distinguish it from other causes of congenital diarrheal disorders (CDDs).
Collapse
|
6
|
Güvenoğlu M, Şimşek-Kiper PÖ, Koşukcu C, Taskiran EZ, Saltık-Temizel İN, Gucer S, Utine E, Boduroğlu K. Homozygous Missense Epithelial Cell Adhesion Molecule Variant in a Patient with Congenital Tufting Enteropathy and Literature Review. Pediatr Gastroenterol Hepatol Nutr 2022; 25:441-452. [PMID: 36451688 PMCID: PMC9679307 DOI: 10.5223/pghn.2022.25.6.441] [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: 04/20/2022] [Revised: 06/28/2022] [Accepted: 09/21/2022] [Indexed: 11/22/2022] Open
Abstract
Congenital diarrheal disorders (CDDs) with genetic etiology are uncommon hereditary intestinal diseases characterized by chronic, life-threatening, intractable watery diarrhea that starts in infancy. CDDs can be mechanistically divided into osmotic and secretory diarrhea. Congenital tufting enteropathy (CTE), also known as intestinal epithelial dysplasia, is a type of secretory CDD. CTE is a rare autosomal recessive enteropathy that presents with intractable neonatal-onset diarrhea, intestinal failure, severe malnutrition, and parenteral nutrition dependence. Villous atrophy of the intestinal epithelium, crypt hyperplasia, and irregularity of surface enterocytes are the specific pathological findings of CTE. The small intestine and occasionally the colonic mucosa include focal epithelial tufts. In 2008, Sivagnanam et al. discovered that mutations in the epithelial cell adhesion molecule (EpCAM, MIM# 185535) were the genetic cause of CTE (MIM# 613217). More than a hundred mutations have been reported to date. Furthermore, mutations in the serine peptidase inhibitor Kunitz type 2 (SPINT2, MIM# 605124) have been linked to syndromic CTE. In this study, we report the case of a 17-month-old male infant with congenital diarrhea. Despite extensive etiological workup, no etiology could be established before admission to our center. The patient died 15 hours after being admitted to our center in a metabolically decompensated state, probably due to a delay in admission and diagnosis. Molecular autopsy with exome sequencing revealed a previously reported homozygous missense variant, c.757G>A, in EpCAM, which was confirmed by histopathological examination.
Collapse
Affiliation(s)
- Merve Güvenoğlu
- Department of Pediatric Genetics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | | | - Can Koşukcu
- Department of Bioinformatics, Institute of Health Sciences, Hacettepe University, Ankara, Turkey
| | - Ekim Z Taskiran
- Department of Medical Genetics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - İnci Nur Saltık-Temizel
- Department of Pediatric Gastroenterology, Hepatology, and Nutrition, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Safak Gucer
- Division of Pediatric Pathology, Department of Pediatrics, Hacettepe University, Ankara, Turkey
| | - Eda Utine
- Department of Pediatric Genetics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Koray Boduroğlu
- Department of Pediatric Genetics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
7
|
Gonzalez-Hakspiel LC, Wilches-Cuadros MA, Nausa-Suárez PA, Fernández F, Patiño-Ascencio P, Manrique-Guerrero A, Díaz-Díaz DD, Castro-Rojas CR. Severe congenital diarrhea secondary to tufting enteropathy. Case report. CASE REPORTS 2022. [DOI: 10.15446/cr.v8n1.90883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Introduction: Tufting enteropathy is a rare cause of congenital diarrhea in neonates. It is characterized by the abnormal distribution of epithelial adhesion molecules, which causes enterocytes to shed into the lumen, forming the characteristic tufts.
Case presentation: A 15-day-old female neonate was taken by her parents to the emergency department of a tertiary care hospital due to diarrheal stools she had been experiencing since birth. The patient presented with dehydration, abnormal weight loss, metabolic acidosis, and acute kidney failure. She received treatment with alizapride, loperamide, zinc sulfate, and probiotics, but after 75 days of treatment she was still symptomatic. An upper tract endoscopy and colonoscopy were performed, finding flattening of the villi and lymphoid cells in the lamina propria. However, the symptoms persisted, and she died at the age of ten months. A post-mortem exome sequencing reported tufting enteropathy.
Conclusions. When congenital diarrhea is present, tufting enteropathy should be considered. An early molecular study would allow to evaluate the possibility of performing an intestinal transplant or modifying the treatment to meet the patient’s palliative care needs.
Collapse
|
8
|
Szabo R, Ward JM, Artunc F, Bugge TH. EPCAM and TROP2 share role in claudin stabilization and development of intestinal and extraintestinal epithelia in mice. Biol Open 2022; 11:275770. [PMID: 35730316 PMCID: PMC9294608 DOI: 10.1242/bio.059403] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/10/2022] [Indexed: 11/20/2022] Open
Abstract
EPCAM (Epithelial Cell Adhesion Molecule) is a transmembrane glycoprotein expressed on the surface of most epithelial and epithelium-derived tumor cells and reported to regulate stability of epithelial tight junction proteins, claudins. Despite its widespread expression, loss of EPCAM function has so far only been reported to prominently affect intestinal development, resulting in severe early onset enteropathy associated with impaired growth and decreased survival in both humans and mice. In this study, we show that the critical role of EPCAM is not limited to intestinal tissues and that it shares its essential function with its only known homolog, TROP2 (Trophoblast cell surface antigen 2). EPCAM-deficient mice show significant growth retardation and die within four weeks after birth. In addition to changes in small and large intestines, loss of EPCAM results in hyperkeratosis in skin and forestomach, hair follicle atrophy leading to alopecia, nephron hypoplasia in kidney, proteinuria, and altered production of digestive enzymes by pancreas. Expression of TROP2 partially, but not completely, overlaps with EPCAM in a number developing epithelia. Although loss of TROP2 had no gross impact on mouse development and survival, TROP2 deficiency generally compounded developmental defects observed in EPCAM-deficient mice, led to about 60% decrease in embryonic viability, and further shortened postnatal lifespan of born pups. Importantly, TROP2 was able to compensate for the loss of EPCAM in stabilizing claudin-7 expression and cell membrane localization in tissues that co-express both proteins. These findings identify overlapping functions of EPCAM and TROP2 as regulators of epithelial development in both intestinal and extraintestinal tissues.
Collapse
Affiliation(s)
- Roman Szabo
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | | | - Ferruh Artunc
- Department of Internal Medicine, Division of Endocrinology, Diabetology and Nephrology, University Hospital Tübingen, Tübingen, Germany.,Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University Tübingen, Germany.,German Center for Diabetes Research (DZD) at the University Tübingen, Germany
| | - Thomas H Bugge
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
9
|
Abstract
Advancements in donor management, organ preservation and operative techniques, as well as immunosuppressive therapies, have provided children with intestinal failure and its complications a chance not only for enteral autonomy but also long-term survival through intestinal transplantation (ITx). First described in the 1960's, experience has grown in managing these complex patients both pre- and post-transplant. The goals of this review are to provide a brief history of intestinal transplantation and intestinal rehabilitation in pediatric patients, followed by focused discussions of the indications for ITx, induction and maintenance immunosuppression therapies, common post-operative complications, and outcomes/quality of life post-transplant.
Collapse
|
10
|
Feakins R, Torres J, Borralho-Nunes P, Burisch J, Cúrdia Gonçalves T, De Ridder L, Driessen A, Lobatón T, Menchén L, Mookhoek A, Noor N, Svrcek M, Villanacci V, Zidar N, Tripathi M. ECCO Topical Review on Clinicopathological Spectrum and Differential Diagnosis of Inflammatory Bowel Disease. J Crohns Colitis 2022; 16:343-368. [PMID: 34346490 DOI: 10.1093/ecco-jcc/jjab141] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Many diseases can imitate inflammatory bowel disease [IBD] clinically and pathologically. This review outlines the differential diagnosis of IBD and discusses morphological pointers and ancillary techniques that assist with the distinction between IBD and its mimics. METHODS European Crohn's and Colitis Organisation [ECCO] Topical Reviews are the result of an expert consensus. For this review, ECCO announced an open call to its members and formed three working groups [WGs] to study clinical aspects, pathological considerations, and the value of ancillary techniques. All WGs performed a systematic literature search. RESULTS Each WG produced a draft text and drew up provisional Current Practice Position [CPP] statements that highlighted the most important conclusions. Discussions and a preliminary voting round took place, with subsequent revision of CPP statements and text and a further meeting to agree on final statements. CONCLUSIONS Clinicians and pathologists encounter a wide variety of mimics of IBD, including infection, drug-induced disease, vascular disorders, diverticular disease, diversion proctocolitis, radiation damage, and immune disorders. Reliable distinction requires a multidisciplinary approach.
Collapse
Affiliation(s)
- Roger Feakins
- Department of Cellular Pathology, Royal Free Hospital, London, and University College London, UK
| | - Joana Torres
- Department of Gastroenterology, Hospital Beatriz Ângelo, Loures, Portugal
| | - Paula Borralho-Nunes
- Department of Pathology, Hospital Cuf Descobertas, Lisboa and Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Johan Burisch
- Gastrounit, Medical Division, Hvidovre Hospital, University of Copenhagen, Denmark
| | - Tiago Cúrdia Gonçalves
- Department of Gastroenterology, Hospital da Senhora da Oliveira, Guimarães, Portugal.,School of Medicine, University of Minho, Braga/Guimarães, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Lissy De Ridder
- Department of Paediatric Gastroenterology, Erasmus MC Sophia Children's Hospital, University Medical Center Rotterdam, The Netherlands
| | - Ann Driessen
- Department of Pathology, University Hospital Antwerp, University Antwerp, Edegem, Belgium
| | - Triana Lobatón
- Department of Gastroenterology, Ghent University Hospital, Ghent, Belgium
| | - Luis Menchén
- Department of Digestive System Medicine, Hospital General Universitario-Insitituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.,Department of Medicine, Universidad Complutense, Madrid, Spain.,Centro de Investigación Biomédica En Red de Enfermedades Hepáticas y Digestivas [CIBEREHD], Madrid, Spain
| | - Aart Mookhoek
- Department of Pathology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Nurulamin Noor
- Department of Gastroenterology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Trust, Cambridge, UK
| | - Magali Svrcek
- Department of Pathology, Sorbonne Université, AP-HP, Saint-Antoine Hospital, Paris, France
| | - Vincenzo Villanacci
- Department of Histopathology, Spedali Civili and University of Brescia, Brescia, Italy
| | - Nina Zidar
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Monika Tripathi
- Department of Histopathology, Cambridge Biomedical Campus, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
11
|
Parente P, Mastracci L, Vanoli A, Fassan M, Pastore M, Bossa F, Francalanci P, Alaggio R, Graziano P, Grillo F. Pattern-based Histologic Approach in Very Early Onset IBD: Main Features and Differential Diagnosis. Adv Anat Pathol 2022; 29:71-80. [PMID: 34620740 DOI: 10.1097/pap.0000000000000323] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Very early onset inflammatory bowel disease (VEO-IBD) is a clinical umbrella classification composed of IBD-like diseases encompassing both classic IBD (Crohn's disease and ulcerative colitis) and monogenic disorder, both arising before 6 years of age. VEO-IBD patients present significant clinical differences from IBD occurring in older children and in adults, including more severe disease, often unresponsive to conventional IBD therapy and a greater proportion of cases featuring an underlying genetic alteration. Histologic findings of gastrointestinal biopsies can show an IBD-like pattern (both Crohn's disease-like and ulcerative colitis-like pattern), an apoptotic-like and enterocolitis-like pattern. Findings of specific morphologic alterations, such as villous blunting, apoptosis, dense eosinophilic infiltrates, lack of plasma cells and severe glandular atrophy, can suggest a monogenic disorder. Moreover, individuals with monogenic disorders may develop significant problems such as primary immunodeficiency, impacting treatment options. Finally, IBD histology in childhood can differ from that in older patients and adults. This complexity makes a differential diagnosis between IBD and other pediatric diseases involving the gastrointestinal tract difficult, especially considering that histologic features can be similar between different diseases. Without an appropriate diagnosis, the clinical course of VEO-IBD has greater potential for escalated treatment regimens involving extensive surgery and more intensive medical therapies rather than specific therapy directed toward the underlying defect. For these reasons, a pattern-based histologic approach correlated with clinical and laboratory findings with a multidisciplinary approach is fundamental to reach a correct diagnosis in an adequate clinical context.
Collapse
Affiliation(s)
| | - Luca Mastracci
- Unit of Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova
- Ospedale Policlinico San Martino, IRCCS, Genova
| | - Alessandro Vanoli
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia and Fondazione IRCCS San Matteo Hospital, Pavia
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padova
- Veneto Institute of Oncology (IOV-IRCCS), Padova
| | | | - Fabrizio Bossa
- Gastroenterology, Fondazione IRCCS Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia
| | - Paola Francalanci
- Pathology Unit, Children's Hospital Bambino Gesù, IRCCS, Roma, Italy
| | - Rita Alaggio
- Pathology Unit, Children's Hospital Bambino Gesù, IRCCS, Roma, Italy
| | | | - Federica Grillo
- Unit of Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova
- Ospedale Policlinico San Martino, IRCCS, Genova
| |
Collapse
|
12
|
Rossi C, Simoncelli G, Arpa G, Stracuzzi A, Parente P, Fassan M, Vanoli A, Villanacci V. Histopathology of intestinal villi in neonatal and paediatric age: main features with clinical correlation - Part I. Pathologica 2022; 114:12-21. [PMID: 34856604 PMCID: PMC9040547 DOI: 10.32074/1591-951x-337] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 07/30/2021] [Indexed: 11/30/2022] Open
Abstract
The neonatal and paediatric spectrum of small bowel disorders encompass a wide variety of conditions, ranging from food allergies to life-threatening surgical emergencies or life-long medical conditions and, as such, it comes with a whole set of diagnostic challenges for the non-paediatric pathologist. Histologic examination is a cornerstone of diagnosis in a large number of diseases and may still provide important diagnostic clues in the appropriate clinical context. In this review, divided in two sections, we aim to provide a comprehensive histopathological summary of paediatric small bowel alteration and their differential diagnoses with a reference to the main clinical aspects required for appropriate interpretation. Specifically, in this first part, we describe congenital and metabolic disorders, intestinal lymphangiectasia, immunodeficiencies, GVHD, and necrotising enterocolitis.
Collapse
Affiliation(s)
- Chiara Rossi
- Unit of Anatomic Pathology, Department of Molecular Medicine, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | | | - Giovanni Arpa
- Unit of Anatomic Pathology, Department of Molecular Medicine, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Alessandra Stracuzzi
- Pathological Anatomy Unit, Department of Diagnostic and Laboratory Medicine, IRCCS Bambino Gesù Children’s Hospital, Rome, Italy
| | - Paola Parente
- Pathology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology Unit, University of Padua, Padua, Italy
- Veneto Institute of Oncology, IOV-IRCCS, Padua, Italy
| | - Alessandro Vanoli
- Unit of Anatomic Pathology, Department of Molecular Medicine, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy
| | | |
Collapse
|
13
|
Montoya-Cerrillo D, Bernieh A, Saad AG. Critical diagnoses in paediatric gastrointestinal diseases. Pathology 2022; 54:195-206. [PMID: 35033374 DOI: 10.1016/j.pathol.2021.09.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/26/2021] [Accepted: 09/30/2021] [Indexed: 12/11/2022]
Abstract
Gastrointestinal biopsies represent an increasing proportion of the paediatric pathologist's workload, an increase fundamentally due to an expansion of the understanding of the basic clinical, molecular, genetic, and histopathological features of paediatric gastrointestinal disorders. The histological interpretation of endoscopically retrieved gastrointestinal biopsies in children requires a unique set of diagnostic expertise and detailed knowledge of various gastrointestinal disorders that have a predilection for the paediatric population. This article's major role is to highlight the unique problems inherent to paediatric gastrointestinal disorders that require immediate communication with the paediatric surgeon or the gastroenterologist. For this, we tried to cover the most important diseases that a paediatric pathologist might encounter in daily practice. Some of these diseases are relatively rare, such as microvillous inclusion disease and tufting enteropathy, but some are more common such as eosinophilic disorders and inflammatory bowel disease. Awareness of the histopathological features of these diseases, particularly those that are relatively uncommon, is crucial to spare the patient a lengthy and costly evaluation. We made a particular effort to abundantly reference this article should the reader wish to expand on the content of any section.
Collapse
Affiliation(s)
| | - Anas Bernieh
- Division of Pathology, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Ali G Saad
- Department of Pathology, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
14
|
Goulet O, Pigneur B, Charbit-Henrion F. Congenital enteropathies involving defects in enterocyte structure or differentiation. Best Pract Res Clin Gastroenterol 2022; 56-57:101784. [PMID: 35331396 DOI: 10.1016/j.bpg.2021.101784] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/15/2021] [Accepted: 12/22/2021] [Indexed: 01/31/2023]
Abstract
Congenital enteropathies (CE) are a group of rare inherited diseases with a typical onset early in life. They involve defects in enterocyte structure or differentiation. They can cause a severe condition of intestinal failure (IF). The diagnostic approach is based first on clinical presentation (consanguinity, prenatal expression, polyhydramnios, early neonatal onset, aspect of stools, persistence at bowel rest, associated extra-digestive manifestations….) and histo-pathological analyses. These rare intestinal diseases cause protracted diarrhea that might resolve, for a few, with a dietetic approach. However, protracted or permanent IF may require long term parenteral nutrition and, in limited cases, intestinal transplantation. With the progresses in both clinical nutrition and genetics, many of these CE are nowadays associated with recognized gene mutations. It improved our knowledge and the understanding in the patho-physiology of these diseases, thus, leading potentially to therapeutic perspectives. These review cover most of the early onset CE and excludes the immune related diarrhea.
Collapse
Affiliation(s)
- Olivier Goulet
- Division of Paediatric Gastroenterology Hepatology and Nutrition, University Paris-Centre, Hôpital Necker-Enfants Malades, 149, Rue de Sèvres, 75743, PARIS Cedex 15, France.
| | - Bénédicte Pigneur
- Division of Paediatric Gastroenterology Hepatology and Nutrition, University Paris-Centre, Hôpital Necker-Enfants Malades, 149, Rue de Sèvres, 75743, PARIS Cedex 15, France
| | - Fabienne Charbit-Henrion
- Department of Genetics, Hôpital Necker-Enfants Malades, 149, Rue de Sèvres, 75743, PARIS Cedex 15, France
| |
Collapse
|
15
|
Leventoğlu E, Büyükkaragöz B, Düztaş DT, Gürkan ÖE. Pseudo-Bartter syndrome and staghorn calculi in an infant with chronic diarrhea: Answers. Pediatr Nephrol 2021; 36:4099-4101. [PMID: 34414498 DOI: 10.1007/s00467-021-05217-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 10/20/2022]
Affiliation(s)
- Emre Leventoğlu
- Faculty of Medicine, Department of Pediatric Nephrology, Gazi University, Ankara, Turkey.
| | - Bahar Büyükkaragöz
- Faculty of Medicine, Department of Pediatric Nephrology, Gazi University, Ankara, Turkey
| | - Demet Teker Düztaş
- Faculty of Medicine, Department of Pediatric Gastroenterology, Gazi University, Ankara, Turkey
| | - Ödül Eğritaş Gürkan
- Faculty of Medicine, Department of Pediatric Gastroenterology, Gazi University, Ankara, Turkey
| |
Collapse
|
16
|
Goulet O, Breton A, Coste ME, Dubern B, Ecochard-Dugelay E, Guimber D, Loras-Duclaux I, Abi Nader E, Marinier E, Peretti N, Lambe C. Pediatric Home Parenteral Nutrition in France: A six years national survey. Clin Nutr 2021; 40:5278-5287. [PMID: 34534896 DOI: 10.1016/j.clnu.2021.08.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/31/2021] [Accepted: 08/05/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND AIMS Home Parenteral Nutrition (HPN) is the cornerstone management for children suffering from chronic intestinal failure (CIF). In France, HPN is organized from a network of 7 certified centers located in University Hospitals spread across the national territory. This study aims to review the data involving children on HPN over a 6-years period in France to outline the global and continuous improvement in care. PATIENTS AND METHODS This cross-sectional study included all children enrolled in any of the 7 French HPN certified centers from January 1st, 2014 to December 31st, 2019. Data was recorded from annual databases provided by each center regarding: age at inclusion, indication and duration of HPN, type of intravenous lipid emulsion (ILE), outcome [PN weaning off, transfer to adult center, death, intestinal transplantation (ITx)], rate of catheter-related bloodstream infections (CRSBIs) for 1000 days of HPN, Taurolidine lock procedure (TLP) use and prevalence of cholestasis defined as conjugated bilirubin ≥20 μmol/l. RESULTS The number of patients increased by 43.6% from 268 in 2014 to 385 in 2019. According to the year of follow up, the indications for HPN were short bowel syndrome (SBS) (42.3-46.6%), congenital enteropathies (CE) (18.5-22.8%), chronic intestinal pseudo-obstruction syndrome (CIPOS) (13.0-16.3%), long segment Hirschsprung's disease (LSHD) (9.7-13.3%), Crohn's disease (CD) (1.6-2.6%) and other non-primary digestive diseases (NPDD) such as immune deficiency, cancer or metabolic disease (4.0-9.2%). The median age at discharge on HPN decreased from 11.7 months in 2014 to 8.3 months in 2019 (p < .001). By December 31st, 2019, 44.8% of children had left the HPN program after a median duration ranging between 39.9 and 66.4 months. Among these patients, 192 (74.2%) were weaned off PN (94.7% SBS), 41 (15.8%) were transferred to adult centers for CIPOS (42%), SBS (31%) or CE (27%), 21 died (8.1%) - mostly in relation to cancer or immune deficiency - and 5 were transplanted (1.9%): 4 underwent combined liver-intestine transplantation for LSHD (n = 2), SBS, CE and one multivisceral Tx for CIPOS. The use of a composite fish-oil based ILE increased from 67.4% in 2014 to 88.3% in 2019 (p < 0.001). CRBSIs dropped from 1.04 CRSBIs per 1000 days HPN in 2014 to 0.61 in 2019 (p < 0.001) while meantime, the percentage of children receiving TLP increased from 29.4% to 63.0% (p < 0.001). The prevalence of cholestasis (conjugated bilirubin ≥ 20 μmol/l) was low and stable between 4.1 and 5.9% of children during the study period. CONCLUSION In France, the number of children enrolled in a HPN program continuously increased over a 6 years period. SBS is the leading cause of CIF requiring HPN. The rate of CRBSIs dropped dramatically as the use of TLP increased. Mortality rate was low and mainly in relation to the underlying disease (cancer, immune deficiency). Cholestasis and intestinal Tx remained very rare.
Collapse
Affiliation(s)
- Olivier Goulet
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Necker-Enfants Malades University of Paris-UFR Paris Descartes, Certified Center for Home Parenteral Nutrition, Reference Center for Rare Digestive Diseases in Children, Paris, France
| | - Anne Breton
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Purpan University Hospital, Certified Center for Home Parenteral Nutrition, Toulouse, France
| | - Marie-Edith Coste
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, La Timone University Hospital, Certified Center for Home Parenteral Nutrition, Marseille, France
| | - Béatrice Dubern
- Division of Pediatric Nutrition and Gastroenterology, Armand Trousseau University Hospital, Certified Center for Home Parenteral Nutrition, Sorbonne University, Paris, France
| | - Emmanuelle Ecochard-Dugelay
- Division of Pediatric Gastroenterology and Nutrition, Robert Debré University Hospital, Certified Center for Home Parenteral Nutrition, Reference Center for Rare Digestive Diseases in Children Paris, France
| | - Dominique Guimber
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Jeanne de Flandre University Hospital, Certified Center for Home Parenteral Nutrition, Reference Center for Rare Digestive Diseases in Children, Lille, France
| | - Irène Loras-Duclaux
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Pediatric University Hospital, Certified Center for Home Parenteral Nutrition, Reference Center for Rare Digestive Diseases in Children, Lyon, France
| | - Elie Abi Nader
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Necker-Enfants Malades University of Paris-UFR Paris Descartes, Certified Center for Home Parenteral Nutrition, Reference Center for Rare Digestive Diseases in Children, Paris, France
| | - Evelyne Marinier
- Division of Pediatric Gastroenterology and Nutrition, Robert Debré University Hospital, Certified Center for Home Parenteral Nutrition, Reference Center for Rare Digestive Diseases in Children Paris, France
| | - Noel Peretti
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Pediatric University Hospital, Certified Center for Home Parenteral Nutrition, Reference Center for Rare Digestive Diseases in Children, Lyon, France
| | - Cecile Lambe
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Necker-Enfants Malades University of Paris-UFR Paris Descartes, Certified Center for Home Parenteral Nutrition, Reference Center for Rare Digestive Diseases in Children, Paris, France.
| |
Collapse
|
17
|
Yan W, Xiao Y, Zhang Y, Tao Y, Cao Y, Liu K, Cai W, Wang Y. Monogenic mutations in four cases of neonatal-onset watery diarrhea and a mutation review in East Asia. Orphanet J Rare Dis 2021; 16:383. [PMID: 34503561 PMCID: PMC8427875 DOI: 10.1186/s13023-021-01995-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 07/25/2021] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Infants with neonatal-onset diarrhea present with intractable diarrhea in the first few weeks of life. A monogenic mutation is one of the disease etiologies and the use of next-generation sequencing (NGS) has made it possible to screen patients for their mutations. MAIN BODY We retrospectively reviewed the clinical data of four children from unrelated families, who presented with neonatal-onset, chronic, watery, non-bloody diarrhea. After genetic whole-exome sequencing, novel mutations were identified in the EPCAM gene of two children. Congenital chloride diarrhea was diagnosed in one case, which was associated with an SLC26A3 mutation, in which the patient presented with watery diarrhea, malnutrition, and hypochloremic alkalosis. Patient 4 was diagnosed with microvillus inclusion disease and possessed novel compound heterozygous mutations in the MYO5B gene. A review of the genetic variants of SLC26A3 reported in East Asia revealed that c.269_270 dupAA (p.G91Kfs*3) is the most frequent SLC26A3 mutation in China, compared with c.2063-1 G > T in Japan and Korea. EPCAM and MYO5B genetic variants were only sporadically reported in East Asia. CONCLUSION This study expands our knowledge of the clinical manifestations and molecular genetics of neonatal-onset watery diarrhea. Early diagnosis could be achieved by genomic analysis in those infants whose histology features are not typical. The discovery of four novel mutations in the EPCAM gene and two novel mutations in the MYO5B gene provides further etiological evidence for the association of genetic mutations with neonatal-onset diarrhea. To date, c.269_270 dupAA is the most frequent SLC26A3 mutation in China.
Collapse
Affiliation(s)
- Weihui Yan
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, People's Republic of China
- Innovative Research Team of High-Level Local Universities in Shanghai, Shanghai, People's Republic of China
| | - Yongtao Xiao
- Shanghai Institute for Pediatric Research, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, People's Republic of China
- Innovative Research Team of High-Level Local Universities in Shanghai, Shanghai, People's Republic of China
| | - Yunyi Zhang
- Shanghai Institute for Pediatric Research, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, People's Republic of China
| | - Yijing Tao
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yi Cao
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Kunhui Liu
- Department of Pediatric Surgery, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Wei Cai
- Shanghai Institute for Pediatric Research, Shanghai, People's Republic of China.
- Department of Pediatric Surgery, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, People's Republic of China.
- Innovative Research Team of High-Level Local Universities in Shanghai, Shanghai, People's Republic of China.
- Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, No.1665, Kong Jiang Road, Yangpu, Shanghai, 200092, People's Republic of China.
| | - Ying Wang
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, People's Republic of China.
- Innovative Research Team of High-Level Local Universities in Shanghai, Shanghai, People's Republic of China.
- Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, No.1665, Kong Jiang Road, Yangpu, Shanghai, 200092, People's Republic of China.
| |
Collapse
|
18
|
Intestinal immunoregulation: lessons from human mendelian diseases. Mucosal Immunol 2021; 14:1017-1037. [PMID: 33859369 DOI: 10.1038/s41385-021-00398-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 02/04/2023]
Abstract
The mechanisms that maintain intestinal homeostasis despite constant exposure of the gut surface to multiple environmental antigens and to billions of microbes have been scrutinized over the past 20 years with the goals to gain basic knowledge, but also to elucidate the pathogenesis of inflammatory bowel diseases (IBD) and to identify therapeutic targets for these severe diseases. Considerable insight has been obtained from studies based on gene inactivation in mice as well as from genome wide screens for genetic variants predisposing to human IBD. These studies are, however, not sufficient to delineate which pathways play key nonredundant role in the human intestinal barrier and to hierarchize their respective contribution. Here, we intend to illustrate how such insight can be derived from the study of human Mendelian diseases, in which severe intestinal pathology results from single gene defects that impair epithelial and or hematopoietic immune cell functions. We suggest that these diseases offer the unique opportunity to study in depth the pathogenic mechanisms leading to perturbation of intestinal homeostasis in humans. Furthermore, molecular dissection of monogenic intestinal diseases highlights key pathways that might be druggable and therapeutically targeted in common forms of IBD.
Collapse
|
19
|
Chen S, Goldsmith JD, Fawaz R, Al-Ibraheemi A, Perez-Atayde AR, Vargas SO. Liver Pathology, Including MOC31 Immunohistochemistry, in Congenital Tufting Enteropathy. Am J Surg Pathol 2021; 45:1091-1097. [PMID: 33756496 DOI: 10.1097/pas.0000000000001710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Congenital tufting enteropathy (CTE) is a rare heritable cause of intractable diarrhea due to EPCAM mutation. Pathologic findings include intestinal villous atrophy, tufted discohesive tear-drop-shaped epithelium, and a normal brush border. In affected patients, absent intestinal epithelial cell adhesion molecule (EpCAM) expression results in loss of MOC31 immunostaining. CTE liver pathology has not yet been described. We identified CTE patients with liver biopsies and reviewed clinicopathologic material including MOC31 immunohistochemistry. Three CTE patients had 4 liver core biopsies (at ages 1, 5, 7, and 16 y), 2 for preintestinal transplant evaluation, and 2 (from a single patient) for pretreatment assessment of chronic hepatitis C; all had received parenteral nutrition (PN). All samples showed loss of biliary epithelial polarization and mild portal and lobular inflammation. Only the hepatitis C patient demonstrated fibrosis. One patient each had lobular neutrophilic microabscesses and macrovesicular steatosis. Proliferative ductular reactions were absent in CTE patients but present in all controls on PN for other reasons. MOC31 was absent in biliary epithelium and hepatocytes of all CTE patients; controls showed consistent strong membranous biliary epithelial and patchy membranous periportal hepatocyte staining. Our data show that, histologically, hepatopathy in CTE can be difficult to separate from comorbid disease including PN effect; however, the absent ductular reaction may be characteristic. MOC31 localization in the biliary epithelium and zone 1 hepatocytes of controls suggests these compartments of the liver might be most susceptible to effects of EpCAM deficiency. In addition, we validate the liver as suitable tissue for CTE diagnosis using MOC31 immunohistochemistry.
Collapse
Affiliation(s)
| | | | - Rima Fawaz
- Medicine, Division of Gastroenterology, Hepatology, and Nutrition, Boston Children's Hospital, Boston, MA
| | | | | | | |
Collapse
|
20
|
Das B, Okamoto K, Rabalais J, Young JA, Barrett KE, Sivagnanam M. Aberrant Epithelial Differentiation Contributes to Pathogenesis in a Murine Model of Congenital Tufting Enteropathy. Cell Mol Gastroenterol Hepatol 2021; 12:1353-1371. [PMID: 34198013 PMCID: PMC8479479 DOI: 10.1016/j.jcmgh.2021.06.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Congenital tufting enteropathy (CTE) is an intractable diarrheal disease of infancy caused by mutations of epithelial cell adhesion molecule (EpCAM). The cellular and molecular basis of CTE pathology has been elusive. We hypothesized that the loss of EpCAM in CTE results in altered lineage differentiation and defects in absorptive enterocytes thereby contributing to CTE pathogenesis. METHODS Intestine and colon from mice expressing a CTE-associated mutant form of EpCAM (mutant mice) were evaluated for specific markers by quantitative real-time polymerase chain reaction, Western blotting, and immunostaining. Body weight, blood glucose, and intestinal enzyme activity were also investigated. Enteroids derived from mutant mice were used to assess whether the decreased census of major secretory cells could be rescued. RESULTS Mutant mice exhibited alterations in brush-border ultrastructure, function, disaccharidase activity, and glucose absorption, potentially contributing to nutrient malabsorption and impaired weight gain. Altered cell differentiation in mutant mice led to decreased enteroendocrine cells and increased numbers of nonsecretory cells, though the hypertrophied absorptive enterocytes lacked key features, causing brush border malfunction. Further, treatment with the Notch signaling inhibitor, DAPT, increased the numbers of major secretory cell types in mutant enteroids (graphical abstract 1). CONCLUSIONS Alterations in intestinal epithelial cell differentiation in mutant mice favor an increase in absorptive cells at the expense of major secretory cells. Although the proportion of absorptive enterocytes is increased, they lack key functional properties. We conclude that these effects underlie pathogenic features of CTE such as malabsorption and diarrhea, and ultimately the failure to thrive seen in patients.
Collapse
Affiliation(s)
- Barun Das
- Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Kevin Okamoto
- Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - John Rabalais
- Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Jocelyn A. Young
- Department of Pediatrics, University of California, San Diego, La Jolla, California,Department of Pediatrics, Rady Children’s Hospital, San Diego, California
| | - Kim E. Barrett
- Department of Medicine, University of California, San Diego, La Jolla, California
| | - Mamata Sivagnanam
- Department of Pediatrics, University of California, San Diego, La Jolla, California,Department of Pediatrics, Rady Children’s Hospital, San Diego, California,Correspondence Address correspondence to: Mamata Sivagnanam, MD, Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, 9500 Gilman Drive, La Jolla, CA 92093. fax: 858-967-8917.
| |
Collapse
|
21
|
Ayyıldız Civan H, Leitner C, Östreicher I, Schneider AM, Cremer M, Mayr JA, Rossi R, Müller T, Janecke AR. Three Novel EPCAM Variants Causing Tufting Enteropathy in Three Families. CHILDREN (BASEL, SWITZERLAND) 2021; 8:503. [PMID: 34198699 PMCID: PMC8232273 DOI: 10.3390/children8060503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/02/2021] [Accepted: 06/10/2021] [Indexed: 11/16/2022]
Abstract
Tufting enteropathy (TE) is caused by recessive EPCAM mutations, and is characterized by intractable diarrhea of congenital onset and disorganization of enterocytes. TE generally requires parenteral nutrition (PN) during childhood or intestinal bowel transplantation. We report three unrelated families with six children with TE. We highlight the high rate of disease-related mortality. We observe adequate weight gain with PN, but low to normal and stunted body length, supporting the recent notion that a short stature might be intrinsic to TE. The diagnosis of TE in the index patients from each family was delayed for months to years, even when clinical data, duodenal biopsies, or exome sequencing data were obtained early on. We identified three novel pathogenic EPCAM variants: a deletion of exon 1 that removes the ATG initiation codon, a missense variant c.326A > G (p.Gln109Arg), and nonsense mutation c.429G > A (p.Trp143*) in a compound heterozygous state with the Mediterranean splice site variant c.556-14A > G (Tyr186Phefs*6). Homozygosity for p.Gln109Arg was associated with absent EPCAM staining, and compound heterozygosity for p.Trp143*/Tyr186Phefs*6 was associated with reduced EPCAM staining in duodenal biopsies; such observations might contribute to a genotype-phenotype correlation in larger cohorts of TE patients. This study extends the clinical and molecular spectrum of TE.
Collapse
Affiliation(s)
- Hasret Ayyıldız Civan
- Department of Pediatric Gastroenterology, Hepatology and Nutrititon, Health Science University, Istanbul Bakırkoy Dr. Sadi Konuk Education and Research Hospital, 34147 Istanbul, Turkey;
| | - Coleen Leitner
- Department of Pediatrics I, Medical University of Innsbruck, 6020 Innsbruck, Austria; (C.L.); (T.M.)
| | - Iris Östreicher
- Department of Pediatrics, Klinikum Neukoelln, 12351 Berlin, Germany; (I.Ö.); (M.C.); (R.R.)
| | - Anna-Maria Schneider
- Department of Pediatrics, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; (A.-M.S.); (J.A.M.)
| | - Malte Cremer
- Department of Pediatrics, Klinikum Neukoelln, 12351 Berlin, Germany; (I.Ö.); (M.C.); (R.R.)
- Department of Neonatology, Charité University Medical Center, 10117 Berlin, Germany
| | - Johannes A. Mayr
- Department of Pediatrics, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; (A.-M.S.); (J.A.M.)
| | - Rainer Rossi
- Department of Pediatrics, Klinikum Neukoelln, 12351 Berlin, Germany; (I.Ö.); (M.C.); (R.R.)
| | - Thomas Müller
- Department of Pediatrics I, Medical University of Innsbruck, 6020 Innsbruck, Austria; (C.L.); (T.M.)
| | - Andreas R. Janecke
- Department of Pediatrics I, Medical University of Innsbruck, 6020 Innsbruck, Austria; (C.L.); (T.M.)
- Division of Human Genetics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
22
|
Das B, Sivagnanam M. Congenital Tufting Enteropathy: Biology, Pathogenesis and Mechanisms. J Clin Med 2020; 10:E19. [PMID: 33374714 PMCID: PMC7793535 DOI: 10.3390/jcm10010019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/17/2020] [Accepted: 12/17/2020] [Indexed: 01/05/2023] Open
Abstract
Congenital tufting enteropathy (CTE) is an autosomal recessive disease of infancy that causes severe intestinal failure with electrolyte imbalances and impaired growth. CTE is typically diagnosed by its characteristic histological features, including villous atrophy, crypt hyperplasia and focal epithelial tufts consisting of densely packed enterocytes. Mutations in the EPCAM and SPINT2 genes have been identified as the etiology for this disease. The significant morbidity and mortality and lack of direct treatments for CTE patients demand a better understanding of disease pathophysiology. Here, the latest knowledge of CTE biology is systematically reviewed, including clinical aspects, disease genetics, and research model systems. Particular focus is paid to the pathogenesis of CTE and predicted mechanisms of the disease as these would provide insight for future therapeutic options. The contribution of intestinal homeostasis, including the role of intestinal cell differentiation, defective enterocytes, disrupted barrier and cell-cell junction, and cell-matrix adhesion, is vividly described here (see Graphical Abstract). Moreover, based on the known dynamics of EpCAM signaling, potential mechanistic pathways are highlighted that may contribute to the pathogenesis of CTE due to either loss of EpCAM function or EpCAM mutation. Although not fully elucidated, these pathways provide an improved understanding of this devastating disease.
Collapse
Affiliation(s)
- Barun Das
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA;
| | - Mamata Sivagnanam
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA;
- Rady Children’s Hospital, San Diego, CA 92123, USA
| |
Collapse
|
23
|
Zhou YQ, Wu GS, Kong YM, Zhang XY, Wang CL. New mutation in EPCAM for congenital tufting enteropathy: A case report. World J Clin Cases 2020; 8:4975-4980. [PMID: 33195669 PMCID: PMC7642537 DOI: 10.12998/wjcc.v8.i20.4975] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/31/2020] [Accepted: 09/09/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Congenital tufting enteropathy (CTE) is a rare cause of diarrhea in children. However, it can result in early-onset of chronic diarrhea and failure to thrive. Children with this disease have to depend on total parenteral nutrition (TPN), and eventually small intestine transplantation. The epithelial cell adhesion molecule (EPCAM) gene was identified to be associated with CTE. Here, we present a case of an infant with CTE due to a mutation not reported in the literature before.
CASE SUMMARY A 1-year and 7-mo infant boy exhibited intractable watery diarrhea and mushy stool within 1 wk after birth, for which he had required medical treatment and hospitalization several times. His sister presented similar symptoms and died at the age of two. On admission, his body weight was 5700 g (-4.8SDS) and measured 66 cm (-5.4SDS) in height. Meanwhile, he cannot speak or climb. He exhibited mild anemia, hypocalcemia, hypomagnesemia, and an infection in the upper respiratory tract. Microvilli sparse and vacuolar degeneration of epithelial cells were reported by small intestine biopsy. Whole-exome sequencing showed a novel homozygous splice mutation (c.657+1[IVS6] G>A) in the EPCAM gene. He was treated with TPN and recombinant human growth hormone. After 2 mo, his body weight was up to 8500 g and he has been waiting for small bowel transplantation.
CONCLUSION CTE is rare but fatal. Patients with CTE require rapid diagnosis and therapy to improve their survival.
Collapse
Affiliation(s)
- Yan-Qiong Zhou
- Department of Pediatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Guo-Sheng Wu
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Yuan-Mei Kong
- Department of Pediatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Xiao-Yuan Zhang
- Department of Pediatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Chun-Lin Wang
- Department of Pediatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|
24
|
Tufting Enteropathy: A Review of Clinical and Histological Presentation, Etiology, Management, and Outcome. Gastroenterol Res Pract 2020; 2020:5608069. [PMID: 33029133 PMCID: PMC7530495 DOI: 10.1155/2020/5608069] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 09/04/2020] [Accepted: 09/15/2020] [Indexed: 02/07/2023] Open
Abstract
Congenital tufting enteropathy (CTE), also named intestinal epithelial dysplasia, is a rare, autosomal recessive enteropathy with persistent and life-threatening intractable diarrhea early in life. Intractable diarrhea is present independent of breast or formula feeding. Most CTE patients require total parenteral nutrition (TPN), and in severe cases, small bowel transplantation is needed. In the last decade, we have seen remarkable progress in certain aspects, such as the pathogenesis and diagnostic methods of the disease. Rapidly developing molecular analysis techniques have improved the diagnostic methods for CTE and reduced invasive and expensive procedures. Mutations in the gene encoding human epithelial cell adhesion molecule (EpCAM) were identified in the typical form of CTE, which usually exhibits isolated refractory diarrhea. Moreover, the syndromic form of CTE features anal and choanal atresias as well as ophthalmologic signs, which are associated with mutations in the gene encoding Serine Peptidase Inhibitor Kunitz Type 2 (SPINT2). This article reviews CTE disease based on its clinical and histological presentation, etiology and pathogenesis, and management and outcome.
Collapse
|
25
|
Fagotto F, Aslemarz A. EpCAM cellular functions in adhesion and migration, and potential impact on invasion: A critical review. Biochim Biophys Acta Rev Cancer 2020; 1874:188436. [PMID: 32976980 DOI: 10.1016/j.bbcan.2020.188436] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/19/2020] [Accepted: 09/19/2020] [Indexed: 12/14/2022]
Abstract
EpCAM has long been known as a cell surface protein highly expressed in carcinomas. It has since become one of the key cancer biomarkers. Despite its high fame, its actual role in cancer development is still controversial. Beyond a flurry of correlative studies, which point either to a positive or a negative link with tumour progression, there has been surprisingly few studies on the actual cellular mechanisms of EpCAM and on their functional consequences. Clearly, EpCAM plays multiple important roles, in cell proliferation as well as in cell adhesion and migration. The two latter functions, directly relevant for metastasis, are the focus of this review. We attempt here to bring together the available experimental data to build a global coherent view of EpCAM functions. We also include in this overview EpCAM2/Trop2, the close relative of EpCAM. At the core of EpCAM (and EpCAM2/Trop2) function stands the ability to repress contractility of the actomyosin cell cortex. This activity appears to involve direct inhibition by EpCAM of members of the novel PKC family and of a specific downstream PKD-Erk cascade. We will discuss how this activity can result in a variety of adhesive and migratory phenotypes, thus potentially explaining at least part of the apparent inconsistencies between different studies. The picture remains fragmented, and we will highlight some of the conflicting evidence and the many unsolved issues, starting with the controversy around its original description as a cell-cell adhesion molecule.
Collapse
Affiliation(s)
- François Fagotto
- CRBM, University of Montpellier and CNRS, Montpellier 34293, France.
| | - Azam Aslemarz
- CRBM, University of Montpellier and CNRS, Montpellier 34293, France; Department of Biology, McGill University, Montreal, QC H3A1B1, Canada
| |
Collapse
|
26
|
Nakato G, Morimura S, Lu M, Feng X, Wu C, Udey MC. Amelioration of Congenital Tufting Enteropathy in EpCAM (TROP1)-Deficient Mice via Heterotopic Expression of TROP2 in Intestinal Epithelial Cells. Cells 2020; 9:cells9081847. [PMID: 32781650 PMCID: PMC7465201 DOI: 10.3390/cells9081847] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 08/04/2020] [Accepted: 08/04/2020] [Indexed: 12/11/2022] Open
Abstract
TROP1 (EpCAM) and TROP2 are homologous cell surface proteins that are widely expressed, and often co-expressed, in developing and adult epithelia. Various functions have been ascribed to EpCAM and TROP2, but responsible mechanisms are incompletely characterized and functional equivalence has not been examined. Adult intestinal epithelial cells (IEC) express high levels of EpCAM, while TROP2 is not expressed. EpCAM deficiency causes congenital tufting enteropathy (CTE) in humans and a corresponding lethal condition in mice. We expressed TROP2 and EpCAM in the IEC of EpCAM-deficient mice utilizing a villin promoter to assess EpCAM and TROP2 function. Expression of EpCAM or TROP2 in the IEC of EpCAM knockout mice prevented CTE. TROP2 rescue (T2R) mice were smaller than controls, while EpCAM rescue (EpR) mice were not. Abnormalities were observed in the diameters and histology of T2R small intestine, and Paneth and stem cell markers were decreased. T2R mice also exhibited enlarged mesenteric lymph nodes, enhanced permeability to 4 kDa FITC-dextran and increased sensitivity to detergent-induced colitis, consistent with compromised barrier function. Studies of IEC organoids and spheroids revealed that stem cell function was also compromised in T2R mice. We conclude that EpCAM and TROP2 exhibit functional redundancy, but they are not equivalent.
Collapse
Affiliation(s)
- Gaku Nakato
- Intestinal Microbiota Project, Intestinal Ecosystem Regulation Group, Kanagawa Institute of Industrial Science and Technology, Kawasaki-shi, Kanagawa 210-0821, Japan
- Correspondence: ; Tel.: +81-44-280-2214
| | - Sohshi Morimura
- Department of Dermatology, International University of Health and Welfare, Narita-shi, Chiba 286-8520, Japan;
| | - Michael Lu
- Experimental Immunology Branch, National Cancer Institute, Bethesda, MD 20892, USA;
| | - Xu Feng
- Retired from National Cancer Institute, Bethesda, MD 20892, USA;
| | - Chuanjin Wu
- Laboratory of Immune Cell Biology, National Cancer Institute, Bethesda, MD 20892, USA;
| | - Mark C. Udey
- Dermatology Division, Department of Medicine, Washington University School of Medicine, Saint Louis, MO 63110, USA;
| |
Collapse
|
27
|
Hassan K, Sher G, Hamid E, Hazima KA, Abdelrahman H, Al Mudahka F, Al-Masri W, Sankar J, Daryaee M, Shawish R, Khan MA, Nawaz Z. Outcome associated with EPCAM founder mutation c.499dup in Qatar. Eur J Med Genet 2020; 63:104023. [PMID: 32735948 DOI: 10.1016/j.ejmg.2020.104023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 05/12/2020] [Accepted: 07/20/2020] [Indexed: 01/16/2023]
Abstract
Tufting enteropathy (TE) is a rare autosomal recessive congenital enteropathy that usually requires long-term parenteral nutrition (PN). In the Arabic Peninsula, four distinct EPCAM mutations have been identified to cause TE. As consanguineous marriages are socially favored, pre-marital and pre-conception testing has become a critical disease prevention strategy. This study aimed to identify the pathogenic EPCAM mutations causing TE in Qatari families and determine possible genotype-phenotype correlations. Twenty-two TE patients from seven multiplex families with TE were identified. Blood samples were collected from patients and first-degree relatives. Exons of the gene were amplified and sequenced. Retrospective chart review and/or family interviews were conducted to determine phenotypic characteristics of the disease. Sequence analysis revealed a single, previously described c.499dup mutation in exon 5 of all families tested, suggesting a founder effect. Of the 18 patients whose full clinical information was available, three patients (17%) were off PN with a good quality of life, without intestinal transplantation, and one (6%) was receiving partial PN. Our patients with TE were severely stunted compared to a similar group of patients receiving long-term PN for short bowel syndrome, suggesting that this could possibly be due to TE rather than secondary to inadequate nutrition. Our study identified the EPCAM mutation c.499dup as the genetic defect causing TE in all the participant Qatari families. This finding should facilitate early diagnosis of TE and genetic counseling. Furthermore, it should aid in the prevention of TE through pre-marital screening, antenatal diagnosis, and pre-implantation genetic diagnosis.
Collapse
Affiliation(s)
- Kamal Hassan
- Pediatric Gastroenterology Section, Hamad General Hospital, P.O. Box 3050, Doha, Qatar; Pediatric Gastroenterology Section, Sidra Medicine, P.O. Box 269999, Doha, Qatar.
| | - Gulab Sher
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, 3050, Qatar
| | - Eman Hamid
- Pediatric Gastroenterology Section, Hamad General Hospital, P.O. Box 3050, Doha, Qatar
| | - Khalid Abou Hazima
- Pediatric Gastroenterology Section, Hamad General Hospital, P.O. Box 3050, Doha, Qatar; Pediatric Gastroenterology Section, Sidra Medicine, P.O. Box 269999, Doha, Qatar
| | - Hatim Abdelrahman
- Pediatric Gastroenterology Section, Hamad General Hospital, P.O. Box 3050, Doha, Qatar; Pediatric Gastroenterology Section, Sidra Medicine, P.O. Box 269999, Doha, Qatar
| | - Fatma Al Mudahka
- Pediatric Gastroenterology Section, Hamad General Hospital, P.O. Box 3050, Doha, Qatar; Pediatric Gastroenterology Section, Sidra Medicine, P.O. Box 269999, Doha, Qatar
| | - Wesam Al-Masri
- Pediatric Gastroenterology Section, Hamad General Hospital, P.O. Box 3050, Doha, Qatar; Pediatric Gastroenterology Section, Sidra Medicine, P.O. Box 269999, Doha, Qatar
| | - Jisha Sankar
- Pediatric Gastroenterology Section, Hamad General Hospital, P.O. Box 3050, Doha, Qatar; Pediatric Gastroenterology Section, Sidra Medicine, P.O. Box 269999, Doha, Qatar
| | - Mahlah Daryaee
- Patient & Family Department, Hamad General Hospital, P.O. Box 3050, Doha, Qatar
| | - Rana Shawish
- Pediatric Gastroenterology Section, Sidra Medicine, P.O. Box 269999, Doha, Qatar; Patient & Family Department, Hamad General Hospital, P.O. Box 3050, Doha, Qatar
| | - Muzammil Ahmad Khan
- Diagnostic Genomic Division, Department of Laboratory Medicine and Pathology, Hamad Medical Corporation, Doha, 3050, Qatar; Gomal Centre of Biochemistry and Biotechnology, Gomal University Dera Ismail Khan, Khyber-Pakhtoonkhwa, 29050, Pakistan
| | - Zafar Nawaz
- Diagnostic Genomic Division, Department of Laboratory Medicine and Pathology, Hamad Medical Corporation, Doha, 3050, Qatar
| |
Collapse
|
28
|
Das B, Okamoto K, Rabalais J, Marchelletta RR, Barrett KE, Das S, Niwa M, Sivagnanam M. Congenital Tufting Enteropathy-Associated Mutant of Epithelial Cell Adhesion Molecule Activates the Unfolded Protein Response in a Murine Model of the Disease. Cells 2020; 9:cells9040946. [PMID: 32290509 PMCID: PMC7226999 DOI: 10.3390/cells9040946] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/31/2020] [Accepted: 04/09/2020] [Indexed: 12/18/2022] Open
Abstract
Congenital tufting enteropathy (CTE) is a rare chronic diarrheal disease of infancy caused by mutations in epithelial cell adhesion molecule (EpCAM). Previously, a murine CTE model showed mis-localization of EpCAM away from the basolateral cell surface in the intestine. Here we demonstrate that mutant EpCAM accumulated in the endoplasmic reticulum (ER) where it co-localized with ER chaperone, GRP78/BiP, revealing potential involvement of ER stress-induced unfolded protein response (UPR) pathway in CTE. To investigate the significance of ER-localized mutant EpCAM in CTE, activation of the three UPR signaling branches initiated by the ER transmembrane protein components IRE1, PERK, and ATF6 was tested. A significant reduction in BLOS1 and SCARA3 mRNA levels in EpCAM mutant intestinal cells demonstrated that regulated IRE1-dependent decay (RIDD) was activated. However, IRE1 dependent XBP1 mRNA splicing was not induced. Furthermore, an increase in nuclear-localized ATF6 in mutant intestinal tissues revealed activation of the ATF6-signaling arm. Finally, an increase in both the phosphorylated form of the translation initiation factor, eIF2α, and ATF4 expression in the mutant intestine provided support for activation of the PERK-mediated pathway. Our results are consistent with a significant role for UPR in gastrointestinal homeostasis and provide a working model for CTE pathophysiology.
Collapse
Affiliation(s)
- Barun Das
- Department of Pediatrics, University of California, San Diego, CA 92093, USA; (B.D.); (K.O.); (J.R.)
| | - Kevin Okamoto
- Department of Pediatrics, University of California, San Diego, CA 92093, USA; (B.D.); (K.O.); (J.R.)
| | - John Rabalais
- Department of Pediatrics, University of California, San Diego, CA 92093, USA; (B.D.); (K.O.); (J.R.)
| | - Ronald R. Marchelletta
- Department of Medicine, University of California, San Diego, CA 92093, USA; (R.R.M.); (K.E.B.)
| | - Kim E. Barrett
- Department of Medicine, University of California, San Diego, CA 92093, USA; (R.R.M.); (K.E.B.)
| | - Soumita Das
- Department of Pathology, University of California, San Diego, CA 92093, USA;
| | - Maho Niwa
- Division of Biological Sciences, University of California, San Diego, CA 92093, USA;
| | - Mamata Sivagnanam
- Department of Pediatrics, University of California, San Diego, CA 92093, USA; (B.D.); (K.O.); (J.R.)
- Rady Children’s Hospital, San Diego, CA 92123, USA
- Correspondence: ; Tel.: +1-858-966-8907
| |
Collapse
|
29
|
Fang Y, Luo Y, Yu J, Chen J. A case of severe malnutrition infant with neonatal onset intractable diarrhea. BMC Pediatr 2020; 20:133. [PMID: 32293360 PMCID: PMC7087373 DOI: 10.1186/s12887-020-1999-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 02/25/2020] [Indexed: 11/13/2022] Open
Abstract
Background Congenital tufting enteropathy (CTE) is a rare disease that manifests as intractable diarrhea during the neonatal period which is associated with mutations of the epithelial cell adhesion molecule (EpCAM) gene. Case presentation A male infant who presented with vomiting, diarrhea, abdominal distention, malnutrition and growth failure was admitted to our department when he was 2 months old. His parents were healthy and nonconsanguineous. Etiologic examinations of stool, inflammatory markers, blood gas and electrolytes levels, serum albumin level, serum immunoglobin levels were all normal. And there was no indication for metabolic diseases. Additionally, gastrointestinal contrast did not reveal abnormality of gastrointestinal. The patient was diagnosed with intestinal malabsorptive syndrome and severe malnutrition without definite cause. He was on supportive treatment and nutritional therapy for 13 months. However, he did not gain weight obviously. He was discharged at the age of 15 months and was fed with partial hydrolyzed formula and rice paste at home. Three months later he developed hypoglycemia and severe respiratory infection. Finally, he died due to sepsis and multiple organs failure. The next generation sequencing revealed one homozygous mutation of EpCAM gene and one complex heterozygous mutation of TTC7A gene. He was diagnosed CTE according to the genetic results and clinical manifestations. Conclusions CTE is rarely reported in Asia. Patients present with congenital diarrhea, poor weight gain and growth failure are recommended to perform endoscopy examination with proper immunohistochemistry study as early as possible, and genetic testing is necessary when suspecting congenital diarrhea and enteropathy.
Collapse
Affiliation(s)
- Youhong Fang
- Department of gastroenterology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Bin Sheng Road, Hangzhou, 310052, Zhejiang Province, China
| | - Youyou Luo
- Department of gastroenterology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Bin Sheng Road, Hangzhou, 310052, Zhejiang Province, China
| | - Jindan Yu
- Department of gastroenterology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Bin Sheng Road, Hangzhou, 310052, Zhejiang Province, China
| | - Jie Chen
- Department of gastroenterology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 3333 Bin Sheng Road, Hangzhou, 310052, Zhejiang Province, China.
| |
Collapse
|
30
|
Holt-Danborg L, Vodopiutz J, Nonboe AW, De Laffolie J, Skovbjerg S, Wolters VM, Müller T, Hetzer B, Querfurt A, Zimmer KP, Jensen JK, Entenmann A, Heinz-Erian P, Vogel LK, Janecke AR. SPINT2 (HAI-2) missense variants identified in congenital sodium diarrhea/tufting enteropathy affect the ability of HAI-2 to inhibit prostasin but not matriptase. Hum Mol Genet 2020; 28:828-841. [PMID: 30445423 DOI: 10.1093/hmg/ddy394] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 11/07/2018] [Accepted: 11/08/2018] [Indexed: 11/13/2022] Open
Abstract
The syndromic form of congenital sodium diarrhea (SCSD) is caused by bi-allelic mutations in SPINT2, which encodes a Kunitz-type serine protease inhibitor (HAI-2). We report three novel SCSD patients, two novel SPINT2 mutations and review published cases. The most common findings in SCSD patients were choanal atresia (20/34) and keratitis of infantile onset (26/34). Characteristic epithelial tufts on intestinal histology were reported in 13/34 patients. Of 13 different SPINT2 variants identified in SCSD, 4 are missense variants and localize to the second Kunitz domain (KD2) of HAI-2. HAI-2 has been implicated in the regulation of the activities of several serine proteases including prostasin and matriptase, which are both important for epithelial barrier formation. No patient with bi-allelic stop mutations was identified, suggesting that at least one SPINT2 allele encoding a protein with residual HAI-2 function is necessary for survival. We show that the SCSD-associated HAI-2 variants p.Phe161Val, p.Tyr163Cys and p.Gly168Ser all display decreased ability to inhibit prostasin-catalyzed cleavage. However, the SCSD-associated HAI-2 variants inhibited matriptase as efficiently as the wild-type HAI-2. Homology modeling indicated limited solvent exposure of the mutated amino acids, suggesting that they induce misfolding of KD2. This suggests that prostasin needs to engage with an exosite motif located on KD2 in addition to the binding loop (Cys47/Arg48) located on the first Kunitz domain in order to inhibit prostasin. In conclusion our data suggests that SCSD is caused by lack of inhibition of prostasin or a similar protease in the secretory pathway or on the plasma membrane.
Collapse
Affiliation(s)
- Lasse Holt-Danborg
- Department of Cellular and Molecular Medicine, The Panum Institute, University of Copenhagen, Denmark
| | - Julia Vodopiutz
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna
| | - Annika W Nonboe
- Department of Cellular and Molecular Medicine, The Panum Institute, University of Copenhagen, Denmark
| | - Jan De Laffolie
- Abteilung Allgemeine Pädiatrie und Neonatologie, Zentrum für Kinderheilkunde und Jugendmedizin, Justus-Liebig-Universität, Gießen, Germany
| | - Signe Skovbjerg
- Department of Cellular and Molecular Medicine, The Panum Institute, University of Copenhagen, Denmark
| | - Victorien M Wolters
- Department of Pediatric Gastroenterology, WKZ/ UMC Utrecht, Utrecht, The Netherlands
| | - Thomas Müller
- Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Benjamin Hetzer
- Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Alexander Querfurt
- Gesundheit Nord gGmbH, Klinikverbund Bremen, Klinik für Kinder und Jugendmedizin, Professor-Hess-Kinderklinik, Klinikum Bremen-Mitte, Bremen, Germany
| | - Klaus-Peter Zimmer
- Abteilung Allgemeine Pädiatrie und Neonatologie, Zentrum für Kinderheilkunde und Jugendmedizin, Justus-Liebig-Universität, Gießen, Germany
| | - Jan K Jensen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Andreas Entenmann
- Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Peter Heinz-Erian
- Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria
| | - Lotte K Vogel
- Department of Cellular and Molecular Medicine, The Panum Institute, University of Copenhagen, Denmark
| | - Andreas R Janecke
- Department of Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria.,Division of Human Genetics, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
31
|
Abstract
Congenital diarrheal disorders are heterogeneous conditions characterized by diarrhea with onset in the first years of life. They range from simple temporary conditions, such as cow's milk protein intolerance to irreversible complications, such as microvillous inclusion disease with significant morbidity and mortality. Advances in genomic medicine have improved our understanding of these disorders, leading to an ever-increasing list of identified causative genes. The diagnostic approach to these conditions consists of establishing the presence of diarrhea by detailed review of the history, followed by characterizing the composition of the diarrhea, the response to fasting, and with further specialized testing.
Collapse
Affiliation(s)
- Abdul Aziz Elkadri
- Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, USA.
| |
Collapse
|
32
|
Das B, Okamoto K, Rabalais J, Kozan PA, Marchelletta RR, McGeough MD, Durali N, Go M, Barrett KE, Das S, Sivagnanam M. Enteroids expressing a disease-associated mutant of EpCAM are a model for congenital tufting enteropathy. Am J Physiol Gastrointest Liver Physiol 2019; 317:G580-G591. [PMID: 31433211 PMCID: PMC6879886 DOI: 10.1152/ajpgi.00098.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Congenital tufting enteropathy (CTE) is an autosomal recessive disease characterized by severe intestinal failure in infancy and mutations in the epithelial cell adhesion molecule (EPCAM) gene. Previous studies of CTE in mice expressing mutant EpCAM show neonatal lethality. Hence, to study the cellular, molecular, and physiological alterations that result from EpCAM mutation, a tamoxifen-inducible mutant EpCAM enteroid model has been generated. The presence of mutant EpCAM in the model was confirmed at both mRNA and protein levels. Immunofluorescence microscopy demonstrated the reduced expression of mutant EpCAM. Mutant enteroids had reduced budding potential as well as significantly decreased mRNA expression for epithelial lineage markers (Mucin 2, lysozyme, sucrase-isomaltase), proliferation marker Ki67, and secretory pathway transcription factors (Atoh1, Hnf1b). Significantly decreased numbers of Paneth and goblet cells were confirmed by staining. These findings were correlated with intestinal tissue from CTE patients and the mutant mice model that had significantly fewer Paneth and goblet cells than in healthy counterparts. FITC-dextran studies demonstrated significantly impaired barrier function in monolayers derived from mutant enteroids compared with control monolayers. In conclusion, we have established an ex vivo CTE model. The role of EpCAM in the budding potential, differentiation, and barrier function of enteroids is noted. Our study establishes new facets of EpCAM biology that will aid in understanding the pathophysiology of CTE and role of EpCAM in health and disease.NEW & NOTEWORTHY Here, we develop a novel ex vivo enteroid model for congenital tufting enteropathy (CTE) based on epithelial cell adhesion molecule (EPCAM) gene mutations found in patients. With this model we demonstrate the role of EpCAM in maintaining the functional homeostasis of the intestinal epithelium, including differentiation, proliferation, and barrier integrity. This study further establishes a new direction in EpCAM biology that will help in understanding the detailed pathophysiology of CTE and role of EpCAM.
Collapse
Affiliation(s)
- Barun Das
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Kevin Okamoto
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - John Rabalais
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Philip A. Kozan
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | | | - Matthew D. McGeough
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Nassim Durali
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Maria Go
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California
| | - Kim E. Barrett
- 2Department of Medicine, University of California, San Diego, La Jolla, California
| | - Soumita Das
- 3Department of Pathology, University of California, San Diego, La Jolla, California
| | - Mamata Sivagnanam
- 1Department of Pediatrics, University of California, San Diego, La Jolla, California,4Rady Children’s Hospital, San Diego, California
| |
Collapse
|
33
|
Tamura K, Kaneda M, Futagawa M, Takeshita M, Kim S, Nakama M, Kawashita N, Tatsumi-Miyajima J. Genetic and genomic basis of the mismatch repair system involved in Lynch syndrome. Int J Clin Oncol 2019; 24:999-1011. [PMID: 31273487 DOI: 10.1007/s10147-019-01494-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 06/17/2019] [Indexed: 12/11/2022]
Abstract
Lynch syndrome is a cancer-predisposing syndrome inherited in an autosomal-dominant manner, wherein colon cancer and endometrial cancer develop frequently in the family, it results from a loss-of-function mutation in one of four different genes (MLH1, MSH2, MSH6, and PMS2) encoding mismatch repair proteins. Being located immediately upstream of the MSH2 gene, EPCAM abnormalities can affect MSH2 and cause Lynch syndrome. Mismatch repair proteins are involved in repairing of incorrect pairing (point mutations and deletion/insertion of simple repetitive sequences, so-called microsatellites) that can arise during DNA replication. MSH2 forms heterodimers with MSH6 or MSH3 (MutSα, MutSβ, respectively) and is involved in mismatch-pair recognition and initiation of repair. MLH1 forms a complex with PMS2, and functions as an endonuclease. If the mismatch repair system is thoroughly working, genome integrity is maintained completely. Lynch syndrome is a state of mismatch repair deficiency due to a monoallelic abnormality of any mismatch repair genes. The phenotype indicating the mismatch repair deficiency can be frequently shown as a microsatellite instability in tumors. Children with germline biallelic mismatch repair gene abnormalities were reported to develop conditions such as gastrointestinal polyposis, colorectal cancer, brain cancer, leukemia, etc., and so on, demonstrating the need to respond with new concepts in genetic counseling. In promoting cancer genome medicine in a new era, such as by utilizing immune checkpoints, it is important to understand the genetic and genomic molecular background, including the status of mismatch repair deficiency.
Collapse
Affiliation(s)
- Kazuo Tamura
- Division of Medical Genetics, Master of Science, Graduate School of Science and Engineering Research, Kindai University, Higashiosaka, Japan.
| | - Motohide Kaneda
- Division of Medical Genetics, Master of Science, Graduate School of Science and Engineering Research, Kindai University, Higashiosaka, Japan
| | - Mashu Futagawa
- Division of Medical Genetics, Master of Science, Graduate School of Science and Engineering Research, Kindai University, Higashiosaka, Japan
| | - Miho Takeshita
- Division of Medical Genetics, Master of Science, Graduate School of Science and Engineering Research, Kindai University, Higashiosaka, Japan
| | - Sanghyuk Kim
- Division of Medical Genetics, Master of Science, Graduate School of Science and Engineering Research, Kindai University, Higashiosaka, Japan
| | - Mina Nakama
- Division of Clinical Genetics, Gifu University Hospital, Gifu, Japan
| | - Norihito Kawashita
- Division of Medical Genetics, Master of Science, Graduate School of Science and Engineering Research, Kindai University, Higashiosaka, Japan
| | - Junko Tatsumi-Miyajima
- Division of Medical Genetics, Master of Science, Graduate School of Science and Engineering Research, Kindai University, Higashiosaka, Japan
| |
Collapse
|
34
|
Pathak SJ, Mueller JL, Okamoto K, Das B, Hertecant J, Greenhalgh L, Cole T, Pinsk V, Yerushalmi B, Gurkan OE, Yourshaw M, Hernandez E, Oesterreicher S, Naik S, Sanderson IR, Axelsson I, Agardh D, Boland CR, Martin MG, Putnam CD, Sivagnanam M. EPCAM mutation update: Variants associated with congenital tufting enteropathy and Lynch syndrome. Hum Mutat 2019; 40:142-161. [PMID: 30461124 PMCID: PMC6328345 DOI: 10.1002/humu.23688] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/24/2018] [Accepted: 11/14/2018] [Indexed: 12/30/2022]
Abstract
The epithelial cell adhesion molecule gene (EPCAM, previously known as TACSTD1 or TROP1) encodes a membrane-bound protein that is localized to the basolateral membrane of epithelial cells and is overexpressed in some tumors. Biallelic mutations in EPCAM cause congenital tufting enteropathy (CTE), which is a rare chronic diarrheal disorder presenting in infancy. Monoallelic deletions of the 3' end of EPCAM that silence the downstream gene, MSH2, cause a form of Lynch syndrome, which is a cancer predisposition syndrome associated with loss of DNA mismatch repair. Here, we report 13 novel EPCAM mutations from 17 CTE patients from two separate centers, review EPCAM mutations associated with CTE and Lynch syndrome, and structurally model pathogenic missense mutations. Statistical analyses indicate that the c.499dupC (previously reported as c.498insC) frameshift mutation was associated with more severe treatment regimens and greater mortality in CTE, whereas the c.556-14A>G and c.491+1G>A splice site mutations were not correlated with treatments or outcomes significantly different than random simulation. These findings suggest that genotype-phenotype correlations may be useful in contributing to management decisions of CTE patients. Depending on the type and nature of EPCAM mutation, one of two unrelated diseases may occur, CTE or Lynch syndrome.
Collapse
Affiliation(s)
- Sagar J. Pathak
- Department of PediatricsUniversity of California, San DiegoLa JollaCalifornia
- Rady Children's HospitalSan DiegoCalifornia
| | - James L. Mueller
- Department of PediatricsUniversity of California, San DiegoLa JollaCalifornia
| | - Kevin Okamoto
- Department of PediatricsUniversity of California, San DiegoLa JollaCalifornia
| | - Barun Das
- Department of PediatricsUniversity of California, San DiegoLa JollaCalifornia
| | - Jozef Hertecant
- Genetics/Metabolics ServiceTawam HospitalAl AinUnited Arab Emirates
| | | | - Trevor Cole
- West Midlands Regional Genetics Service and Birmingham Health PartnersBirmingham Women's HospitalBirminghamUK
| | - Vered Pinsk
- Division of Pediatrics, Pediatric Gastroenterology UnitSoroka University Medical Center and Faculty of Health SciencesBen‐Gurion University of the NegevBeer‐ShevaIsrael
| | - Baruch Yerushalmi
- Division of Pediatrics, Pediatric Gastroenterology UnitSoroka University Medical Center and Faculty of Health SciencesBen‐Gurion University of the NegevBeer‐ShevaIsrael
| | - Odul E. Gurkan
- Department of PediatricsGazi University School of MedicineAnkaraTurkey
| | - Michael Yourshaw
- Department of Human GeneticsUniversity of California, Los AngelesLos AngelesCalifornia
| | - Erick Hernandez
- Pediatric GastroenterologyMiami Children's Health SystemMiamiFlorida
| | | | - Sandhia Naik
- Paediatric GastroenterologyBarts and the London School of MedicineLondonUK
| | - Ian R. Sanderson
- Paediatric GastroenterologyBarts and the London School of MedicineLondonUK
| | - Irene Axelsson
- Department of PediatricsSkane University HospitalMalmoSweden
| | - Daniel Agardh
- Department of Clinical SciencesLund University, Skane University HospitalMalmoSweden
| | - C. Richard Boland
- Department of MedicineUniversity of California, San DiegoLa JollaCalifornia
| | - Martin G. Martin
- Department of PediatricsUniversity of California, Los AngelesLos AngelesCalifornia
| | - Christopher D. Putnam
- Department of MedicineUniversity of California, San DiegoLa JollaCalifornia
- San Diego BranchLudwig Institute for Cancer ResearchLa JollaCalifornia
| | - Mamata Sivagnanam
- Department of PediatricsUniversity of California, San DiegoLa JollaCalifornia
- Rady Children's HospitalSan DiegoCalifornia
| |
Collapse
|
35
|
Bilateral Total Hip and Unilateral Knee Arthroplasties in a Young Adult with Arthropathy-Associated Intestinal Epithelial Dysplasia (Tufting Enteropathy). Case Rep Orthop 2018; 2018:4630759. [PMID: 30510828 PMCID: PMC6232787 DOI: 10.1155/2018/4630759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 10/15/2018] [Indexed: 11/17/2022] Open
Abstract
Intestinal epithelial dysplasia (tufting enteropathy) is an uncommon congenital disorder. Furthermore, its association with chronic inflammatory arthropathy is rarely documented in the literature. Low prevalence rates of 1 in 100,000 live births in Western Europe exist, with higher rates in North Africa and Middle Eastern countries. Malta, being a small Mediterranean island at the cusp between Europe and North Africa, has an anecdotal sevenfold prevalence rate. This is the first documented case report of a patient with both intestinal epithelial dysplasia and severe bilateral hip and knee arthropathy that required simultaneous bilateral hip followed by, after a short interval, unilateral knee arthroplasties. Our aim is to highlight the rapid progression of associated arthropathy as well as the successful treatment with joint arthroplasties in such extreme cases. Surgical treatment may be a necessity despite best medical efforts to halt the disease.
Collapse
|
36
|
Huang L, Yang Y, Yang F, Liu S, Zhu Z, Lei Z, Guo J. Functions of EpCAM in physiological processes and diseases (Review). Int J Mol Med 2018; 42:1771-1785. [PMID: 30015855 PMCID: PMC6108866 DOI: 10.3892/ijmm.2018.3764] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 07/04/2018] [Indexed: 12/14/2022] Open
Abstract
EpCAM (epithelial cell adhesion molecule) is a type I transmembrane glycoprotein, which was originally identified as a tumor-associated antigen due to its high expression level in rapidly growing epithelial tumors. Germ line mutations of the human EpCAM gene have been indicated as the cause of congenital tufting enteropathy. Previous studies based on cell models have revealed that EpCAM contributes to various biological processes including cell adhesion, signaling, migration and proliferation. Due to the previous lack of genetic animal models, the in vivo functions of EpCAM remain largely unknown. However, EpCAM genetic animal models have recently been generated, and are useful for understanding the functions of EpCAM. The authors here briefly review the functions and mechanisms of EpCAM in physiological processes and different diseases.
Collapse
Affiliation(s)
- Li Huang
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong 510006, P.R. China
| | - Yanhong Yang
- The First Affiliated Hospital, School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong 510080, P.R. China
| | - Fei Yang
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong 510006, P.R. China
| | - Shaomin Liu
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong 510006, P.R. China
| | - Ziqin Zhu
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong 510006, P.R. China
| | - Zili Lei
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong 510006, P.R. China
| | - Jiao Guo
- Guangdong Metabolic Disease Research Center of Integrated Chinese and Western Medicine, Guangdong Pharmaceutical University, Guangzhou Higher Education Mega Center, Guangzhou, Guangdong 510006, P.R. China
| |
Collapse
|
37
|
Reversal of Intestinal Failure in Children With Tufting Enteropathy Supported With Parenteral Nutrition at Home. J Pediatr Gastroenterol Nutr 2018; 66:967-971. [PMID: 29334565 DOI: 10.1097/mpg.0000000000001894] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES The aim of the study was to review long-term outcome of intestinal epithelial dysplasia (IED)/tufting enteropathy (TE) patients treated with parenteral nutrition (PN) at home managed by an intestinal failure (IF) rehabilitation service. METHODS Infants presenting from 1986 to 2010 with IF, and TE histology were retrospectively reviewed for up to 30 years. Data collected included outcome, presentation, nutrition (parenteral/enteral), country of residence, race, EpCAM gene, growth, bone age, and occupation. RESULTS Thirteen patients (6 boys) in Malta and the UK with TE histology were established on home PN. Survival was 100% for UK children and 92% overall (1 death aged 13 months). Six patients (50% of the surviving 12) weaned off PN. Overall PN requirements reduced with increasing age and <7 infusions/week were needed by 10/12, 83% by 10 years, 6/8, 75% who had reached 15 years, 5/7, 71% who had reached 20, and all 4, 100% >25 years. Two of 12 cases weaned from PN by 10 years, 1 of 8 by 15 years, 3 of 7 by 20 years, and 3 of 4 or 75% >25 years. Seven Maltese patients homozygous for the same EPCAM gene abnormality had a similar outcome to the other cases. Weight, height, bone mineralization, bone age, and insulin-like growth factor-1 (IGF-1) levels were low, but improved with age. Patients achieved educational levels of parents and were employed. CONCLUSIONS IED cases should have >92% chance of long-term survival and >50% chance of enteral autonomy by/in early adult life and 75% by 25 years. Even if PN dependent s/he can gain employment. Patients with IED managed on PN at home by an IF rehabilitation service should avoid intestinal transplant.
Collapse
|
38
|
Sheikh IA, Ammoury R, Ghishan FK. Pathophysiology of Diarrhea and Its Clinical Implications. PHYSIOLOGY OF THE GASTROINTESTINAL TRACT 2018:1669-1687. [DOI: 10.1016/b978-0-12-809954-4.00068-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
39
|
Abstract
BACKGROUND AND AIMS Congenital tufting enteropathy (CTE) is a rare autosomal recessive form of intractable diarrhea of infancy. Patients develop chronic diarrhea within days after birth, leading to severe malabsorption and significant mortality. CTE is characterized by subtotal villous atrophy with crypt hyperplasia. Typical features include abnormal villi in the intestinal epithelium and disorganization of surface enterocytes with focal crowding, resembling tufts. The pathogenesis of CTE remains poorly understood. CTE has been reported in Western populations, but until now had not been reported in China. The objective of this study was to identify the gene responsible for CTE in a Chinese individual. METHODS A 13-year-old girl with suspected CTE, whose parents were both healthy, was evaluated in our clinic. Tissues were obtained by endoscopy and examined by electron microscopy. Genomic DNA, extracted from the peripheral blood of the child and parents, was subjected to whole-exome sequencing. After mutations in the gene encoding epithelial cell adhesion molecule (EPCAM) were identified, expression of EPCAM was examined by immunohistochemistry staining. RESULTS Whole-exome sequencing revealed compound heterozygous mutations in EPCAM in the patient, with immunohistochemical analysis showing complete loss of EPCAM expression in the intestinal villi and crypts. CONCLUSIONS We identified compound heterozygous mutations in EPCAM, with loss of EPCAM expression in duodenal enterocytes, in a patient with intractable diarrhea since infancy who was subsequently diagnosed with CTE. This is the first case of CTE to be reported in a Chinese patient.
Collapse
Affiliation(s)
- Wenjuan Tang
- Department of Gastroenterology, Children's Hospital of Fudan University, Shanghai
| | - Taosheng Huang
- Divisions of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | | | - Ying Huang
- Department of Gastroenterology, Children's Hospital of Fudan University, Shanghai
| |
Collapse
|
40
|
Gaston C, Salomon J, Goulet O, Delacour D. [The congenital tufting enteropathy, or when the intestine is under low cellular tension]. Med Sci (Paris) 2017; 33:694-697. [PMID: 28945548 DOI: 10.1051/medsci/20173308003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Cécile Gaston
- Adhérence cellulaire et mécanique, Institut Jacques Monod, CNRS UMR7592, Université Paris Diderot, 15, rue Hélène Brion, 75013 Paris, France
| | - Julie Salomon
- Adhérence cellulaire et mécanique, Institut Jacques Monod, CNRS UMR7592, Université Paris Diderot, 15, rue Hélène Brion, 75013 Paris, France - Département de gastroentérologie pédiatrique et nutrition, Hôpital Necker-Enfants Malades, Sorbonne Paris Cité, 149, rue de Sèvres, 75015 Paris, France
| | - Olivier Goulet
- Département de gastroentérologie pédiatrique et nutrition, Hôpital Necker-Enfants Malades, Sorbonne Paris Cité, 149, rue de Sèvres, 75015 Paris, France
| | - Delphine Delacour
- Adhérence cellulaire et mécanique, Institut Jacques Monod, CNRS UMR7592, Université Paris Diderot, 15, rue Hélène Brion, 75013 Paris, France
| |
Collapse
|
41
|
Ensari A, Kelsen J, Russo P. Newcomers in paediatric GI pathology: childhood enteropathies including very early onset monogenic IBD. Virchows Arch 2017; 472:111-123. [PMID: 28718031 DOI: 10.1007/s00428-017-2197-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 07/02/2017] [Accepted: 07/06/2017] [Indexed: 12/16/2022]
Abstract
Childhood enteropathies are a group of diseases causing severe chronic (>2-3 weeks) diarrhoea often starting in the first week of life with the potential for fatal complications for the affected infant. Early identification and accurate classification of childhood enteropathies are, therefore, crucial for making treatment decisions to prevent life-threatening complications. Childhood enteropathies are classified into four groups based on the underlying pathology: (i) conditions related to defective digestion, absorption and transport of nutrients and electrolytes; (ii) disorders related to enterocyte differentiation and polarization; (iii) defects of enteroendocrine cell differentiation; and (iv) disorders associated with defective modulation of intestinal immune response. While the intestinal mucosa is usually normal in enteropathies related to congenital transport or enzyme deficiencies, the intestinal biopsy in other disorders may reveal a wide range of abnormalities varying from normal villous architecture to villous atrophy and/or inflammation, or features specific to the underlying disorder including epithelial abnormalities, lipid vacuolization in the enterocytes, absence of plasma cells, lymphangiectasia, microorganisms, and mucosal eosinophilic or histiocytic infiltration. This review intends to provide an update on small intestinal biopsy findings in childhood enteropathies, the "newcomers", including very early onset monogenic inflammatory bowel disease (IBD), in particular, for the practicing pathologist.
Collapse
Affiliation(s)
- Arzu Ensari
- Department of Pathology, Ankara University Medical School, Sihhiye, 06100, Ankara, Turkey.
| | - Judith Kelsen
- Division of Gastroenterology, Hepatology and Nutrition, The Children's Hospital of Philadelphia, Perelman School at the University of Pennsylvania, 3401 Civic Center Boulevard, 5 NW26, Philadelphia, PA, 19104, USA
| | - Pierre Russo
- Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia, Perelman School of Medicine at The University of Pennsylvania, 3401 Civic Center Boulevard, 5 NW26, Philadelphia, PA, 19104, USA
| |
Collapse
|
42
|
Tan QKG, Cardona DM, Rehder CW, McDonald MT. Identification of EPCAM mutation: clinical use of microarray. Clin Case Rep 2017; 5:980-985. [PMID: 28588851 PMCID: PMC5457984 DOI: 10.1002/ccr3.914] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 01/30/2017] [Accepted: 03/02/2017] [Indexed: 11/30/2022] Open
Abstract
We report a case of an infant with congenital tufting enteropathy (CTE) who presented with severe failure to thrive despite multiple interventions. This study illustrates that CTE may be missed by endoscopy, and the use of chromosomal microarray and immunohistological analysis may be integral to diagnosis.
Collapse
Affiliation(s)
- Queenie K-G Tan
- Division of Medical Genetics Department of Pediatrics Duke University Health System Durham North Carolina
| | - Diana M Cardona
- Department of Pathology Duke University Health System Durham North Carolina
| | - Catherine W Rehder
- Department of Pathology Duke University Health System Durham North Carolina
| | - Marie T McDonald
- Division of Medical Genetics Department of Pediatrics Duke University Health System Durham North Carolina
| |
Collapse
|
43
|
Salomon J, Gaston C, Magescas J, Duvauchelle B, Canioni D, Sengmanivong L, Mayeux A, Michaux G, Campeotto F, Lemale J, Viala J, Poirier F, Minc N, Schmitz J, Brousse N, Ladoux B, Goulet O, Delacour D. Contractile forces at tricellular contacts modulate epithelial organization and monolayer integrity. Nat Commun 2017; 8:13998. [PMID: 28084299 PMCID: PMC5241865 DOI: 10.1038/ncomms13998] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 11/17/2016] [Indexed: 12/31/2022] Open
Abstract
Monolayered epithelia are composed of tight cell assemblies that ensure polarized exchanges. EpCAM, an unconventional epithelial-specific cell adhesion molecule, is assumed to modulate epithelial morphogenesis in animal models, but little is known regarding its cellular functions. Inspired by the characterization of cellular defects in a rare EpCAM-related human intestinal disease, we find that the absence of EpCAM in enterocytes results in an aberrant apical domain. In the course of this pathological state, apical translocation towards tricellular contacts (TCs) occurs with striking tight junction belt displacement. These unusual cell organization and intestinal tissue defects are driven by the loss of actomyosin network homoeostasis and contractile activity clustering at TCs, yet is reversed by myosin-II inhibitor treatment. This study reveals that adequate distribution of cortical tension is crucial for individual cell organization, but also for epithelial monolayer maintenance. Our data suggest that EpCAM modulation protects against epithelial dysplasia and stabilizes human tissue architecture.
Collapse
Affiliation(s)
- Julie Salomon
- Cell Adhesion and Mechanics, Institut Jacques Monod, CNRS UMR7592, Paris Diderot University, 75205 Paris, France.,Department of Paediatric Gastroenterology, Hôpital Necker-Enfants Malades, Sorbonne Paris Cité, 75015 Paris, France
| | - Cécile Gaston
- Cell Adhesion and Mechanics, Institut Jacques Monod, CNRS UMR7592, Paris Diderot University, 75205 Paris, France
| | - Jérémy Magescas
- Cell Adhesion and Mechanics, Institut Jacques Monod, CNRS UMR7592, Paris Diderot University, 75205 Paris, France
| | - Boris Duvauchelle
- Morphogenesis, Homoeostasis and Pathologies, Institut Jacques Monod, CNRS UMR7592, Paris Diderot University, 75013 Paris, France
| | - Danielle Canioni
- Department of Paediatric Anatomo-Pathology, Hôpital Necker-Enfants Malades, Sorbonne Paris Cité, 75015 Paris, France
| | - Lucie Sengmanivong
- Membrane Dynamics and Mechanics of Intracellular Signaling Laboratory, Institut Curie-Centre de Recherche, PSL Research University, 75005 Paris, France
| | - Adeline Mayeux
- Cell Adhesion and Mechanics, Institut Jacques Monod, CNRS UMR7592, Paris Diderot University, 75205 Paris, France
| | - Grégoire Michaux
- Institut de Génétique et Développement de Rennes, CNRS UMR6290, 35000 Rennes, France
| | - Florence Campeotto
- Department of Paediatric Gastroenterology, Hôpital Necker-Enfants Malades, Sorbonne Paris Cité, 75015 Paris, France.,Laboratoire de Microbiologie EA 4065, Faculté de Pharmacie, Université Paris Descartes, 75005 Paris, France
| | - Julie Lemale
- Department of Pediatric Nutrition and Gastroenterology, Armand-Trousseau Hospital, Assistance Publique-Hôpitaux de Paris, Institute of Cardiometabolism and Nutrition, Pierre et Marie Curie University, 75012 Paris, France
| | - Jérôme Viala
- Department of Pediatric Gastroenterology, Assistance Publique-Hôpitaux de Paris, Robert Debré Hospital, Université Paris Diderot, Sorbonne Paris Cité, UMR843, 75019 Paris, France
| | - Françoise Poirier
- Morphogenesis, Homoeostasis and Pathologies, Institut Jacques Monod, CNRS UMR7592, Paris Diderot University, 75013 Paris, France
| | - Nicolas Minc
- Cellular Spatial Organization, Institut Jacques Monod, CNRS UMR7592, Paris Diderot University, 75205 Paris, France
| | - Jacques Schmitz
- Department of Paediatric Gastroenterology, Hôpital Necker-Enfants Malades, Sorbonne Paris Cité, 75015 Paris, France
| | - Nicole Brousse
- Department of Paediatric Anatomo-Pathology, Hôpital Necker-Enfants Malades, Sorbonne Paris Cité, 75015 Paris, France
| | - Benoit Ladoux
- Cell Adhesion and Mechanics, Institut Jacques Monod, CNRS UMR7592, Paris Diderot University, 75205 Paris, France.,Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Olivier Goulet
- Department of Paediatric Gastroenterology, Hôpital Necker-Enfants Malades, Sorbonne Paris Cité, 75015 Paris, France
| | - Delphine Delacour
- Cell Adhesion and Mechanics, Institut Jacques Monod, CNRS UMR7592, Paris Diderot University, 75205 Paris, France
| |
Collapse
|
44
|
Kammermeier J, Dziubak R, Pescarin M, Drury S, Godwin H, Reeve K, Chadokufa S, Huggett B, Sider S, James C, Acton N, Cernat E, Gasparetto M, Noble-Jamieson G, Kiparissi F, Elawad M, Beales PL, Sebire NJ, Gilmour K, Uhlig HH, Bacchelli C, Shah N. Phenotypic and Genotypic Characterisation of Inflammatory Bowel Disease Presenting Before the Age of 2 years. J Crohns Colitis 2017; 11:60-69. [PMID: 27302973 PMCID: PMC5885808 DOI: 10.1093/ecco-jcc/jjw118] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Inflammatory bowel disease [IBD] presenting in early childhood is extremely rare. More recently, progress has been made to identify children with monogenic forms of IBD predominantly presenting very early in life. In this study, we describe the heterogeneous phenotypes and genotypes of patients with IBD presenting before the age of 2 years and establish phenotypic features associated with underlying monogenicity. METHODS Phenotype data of 62 children with disease onset before the age of 2 years presenting over the past 20 years were reviewed. Children without previously established genetic diagnosis were prospectively recruited for next-generation sequencing. RESULTS In all, 62 patients [55% male] were identified. The median disease onset was 3 months of age (interquartile range [IQR]: 1 to 11). Conventional IBD classification only applied to 15 patients with Crohn's disease [CD]-like [24%] and three with ulcerative colitis [UC]-like [5%] phenotype; 44 patients [71%] were diagnosed with otherwise unclassifiable IBD. Patients frequently required parenteral nutrition [40%], extensive immunosuppression [31%], haematopoietic stem-cell transplantation [29%], and abdominal surgery [19%]. In 31% of patients, underlying monogenic diseases were established [EPCAM, IL10, IL10RA, IL10RB, FOXP3, LRBA, SKIV2L, TTC37, TTC7A]. Phenotypic features significantly more prevalent in monogenic IBD were: consanguinity, disease onset before the 6th month of life, stunting, extensive intestinal disease and histological evidence of epithelial abnormalities. CONCLUSIONS IBD in children with disease onset before the age of 2 years is frequently unclassifiable into Crohn's disease and ulcerative colitis, particularly treatment resistant, and can be indistinguishable from monogenic diseases with IBD-like phenotype.
Collapse
Affiliation(s)
- Jochen Kammermeier
- Genetics and Genomic Medicine, Institute of Child Health, University College London, London, UK,Department of Gastroenterology, Great Ormond Street Hospital, London, UK
| | - Robert Dziubak
- Department of Gastroenterology, Great Ormond Street Hospital, London, UK
| | - Matilde Pescarin
- Department of Gastroenterology, Great Ormond Street Hospital, London, UK
| | - Suzanne Drury
- Genetics and Genomic Medicine, Institute of Child Health, University College London, London, UK,NE Thames Regional Genetics Laboratory, Great Ormond Street Hospital, London, UK
| | - Heather Godwin
- Department of Gastroenterology, Great Ormond Street Hospital, London, UK
| | - Kate Reeve
- Department of Gastroenterology, Great Ormond Street Hospital, London, UK
| | | | - Bonita Huggett
- Department of Gastroenterology, Great Ormond Street Hospital, London, UK
| | - Sara Sider
- Department of Gastroenterology, Great Ormond Street Hospital, London, UK
| | - Chela James
- Genetics and Genomic Medicine, Institute of Child Health, University College London, London, UK
| | - Nikki Acton
- Department of Gastroenterology, Great Ormond Street Hospital, London, UK
| | - Elena Cernat
- Department of Gastroenterology, Great Ormond Street Hospital, London, UK
| | - Marco Gasparetto
- Department of Paediatric Gastroenterology, Addenbrookes Hospital, Cambridge, UK
| | - Gabi Noble-Jamieson
- Department of Paediatric Gastroenterology, Addenbrookes Hospital, Cambridge, UK
| | - Fevronia Kiparissi
- Department of Gastroenterology, Great Ormond Street Hospital, London, UK
| | - Mamoun Elawad
- Department of Gastroenterology, Great Ormond Street Hospital, London, UK
| | - Phil L. Beales
- Genetics and Genomic Medicine, Institute of Child Health, University College London, London, UK
| | - Neil J. Sebire
- Department of Histopathology, Great Ormond Street Hospital, London, UK
| | - Kimberly Gilmour
- Department of Immunology, Great Ormond Street Hospital, London, UK
| | - Holm H. Uhlig
- Transitional Gastroenterology Unit, Nuffield Department of Medicine and Department of Paediatrics, University of Oxford, UK
| | - Chiara Bacchelli
- Genetics and Genomic Medicine, Institute of Child Health, University College London, London, UK
| | - Neil Shah
- Department of Gastroenterology, Great Ormond Street Hospital, London, UK
| |
Collapse
|
45
|
Abstract
BACKGROUND/AIM Tufting enteropathy (TE) is a rare cause of congenital intractable diarrhea in children. It often results in an irreversible intestinal failure and total parenteral nutrition (TPN) dependency; eventually, intestinal transplantation may be necessary. Data on TE from the Middle East are scarce; therefore, our aim of conducting this study was to clarify the clinical, histopathologic, and molecular features of TE in Saudi children. PATIENTS AND METHODS This was a retrospective chart review of four children with TE who presented between January 2011 and December 2013 to King Fahad Specialist Hospital-Dammam (KFSH-D). The diagnosis of TE was suspected based on characteristic histopathologic intestinal biopsy findings and confirmed by EpCAM gene testing. RESULTS Molecular testing identified two novel mutations in the EpCAM gene in our patients. These mutations were associated with severe phenotype of the disease characterized by very early onset (median of 2 weeks of life), TPN dependency, and death during early childhood. Two patients died due to central line-related complications. Two patients were referred for intestinal transplantation due to loss of intravenous access in one and progressive liver disease in the other. CONCLUSION Mutations in EpCAM gene in Saudi children are characterized by severe phenotype and poor outcome.
Collapse
Affiliation(s)
- Shaden AlMahamed
- Department of Pediatrics, Division of Pediatric Gastroenterology and Hepatology, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Abdelhai Hammo
- Department of Pediatrics, Division of Pediatric Gastroenterology and Hepatology, King Fahad Specialist Hospital, Dammam, Saudi Arabia,Address for correspondence: Dr. Abdelhai Hammo, Department of Pediatrics, Division of Pediatric Gastroenterology and Hepatology, King Fahad Specialist Hospital, Dammam - 314444, Saudi Arabia. E-mail:
| |
Collapse
|
46
|
Overeem AW, Posovszky C, Rings EHMM, Giepmans BNG, van IJzendoorn SCD. The role of enterocyte defects in the pathogenesis of congenital diarrheal disorders. Dis Model Mech 2016; 9:1-12. [PMID: 26747865 PMCID: PMC4728335 DOI: 10.1242/dmm.022269] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Congenital diarrheal disorders are rare, often fatal, diseases that are difficult to diagnose (often requiring biopsies) and that manifest in the first few weeks of life as chronic diarrhea and the malabsorption of nutrients. The etiology of congenital diarrheal disorders is diverse, but several are associated with defects in the predominant intestinal epithelial cell type, enterocytes. These particular congenital diarrheal disorders (CDDENT) include microvillus inclusion disease and congenital tufting enteropathy, and can feature in other diseases, such as hemophagocytic lymphohistiocytosis type 5 and trichohepatoenteric syndrome. Treatment options for most of these disorders are limited and an improved understanding of their molecular bases could help to drive the development of better therapies. Recently, mutations in genes that are involved in normal intestinal epithelial physiology have been associated with different CDDENT. Here, we review recent progress in understanding the cellular mechanisms of CDDENT. We highlight the potential of animal models and patient-specific stem-cell-based organoid cultures, as well as patient registries, to integrate basic and clinical research, with the aim of clarifying the pathogenesis of CDDENT and expediting the discovery of novel therapeutic strategies. Summary: Overview of the recent progress in our understanding of congenital diarrheal disorders, and the available models to study these diseases.
Collapse
Affiliation(s)
- Arend W Overeem
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Carsten Posovszky
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, 89075 Ulm, Germany
| | - Edmond H M M Rings
- Department of Pediatrics, Erasmus Medical Center Rotterdam, Erasmus University Rotterdam, 3000 CB Rotterdam, The Netherlands Department of Pediatrics, Leiden University Medical Center, Leiden University, 2300 RC Leiden, The Netherlands
| | - Ben N G Giepmans
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Sven C D van IJzendoorn
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| |
Collapse
|
47
|
Azzopardi C, Pullicino E, Coleiro B, Galea Soler S. Congenital tufting enteropathy and chronic arthritis: a clinical and radiological perspective. BMJ Case Rep 2016; 2016:bcr-2016-215252. [PMID: 27558188 DOI: 10.1136/bcr-2016-215252] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Congenital tufting enteropathy is a rare condition which presents in early infancy. It is a condition which should be suspected in infants who present with diarrhoea soon after birth. A rare association with arthritis has been observed with a handful of cases documented in the literature. Our case differs as the arthritis described is erosive in nature, a feature which is not present in other cases.
Collapse
Affiliation(s)
| | - Edgar Pullicino
- Department of Gastroenterology, Mater Dei Hospital, Msida, Malta
| | | | | |
Collapse
|
48
|
Genetic analysis of Italian patients with congenital tufting enteropathy. World J Pediatr 2016; 12:219-24. [PMID: 26684320 DOI: 10.1007/s12519-015-0070-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 12/02/2014] [Indexed: 10/22/2022]
Abstract
BACKGROUND Congenital tufting enteropathy (CTE), an inherited autosomal recessive rare disease, is a severe diarrhea of infancy which is clinically characterized by absence of inflammation and presence of intestinal villous atrophy. Mutations in the EpCAM gene were identified to cause CTE. Recent cases of syndromic tufting enteropathy harboring the SPINT2 (19q13.2) mutation were described. METHODS Four CTE Italian patients were clinically and immunohistochemically characterized. Direct DNA sequencing of EpCAM and SPINT2 genes was performed. RESULTS All patients were of Italian origin. Three different mutations were detected (p.Asp219Metfs*15, Tyr186Phefs*6 and p.Ile146Asn) in the EpCAM gene; one of them is novel (p.Ile146Asn). Two patients (P1 and P2) showed compound heterozygosity revealing two mutations in separate alleles. A third patient (P3) was heterozygous for only one novel EpCAM missense mutation (p.Ile146Asn). In a syndromic patient (P4), no deleterious EpCAM mutation was found. Additional SPINT2 mutational analysis was performed. P4 showed a homozygous SPINT2 mutation (p.Y163C). No SPINT2 mutation was found in P3. CLDN7 was also evaluated as a candidate gene by mutational screening in P3 but no mutation was identified. CONCLUSION This study presented a molecular characterization of CTE Italian patients, and identified three mutations in the EpCAM gene and one in the SPINT2 gene. One of EpCAM mutations was novel, therefore increasing the mutational spectrum of allelic variants of the EpCAM gene. Molecular analysis of the SPINT2 gene also allowed us to identify a SPINT2 substitution mutation (c.488A>G) recently found to be associated with syndromic CTE subjects.
Collapse
|
49
|
Congenital intestinal diarrhoeal diseases: A diagnostic and therapeutic challenge. Best Pract Res Clin Gastroenterol 2016; 30:187-211. [PMID: 27086885 DOI: 10.1016/j.bpg.2016.03.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 03/05/2016] [Indexed: 01/31/2023]
Abstract
Congenital diarrhoeal disorders are a heterogeneous group of inherited malabsorptive or secretory diseases typically appearing in the first weeks of life, which may be triggered by the introduction of distinct nutrients. However, they may also be unrecognised for a while and triggered by exogenous factors later on. In principle, they can be clinically classified as osmotic, secretory or inflammatory diarrhoea. In recent years the disease-causing molecular defects of these congenital disorders have been identified. According to the underlying pathophysiology they can be classified into four main groups: 1) Defects of digestion, absorption and transport of nutrients or electrolytes 2) Defects of absorptive enterocyte differentiation or polarisation 3) Defects of the enteroendocrine cells 4) Defects of the immune system affecting the intestine. Here, we describe the clinical presentation of congenital intestinal diarrhoeal diseases, the diagnostic work-up and specific treatment aspects.
Collapse
|
50
|
Weaning Off Prognosis Factors of Home Parenteral Nutrition for Children With Primary Digestive Disease. J Pediatr Gastroenterol Nutr 2016; 62:462-8. [PMID: 26398153 DOI: 10.1097/mpg.0000000000000980] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
OBJECTIVES The aim of the present study was to describe the indications for home parenteral nutrition (HPN) in children with primary digestive diseases and to identify factors associated with weaning off. METHODS All the children initially discharged on HPN between January 1, 2000, and December 31, 2009, for chronic intestinal failure (IF) were included. The associations between clinical factors and weaning off of HPN were assessed using a multivariable Cox regression model. RESULTS Among the 151 children (boys = 58%) included in this study, 98 (65%) presented with short bowel syndrome (SBS), 17 (11%) with digestive neuromuscular disorders, 14 (9%) with mucosal diseases, 13 (9%) with inflammatory bowel disease, and 9 (6%) with other primary digestive diseases. The probability of survival was ∼100%. At the end of the follow-up, the probability for weaning off of HPN was 0.73 (95% confidence interval 0.54-0.84) but varied according to the underlying cause of IF (for example, SBS and inflammatory bowel disease had a better prognosis). The median time until weaning off was 21 months (95% confidence interval 18-38 months). Unfavourable prognostic factors for weaning off of HPN included a bowel remnant of <40 cm, the presence of <50% of the colon, and daily lipid intakes >1.5 g · kg · day. Underlying disease was also associated with weaning off. CONCLUSIONS HPN is a safe therapeutic option for children with chronic IF requiring long-term nutritional management. Prognostic factors for weaning off of HPN were identified, and they highlight the relevance of SBS anatomy and parenteral nutrition caloric intake. The outcome of children on HPN was primarily dependent on the underlying disease.
Collapse
|