1
|
Ravi N, Kumar S, Ramachandran A. Prenatal Diagnosis of ACTG2-Related Megacystis-Microcolon-Intestinal Hypoperistalsis Syndrome-Case Report and Systematic Review. J Clin Med 2025; 14:3204. [PMID: 40364235 PMCID: PMC12072778 DOI: 10.3390/jcm14093204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/26/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Background/Objectives: Megacystis-microcolon-intestinal hypoperistalsis syndrome (MMIHS) is characterized by smooth muscle dysfunction and results in severe bladder dilatation and intestinal dysmotility. Prenatal diagnosis is challenging due to the non-specific nature of ultrasound findings and the limitations of current genetic testing. We present a case of persistent fetal megacystis, with genetic testing confirming MMIHS, and a systematic review of prenatally diagnosed cases. Methods: An electronic search of the PubMed, Medline, Web of Science and CORE databases was conducted to identify reports of genetic prenatal diagnoses of MMIHS. The inclusion criteria were cases of prenatally suspected MMIHS with a genetic diagnosis based on prenatal samples. Reports that described neonatal or paediatric cases or lacked clinical details or genetic testing results were excluded, and the clinical details for the included cases were collected. Results: We identified six publications describing 17 cases of MMIHS confirmed on genetic testing. Including our case, 18 cases are described in this manuscript. Most cases (72.2%) presented in the second or third trimester of pregnancy; the majority (55.6%) were due to ACTG2 mutations. All cases had fetal megacystis detected on ultrasound. Five cases (27.8%) also had a known family history of MMIHS. The majority of the cases (77.8%) resulted in the termination of pregnancy. Conclusions: MMIHS is a rare condition with significant morbidity and mortality and prenatal diagnosis remains challenging. ACTG2 mutations are described in over half of these cases. These data contribute to the limited literature on its prenatal presentation and the evolving role of prenatal molecular genetic testing.
Collapse
Affiliation(s)
- Neha Ravi
- Centre for Maternal and Fetal Medicine, Mater Mothers’ Hospital, Brisbane 4101, Australia
| | - Sailesh Kumar
- Centre for Maternal and Fetal Medicine, Mater Mothers’ Hospital, Brisbane 4101, Australia
- Mater Research Institute, The University of Queensland, Brisbane 4101, Australia
- School of Medicine, The University of Queensland, Brisbane 4072, Australia
| | - Aparna Ramachandran
- Centre for Maternal and Fetal Medicine, Mater Mothers’ Hospital, Brisbane 4101, Australia
| |
Collapse
|
2
|
Yu Y, Li J, Wang X, Li X, Lyu C, Yang L, Bai Y. Actin Gamma Smooth Muscle 2 Promotes Epithelial Ovarian Cancer Cell Proliferation via the AKT1/NF-κB Signaling Pathway. Cell Biochem Funct 2025; 43:e70077. [PMID: 40289545 DOI: 10.1002/cbf.70077] [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: 10/07/2024] [Revised: 02/15/2025] [Accepted: 04/11/2025] [Indexed: 04/30/2025]
Abstract
Epithelial ovarian cancer (EOC) is associated with high mortality rates worldwide and is characterized as the most lethal gynecological cancer. The study aimed to investigate the functional role and underlying molecular mechanism of actin gamma smooth muscle 2 (ACTG2) in the progression of EOC. Data mining from The Cancer Genome Atlas (TCGA) databases showed the expression of ACTG2 was significantly upregulated in EOC and negatively associated with longer overall survival and better prognosis of patients. By using of gain-of-function and loss-of-function experiments in vitro and in vivo, we found that ACTG2 promoted EOC cell proliferation and suppressed cell apoptosis. Mechanistic study revealed that ACTG2 regulates EOC cell proliferation by activating the AKT serine/threonine kinase 1 (AKT1)/nuclear factor-κB (NF-κB) signaling pathway. Importantly, p65 plays a crucial role in this newly identified regulatory mechanism. Our findings demonstrate that ACTG2 may play an oncogenic role in EOC, suggesting its potential as a therapeutic target.
Collapse
Affiliation(s)
- Yinjue Yu
- Department of Radiotherapy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Central Laboratory, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Jiangxia Li
- Central Laboratory, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Xiaohang Wang
- Department of Radiotherapy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoxiao Li
- Department of Anesthesiology, Zhongshan Hospital Fudan University, Shanghai, China
| | - Cuiting Lyu
- Central Laboratory, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Lina Yang
- Central Laboratory, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Yongrui Bai
- Department of Radiotherapy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
3
|
Kaenkumchorn TK, Lampone O, Huebner K, Cramer J, Karls C. When parenteral nutrition is the answer: The case of pediatric intestinal rehabilitation. Nutr Clin Pract 2024; 39:991-1002. [PMID: 38961658 DOI: 10.1002/ncp.11179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/09/2024] [Accepted: 06/03/2024] [Indexed: 07/05/2024] Open
Abstract
In pediatric patients with intestinal failure, parenteral nutrition is lifesaving but also has several associated risks. The goals of intestinal rehabilitation include promoting growth, minimizing complications associated with intestinal failure, and reaching enteral autonomy, if possible. Pediatric intestinal rehabilitation programs are interdisciplinary teams that strive to provide optimal care for children dependent on parenteral nutrition. The provision of parenteral nutrition requires close monitoring of patients' growth, nutrition concerns, clinical status, and laboratory parameters. Recent advances in the field of intestinal rehabilitation include new lipid emulsions, considerations regarding enteral feeding, advances in micronutrient provision, and central venous catheter preservation techniques. Challenges in the field remain, including improving overall quality of life with home parenteral nutrition administration and preventing recently recognized complications such as chronic intestinal inflammation.
Collapse
Affiliation(s)
- Tanyaporn K Kaenkumchorn
- Division of Gastroenterology, Hepatology, and Nutrition, Children's Wisconsin, Milwaukee, Wisconsin, USA
| | - Olivia Lampone
- Clinical Nutrition, Children's Wisconsin, Milwaukee, Wisconsin, USA
| | - Kayla Huebner
- Department of Pharmacy, Children's Wisconsin, Milwaukee, Wisconsin, USA
| | - Jesse Cramer
- Department of Pharmacy, Children's Wisconsin, Milwaukee, Wisconsin, USA
| | - Catherine Karls
- Clinical Nutrition, Children's Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
4
|
Choi Y, Park H, Kim J, Lee H, Kim M. Heat Stress Induces Alterations in Gene Expression of Actin Cytoskeleton and Filament of Cellular Components Causing Gut Disruption in Growing-Finishing Pigs. Animals (Basel) 2024; 14:2476. [PMID: 39272260 PMCID: PMC11394201 DOI: 10.3390/ani14172476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/17/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
We aimed to investigate the impact of heat stress (HS) on the expression of tight junction (TJ) proteins and the interaction between genes affecting intestinal barrier function using transcriptomics in the porcine jejunum. Twenty-four barrows (crossbred Yorkshire × Landrace × Duroc; average initial body weight, 56.71 ± 1.74 kg) were placed in different temperatures (normal temperature [NT]; HS) and reared for 56 days. At the end of the experiment, jejunal samples were collected from three pigs per treatment for transcriptome and reverse-transcription quantitative polymerase chain reaction (RT-qPCR) analyses. We identified 43 differentially expressed genes, involving five Kyoto Encyclopedia of Genes and Genomes pathways, eight molecular functions, seven cellular components (CCs), and nine biological processes, using gene ontology enrichment analysis. Genes associated with the actin cytoskeleton, filament-binding pathways, and TJ proteins were selected and analyzed by RT-qPCR. Significant differences in relative mRNA expression showed that downregulated genes in the HS group included ZO1, CLDN1, OCLN, PCK1, and PCK2, whereas ACTG2, DES, MYL9, MYLK, TPM1, TPM2, CNN1, PDLIM3, and PCP4 were upregulated by HS (p < 0.05). These findings indicate that HS in growing-finishing pigs induces depression in gut integrity, which may be related to genes involved in the actin cytoskeleton and filaments of CC.
Collapse
Affiliation(s)
- Yohan Choi
- Swine Science Division, National Institute of Animal Science, Rural Development Administration, Cheonan 31000, Republic of Korea
| | - Hyunju Park
- Swine Science Division, National Institute of Animal Science, Rural Development Administration, Cheonan 31000, Republic of Korea
| | - Joeun Kim
- Swine Science Division, National Institute of Animal Science, Rural Development Administration, Cheonan 31000, Republic of Korea
| | - Hyunseo Lee
- School of Animal Life Convergence Science, Hankyong National University, Anseong 17579, Republic of Korea
| | - Minju Kim
- School of Animal Life Convergence Science, Hankyong National University, Anseong 17579, Republic of Korea
- Institute of Applied Humanimal Science, Hankyong National University, Anseong 17579, Republic of Korea
| |
Collapse
|
5
|
Devavarapu PKV, Uppaluri KR, Nikhade VA, Palasamudram K, Sri Manjari K. Exploring the complexities of megacystis-microcolon-intestinal hypoperistalsis syndrome: insights from genetic studies. Clin J Gastroenterol 2024; 17:383-395. [PMID: 38461165 DOI: 10.1007/s12328-024-01934-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 01/30/2024] [Indexed: 03/11/2024]
Abstract
Megacystis microcolon intestinal hypoperistalsis syndrome (MMIHS) is an uncommon genetic disorder inherited in an autosomal recessive pattern that affects the muscles that line the bladder and intestines. The most common genes associated with MMIHS mutations are ACTG2, LMOD1, MYH11, MYL9, MYLK, and PDCL3. However, the complete genetic landscape of MMIHS still needs to be fully understood. The diagnosis of MMIHS can be challenging. However, advances in prenatal and diagnostic techniques, such as ultrasound and fetal urine analysis, have improved the ability to detect the syndrome early. Targeted next-generation sequencing (NGS) and other diagnostic tests can also diagnose MMIHS. The management of MMIHS involves addressing severe intestinal dysmotility, which often necessitates total parenteral nutrition (TPN), which can lead to complications such as hepatotoxicity and nutritional deficiencies. Multivisceral and intestinal transplantation has emerged as therapeutic options, offering the potential for improved outcomes and enteral autonomy. Understanding the genetic underpinnings of MMIHS is crucial for personalized care. While the prognosis varies, timely interventions and careful monitoring enhance patient outcomes. Genetic studies have given us valuable insights into the molecular mechanisms of MMIHS. These studies have identified mutations in genes involved in the development and function of smooth muscle cells. They have also shown that MMIHS is associated with defects in the signaling pathways that control muscle contraction. Continued research in the genetics of MMIHS holds promise for unraveling the complexities of MMIHS and improving the lives of affected individuals.
Collapse
Affiliation(s)
- Prasad K V Devavarapu
- Department of Biochemistry, GITAM Institute of Medical Sciences & Research, Rushikonda, Visakhapatnam, Andhra Pradesh, 530045, India
| | - Kalyan Ram Uppaluri
- GenepoweRx, Suit #2B, Plot No. 240, Nirvana, Road No. 36, Jawahar Colony, Jubilee Hills, Hyderabad, Telangana, 500033, India
| | - Vrushabh Anil Nikhade
- KIT's College of Engineering (Autonomous), Kolhapur, Maharashtra, 416234, India
- GenepoweRx, Suit #2B, Plot No. 240, Nirvana, Road No. 36, Jawahar Colony, Jubilee Hills, Hyderabad, Telangana, 500033, India
| | - Kalyani Palasamudram
- GenepoweRx, Suit #2B, Plot No. 240, Nirvana, Road No. 36, Jawahar Colony, Jubilee Hills, Hyderabad, Telangana, 500033, India
| | - Kavutharapu Sri Manjari
- GenepoweRx, Suit #2B, Plot No. 240, Nirvana, Road No. 36, Jawahar Colony, Jubilee Hills, Hyderabad, Telangana, 500033, India.
| |
Collapse
|
6
|
Ceron RH, Báez-Cruz FA, Palmer NJ, Carman PJ, Boczkowska M, Heuckeroth RO, Ostap EM, Dominguez R. Molecular mechanisms linking missense ACTG2 mutations to visceral myopathy. SCIENCE ADVANCES 2024; 10:eadn6615. [PMID: 38820162 PMCID: PMC11141634 DOI: 10.1126/sciadv.adn6615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 04/29/2024] [Indexed: 06/02/2024]
Abstract
Visceral myopathy is a life-threatening disease characterized by muscle weakness in the bowel, bladder, and uterus. Mutations in smooth muscle γ-actin (ACTG2) are the most common cause of the disease, but the mechanisms by which the mutations alter muscle function are unknown. Here, we examined four prevalent ACTG2 mutations (R40C, R148C, R178C, and R257C) that cause different disease severity and are spread throughout the actin fold. R178C displayed premature degradation, R148C disrupted interactions with actin-binding proteins, R40C inhibited polymerization, and R257C destabilized filaments. Because these mutations are heterozygous, we also analyzed 50/50 mixtures with wild-type (WT) ACTG2. The WT/R40C mixture impaired filament nucleation by leiomodin 1, and WT/R257C produced filaments that were easily fragmented by smooth muscle myosin. Smooth muscle tropomyosin isoform Tpm1.4 partially rescued the defects of R40C and R257C. Cryo-electron microscopy structures of filaments formed by R40C and R257C revealed disrupted intersubunit contacts. The biochemical and structural properties of the mutants correlate with their genotype-specific disease severity.
Collapse
Affiliation(s)
- Rachel H. Ceron
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- The Children’s Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, USA
| | - Faviolla A. Báez-Cruz
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicholas J. Palmer
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Peter J. Carman
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Malgorzata Boczkowska
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert O. Heuckeroth
- The Children’s Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - E. Michael Ostap
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Roberto Dominguez
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
7
|
Billon C, Piccoli GB, de Sainte Agathe JM, Stoeva R, Derive N, Heidet L, Berrebi D, Bruneval P, Jeunemaitre X, Hureaux M. Genome-wide analysis identifies MYH11 compound heterozygous variants leading to visceral myopathy corresponding to late-onset form of megacystis-microcolon-intestinal hypoperistalsis syndrome. Mol Genet Genomics 2024; 299:44. [PMID: 38625590 DOI: 10.1007/s00438-024-02136-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 03/26/2024] [Indexed: 04/17/2024]
Abstract
Megacystis-microcolon-hypoperistalsis-syndrome (MMIHS) is a rare and early-onset congenital disease characterized by massive abdominal distension due to a large non-obstructive bladder, a microcolon and decreased or absent intestinal peristalsis. While in most cases inheritance is autosomal dominant and associated with heterozygous variant in ACTG2 gene, an autosomal recessive transmission has also been described including pathogenic bialellic loss-of-function variants in MYH11. We report here a novel family with visceral myopathy related to MYH11 gene, confirmed by whole genome sequencing (WGS). WGS was performed in two siblings with unusual presentation of MMIHS and their two healthy parents. The 38 years-old brother had severe bladder dysfunction and intestinal obstruction, whereas the 30 years-old sister suffered from end-stage kidney disease with neurogenic bladder and recurrent sigmoid volvulus. WGS was completed by retrospective digestive pathological analyses. Compound heterozygous variants of MYH11 gene were identified, associating a deletion of 1.2 Mb encompassing MYH11 inherited from the father and an in-frame variant c.2578_2580del, p.Glu860del inherited from the mother. Pathology analyses of the colon and the rectum revealed structural changes which significance of which is discussed. Cardiac and vascular assessment of the mother was normal. This is the second report of a visceral myopathy corresponding to late-onset form of MMIHS related to compound heterozygosity in MYH11; with complete gene deletion and a hypomorphic allele in trans. The hypomorphic allele harbored by the mother raised the question of the risk of aortic disease in adults. This case shows the interest of WGS in deciphering complex phenotypes, allowing adapted diagnosis and genetic counselling.
Collapse
Affiliation(s)
- Clarisse Billon
- Université Paris Cité, Paris, France
- Service de Médecine Génomique des Maladies Rares, Groupe Hospitalier Universitaire Centre, Assistance Publique Hôpitaux de Paris, 75015, Paris, France
| | | | - Jean-Madeleine de Sainte Agathe
- Laboratoire de Biologie Médicale MultiSites SeqOIA, Paris, France
- Département de Génétique Médicale, Groupe Hospitalier Universitaire Pitié Salpêtrière, Assistance Publique Hôpitaux de Paris, Sorbonne Université, Paris, France
| | - Radka Stoeva
- Service de Génétique Médicale, Centre Hospitalier du Mans, Le Mans, France
| | - Nicolas Derive
- Laboratoire de Biologie Médicale MultiSites SeqOIA, Paris, France
| | - Laurence Heidet
- Centre de référence des Maladies Rénales Héréditaires de L'Enfant Et de L'Adulte, MARHEA, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, 75015, Paris, France
- Service de Néphrologie Pédiatrique, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, 75015, Paris, France
| | - Dominique Berrebi
- Université Paris Cité, Paris, France
- Service de Pathologie, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, 75015, Paris, France
| | - Patrick Bruneval
- Université Paris Cité, Paris, France
- Service de Cardiologie, Hôpital Européen Georges Pompidou, Assistance Publique Hôpitaux de Paris, 75015, Paris, France
| | - Xavier Jeunemaitre
- Université Paris Cité, Paris, France
- Service de Médecine Génomique des Maladies Rares, Groupe Hospitalier Universitaire Centre, Assistance Publique Hôpitaux de Paris, 75015, Paris, France
| | - Marguerite Hureaux
- Université Paris Cité, Paris, France.
- Service de Médecine Génomique des Maladies Rares, Groupe Hospitalier Universitaire Centre, Assistance Publique Hôpitaux de Paris, 75015, Paris, France.
- Centre de référence des Maladies Rénales Héréditaires de L'Enfant Et de L'Adulte, MARHEA, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, 75015, Paris, France.
- INSERM, PARCC U970, 75015, Paris, France.
| |
Collapse
|
8
|
Mejia M, Royero Arias M, Pimiento Figueroa J, Romero Espitia W. Megacystis-Microcolon-Intestinal Hypoperistalsis Syndrome: A Case Report of an Uncommon Condition. Cureus 2024; 16:e54255. [PMID: 38496087 PMCID: PMC10944333 DOI: 10.7759/cureus.54255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2024] [Indexed: 03/19/2024] Open
Abstract
The megacystis-microcolon-intestinal hypoperistalsis syndrome (MMIHS), also known as Berdon syndrome, is a rare congenital condition that falls within the spectrum of visceral myopathies. It is characterized by the presence of megacystis, microcolon, and hypoperistalsis, which are secondary to gastrointestinal and urinary system dysmotility. It is frequently associated with other alterations in the gastrointestinal and genitourinary tracts. Although it is possible to make the diagnosis in the prenatal period, most cases are diagnosed after birth through genetic and imaging studies. Advances in treatment have led to a progressive increase in survival rates. We present the case of a newborn with congenital alterations described prenatally and with imaging findings characteristic of the syndrome.
Collapse
Affiliation(s)
- Marcia Mejia
- Radiology, Universidad de Antioquia, Medellín, COL
| | - Mónica Royero Arias
- Pediatric Radiology, Hospital Universitario San Vicente Fundación, Medellín, COL
| | | | | |
Collapse
|
9
|
Chen CP. Syndromic and single gene disorders associated with fetal megacystis (I): Megacystis-microcolon-intestinal hypoperistalsis syndrome (MMIHS). Taiwan J Obstet Gynecol 2024; 63:19-21. [PMID: 38216263 DOI: 10.1016/j.tjog.2023.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2023] [Indexed: 01/14/2024] Open
Abstract
Fetal megacystis has been reported to be associated with chromosomal abnormalities, megacystis-microcolon-intestinal hypoperistalsis syndrome (MMIHS), obstructive uropathy, prune belly syndrome, cloacal anomalies, limb-body wall complex, amniotic band syndrome, anorectal malformations, VACTERL association (vertebral anomalies, anal atresia, cardiac malformations, tracheo-esophageal fistula, renal anomalies and limb abnormalities) and fetal overgrowth syndrome such as Bechwith-Wiedemann syndrome and Sotos syndrome. This review provides an overview of syndromic and single gene disorders associated with fetal megacystis which is useful for genetic counseling at prenatal diagnosis of fetal megacystis.
Collapse
Affiliation(s)
- Chih-Ping Chen
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan; Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan; School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan; Institute of Clinical and Community Health Nursing, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Obstetrics and Gynecology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan.
| |
Collapse
|
10
|
Viti F, De Giorgio R, Ceccherini I, Ahluwalia A, Alves MM, Baldo C, Baldussi G, Bonora E, Borrelli O, Dall'Oglio L, De Coppi P, De Filippo C, de Santa Barbara P, Diamanti A, Di Lorenzo C, Di Maulo R, Galeone A, Gandullia P, Hashmi SK, Lacaille F, Lancon L, Leone S, Mahé MM, Molnar MJ, Palmitelli A, Perin S, Prato AP, Thapar N, Vassalli M, Heuckeroth RO. Multi-disciplinary Insights from the First European Forum on Visceral Myopathy 2022 Meeting. Dig Dis Sci 2023; 68:3857-3871. [PMID: 37650948 PMCID: PMC10517037 DOI: 10.1007/s10620-023-08066-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 07/28/2023] [Indexed: 09/01/2023]
Abstract
Visceral myopathy is a rare, life-threatening disease linked to identified genetic mutations in 60% of cases. Mostly due to the dearth of knowledge regarding its pathogenesis, effective treatments are lacking. The disease is most commonly diagnosed in children with recurrent or persistent disabling episodes of functional intestinal obstruction, which can be life threatening, often requiring long-term parenteral or specialized enteral nutritional support. Although these interventions are undisputedly life-saving as they allow affected individuals to avoid malnutrition and related complications, they also seriously compromise their quality of life and can carry the risk of sepsis and thrombosis. Animal models for visceral myopathy, which could be crucial for advancing the scientific knowledge of this condition, are scarce. Clearly, a collaborative network is needed to develop research plans to clarify genotype-phenotype correlations and unravel molecular mechanisms to provide targeted therapeutic strategies. This paper represents a summary report of the first 'European Forum on Visceral Myopathy'. This forum was attended by an international interdisciplinary working group that met to better understand visceral myopathy and foster interaction among scientists actively involved in the field and clinicians who specialize in care of people with visceral myopathy.
Collapse
Affiliation(s)
- Federica Viti
- Institute of Biophysics, National Research Council, Via De Marini, 6, 16149, Genoa, Italy.
| | - Roberto De Giorgio
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | | | - Arti Ahluwalia
- Centro di Ricerca 'E. Piaggio' and Department of Information Engineering, University of Pisa, Pisa, Italy
| | - Maria M Alves
- Department of Clinical Genetics, Erasmus University Medical Center - Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Chiara Baldo
- IRCCS Istituto Giannina Gaslini Pediatric Hospital, Genoa, Italy
| | - Giannina Baldussi
- 'Uniti per la P.I.P.O.' Patient Advocacy Organization, Brescia, Italy
| | - Elena Bonora
- Unit of Medical Genetics, Department of Medical and Surgical Sciences, University of Bologna, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Osvaldo Borrelli
- Department of Gastroenterology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Luigi Dall'Oglio
- Digestive Surgery and Endoscopy, Bambino Gesù Children's Research Hospital IRCCS, Rome, Italy
| | - Paolo De Coppi
- Pediatric Surgery, Great Ormond Street Hospital for Children, London, UK
| | - Carlotta De Filippo
- Institute of Agricultural Biology and Biotechnology of the National Research Council, Pisa, Italy
| | - Pascal de Santa Barbara
- Physiology and Experimental Medicine of the Heart and Muscles (PhyMedExp), University of Montpellier, INSERM, CNRS, Montpellier, France
| | | | - Carlo Di Lorenzo
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Nationwide Children's Hospital, Columbus, OH, USA
| | | | | | - Paolo Gandullia
- IRCCS Istituto Giannina Gaslini Pediatric Hospital, Genoa, Italy
| | - Sohaib K Hashmi
- Department of Pediatrics, The Children's Hospital of Philadelphia Research Institute and the Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, Philadelphia, PA, USA
| | - Florence Lacaille
- Pediatric Gastroenterology-Hepatology-Nutrition, Necker-Enfants Malades Hospital, Paris, France
| | - Laurence Lancon
- 'Association des POIC' Patient Advocacy Organization, Marseille, France
| | - Salvatore Leone
- AMICI ETS, Associazione Nazionale per le Malattie Infiammatorie Croniche dell'Intestino, Milan, Italy
| | - Maxime M Mahé
- Nantes Université, INSERM, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Nantes, France
| | | | | | - Silvia Perin
- Unit of Pediatric Surgery, Department of Women and Child Health, University of Padua, Padua, Italy
| | - Alessio Pini Prato
- Unit of Pediatric Surgery, 'St. Antonio e Biagio e Cesare Arrigo' Hospital, Alessandria, Italy
| | - Nikhil Thapar
- Stem Cell and Regenerative Medicine, GOS Institute of Child Health, University College London, London, UK
- Gastroenterology, Hepatology and Liver Transplant, Queensland Children's Hospital, Brisbane, Australia
- School of Medicine, University of Queensland, Brisbane, Australia
- Woolworths Centre for Child Nutrition Research, Queensland University of Technology, Brisbane, Australia
| | - Massimo Vassalli
- James Watt School of Engineering, University of Glasgow, Glasgow, UK
| | - Robert O Heuckeroth
- Department of Pediatrics, The Children's Hospital of Philadelphia Research Institute and the Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, Philadelphia, PA, USA
| |
Collapse
|
11
|
He X, Dong K, Shen J, Hu G, Mintz JD, Atawia RT, Zhao J, Chen X, Caldwell RW, Xiang M, Stepp DW, Fulton DJ, Zhou J. The Long Noncoding RNA Cardiac Mesoderm Enhancer-Associated Noncoding RNA (Carmn) Is a Critical Regulator of Gastrointestinal Smooth Muscle Contractile Function and Motility. Gastroenterology 2023; 165:71-87. [PMID: 37030336 PMCID: PMC10330198 DOI: 10.1053/j.gastro.2023.03.229] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 03/15/2023] [Accepted: 03/20/2023] [Indexed: 04/10/2023]
Abstract
BACKGROUND & AIMS Visceral smooth muscle cells (SMCs) are an integral component of the gastrointestinal (GI) tract that regulate GI motility. SMC contraction is regulated by posttranslational signaling and the state of differentiation. Impaired SMC contraction is associated with significant morbidity and mortality, but the mechanisms regulating SMC-specific contractile gene expression, including the role of long noncoding RNAs (lncRNAs), remain largely unexplored. Herein, we reveal a critical role of Carmn (cardiac mesoderm enhancer-associated noncoding RNA), an SMC-specific lncRNA, in regulating visceral SMC phenotype and contractility of the GI tract. METHODS Genotype-Tissue Expression and publicly available single-cell RNA sequencing (scRNA-seq) data sets from embryonic, adult human, and mouse GI tissues were interrogated to identify SMC-specific lncRNAs. The functional role of Carmn was investigated using novel green fluorescent protein (GFP) knock-in (KI) reporter/knock-out (KO) mice. Bulk RNA-seq and single nucleus RNA sequencing (snRNA-seq) of colonic muscularis were used to investigate underlying mechanisms. RESULTS Unbiased in silico analyses and GFP expression patterns in Carmn GFP KI mice revealed that Carmn is highly expressed in GI SMCs in humans and mice. Premature lethality was observed in global Carmn KO and inducible SMC-specific KO mice due to GI pseudo-obstruction and severe distension of the GI tract, with dysmotility in cecum and colon segments. Histology, GI transit, and muscle myography analysis revealed severe dilation, significantly delayed GI transit, and impaired GI contractility in Carmn KO vs control mice. Bulk RNA-seq of GI muscularis revealed that loss of Carmn promotes SMC phenotypic switching, as evidenced by up-regulation of extracellular matrix genes and down-regulation of SMC contractile genes, including Mylk, a key regulator of SMC contraction. snRNA-seq further revealed SMC Carmn KO not only compromised myogenic motility by reducing contractile gene expression but also impaired neurogenic motility by disrupting cell-cell connectivity in the colonic muscularis. These findings may have translational significance, because silencing CARMN in human colonic SMCs significantly attenuated contractile gene expression, including MYLK, and decreased SMC contractility. Luciferase reporter assays showed that CARMN enhances the transactivation activity of the master regulator of SMC contractile phenotype, myocardin, thereby maintaining the GI SMC myogenic program. CONCLUSIONS Our data suggest that Carmn is indispensable for maintaining GI SMC contractile function in mice and that loss of function of CARMN may contribute to human visceral myopathy. To our knowledge this is the first study showing an essential role of lncRNA in the regulation of visceral SMC phenotype.
Collapse
Affiliation(s)
- Xiangqin He
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Kunzhe Dong
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia; Immunology Center of Georgia, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Jian Shen
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia; Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Guoqing Hu
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - James D Mintz
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Reem T Atawia
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Juanjuan Zhao
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Xiuxu Chen
- Department of Pathology and Laboratory Medicine, Loyola University Health System, Maywood, Illinois
| | - Robert W Caldwell
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - David W Stepp
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - David J Fulton
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia; Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Jiliang Zhou
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia.
| |
Collapse
|
12
|
Geraghty RM, Orr S, Olinger E, Neatu R, Barroso-Gil M, Mabillard H, Consortium GER, Wilson I, Sayer JA. Use of whole genome sequencing to determine the genetic basis of visceral myopathies including Prune Belly syndrome. JOURNAL OF RARE DISEASES (BERLIN, GERMANY) 2023; 2:9. [PMID: 37288276 PMCID: PMC10241726 DOI: 10.1007/s44162-023-00012-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 05/21/2023] [Indexed: 06/09/2023]
Abstract
Objectives/aims The visceral myopathies (VM) are a group of disorders characterised by poorly contractile or acontractile smooth muscle. They manifest in both the GI and GU tracts, ranging from megacystis to Prune Belly syndrome. We aimed to apply a bespoke virtual genetic panel and describe novel variants associated with this condition using whole genome sequencing data within the Genomics England 100,000 Genomes Project. Methods We screened the Genomics England 100,000 Genomes Project rare diseases database for patients with VM-related phenotypes. These patients were screened for sequence variants and copy number variants (CNV) in ACTG2, ACTA2, MYH11, MYLK, LMOD1, CHRM3, MYL9, FLNA and KNCMA1 by analysing whole genome sequencing data. The identified variants were analysed using variant effect predictor online tool, and any possible segregation in other family members and novel missense mutations was modelled using in silico tools. The VM cohort was also used to perform a genome-wide variant burden test in order to identify confirm gene associations in this cohort. Results We identified 76 patients with phenotypes consistent with a diagnosis of VM. The range of presentations included megacystis/microcolon hypoperistalsis syndrome, Prune Belly syndrome and chronic intestinal pseudo-obstruction. Of the patients in whom we identified heterozygous ACTG2 variants, 7 had likely pathogenic variants including 1 novel likely pathogenic allele. There were 4 patients in whom we identified a heterozygous MYH11 variant of uncertain significance which leads to a frameshift and a predicted protein elongation. We identified one family in whom we found a heterozygous variant of uncertain significance in KCNMA1 which in silico models predicted to be disease causing and may explain the VM phenotype seen. We did not find any CNV changes in known genes leading to VM-related disease phenotypes. In this phenotype selected cohort, ACTG2 is the largest monogenic cause of VM-related disease accounting for 9% of the cohort, supported by a variant burden test approach, which identified ACTG2 variants as the largest contributor to VM-related phenotypes. Conclusions VM are a group of disorders that are not easily classified and may be given different diagnostic labels depending on their phenotype. Molecular genetic analysis of these patients is valuable as it allows precise diagnosis and aids understanding of the underlying disease manifestations. We identified ACTG2 as the most frequent genetic cause of VM. We recommend a nomenclature change to 'autosomal dominant ACTG2 visceral myopathy' for patients with pathogenic variants in ACTG2 and associated VM phenotypes. Supplementary Information The online version contains supplementary material available at 10.1007/s44162-023-00012-z.
Collapse
Affiliation(s)
- Robert M. Geraghty
- Renal Services, The Newcastle Upon Tyne Hospitals NHS Foundation Trust, Freeman Road, Newcastle Upon Tyne, NE7 7DN UK
- Faculty of Medical Sciences, Translational and Clinical Institute, Newcastle University, Central Parkway, Newcastle Upon Tyne, NE1 3BZ UK
| | - Sarah Orr
- Faculty of Medical Sciences, Translational and Clinical Institute, Newcastle University, Central Parkway, Newcastle Upon Tyne, NE1 3BZ UK
| | - Eric Olinger
- Faculty of Medical Sciences, Translational and Clinical Institute, Newcastle University, Central Parkway, Newcastle Upon Tyne, NE1 3BZ UK
| | - Ruxandra Neatu
- Faculty of Medical Sciences, Translational and Clinical Institute, Newcastle University, Central Parkway, Newcastle Upon Tyne, NE1 3BZ UK
| | - Miguel Barroso-Gil
- Faculty of Medical Sciences, Translational and Clinical Institute, Newcastle University, Central Parkway, Newcastle Upon Tyne, NE1 3BZ UK
| | - Holly Mabillard
- Faculty of Medical Sciences, Translational and Clinical Institute, Newcastle University, Central Parkway, Newcastle Upon Tyne, NE1 3BZ UK
| | - Genomics England Research Consortium
- Renal Services, The Newcastle Upon Tyne Hospitals NHS Foundation Trust, Freeman Road, Newcastle Upon Tyne, NE7 7DN UK
- Faculty of Medical Sciences, Translational and Clinical Institute, Newcastle University, Central Parkway, Newcastle Upon Tyne, NE1 3BZ UK
- Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Central Parkway, Newcastle Upon Tyne, NE1 3BZ UK
- National Institute for Health Research Newcastle Biomedical Research Centre, Newcastle Upon Tyne, NE4 5PL UK
| | - Ian Wilson
- Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Central Parkway, Newcastle Upon Tyne, NE1 3BZ UK
| | - John A. Sayer
- Renal Services, The Newcastle Upon Tyne Hospitals NHS Foundation Trust, Freeman Road, Newcastle Upon Tyne, NE7 7DN UK
- Faculty of Medical Sciences, Translational and Clinical Institute, Newcastle University, Central Parkway, Newcastle Upon Tyne, NE1 3BZ UK
- National Institute for Health Research Newcastle Biomedical Research Centre, Newcastle Upon Tyne, NE4 5PL UK
| |
Collapse
|
13
|
Viti F, Pramotton FM, Martufi M, Magrassi R, Pedemonte N, Nizzari M, Zanacchi FC, De Michele B, Alampi M, Zambito M, Santamaria G, Bajetto A, Sardar S, Tomati V, Gandullia P, Giampietro C, Florio T, Beltrame F, Vassalli M, Ceccherini I. Patient's dermal fibroblasts as disease markers for visceral myopathy. BIOMATERIALS ADVANCES 2023; 148:213355. [PMID: 36893487 DOI: 10.1016/j.bioadv.2023.213355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 02/09/2023] [Accepted: 02/19/2023] [Indexed: 06/18/2023]
Abstract
Visceral myopathy (VSCM) is a rare genetic disease, orphan of pharmacological therapy. VSCM diagnosis is not always straightforward due to symptomatology similarities with mitochondrial or neuronal forms of intestinal pseudo-obstruction. The most prevalent form of VSCM is associates with variants in the gene ACTG2, encoding the protein gamma-2 actin. Overall, VSCM is a mechano-biological disorder, in which different genetic variants lead to similar alterations to the contractile phenotype of enteric smooth muscles, resulting in the emergence of life-threatening symptoms. In this work we analyzed the morpho-mechanical phenotype of human dermal fibroblasts from patients affected with VSCM, demonstrating that they retain a clear signature of the disease when compared with different controls. We evaluated several biophysical traits of fibroblasts, and we show that a measure of cellular traction forces can be used as a non-specific biomarker of the disease. We propose that a simple assay based on traction forces could be designed to provide a valuable support for clinical decision or pre-clinical research.
Collapse
Affiliation(s)
- Federica Viti
- Istituto di Biofisica - Consiglio Nazionale delle Ricerche, Via De Marini 16, 16149 Genova, Italy.
| | - Francesca Micaela Pramotton
- EMPA, Swiss Federal Laboratories for Materials Science and Technology, Ueberlandstrasse 129, 8600 Dübendorf, Switzerland; ETH Zurich, The Institute for Mechanical Systems, Leonhardstrasse 21, 8092 Zürich, Switzerland
| | - Michela Martufi
- Istituto di Biofisica - Consiglio Nazionale delle Ricerche, Via De Marini 16, 16149 Genova, Italy; Dipartimento Medicina Interna, Sezione di Farmacologia, Università di Genova, viale Benedetto XV, 2, 16132 Genova, Italy
| | - Raffaella Magrassi
- Istituto di Biofisica - Consiglio Nazionale delle Ricerche, Via De Marini 16, 16149 Genova, Italy
| | - Nicoletta Pedemonte
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, Via Gaslini 5, 16147 Genova, Italy
| | - Mario Nizzari
- Istituto di Biofisica - Consiglio Nazionale delle Ricerche, Via De Marini 16, 16149 Genova, Italy
| | | | - Benedetta De Michele
- Istituto di Biofisica - Consiglio Nazionale delle Ricerche, Via De Marini 16, 16149 Genova, Italy
| | - Manuela Alampi
- Department of Informatics, Bioengineering, Robotics and Systems Engineering, University of Genoa, Viale Causa, 13, 16145 Genova, Italy
| | - Martina Zambito
- Dipartimento Medicina Interna, Sezione di Farmacologia, Università di Genova, viale Benedetto XV, 2, 16132 Genova, Italy
| | - Giuseppe Santamaria
- UOSD Laboratorio di Genetica e Genomica delle Malattie Rare, IRCCS Istituto Giannina Gaslini, Via Gaslini 5, 16147 Genova, Italy
| | - Adriana Bajetto
- Dipartimento Medicina Interna, Sezione di Farmacologia, Università di Genova, viale Benedetto XV, 2, 16132 Genova, Italy
| | - Sabah Sardar
- Centre for the Cellular Microenvironment, James Watt School of Engineering, University of Glasgow, Oakfield avenue, G128LT Glasgow, UK
| | - Valeria Tomati
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, Via Gaslini 5, 16147 Genova, Italy
| | - Paolo Gandullia
- UOC Pediatric Gastroenterology and Digestive Endoscopy, IRCCS Istituto Giannina Gaslini, Via Gaslini 5, 16147 Genova, Italy
| | - Costanza Giampietro
- EMPA, Swiss Federal Laboratories for Materials Science and Technology, Ueberlandstrasse 129, 8600 Dübendorf, Switzerland; ETH Zurich, The Institute for Mechanical Systems, Leonhardstrasse 21, 8092 Zürich, Switzerland
| | - Tullio Florio
- Dipartimento Medicina Interna, Sezione di Farmacologia, Università di Genova, viale Benedetto XV, 2, 16132 Genova, Italy; IRCCS Ospedale Policlinico San Martino, Largo rosanna benzi 10, 16132 Genova, Italy
| | - Francesco Beltrame
- Department of Informatics, Bioengineering, Robotics and Systems Engineering, University of Genoa, Viale Causa, 13, 16145 Genova, Italy
| | - Massimo Vassalli
- Centre for the Cellular Microenvironment, James Watt School of Engineering, University of Glasgow, Oakfield avenue, G128LT Glasgow, UK
| | - Isabella Ceccherini
- UOSD Laboratorio di Genetica e Genomica delle Malattie Rare, IRCCS Istituto Giannina Gaslini, Via Gaslini 5, 16147 Genova, Italy
| |
Collapse
|
14
|
Cai H, Xiao Y, Chen S, Lu Y, Du J, You Y, Zhu J, Zhou J, Cai W, Wang Y. Heterozygous Actg2 R257C mice mimic the phenotype of megacystis microcolon intestinal hypoperistalsis syndrome. Neurogastroenterol Motil 2023; 35:e14472. [PMID: 36264152 DOI: 10.1111/nmo.14472] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/27/2022] [Accepted: 09/08/2022] [Indexed: 01/01/2023]
Abstract
BACKGROUND Megacystis microcolon intestinal hypoperistalsis syndrome (MMIHS) is a rare and serious congenital disorder with poor outcomes, where a heterozygous missense mutation is present in the ACTG2 gene. Here, we aimed to investigate the pathogenesis of ACTG2 in MMIHS. METHODS A cohort with 20 patients with MMIHS was screened. Actg2R257C heterozygous mutant mice were generated using the CRISPR/Cas9 system. Gastrointestinal (GI) motility, voluntary urination, collagen gel contraction, and G-actin/F-actin analysis were performed. KEY RESULTS The R257C variant of ACTG2 most frequently occurred in patients with MMIHS and demonstrated the typical symptoms of MMIHS. Actg2R257C heterozygous mutant mice had dilated intestines and bladders. The functional assay showed a prolonged total time of GI transit and decreased urine spot area. Collagen gel contraction assay and G-actin/F-actin analysis indicated that mutant mice showed reduced area of contraction of smooth muscle cells (SMCs) and impaired actin polymerization. CONCLUSIONS & INFERENCES A mouse model demonstrating MMIHS-like symptoms was generated. The Actg2R257C heterozygous variant impairs SMCs contraction by interfering with actin polymerization, leading to GI motility disorders.
Collapse
Affiliation(s)
- Hui Cai
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yongtao Xiao
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.,Shanghai Institute of Pediatric Research, Shanghai, China
| | - Shanshan Chen
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Lu
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.,Shanghai Institute of Pediatric Research, Shanghai, China
| | - Jun Du
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.,Shanghai Institute of Pediatric Research, Shanghai, China
| | - Yaying You
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Zhu
- Shanghai Institute of Pediatric Research, Shanghai, China
| | - Jie Zhou
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Cai
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.,Shanghai Institute of Pediatric Research, Shanghai, China
| | - Ying Wang
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.,Shanghai Institute of Pediatric Research, Shanghai, China
| |
Collapse
|
15
|
Sha Y, He Y, Liu X, Zhao S, Hu J, Wang J, Li S, Li W, Shi B, Hao Z. Rumen Epithelial Development- and Metabolism-Related Genes Regulate Their Micromorphology and VFAs Mediating Plateau Adaptability at Different Ages in Tibetan Sheep. Int J Mol Sci 2022; 23:ijms232416078. [PMID: 36555715 PMCID: PMC9786296 DOI: 10.3390/ijms232416078] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
The rumen is an important hallmark organ of ruminants and plays an important role in the metabolism and immune barrier of Tibetan sheep on the Plateau. However, there are few studies on rumen development and metabolism regulation in Tibetan sheep at different ages. Here, we comprehensively analyzed the immune function, fermentation function, rumen epithelial micromorphology and transcriptome profile of Tibetan sheep at different ages. The results showed that the concentration of IgG decreased and the concentration of IgM increased with age (p < 0.05), and the highest concentration of IgA was observed at 1.5 and 3.5 years of age. In terms of rumen fermentation characteristics, VFAs of 4-month-old lambs were the highest, followed by VFAs and NH3-N of Tibetan sheep at 3.5 years of age. Hematoxylin-eosin staining and transmission electron microscopy section examination of rumen epithelial tissue showed that the rumen papilla width increased with age (p < 0.001), the thickness of the stratum corneum decreased, the cells in the stratum corneum showed accelerated migration and the thickness of the rumen muscle layer increased (p < 0.001). Desmosomal junctions between the layers of rumen epithelium increased at 1.5 and 3.5 years old, forming a compact barrier structure, and the basal layer had more mitochondria involved in the regulation of energy metabolism. RNA-seq analysis revealed that a total of 1006 differentially expressed genes (DEGs) were identified at four ages. The DEGs of Tibetan sheep aged 4 months and 6 years were mainly enriched in the oxidation−reduction process and ISG15-protein conjugation pathway. The 1.5 and 3.5-year-olds were mainly enriched in skeletal muscle thin filament assembly, mesenchyme migration and the tight junction pathway. WGCNA showed that DEGs related to rumen microbiota metabolite VFAs and epithelial morphology were enriched in “Metabolism of xenobiotics by cytochrome P450, PPAR signaling pathway, Butanoate metabolism pathways” and participated in the regulation of rumen epithelial immune and fermentation metabolism functions of Tibetan sheep at different ages. This study systematically revealed the regulatory mechanism of rumen epithelial development and metabolism in the plateau adaptation of Tibetan sheep, providing a new approach for the study of plateau adaptation.
Collapse
Affiliation(s)
- Yuzhu Sha
- College of Animal Science and Technology/Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Yanyu He
- School of Fundamental Sciences, Massey University, Palmerston North 4410, New Zealand
| | - Xiu Liu
- College of Animal Science and Technology/Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
- Correspondence: ; Tel.: +86-931-763-1870
| | - Shengguo Zhao
- College of Animal Science and Technology/Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Jiang Hu
- College of Animal Science and Technology/Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Jiqing Wang
- College of Animal Science and Technology/Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Shaobin Li
- College of Animal Science and Technology/Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Wenhao Li
- Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining 810000, China
| | - Bingang Shi
- College of Animal Science and Technology/Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| | - Zhiyun Hao
- College of Animal Science and Technology/Gansu Key Laboratory of Herbivorous Animal Biotechnology, Gansu Agricultural University, Lanzhou 730070, China
| |
Collapse
|
16
|
Kapur RP, Goldstein AM, Loeff DS, Myers CT, Paschal CR. Intestinal Pathology in Patients With Pathogenic ACTG2-Variant Visceral Myopathy: 16 Patients From 12 Families and Review of the Literature. Pediatr Dev Pathol 2022; 25:581-597. [PMID: 35695198 DOI: 10.1177/10935266221107449] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Dominant gamma-smooth muscle actin gene (ACTG2) variants cause clinically diverse forms of visceral myopathy. Many patients undergo intestinal resection or biopsy before identification of their genetic defect. The pathology of ACTG2-variant visceral myopathy has not been evaluated systematically. METHODS Glass slides, ultrastructural images, molecular genetic reports, and clinical records from 16 patients with pathogenic (15) or likely pathogenic (1) ACTG2 variants were reviewed and compared with surgical specimens from controls (no evidence of a primary myopathy or pseudo-obstruction due to Hirschsprung disease) and published descriptions. RESULTS The variable clinical manifestations in our cohort matched those in the literature. Only non-specific light and electron microscopic findings observed in non-myopathic controls were encountered in 13 of 16 patients. The remaining 3 patients harbored hyalinized cytoplasmic inclusions in smooth muscle cells and 1 of them had polyglucosan bodies in the muscularis propria. CONCLUSIONS Apart from hyalinized inclusions, which were only observed in 3/16 patients, intestinal pathology in the majority of patients with ACTG2 variants is not indicative of an underlying visceral myopathy. Molecular testing should be considered even when no diagnostic intestinal pathology is identified.
Collapse
Affiliation(s)
- Raj P Kapur
- Department of Laboratories, 7274Seattle Children's Hospital, Seattle, WA, USA.,Department of Laboratory Medicine and Pathology, 7284University of Washington, Seattle, WA, USA
| | - Allan M Goldstein
- Department of Pediatric Surgery, 2348Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Deborah S Loeff
- Department of Pediatric Surgery, Comer Children's Hospital, University of Chicago Medicine, Chicago, IL, USA.,Advocate Aurora Children's Hospital, Park Ridge, IL, USA
| | - Candace T Myers
- Department of Laboratories, 7274Seattle Children's Hospital, Seattle, WA, USA
| | - Cate R Paschal
- Department of Laboratories, 7274Seattle Children's Hospital, Seattle, WA, USA.,Department of Laboratory Medicine and Pathology, 7284University of Washington, Seattle, WA, USA
| |
Collapse
|
17
|
Connaughton DM, Hildebrandt F. Disease mechanisms of monogenic congenital anomalies of the kidney and urinary tract American Journal of Medical Genetics Part C. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2022; 190:325-343. [PMID: 36208064 PMCID: PMC9618346 DOI: 10.1002/ajmg.c.32006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/14/2022] [Accepted: 09/16/2022] [Indexed: 11/05/2022]
Abstract
Congenital Anomalies of the Kidney and Urinary Tract (CAKUT) is a developmental disorder of the kidney and/or genito-urinary tract that results in end stage kidney disease (ESKD) in up to 50% of children. Despite the congenital nature of the disease, CAKUT accounts for almost 10% of adult onset ESKD. Multiple lines of evidence suggest that CAKUT is a Mendelian disorder, including the observation of familial clustering of CAKUT. Pathogenesis in CAKUT is embryonic in origin, with disturbances of kidney and urinary tract development resulting in a heterogeneous range of disease phenotypes. Despite polygenic and environmental factors being implicated, a significant proportion of CAKUT is monogenic in origin, with studies demonstrating single gene defects in 10%-20% of patients with CAKUT. Here, we review monogenic disease causation with emphasis on the etiological role of gene developmental pathways in CAKUT.
Collapse
Affiliation(s)
- Dervla M Connaughton
- Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Medicine, Division of Nephrology, London Health Sciences Centre, London, Ontario, Canada
| | - Friedhelm Hildebrandt
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
18
|
Fournier N, Fabre A. Smooth muscle motility disorder phenotypes: A systematic review of cases associated with seven pathogenic genes ( ACTG2, MYH11, FLNA, MYLK, RAD21, MYL9 and LMOD1). Intractable Rare Dis Res 2022; 11:113-119. [PMID: 36200034 PMCID: PMC9437995 DOI: 10.5582/irdr.2022.01060] [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: 05/25/2022] [Revised: 08/15/2022] [Accepted: 08/25/2022] [Indexed: 11/05/2022] Open
Abstract
Smooth muscle disorders affecting both the intestine and the bladder have been known for a decade. However, the recent discovery of genes associated with these dysfunctions has led to the description of several clinical phenotypes. We performed a systematic review of all published cases involving seven genes with pathogenic variants, ACTG2, MYH11, FLNA, MYLK, RAD21, MYL9 and LMOD1, and included 28 articles describing 112 patients and 5 pregnancies terminated before birth. The most commonly described mutations involved ACTG2 (75/112, 67% of patients), MYH11 (14%) and FLNA (13%). Twenty-seven patients (28%) died at a median age of 14.5 months. Among the 76 patients for whom this information was available, 10 (13%) had isolated chronic intestinal pseudo-obstruction (CIPO), 17 (22%) had isolated megacystis, and 48 (63%) had combined CIPO and megacystis. The respective proportions of these phenotypes were 9%, 20% and 71% among the 56 patients with ACTG2 mutations, 20%, 20% and 60% among the 10 patients with MYH11 mutations and 50%, 50% and 0% among the 7 patients with FLNA mutations.
Collapse
Affiliation(s)
- Ninon Fournier
- APHM, Timone Enfant, Pediatric Multidisciplinary Department, Marseille, France
| | - Alexandre Fabre
- APHM, Timone Enfant, Pediatric Multidisciplinary Department, Marseille, France
- Aix-Marseille Université, INSERM, GMGF, Marseille, France
- Address correspondence to:Alexandre Fabre, Pediatric Multidisciplinary Department, Timone Enfant Hospital, APHM, Aix-Marseille University, 264 Rue Saint Pierre 13005 Marseille, France. E-mail:
| |
Collapse
|
19
|
Huang CH, Schuring J, Skinner JP, Mok L, Chong MMW. MYL9 deficiency is neonatal lethal in mice due to abnormalities in the lung and the muscularis propria of the bladder and intestine. PLoS One 2022; 17:e0270820. [PMID: 35802750 PMCID: PMC9269942 DOI: 10.1371/journal.pone.0270820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 06/20/2022] [Indexed: 11/19/2022] Open
Abstract
Class II myosin complexes are responsible for muscle contraction as well as other non-sarcomeric contractile functions in cells. Myosin heavy chain molecules form the core of these structures, while light chain molecules regulate their stability and function. MYL9 is a light chain isoform that is thought to regulate non-sarcomeric myosin. However, whether this in only in specific cell types or in all cells remains unclear. To address this, we generated MYL9 deficient mice. These mice die soon after birth with abnormalities in multiple organs. All mice exhibited a distended bladder, shortening of the small intestine and alveolar overdistension in the lung. The Myl9 allele in these mice included a LacZ reporter knockin that allowed for mapping of Myl9 gene expression. Using this reporter, we show that MYL9 expression is restricted to the muscularis propria of the small intestine and bladder, as well as in the smooth muscle layer of the bronchi in the lung and major bladder vessels in all organs. This suggests that MYL9 is important for the function of smooth muscle cells in these organs. Smooth muscle dysfunction is therefore likely to be the cause of the abnormalities observed in the intestine, bladder and lung of MYL9 deficient mice and the resulting neonatal lethality.
Collapse
Affiliation(s)
- Chu-Han Huang
- St Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
- Department of Medicine (St Vincent’s), University of Melbourne, Fitzroy, VIC, Australia
| | - Joyce Schuring
- St Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
- HAN University of Applied Sciences, Nijmegen, The Netherlands
| | | | - Lawrence Mok
- St Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
- Department of Medicine (St Vincent’s), University of Melbourne, Fitzroy, VIC, Australia
| | - Mark M. W. Chong
- St Vincent’s Institute of Medical Research, Fitzroy, VIC, Australia
- Department of Medicine (St Vincent’s), University of Melbourne, Fitzroy, VIC, Australia
| |
Collapse
|
20
|
Sandy NS, Huysentruyt K, Mulder DJ, Warner N, Chong K, Morel C, AlQahtani S, Wales PW, Martin MG, Muise AM, Avitzur Y. The Diverse Phenotype of Intestinal Dysmotility Secondary to ACTG2-related Disorders. J Pediatr Gastroenterol Nutr 2022; 74:575-581. [PMID: 35149643 PMCID: PMC9632465 DOI: 10.1097/mpg.0000000000003400] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS The initial description of a heterozygous dominant ACTG2 variant in familial visceral myopathy was followed by the identification of additional variants in other forms of intestinal dysmotility disorders. we aimed to describe the diverse phenotype of this newly reported and rare disease. METHODS Report of 4 new patients, and a systematic review of ACTG2-related disorders. we analyzed the population frequency and used in silico gene damaging predictions. Genotype-phenotype correlations were explored. RESULTS One hundred three patients (52% girls), from 14 publications, were included. Twenty-eight unique variants were analyzed, all exceedingly rare, and 27 predicted to be highly damaging. The median Combined Annotation Dependent Depletion (CADD) score was 29.2 (Interquartile range 26.3-29.4). Most patients underwent abdominal surgery (66%), about half required intermittent bladder catheterization (48.5%), and more than half were parenteral nutrition (PN)-dependent (53%). One-quarter of the patients died (25.7%), and 6 required transplant (5.8%). Girls had a higher rate of microcolon (P = 0.009), PN dependency (P = 0.003), and death/transplant (P = 0.029) compared with boys, and early disease onset (<2 years of age) was associated with megacystis-microcolon-intestinal hypoperistalsis syndrome (MMIHS) features. There was no statistical association between disease characteristics and CADD scores. CONCLUSIONS Damaging ACTG2 variants are rare, often associated with MMIHS phenotype, and overall have a wide phenotypic variation. Symptoms usually present in the perinatal period but can also appear at a later age. The course of the disease is marked by frequent need for surgical interventions, PN support, and mortality. Poor outcomes are more common among girls with ACTG2 variants.
Collapse
Affiliation(s)
- Natascha S. Sandy
- Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
- Group for Improvement of Intestinal Function and Treatment (GIFT), The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Koen Huysentruyt
- Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
- Group for Improvement of Intestinal Function and Treatment (GIFT), The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
- Department of Pediatric Gastroenterology, Universitair Ziekenhuis Brussel, vrije Universiteit Brussel (vUB), Brussels, Belgium
| | - Daniel J. Mulder
- Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Neil Warner
- Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Karen Chong
- The Prenatal Diagnosis and Medical Genetics Program. Mount Sinai Hospital, Toronto, ON
| | - Chantal Morel
- Cancer Clinical Research Unit (CCRU), Princess Margaret Cancer Centre, The Hospital for Sick Children, University ofToronto, Toronto, Ontario, Canada
| | - Saleh AlQahtani
- Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Paul W. Wales
- Group for Improvement of Intestinal Function and Treatment (GIFT), The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
- Division of general and thoracic Surgery, The Hospital for Sick Children, University ofToronto, Toronto, Ontario, Canada
| | - Martin G. Martin
- # Division of Gastroenterology and Nutrition, Department of Pediatrics, Mattel Children’s Hospital and the David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Aleixo M. Muise
- Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Yaron Avitzur
- Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
- Group for Improvement of Intestinal Function and Treatment (GIFT), The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
21
|
Liu K, Lu L, Chen S, Gu B, Cai H, Wang Y, Cai W. Loss-of-function variants within LMOD1 actin-binding site 2 cause pediatric intestinal pseudo-obstruction by impairing protein stability and actin nucleation. FASEB J 2022; 36:e22194. [PMID: 35170814 DOI: 10.1096/fj.202101395r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 01/12/2022] [Accepted: 01/21/2022] [Indexed: 11/11/2022]
Abstract
The leiomodin1 (LMOD1) gene, encoding a potent actin nucleator, was recently reported as a potential pathogenic gene of megacystis-microcolon-intestinal hypoperistalsis syndrome (MMIHS, OMIM 619362). However, only a single patient has been reported to have LMOD1 mutations, and the underlying pathogenic mechanism remains unknown. Here, we described a male infant with LMOD1 mutations presenting typical symptoms of pediatric intestinal pseudo-obstruction (PIPO) but without megacystis and microcolon. Two compound heterozygous missense variants (c.1106C>T, p.T369M; c.1262G>A, p.R421H) were identified, both affecting highly conserved amino acid residues within the second actin-binding site (ABS2) domain of LMOD1. Expression analysis showed that both variants resulted in significantly reduced protein amounts, especially for p.T369M, which was almost undetectable. The reduction was only partially rescued by the proteasome inhibitor MG-132, indicating that there might be proteasome-independent pathways involved in the degradation of the mutant proteins. Molecular modeling showed that variant p.T369M impaired the local protein conformation of the ABS2 domain, while variant p.R421H directly impaired the intermolecular interaction between ABS2 and actin. Accordingly, both variants significantly damaged LMOD1-mediated actin nucleation. These findings provide further human genetic evidence supporting LMOD1 as a pathogenic gene underlying visceral myopathy including PIPO and MMIHS, strengthen the critical role of ABS2 domain in LMOD1-mediated actin nucleation, and moreover, reveal an unrecognized role of ABS2 in protein stability.
Collapse
Affiliation(s)
- Keqiang Liu
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute for Pediatric Research, Shanghai, China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Lina Lu
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shanshan Chen
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Beilin Gu
- Shanghai Institute for Pediatric Research, Shanghai, China
| | - Hui Cai
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Wang
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Wei Cai
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute for Pediatric Research, Shanghai, China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.,Department of Pediatric Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
22
|
Hahn JW, Moon SY, Kim MS, Woo MH, Sohn MJ, Kim HY, Seong MW, Park SS, Park SH, Moon JS, Ko JS. ACTG2 Variants in Pediatric Chronic Intestinal Pseudo-obstruction With Megacystis. J Neurogastroenterol Motil 2022; 28:104-110. [PMID: 34980693 PMCID: PMC8748860 DOI: 10.5056/jnm20243] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/23/2021] [Accepted: 03/08/2021] [Indexed: 12/11/2022] Open
Abstract
Background/Aims Chronic intestinal pseudo-obstruction (CIPO) is a clinically heterogeneous syndrome characterized by compromised peristalsis and intestinal obstruction. Variants of actin gamma 2 (ACTG2), a protein crucial for correct enteric muscle contraction, have been found in CIPO patients. The aim of this study is to examine the clinical features and ACTG2 variants in Korean patients with CIPO. Methods From January 1995 to August 2020, 12 patients diagnosed with CIPO were included and genetic analysis testing of ACTG2 was performed. Results Heterozygous ACTG2 missense variants were found in 6 patients (50.0%). The p.Arg257Cys variant was found in 3 patients, and p.Arg63Gln and p.Arg178His variants were found in 1 patient each. A novel variant, p.Ile193Phe, was found in 1 patient. Three patients were diagnosed at birth, 2 at the age of 1 year, and 1 at 3 years of age. Abnormal prenatal genitourinary ultrasonographic findings were found in all 6 patients; microcolon was found in 4 patients (66.7%), and megacystis in all 6 patients. The pathology showed abnormal ganglion cells as well as myopathic findings. All patients are dependent on total parenteral nutrition and are to date alive. Conclusions ACTG2 variants are commonly found in Korean patients with CIPO. In CIPO patients with megacystis and abnormal prenatal ultrasonography, genetic testing of ACTG2 should be considered. Molecular diagnosis of CIPO is more important than pathologic diagnosis.
Collapse
Affiliation(s)
- Jong Woo Hahn
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | - Soo Young Moon
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | - Min Soo Kim
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | - Min Hyung Woo
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | - Min Ji Sohn
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun-Young Kim
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Moon-Woo Seong
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Sung Sup Park
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Sung-Hye Park
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Jin Soo Moon
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| | - Jae Sung Ko
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
23
|
James KN, Lau M, Shayan K, Lenberg J, Mardach R, Ignacio R, Halbach J, Choi L, Kumar S, Ellsworth KA. Expanding the genotypic spectrum of ACTG2-related visceral myopathy. Cold Spring Harb Mol Case Stud 2021; 7:mcs.a006085. [PMID: 33883208 PMCID: PMC8208046 DOI: 10.1101/mcs.a006085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/30/2021] [Indexed: 12/13/2022] Open
Abstract
Visceral myopathies (VMs) encompass a spectrum of disorders characterized by chronic disruption of gastrointestinal function, with or without urinary system involvement. Pathogenic missense variation in smooth muscle γ-actin gene (ACTG2) is associated with autosomal dominant VM. Whole-genome sequencing of an infant presenting with chronic intestinal pseudo-obstruction revealed a homozygous 187 bp (c.589_613 + 163del188) deletion spanning the exon 6–intron 6 boundary within ACTG2. The patient's clinical course was marked by prolonged hospitalizations, multiple surgeries, and intermittent total parenteral nutrition dependence. This case supports the emerging understanding of allelic heterogeneity in ACTG2-related VM, in which both biallelic and monoallelic variants in ACTG2 are associated with gastrointestinal dysfunction of similar severity and overlapped clinical presentation. Moreover, it illustrates the clinical utility of rapid whole-genome sequencing, which can comprehensively and precisely detect different types of genomic variants including small deletions, leading to guidance of clinical care decisions.
Collapse
Affiliation(s)
- Kiely N James
- Rady Children's Institute for Genomic Medicine, San Diego, California 92123, USA
| | - Megan Lau
- UC San Diego School of Medicine, La Jolla, California 92093, USA
| | - Katayoon Shayan
- Pathology Department, Hepatology and Nutrition, Rady Children's Hospital, San Diego, California 92123, USA
| | - Jerica Lenberg
- Rady Children's Institute for Genomic Medicine, San Diego, California 92123, USA
| | - Rebecca Mardach
- Rady Children's Institute for Genomic Medicine, San Diego, California 92123, USA
| | - Romeo Ignacio
- Division of Pediatric Surgery, Hepatology and Nutrition, Rady Children's Hospital, San Diego, California 92123, USA
| | - Jonathan Halbach
- Division of Pediatric Surgery, Hepatology and Nutrition, Rady Children's Hospital, San Diego, California 92123, USA
| | - Lillian Choi
- Division of Gastroenterology, Hepatology and Nutrition, Rady Children's Hospital, San Diego, California 92123, USA
| | - Soma Kumar
- Division of Gastroenterology, Hepatology and Nutrition, Rady Children's Hospital, San Diego, California 92123, USA
| | | |
Collapse
|
24
|
Hashmi SK, Ceron RH, Heuckeroth RO. Visceral myopathy: clinical syndromes, genetics, pathophysiology, and fall of the cytoskeleton. Am J Physiol Gastrointest Liver Physiol 2021; 320:G919-G935. [PMID: 33729000 PMCID: PMC8285581 DOI: 10.1152/ajpgi.00066.2021] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Visceral smooth muscle is a crucial component of the walls of hollow organs like the gut, bladder, and uterus. This specialized smooth muscle has unique properties that distinguish it from other muscle types and facilitate robust dilation and contraction. Visceral myopathies are diseases where severe visceral smooth muscle dysfunction prevents efficient movement of air and nutrients through the bowel, impairs bladder emptying, and affects normal uterine contraction and relaxation, particularly during pregnancy. Disease severity exists along a spectrum. The most debilitating defects cause highly dysfunctional bowel, reduced intrauterine colon growth (microcolon), and bladder-emptying defects requiring catheterization, a condition called megacystis-microcolon-intestinal hypoperistalsis syndrome (MMIHS). People with MMIHS often die early in childhood. When the bowel is the main organ affected and microcolon is absent, the condition is known as myopathic chronic intestinal pseudo-obstruction (CIPO). Visceral myopathies like MMIHS and myopathic CIPO are most commonly caused by mutations in contractile apparatus cytoskeletal proteins. Here, we review visceral myopathy-causing mutations and normal functions of these disease-associated proteins. We propose molecular, cellular, and tissue-level models that may explain clinical and histopathological features of visceral myopathy and hope these observations prompt new mechanistic studies.
Collapse
Affiliation(s)
- Sohaib Khalid Hashmi
- 1Department of Pediatrics, The Children’s Hospital
of Philadelphia Research Institute and the Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, Philadelphia, Pennsylvania,2Department of Bioengineering, The University of Pennsylvania School of Engineering and Applied Science, Philadelphia, Pennsylvania
| | - Rachel Helen Ceron
- 1Department of Pediatrics, The Children’s Hospital
of Philadelphia Research Institute and the Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, Philadelphia, Pennsylvania,3Department of Physiology, The University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Robert O. Heuckeroth
- 1Department of Pediatrics, The Children’s Hospital
of Philadelphia Research Institute and the Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, Philadelphia, Pennsylvania
| |
Collapse
|
25
|
Holland AM, Bon-Frauches AC, Keszthelyi D, Melotte V, Boesmans W. The enteric nervous system in gastrointestinal disease etiology. Cell Mol Life Sci 2021; 78:4713-4733. [PMID: 33770200 PMCID: PMC8195951 DOI: 10.1007/s00018-021-03812-y] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/20/2021] [Accepted: 03/10/2021] [Indexed: 02/06/2023]
Abstract
A highly conserved but convoluted network of neurons and glial cells, the enteric nervous system (ENS), is positioned along the wall of the gut to coordinate digestive processes and gastrointestinal homeostasis. Because ENS components are in charge of the autonomous regulation of gut function, it is inevitable that their dysfunction is central to the pathophysiology and symptom generation of gastrointestinal disease. While for neurodevelopmental disorders such as Hirschsprung, ENS pathogenesis appears to be clear-cut, the role for impaired ENS activity in the etiology of other gastrointestinal disorders is less established and is often deemed secondary to other insults like intestinal inflammation. However, mounting experimental evidence in recent years indicates that gastrointestinal homeostasis hinges on multifaceted connections between the ENS, and other cellular networks such as the intestinal epithelium, the immune system, and the intestinal microbiome. Derangement of these interactions could underlie gastrointestinal disease onset and elicit variable degrees of abnormal gut function, pinpointing, perhaps unexpectedly, the ENS as a diligent participant in idiopathic but also in inflammatory and cancerous diseases of the gut. In this review, we discuss the latest evidence on the role of the ENS in the pathogenesis of enteric neuropathies, disorders of gut-brain interaction, inflammatory bowel diseases, and colorectal cancer.
Collapse
Affiliation(s)
- Amy Marie Holland
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
- Biomedical Research Institute (BIOMED), Hasselt University, Diepenbeek, Belgium
| | - Ana Carina Bon-Frauches
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Daniel Keszthelyi
- Department of Internal Medicine, Division of Gastroenterology-Hepatology, NUTRIM-School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Veerle Melotte
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Werend Boesmans
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands.
- Biomedical Research Institute (BIOMED), Hasselt University, Diepenbeek, Belgium.
| |
Collapse
|
26
|
Prathapan KM, King DE, Raghu VK, Ackerman K, Presel T, Yaworski JA, Ganoza A, Bond G, Sevilla WMA, Rudolph JA, Alissa F. Megacystis Microcolon Intestinal Hypoperistalsis Syndrome: A Case Series With Long-term Follow-up and Prolonged Survival. J Pediatr Gastroenterol Nutr 2021; 72:e81-e85. [PMID: 33264186 PMCID: PMC9124153 DOI: 10.1097/mpg.0000000000003008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Describe clinical characteristics, management, and outcome in a cohort of megacystis microcolon intestinal hypoperistalsis syndrome (MMIHS) patients. METHODS We conducted a retrospective chart review of MMIHS patients followed at a large transplant and intestinal rehabilitation center over a period of 17 years. RESULTS We identified 25 patients with MMIHS (68% girls, 13 transplanted). One transplanted and 1 nontransplanted patient were lost to follow-up. We estimated 100, 100, and 86% for 5-, 10-, and 20-year survival, respectively, with only 1 death. Of the 22 patients alive at the time of study (11 transplanted, 11 nontransplanted), median age was 9.2 years (range 2.7-22.9 years). Longest posttransplant follow-up was 16 years. Seventeen patients had available prenatal imaging reports; all showed distended bladder. Eight had genetic testing (5, ACTG2; 2, MYH11; 1, MYL9). Almost all patients had normal growth with median weight z-score -0.77 (interquartile range -1.39 to 0.26), height z score -1.2 (-2.04 to -0.48) and body mass index z-score 0.23 (-0.37 to 0.93) with no statistical difference between transplanted and nontransplanted patients. All nontransplanted patients were on parenteral nutrition with minimal/no feeds, and all except 1 of the transplanted patients were on full enteral feeds. Recent average bilirubin, INR, albumin, and creatinine fell within the reference ranges. CONCLUSIONS This is the largest single-center case series with the longest duration of follow-up for MMIHS patients. In the current era of improved intestinal rehabilitation and transplantation, MMIHS patients have excellent outcomes in survival, growth, and liver function. This observation contradicts previous reports and should alter counselling and management decisions in these patients at diagnosis.
Collapse
Affiliation(s)
- Krishnapriya Marangattu Prathapan
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | - Dale E. King
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | - Vikram Kalathur Raghu
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | - Kimberly Ackerman
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | - Tracey Presel
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | - Jane Anne Yaworski
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | - Armando Ganoza
- Hillman Center for Pediatric Liver Transplantation, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | - Geoffrey Bond
- Hillman Center for Pediatric Liver Transplantation, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | - Wednesday Marie A. Sevilla
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | - Jeffrey A. Rudolph
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | - Feras Alissa
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
27
|
Xiong X, Li J, Liu C, Xu F. Visceral myopathy diagnosed by a de novo ACTG2 mutation in a patient with chronic intestinal pseudo-obstruction-a case report. Transl Pediatr 2021; 10:679-685. [PMID: 33880338 PMCID: PMC8041608 DOI: 10.21037/tp-20-316] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/31/2020] [Indexed: 12/13/2022] Open
Abstract
Visceral myopathy is a rare genetic disorder that commonly affects the digestive and renal systems. Manifestations include a clinical spectrum covering chronic intestinal pseudo-obstruction (CIPO) and megacystis-microcolon-intestinal hypoperistalsis syndrome (MMIHS). The smooth muscle actin γ-2 gene (ACTG2) is one of the most common disease-causing genes. Here, we present a case of pediatric intestinal pseudo-obstruction associated with a novel missense ACTG2 mutation, c.588G>C/p.E196D. His parents had no this mutation, which suggested the possibility of spontaneous mutation. Amino acid conservation analysis of γ-2 actin showed replacement of glutamate at position 196 by aspartate. The patient suffered from recurrent episodes of abdominal bloating, undergone repeated gastrointestinal surgery, had feeding difficulties, and required long-term parenteral nutrition support. The patient had no other specific symptoms or underlying diseases. X-ray of the abdomen showed dilation of the intestine as well as an air-fluid pattern. The manifestations of biopsy were various. All biochemical tests were normal, and the possibility of secondary intestinal pseudo-obstruction was excluded. The mutation site of ACTG2 in the present study has not been previously described in patients with visceral myopathy, and thus, our study reveals a novel mutation of ACTG2-associated visceral myopathy in a patient with CIPO. This report can serve as a reference for future research and further expands the map of genetic variation for visceral myopathy.
Collapse
Affiliation(s)
- Xiaoyu Xiong
- Department of Pediatric Intensive Care Unit, Chongqing Medical University Affiliated Children's Hospital; National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Jing Li
- Department of Pediatric Intensive Care Unit, Chongqing Medical University Affiliated Children's Hospital; National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Chengjun Liu
- Department of Pediatric Intensive Care Unit, Chongqing Medical University Affiliated Children's Hospital; National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Feng Xu
- Department of Pediatric Intensive Care Unit, Chongqing Medical University Affiliated Children's Hospital; National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
28
|
Matera I, Bordo D, Di Duca M, Lerone M, Santamaria G, Pongiglione M, Lezo A, Diamanti A, Spagnuolo MI, Pini Prato A, Alberti D, Mattioli G, Gandullia P, Ceccherini I. Novel ACTG2 variants disclose allelic heterogeneity and bi-allelic inheritance in pediatric chronic intestinal pseudo-obstruction. Clin Genet 2020; 99:430-436. [PMID: 33294969 DOI: 10.1111/cge.13895] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 12/13/2022]
Abstract
Variants in the ACTG2 gene, encoding a protein crucial for correct enteric muscle contraction, have been found in patients affected with chronic intestinal pseudo-obstruction, either congenital or late-onset visceral myopathy, and megacystis-microcolon-intestinal hypoperistalsis syndrome. Here we report about ten pediatric and one adult patients, from nine families, carrying ACTG2 variants: four show novel still unpublished missense variants, including one that is apparently transmitted according to a recessive mode of inheritance. Four of the remaining five probands carry variants affecting arginine residues, that have already been associated with a severe phenotype. A de novo occurrence of the variants could be confirmed in six of these families. Since a genotype-phenotype correlation is affected by extrinsic factors, such as, diagnosis delay, quality of clinical management, and intra-familial variability, we have undertaken 3D molecular modeling to get further insights into the effects of the variants here described. The present findings and further ACTG2 testing of patients presenting with intestinal pseudo-obstruction, will improve our understanding of visceral myopathies, including implications in the prognosis and genetic counseling of this set of severe disorders.
Collapse
Affiliation(s)
- Ivana Matera
- UOSD Genetica e Genomica delle Malattie Rare, IRCCS Istituto Giannina Gaslini, Genoa, Italia, Italy
| | | | - Marco Di Duca
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, Genoa, Italia, Italy
| | - Margherita Lerone
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, Genoa, Italia, Italy
| | - Giuseppe Santamaria
- UOSD Genetica e Genomica delle Malattie Rare, IRCCS Istituto Giannina Gaslini, Genoa, Italia, Italy
| | - Marta Pongiglione
- UOC Radiologia, IRCCS Istituto Giannina Gaslini, Genoa, Italia, Italy
| | - Antonella Lezo
- Dietetics and Clinical Nutrition Unit, Children's Hospital Regina Margherita, Torino, Italy
| | - Antonella Diamanti
- UOS Nutrizione Artificiale, IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | | | - Alessio Pini Prato
- UO Chirurgia Pediatrica, AON SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Daniele Alberti
- UO Chirurgia Pediatrica, ASST- Spedali Civili di Brescia, Brescia, Italy
| | | | - Paolo Gandullia
- UOC Gastroenterologia. IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Isabella Ceccherini
- UOSD Genetica e Genomica delle Malattie Rare, IRCCS Istituto Giannina Gaslini, Genoa, Italia, Italy
| |
Collapse
|
29
|
Zhu CZ, Zhao HW, Lin HW, Wang F, Li YX. Latest developments in chronic intestinal pseudo-obstruction. World J Clin Cases 2020. [DOI: 10.12998/wjcc.v8.i23.5850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
|
30
|
Zhu CZ, Zhao HW, Lin HW, Wang F, Li YX. Latest developments in chronic intestinal pseudo-obstruction. World J Clin Cases 2020; 8:5852-5865. [PMID: 33344584 PMCID: PMC7723695 DOI: 10.12998/wjcc.v8.i23.5852] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 10/02/2020] [Accepted: 10/19/2020] [Indexed: 02/05/2023] Open
Abstract
Chronic intestinal pseudo-obstruction (CIPO) is a type of intestinal dysfunction presenting as symptoms of intestinal obstruction but without actual mechanical obstruction. An extremely low incidence, non-specific clinical symptoms, strong heterogeneity, and no definitive cause in some patients make CIPO very difficult to diagnose correctly. Imaging and gastrointestinal manometry are commonly used. Most patients have progressive worsening of their symptoms and require intervention, and nutritional assessment and treatment are very important to determine the prognosis. With improvements in surgical techniques, small bowel transplantation is a feasible treatment option for patients with advanced CIPO; however, the long-term prognosis for CIPO patients remains unsatisfactory. Generally, the disease is rare and difficult to diagnose, which leads to clinicians' lack of understanding of the disease and results in a high rate of misdiagnosis. This review describes the characteristics of CIPO and the latest developments in diagnosis and treatment, in detail. The goal of our review is to improve clinicians' understanding of CIPO so that the disease is identified quickly and accurately, and treated as early as possible to improve patients' quality of life.
Collapse
Affiliation(s)
- Chang-Zhen Zhu
- Department of Gastrointestinal Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Hong-Wei Zhao
- Department of Gastrointestinal Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Hong-Wei Lin
- Department of Gastrointestinal Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Feng Wang
- Department of Gastrointestinal Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Yuan-Xin Li
- Department of Gastrointestinal Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| |
Collapse
|
31
|
Kandler JL, Sklirou E, Woerner A, Walsh L, Cox E, Xue Y. Compound heterozygous loss of function variants in MYL9 in a child with megacystis-microcolon-intestinal hypoperistalsis syndrome. Mol Genet Genomic Med 2020; 8:e1516. [PMID: 33031641 PMCID: PMC7667357 DOI: 10.1002/mgg3.1516] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/10/2020] [Indexed: 12/13/2022] Open
Abstract
Megacystis–microcolon–intestinal hypoperistalsis syndrome (MMIHS), or “visceral myopathy,” is a severe early onset disorder characterized by impaired muscle contractility in the bladder and intestines. Five genes are linked to MMIHS: primarily ACTG2, but also LMOD1, MYH11, MYLK, and MYL9. Here we describe a three‐year‐old girl with bilateral hydronephrosis diagnosed at 20 weeks gestation and congenital mydriasis (both of which have been previously observed among individuals with MMIHS). A clinical diagnosis of MMIHS was made based upon the presence of megacystis, lack of urinary bladder peristalsis, and intestinal pseudo‐obstruction. After initial testing of ACTG2 was negative, further sequencing and deletion/duplication testing was performed on the LMOD1, MYH11,MYLK, and MYL9 genes. We identified two heterozygous loss of function variants in MYL9: an exon 4 deletion and a nine base pair deletion that removes the canonical splicing donor site at exon 2 (NM_006097.5:c.184+2_184+10del). Parental testing confirmed these variants to be in trans in our proband. To our knowledge, only one other individual with MMIHS has biallelic mutations in MYL9 (a homozygous deletion encompassing exon 4). We suggest MYL9 be targeted on genetic testing panels for MMIHS, smooth muscle myopathies, and cardiovascular phenotypes.
Collapse
Affiliation(s)
| | - Evgenia Sklirou
- Division of Medical Genetics, Department of Pediatrics, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Audrey Woerner
- Division of Medical Genetics, Department of Pediatrics, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Leslie Walsh
- Division of Medical Genetics, Department of Pediatrics, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Yuan Xue
- Fulgent Genetics, Atlanta, Georgia, USA
| |
Collapse
|
32
|
Hashmi SK, Barka V, Yang C, Schneider S, Svitkina TM, Heuckeroth RO. Pseudo-obstruction-inducing ACTG2R257C alters actin organization and function. JCI Insight 2020; 5:140604. [PMID: 32814715 PMCID: PMC7455133 DOI: 10.1172/jci.insight.140604] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/15/2020] [Indexed: 12/14/2022] Open
Abstract
Actin γ 2, smooth muscle (ACTG2) R257C mutation is the most common genetic cause of visceral myopathy. Individuals with ACTG2 mutations endure prolonged hospitalizations and surgical interventions, become dependent on intravenous nutrition and bladder catheterization, and often die in childhood. Currently, we understand little about how ACTG2 mutations cause disease, and there are no mechanism-based treatments. Our goal was to characterize the effects of ACTG2R257C on actin organization and function in visceral smooth muscle cells. We overexpressed ACTG2WT or ACTG2R257C in primary human intestinal smooth muscle cells (HISMCs) and performed detailed quantitative analyses to examine effects of ACTG2R257C on (a) actin filament formation and subcellular localization, (b) actin-dependent HISMC functions, and (c) smooth muscle contractile gene expression. ACTG2R257C resulted in 41% fewer, 13% thinner, 33% shorter, and 40% less branched ACTG2 filament bundles compared with ACTG2WT. Curiously, total F-actin probed by phalloidin and a pan-actin antibody was unchanged between ACTG2WT- and ACTG2R257C-expressing HISMCs, as was ultrastructural F-actin organization. ACTG2R257C-expressing HISMCs contracted collagen gels similar to ACTG2WT-expressing HISMCs but spread 21% more and were 11% more migratory. In conclusion, ACTG2R257C profoundly affects ACTG2 filament bundle structure, without altering global actin cytoskeleton in HISMCs.
Collapse
Affiliation(s)
- Sohaib Khalid Hashmi
- Department of Pediatrics, Children's Hospital of Philadelphia Research Institute, and Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, Philadelphia, Pennsylvania, USA.,Department of Bioengineering, University of Pennsylvania School of Engineering and Applied Science, Philadelphia, Pennsylvania, USA
| | - Vasia Barka
- Department of Pediatrics, Children's Hospital of Philadelphia Research Institute, and Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, Philadelphia, Pennsylvania, USA
| | - Changsong Yang
- Department of Biology, University of Pennsylvania School of Arts and Sciences, Philadelphia, Pennsylvania, USA
| | - Sabine Schneider
- Department of Pediatrics, Children's Hospital of Philadelphia Research Institute, and Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, Philadelphia, Pennsylvania, USA
| | - Tatyana M Svitkina
- Department of Biology, University of Pennsylvania School of Arts and Sciences, Philadelphia, Pennsylvania, USA
| | - Robert O Heuckeroth
- Department of Pediatrics, Children's Hospital of Philadelphia Research Institute, and Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
33
|
Billon C, Molin A, Poirsier C, Clemenson A, Dauge C, Grelet M, Sigaudy S, Patrier S, Goldenberg A, Layet V, Tantau J, Fleury C, Liard A, Diguet A, Fritih R, Verspyck E, Rendu J, Boutaud L, Tessier A, Thomas S, Razavi F, Achaiaa A, Elkhartoufi N, Hakkakian L, Magnin E, Bôle-Feysot C, Masson C, Ville Y, Roth P, Prieur F, Bessieres B, Bonniere M, Attie-Bitach T. Fetal megacystis-microcolon: Genetic mutational spectrum and identification of PDCL3 as a novel candidate gene. Clin Genet 2020; 98:261-273. [PMID: 32621347 DOI: 10.1111/cge.13801] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/24/2020] [Accepted: 06/25/2020] [Indexed: 12/13/2022]
Abstract
Megacystis-microcolon-intestinal-hypoperistalsis syndrome (MMIHS) is a severe congenital visceral myopathy characterized by an abdominal distension due to a large non-obstructed urinary bladder, a microcolon and intestinal hypo- or aperistalsis. Most of the patients described to date carry a sporadic heterozygous variant in ACTG2. More recently, recessive forms have been reported and mutations in MYH11, LMOD1, MYLK and MYL9 have been described at the molecular level. In the present report, we describe five patients carrying a recurrent heterozygous variant in ACTG2. Exome sequencing performed in four families allowed us to identify the genetic cause in three. In two families, we identified variants in MMIHS causal genes, respectively a nonsense homozygous variant in MYH11 and a previously described homozygous deletion in MYL9. Finally, we identified compound heterozygous variants in a novel candidate gene, PDCL3, c.[143_144del];[380G>A], p.[(Tyr48Ter)];[(Cys127Tyr)]. After cDNA analysis, a complete absence of PDLC3 expression was observed in affected individuals, indicating that both mutated transcripts were unstable and prone to mediated mRNA decay. PDCL3 encodes a protein involved in the folding of actin, a key step in thin filament formation. Presumably, loss-of-function of this protein affects the contractility of smooth muscle tissues, making PDCL3 an excellent candidate gene for autosomal recessive forms of MMIHS.
Collapse
Affiliation(s)
- Clarisse Billon
- Service d'Histologie-Embryologie-Cytogénétique, Unité d'Embryofoetopathologie, Hôpital Necker-Enfants Malades, APHP, Paris, France.,Département de Génétique, Hôpital Européen Georges Pompidou, APHP, Paris, France
| | - Arnaud Molin
- Département de Génétique, Normandie Université, UNICAEN, CHU de Caen Normandie, Caen, France
| | | | - Alix Clemenson
- Service d'Anatomie et Cytotologie Pathologique, CHU de Saint Etienne, Saint Etienne, France
| | - Coralie Dauge
- Department of Pathology, University Hospital, Caen, France
| | - Maude Grelet
- Département de Génétique Médicale, Hôpital de la Timone, APHM, Marseille, France
| | - Sabine Sigaudy
- Département de Génétique Médicale, Hôpital de la Timone, APHM, Marseille, France
| | - Sophie Patrier
- Service d'Anatomie Pathologique, CHU Ch. Nicolle, Rouen, France
| | - Alice Goldenberg
- centre de référence anomalies du développement et syndromes malformatifs, CHU de Rouen, Centre Normand de Génomique et de Médecine Personnalisée, France
| | - Valérie Layet
- Consultations de génétique, Groupe Hospitalier du Havre, Le Havre, France
| | - Julia Tantau
- Service d'Histologie-Embryologie-Cytogénétique, Unité d'Embryofoetopathologie, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Clémence Fleury
- Department of Pathology, Robert-Debré University Hospital, Reims, France
| | - Agnès Liard
- Département de chirurgie infantile, Chu de Rouen, Rouen, France
| | - Alain Diguet
- Laboratoire d'anatomie pathologique, pavillon Jacques-Delarue, CHU de Rouen, Rouen, France
| | - Radia Fritih
- Pathology Department, Hôpital de la Timone, APHM, Marseille, France
| | - Eric Verspyck
- Department of Obstetrics and Gynecology, Rouen University Hospital, Rouen, France
| | - John Rendu
- Unité Médicale de Génétique Moléculaire, Inserm U1216, CHU de Grenoble, Grenoble, France
| | - Lucile Boutaud
- Service d'Histologie-Embryologie-Cytogénétique, Unité d'Embryofoetopathologie, Hôpital Necker-Enfants Malades, APHP, Paris, France.,INSERM UMR 1163, Université de Paris, Imagine Institute, Paris, France
| | - Aude Tessier
- Service d'Histologie-Embryologie-Cytogénétique, Unité d'Embryofoetopathologie, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Sophie Thomas
- INSERM UMR 1163, Université de Paris, Imagine Institute, Paris, France
| | - Ferechté Razavi
- Service d'Histologie-Embryologie-Cytogénétique, Unité d'Embryofoetopathologie, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Amale Achaiaa
- Service d'Histologie-Embryologie-Cytogénétique, Unité d'Embryofoetopathologie, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Nadia Elkhartoufi
- Service d'Histologie-Embryologie-Cytogénétique, Unité d'Embryofoetopathologie, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Leila Hakkakian
- Service d'Histologie-Embryologie-Cytogénétique, Unité d'Embryofoetopathologie, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Eglantine Magnin
- Service d'Histologie-Embryologie-Cytogénétique, Unité d'Embryofoetopathologie, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | | | - Cécile Masson
- Bioinformatics Platform, INSERM UMR 1163, Institut Imagine, Paris, France
| | - Yves Ville
- Service d'Obstétrique, Maternité, Chirurgie, Médecine et Imagerie Fœtales, Hôpital Necker-Enfants Malades, AP-HP, Centre - Université de Paris, Paris, France
| | - Philippe Roth
- Service d'Obstétrique, Maternité, Chirurgie, Médecine et Imagerie Fœtales, Hôpital Necker-Enfants Malades, AP-HP, Centre - Université de Paris, Paris, France
| | - Fabienne Prieur
- Service de génétique, Hôpital Nord CHU Saint-Etienne, Saint Etienne, France
| | - Bettina Bessieres
- Service d'Histologie-Embryologie-Cytogénétique, Unité d'Embryofoetopathologie, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Maryse Bonniere
- Service d'Histologie-Embryologie-Cytogénétique, Unité d'Embryofoetopathologie, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Tania Attie-Bitach
- Service d'Histologie-Embryologie-Cytogénétique, Unité d'Embryofoetopathologie, Hôpital Necker-Enfants Malades, APHP, Paris, France.,INSERM UMR 1163, Université de Paris, Imagine Institute, Paris, France
| |
Collapse
|
34
|
He X, Song J, Cai Z, Chi X, Wang Z, Yang D, Xie S, Zhou J, Fu Y, Li W, Kong W, Zhan J, Zhang H. Kindlin-2 deficiency induces fatal intestinal obstruction in mice. Am J Cancer Res 2020; 10:6182-6200. [PMID: 32483447 PMCID: PMC7255029 DOI: 10.7150/thno.46553] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 04/22/2020] [Indexed: 02/06/2023] Open
Abstract
Rationale: Smooth muscle-motility disorders are mainly characterized by impaired contractility and functional intestinal obstruction. Some of these cases are caused by genetic mutations of smooth muscle genes ACTA2, ACTG2, MYH11, MYLK and LMOD1. Still the etiology is complex and multifactorial and the underlying pathology is poorly understood. Integrin interaction protein Kindlin-2 is widely expressed in striated and smooth muscle cells (SMC). However, the function of Kindlin-2 in the smooth muscle remains elusive. Methods: We generated two mouse models using different cre promoter transgenic mice, Kindlin-2fl/fl SM22α-cre+ (cKO mice) and Kindlin-2fl/fl; MYH-cre+ (iKO mice). Embryos and adult tissues were prepared for hematoxylin and eosin (H&E) staining, immunohistochemistry (IHC) and terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) apoptosis assay. We investigated ultrastructure changes of mouse smooth muscle using transmission electron microscopy (TEM) and measured smooth muscle contractile force in mounting aortic and intestinal rings using the multiwire myograph system (DMT 620M). In addition, cell traction force microscopy (CTFM) was applied to observe the functional change of primary SMC after Kindlin-2 depletion by RNAi. Results: Depletion of Kindlin-2 encoding gene Fermt2 in embryonic smooth muscles leads to apoptosis, downregulates the key components of SMC, impairs smooth muscle development, and finally causes embryonic death at E14.5. Tamoxifen-induced Kindlin-2-specific knockout in adult mouse smooth muscle showed decreased blood pressure, intestinal hypoperistalsis, and eventually died of intestinal obstruction. Kindlin-2 depletion also leads to downregulated Myh11, α-SMA, and CNN, shortened myofilament, broken myofibrils, and impaired contractility of the smooth muscles in iKO mice. Mechanistically, loss of Kindlin-2 decreases Ca2+ influx in primary vascular smooth muscle cells (PVSMC) by downregulating the expression of calcium-binding protein S100A14 and STIM1. Conclusion: We demonstrated that Kindlin-2 is essential for maintaining the normal structure and function of smooth muscles. Loss of Kindlin-2 impairs smooth muscle formation during embryonic development by inducing apoptosis and jeopardizes the contraction of adult smooth muscle by blocking Ca2+ influx that leads to intestinal obstruction. Mice with Kindlin-2 depletion in adult smooth muscle could be a potent animal model of intestinal obstruction for disease research, drug treatment and prognosis.
Collapse
|
35
|
Assia Batzir N, Kishor Bhagwat P, Larson A, Coban Akdemir Z, Bagłaj M, Bofferding L, Bosanko KB, Bouassida S, Callewaert B, Cannon A, Enchautegui Colon Y, Garnica AD, Harr MH, Heck S, Hurst ACE, Jhangiani SN, Isidor B, Littlejohn RO, Liu P, Magoulas P, Mar Fan H, Marom R, McLean S, Nezarati MM, Nugent KM, Petersen MB, Rocha ML, Roeder E, Smigiel R, Tully I, Weisfeld-Adams J, Wells KO, Posey JE, Lupski JR, Beaudet AL, Wangler MF. Recurrent arginine substitutions in the ACTG2 gene are the primary driver of disease burden and severity in visceral myopathy. Hum Mutat 2020; 41:641-654. [PMID: 31769566 PMCID: PMC7720429 DOI: 10.1002/humu.23960] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/04/2019] [Accepted: 11/06/2019] [Indexed: 02/06/2023]
Abstract
Visceral myopathy with abnormal intestinal and bladder peristalsis includes a clinical spectrum with megacystis-microcolon intestinal hypoperistalsis syndrome and chronic intestinal pseudo-obstruction. The vast majority of cases are caused by dominant variants in ACTG2; however, the overall genetic architecture of visceral myopathy has not been well-characterized. We ascertained 53 families, with visceral myopathy based on megacystis, functional bladder/gastrointestinal obstruction, or microcolon. A combination of targeted ACTG2 sequencing and exome sequencing was used. We report a molecular diagnostic rate of 64% (34/53), of which 97% (33/34) is attributed to ACTG2. Strikingly, missense mutations in five conserved arginine residues involving CpG dinucleotides accounted for 49% (26/53) of disease in the cohort. As a group, the ACTG2-negative cases had a more favorable clinical outcome and more restricted disease. Within the ACTG2-positive group, poor outcomes (characterized by total parenteral nutrition dependence, death, or transplantation) were invariably due to one of the arginine missense alleles. Analysis of specific residues suggests a severity spectrum of p.Arg178>p.Arg257>p.Arg40 along with other less-frequently reported sites p.Arg63 and p.Arg211. These results provide genotype-phenotype correlation for ACTG2-related disease and demonstrate the importance of arginine missense changes in visceral myopathy.
Collapse
Affiliation(s)
- Nurit Assia Batzir
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
- Texas Children's Hospital, Houston, Texas
| | - Pranjali Kishor Bhagwat
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas
| | - Austin Larson
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado School of Medicine, Aurora, Colorado
| | - Zeynep Coban Akdemir
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Maciej Bagłaj
- Department of Pediatric Surgery and Urology, Wroclaw Medical University, Wroclaw, Poland
| | - Leon Bofferding
- Département de Pédiatrie Néonatologie, Kannerklinik, Centre Hospitalier de Luxembourg, Luxembourg City, Luxembourg
| | - Katherine B Bosanko
- Section of Genetics and Metabolism, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Skander Bouassida
- Humboldt Clinic, Vivantes Health Network GmbH, Charité Academic Teaching Hospital, Medical University of Berlin, Berlin, Germany
| | - Bert Callewaert
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University Hospital, Ghent, Belgium
| | - Ashley Cannon
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Yazmin Enchautegui Colon
- Inherited Metabolic Diseases Clinic, Section of Clinical Genetics and Metabolism, University of Colorado Denver, Aurora, Colorado
| | - Adolfo D Garnica
- Section of Genetics and Metabolism, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Margaret H Harr
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Sandra Heck
- Département de Pédiatrie Néonatologie, Kannerklinik, Centre Hospitalier de Luxembourg, Luxembourg City, Luxembourg
| | - Anna C E Hurst
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama
| | | | - Bertrand Isidor
- CHU de Nantes, Service de Génétique Médicale, Nantes 44093 Cedex 1, Nantes, France
| | - Rebecca O Littlejohn
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, San Antonio, Texas
| | - Pengfei Liu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Pilar Magoulas
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
- Texas Children's Hospital, Houston, Texas
| | - Helen Mar Fan
- Genetic Health Queensland, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Ronit Marom
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
- Texas Children's Hospital, Houston, Texas
| | - Scott McLean
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, San Antonio, Texas
| | - Marjan M Nezarati
- Genetics Program, North York General Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Kimberly M Nugent
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, San Antonio, Texas
| | | | - Maria L Rocha
- Humboldt Clinic, Vivantes Health Network GmbH, Charité Academic Teaching Hospital, Medical University of Berlin, Berlin, Germany
| | - Elizabeth Roeder
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, San Antonio, Texas
| | - Robert Smigiel
- Department of Pediatrics, Division of Pediatrics and Rare Disorders, Wroclaw Medical University, Wroclaw, Poland
| | - Ian Tully
- Genetic Health Queensland, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - James Weisfeld-Adams
- Inherited Metabolic Diseases Clinic, Section of Clinical Genetics and Metabolism, University of Colorado Denver, Aurora, Colorado
| | - Katerina O Wells
- Department of Surgery, Division of Colorectal Surgery, Baylor University Medical Center, Dallas, Texas
| | - Jennifer E Posey
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - James R Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
- Texas Children's Hospital, Houston, Texas
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Arthur L Beaudet
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
- Texas Children's Hospital, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| | - Michael F Wangler
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
- Texas Children's Hospital, Houston, Texas
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
36
|
Gilbert MA, Schultz-Rogers L, Rajagopalan R, Grochowski CM, Wilkins BJ, Biswas S, Conlin LK, Fiorino KN, Dhamija R, Pack MA, Klee EW, Piccoli DA, Spinner NB. Protein-elongating mutations in MYH11 are implicated in a dominantly inherited smooth muscle dysmotility syndrome with severe esophageal, gastric, and intestinal disease. Hum Mutat 2020; 41:973-982. [PMID: 31944481 DOI: 10.1002/humu.23986] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 12/17/2019] [Accepted: 01/13/2020] [Indexed: 12/12/2022]
Abstract
Gastrointestinal motility disorders include a spectrum of mild to severe clinical phenotypes that are caused by smooth muscle dysfunction. We investigated the genetic etiology of severe esophageal, gastric, and colonic dysmotility in two unrelated families with autosomal dominant disease presentation. Using exome sequencing, we identified a 2 base pair insertion at the end of the myosin heavy chain 11 (MYH11) gene in all affected members of Family 1 [NM_001040113:c.5819_5820insCA(p.Gln1941Asnfs*91)] and a 1 base pair deletion at the same genetic locus in Proband 2 [NM_001040113:c.5819del(p.Pro1940Hisfs*91)]. Both variants are predicted to result in a similarly elongated protein product. Heterozygous dominant negative MYH11 pathogenic variants have been associated with thoracic aortic aneurysm and dissection while biallelic null alleles have been associated with megacystis microcolon intestinal hypoperistalsis syndrome. This report highlights heterozygous protein-elongating MYH11 variants affecting the SM2 isoforms of MYH11 as a cause for severe gastrointestinal dysmotility, and we hypothesize that the mechanistic pathogenesis of this disease, dominant hypercontractile loss-of-function, is distinct from those implicated in other diseases involving MYH11 dysfunction.
Collapse
Affiliation(s)
- Melissa A Gilbert
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia and The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Laura Schultz-Rogers
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota.,Department of Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Ramakrishnan Rajagopalan
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia and The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Christopher M Grochowski
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia and The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Benjamin J Wilkins
- Division of Anatomic Pathology, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia and The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sawona Biswas
- Division of Human Genetics, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Laura K Conlin
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia and The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kristin N Fiorino
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, The Children's Hospital of Philadelphia and The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,The Suzi and Scott Lustgarten Center for GI Motility, Division of Gastroenterology, Hepatology and Nutrition, The Children's Hospital of Philadelphia and The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Radhika Dhamija
- Department of Medical Genetics, Mayo Clinic, Phoenix, Arizona
| | - Michael A Pack
- Division of Gastroenterology, Department of Medicine, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Cell and Developmental Biology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Eric W Klee
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota.,Department of Health Sciences, Mayo Clinic, Rochester, Minnesota.,Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota
| | - David A Piccoli
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, The Children's Hospital of Philadelphia and The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,The Suzi and Scott Lustgarten Center for GI Motility, Division of Gastroenterology, Hepatology and Nutrition, The Children's Hospital of Philadelphia and The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nancy B Spinner
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, The Children's Hospital of Philadelphia and The Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
37
|
Beaman GM, Galatà G, Teik KW, Urquhart JE, Aishah A, O'Sullivan J, Bhaskar SS, Wood KA, Thomas HB, O'Keefe RT, Woolf AS, Stuart HM, Newman WG. A homozygous missense variant in CHRM3 associated with familial urinary bladder disease. Clin Genet 2019; 96:515-520. [PMID: 31441039 PMCID: PMC6899476 DOI: 10.1111/cge.13631] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/19/2019] [Accepted: 08/02/2019] [Indexed: 12/13/2022]
Abstract
CHRM3 codes for the M3 muscarinic acetylcholine receptor that is located on the surface of smooth muscle cells of the detrusor, the muscle that effects urinary voiding. Previously, we reported brothers in a family affected by a congenital prune belly‐like syndrome with mydriasis due to homozygous CHRM3 frameshift variants. In this study, we describe two sisters with bladders that failed to empty completely and pupils that failed to constrict fully in response to light, who are homozygous for the missense CHRM3 variant c.352G > A; p.(Gly118Arg). Samples were not available for genotyping from their brother, who had a history of multiple urinary tract infections and underwent surgical bladder draining in the first year of life. He died at the age of 6 years. This is the first independent report of biallelic variants in CHRM3 in a family with a rare serious bladder disorder associated with mydriasis and provides important evidence of this association.
Collapse
Affiliation(s)
- Glenda M Beaman
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Health Innovation Manchester, Manchester, UK.,Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Human Sciences, University of Manchester, Manchester, UK
| | - Gabriella Galatà
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Health Innovation Manchester, Manchester, UK.,Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Human Sciences, University of Manchester, Manchester, UK
| | - Keng W Teik
- Department of Genetics, Hospital Kuala Lumpur, Kuala Lumpur, Malaysia
| | - Jill E Urquhart
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Health Innovation Manchester, Manchester, UK.,Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Human Sciences, University of Manchester, Manchester, UK
| | - Ali Aishah
- Department of Genetics, Hospital Kuala Lumpur, Kuala Lumpur, Malaysia
| | - James O'Sullivan
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Health Innovation Manchester, Manchester, UK.,Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Human Sciences, University of Manchester, Manchester, UK
| | - Sanjeev S Bhaskar
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Health Innovation Manchester, Manchester, UK.,Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Human Sciences, University of Manchester, Manchester, UK
| | - Katherine A Wood
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Health Innovation Manchester, Manchester, UK.,Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Human Sciences, University of Manchester, Manchester, UK
| | - Huw B Thomas
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Human Sciences, University of Manchester, Manchester, UK
| | - Raymond T O'Keefe
- Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Human Sciences, University of Manchester, Manchester, UK
| | - Adrian S Woolf
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester, UK.,Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Helen M Stuart
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Health Innovation Manchester, Manchester, UK.,Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Human Sciences, University of Manchester, Manchester, UK
| | - William G Newman
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Health Innovation Manchester, Manchester, UK.,Division of Evolution and Genomic Sciences, Faculty of Biology, Medicine and Human Sciences, University of Manchester, Manchester, UK.,Peking University Health Sciences Center, Beijing, China
| |
Collapse
|
38
|
Compound heterozygous variants in MYH11 underlie autosomal recessive megacystis-microcolon-intestinal hypoperistalsis syndrome in a Chinese family. J Hum Genet 2019; 64:1067-1073. [PMID: 31427716 PMCID: PMC6760584 DOI: 10.1038/s10038-019-0651-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 07/24/2019] [Accepted: 07/25/2019] [Indexed: 12/13/2022]
Abstract
Megacystis-microcolon-intestinal-hypoperistalsis syndrome (MMIHS) is a rare and severe disorder characterized by functional obstruction in the urinary and gastrointestinal tract. The molecular basis of this condition has been defined recently. Heterozygous variants in ACTG2, homozygous mutations in LMOD1, MYLK, and MYH9 were related to the pathogenesis of the syndrome, which encodes proteins involved in the process of smooth muscle contraction, supporting a myopathic basis for the disease. Recent studies have identified homozygous or compound heterozygous variants in MYH11 as a candidate gene of MMIHS. In this report, we described a nonconsanguineous Chinese family with three male fetuses affected with megacystis. Trio-targeted exome sequencing identified compound heterozygous variants, c.2051 G > A (p.R684H) and c.3540_3541delinsTT (p.(E1180D, Q1181Ter)), in MYH11 (NM_001040114). The variants were inherited from the parents, respectively. Western blotting showed a marked decrease in MYH11 protein in the proband's umbilical cord tissue compared with the control sample. The study's results confirmed that MYH11 is a candidate gene for MMIHS with autosomal recessive (AR) inheritance and expanded the mutation spectrum for this clinical condition. Combining clinical phenotype with molecular diagnosis may enable the identification of candidate genes for potential monogenic diseases and facilitate accurate genetic counseling, informed decision-making, and prenatal diagnosis.
Collapse
|
39
|
Kloth K, Renner S, Burmester G, Steinemann D, Pabst B, Lorenz B, Simon R, Kolbe V, Hempel M, Rosenberger G. 16p13.11 microdeletion uncovers loss‐of‐function of a
MYH11
missense variant in a patient with megacystis‐microcolon‐intestinal‐hypoperistalsis syndrome. Clin Genet 2019; 96:85-90. [DOI: 10.1111/cge.13557] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/18/2019] [Accepted: 04/28/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Katja Kloth
- Institute of Human GeneticsUniversity Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Sina Renner
- Institute of Human GeneticsUniversity Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Gunter Burmester
- Department of PediatricsAltonaer Kinderkrankenhaus Hamburg Germany
| | - Doris Steinemann
- Department of Human GeneticsMedical Center Hannover Hannover Germany
| | - Brigitte Pabst
- Department of Human GeneticsMedical Center Hannover Hannover Germany
| | | | - Ronald Simon
- Institute of PathologyUniversity Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Verena Kolbe
- Institute of Human GeneticsUniversity Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Maja Hempel
- Institute of Human GeneticsUniversity Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Georg Rosenberger
- Institute of Human GeneticsUniversity Medical Center Hamburg‐Eppendorf Hamburg Germany
| |
Collapse
|
40
|
Huang J, Liu F, Wang B, Tang H, Teng Z, Li L, Qiu Y, Wu H, Chen J. Central and Peripheral Changes in FOS Expression in Schizophrenia Based on Genome-Wide Gene Expression. Front Genet 2019; 10:232. [PMID: 30967896 PMCID: PMC6439315 DOI: 10.3389/fgene.2019.00232] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 03/04/2019] [Indexed: 01/19/2023] Open
Abstract
Schizophrenia is a chronic, debilitating neuropsychiatric disorder. Multiple transcriptomic gene expression profiling analysis has been used to identify schizophrenia-associated genes, unravel disease-associated biomarkers, and predict clinical outcomes. We aimed to identify gene expression regulation, underlying pathways, and their roles in schizophrenia pathogenesis. We searched the Gene Expression Omnibus (GEO) database for microarray studies of fibroblasts, lymphoblasts, and post-mortem brains of schizophrenia patients. Our analysis demonstrated high FOS expression in non-neural peripheral samples and low FOS expression in brain tissues of schizophrenia patients compared with healthy controls. FOS exhibited predictive value for schizophrenia patients in these datasets. Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis revealed that “amphetamine addiction” was among the top 10 significantly enriched KEGG pathways. FOS and FOSB, which are implicated in the amphetamine addiction pathway, were up-regulated in schizophrenia fibroblast samples. Protein–protein interaction (PPI) network analysis revealed that proteins closely interacting with FOS-encoded protein were also involved in the amphetamine addiction pathway. Pearson correlation test indicated that FOS showed positive correlation with genes in the amphetamine pathway. The results revealed that FOS was acceptable as a biomarker for schizophrenia and may be involved in schizophrenia pathogenesis.
Collapse
Affiliation(s)
- Jing Huang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.,Mental Health Institute of the Second Xiangya Hospital, Central South University, Chinese National Clinical Research Center for Mental Disorders (Xiangya), Chinese National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
| | - Fangkun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Bolun Wang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hui Tang
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.,Mental Health Institute of the Second Xiangya Hospital, Central South University, Chinese National Clinical Research Center for Mental Disorders (Xiangya), Chinese National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
| | - Ziwei Teng
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.,Mental Health Institute of the Second Xiangya Hospital, Central South University, Chinese National Clinical Research Center for Mental Disorders (Xiangya), Chinese National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
| | - Lehua Li
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.,Mental Health Institute of the Second Xiangya Hospital, Central South University, Chinese National Clinical Research Center for Mental Disorders (Xiangya), Chinese National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
| | - Yan Qiu
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.,Mental Health Institute of the Second Xiangya Hospital, Central South University, Chinese National Clinical Research Center for Mental Disorders (Xiangya), Chinese National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
| | - Haishan Wu
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.,Mental Health Institute of the Second Xiangya Hospital, Central South University, Chinese National Clinical Research Center for Mental Disorders (Xiangya), Chinese National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
| | - Jindong Chen
- Department of Psychiatry, The Second Xiangya Hospital, Central South University, Changsha, China.,Mental Health Institute of the Second Xiangya Hospital, Central South University, Chinese National Clinical Research Center for Mental Disorders (Xiangya), Chinese National Technology Institute on Mental Disorders, Hunan Key Laboratory of Psychiatry and Mental Health, Changsha, China
| |
Collapse
|
41
|
Camilleri M, Wieben E, Eckert D, Carlson P, O’Dwyer RH, Gibbons D, Acosta A, Klee EW. Familial chronic megacolon presenting in childhood or adulthood: Seeking the presumed gene association. Neurogastroenterol Motil 2019; 31:e13550. [PMID: 30663199 PMCID: PMC6432647 DOI: 10.1111/nmo.13550] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 12/14/2018] [Accepted: 12/18/2018] [Indexed: 12/18/2022]
Abstract
OBJECTIVE We identified a pedigree over five generations with 49 members, some of whom had chronic megacolon presenting in adolescence or adulthood. We aimed to assess the genetic cause of chronic megacolon through clinical and DNA studies. DESIGN After ethical approval and informed consent, family members provided answers to standard bowel disease questionnaires, radiological or surgical records, and DNA (buccal mucosal scraping). Exome DNA sequencing of colon tissue or blood DNA from seven family members with colon or duodenal dilatation, or no megacolon (n = 1) was carried out. Sanger sequencing was performed in 22 additional family members to further evaluate candidate variants. The study focused on genes of potential relevance to enteric nerve (ENS) maturation and Hirschsprung's disease or megacolon, based on the literature (GFRA1, NKX2-1, KIF26A, TPM3, ACTG2, SCN10A, and C17orf107 [CHRNE]) and other genetic variants that co-segregated with megacolon in the six affected family members. RESULTS Information was available in all except five members alive at time of study; among 30 members who provided DNA, six had definite megacolon, one megaduodenum, seven significant constipation without bowel dilatation, and 16 normal bowel function by questionnaire. Among genes studied, SEMA3F (g.3:50225360A>G; c1873A>G) was found in 6/6 family members with megacolon. The SEMA3F gene variant was assessed as potentially pathogenic, based on M-CAP in silico prediction. SEMA3F function is associated with genes (KIT and PDGFRB) that impact intestinal pacemaker function. CONCLUSION Familial chronic megacolon appears to be associated with SEMA3F, which is associated with genes impacting enteric nerve or pacemaker function.
Collapse
Affiliation(s)
- Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Mayo Clinic, Rochester, Minnesota
| | - Eric Wieben
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota
| | - Deborah Eckert
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Mayo Clinic, Rochester, Minnesota
| | - Paula Carlson
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Mayo Clinic, Rochester, Minnesota
| | - Ralph Hurley O’Dwyer
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Mayo Clinic, Rochester, Minnesota
| | - Denys Gibbons
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Mayo Clinic, Rochester, Minnesota
| | - Andres Acosta
- Clinical Enteric Neuroscience Translational and Epidemiological Research (C.E.N.T.E.R.), Mayo Clinic, Rochester, Minnesota
| | - Eric W. Klee
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
42
|
Roberts NA, Hilton EN, Lopes FM, Singh S, Randles MJ, Gardiner NJ, Chopra K, Coletta R, Bajwa Z, Hall RJ, Yue WW, Schaefer F, Weber S, Henriksson R, Stuart HM, Hedman H, Newman WG, Woolf AS. Lrig2 and Hpse2, mutated in urofacial syndrome, pattern nerves in the urinary bladder. Kidney Int 2019; 95:1138-1152. [PMID: 30885509 PMCID: PMC6481288 DOI: 10.1016/j.kint.2018.11.040] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 11/06/2018] [Accepted: 11/21/2018] [Indexed: 12/29/2022]
Abstract
Mutations in leucine-rich-repeats and immunoglobulin-like-domains 2 (LRIG2) or in heparanase 2 (HPSE2) cause urofacial syndrome, a devastating autosomal recessive disease of functional bladder outlet obstruction. It has been speculated that urofacial syndrome has a neural basis, but it is unknown whether defects in urinary bladder innervation are present. We hypothesized that urofacial syndrome features a peripheral neuropathy of the bladder. Mice with homozygous targeted Lrig2 mutations had urinary defects resembling those found in urofacial syndrome. There was no anatomical blockage of the outflow tract, consistent with a functional bladder outlet obstruction. Transcriptome analysis revealed differential expression of 12 known transcripts in addition to Lrig2, including 8 with established roles in neurobiology. Mice with homozygous mutations in either Lrig2 or Hpse2 had increased nerve density within the body of the urinary bladder and decreased nerve density around the urinary outflow tract. In a sample of 155 children with chronic kidney disease and urinary symptoms, we discovered novel homozygous missense LRIG2 variants that were predicted to be pathogenic in 2 individuals with non-syndromic bladder outlet obstruction. These observations provide evidence that a peripheral neuropathy is central to the pathobiology of functional bladder outlet obstruction in urofacial syndrome, and emphasize the importance of LRIG2 and heparanase 2 for nerve patterning in the urinary tract.
Collapse
Affiliation(s)
- Neil A Roberts
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, UK.
| | - Emma N Hilton
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, UK
| | - Filipa M Lopes
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, UK
| | - Subir Singh
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, UK
| | - Michael J Randles
- School of Allied Health Sciences, De Montfort University, Leicester, UK
| | - Natalie J Gardiner
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Karl Chopra
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, UK
| | - Riccardo Coletta
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, UK; Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Zunera Bajwa
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, UK
| | - Robert J Hall
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK; Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Wyatt W Yue
- Structural Genomics Consortium, Nuffield Department of Clinical Medicine, University of Oxford, UK
| | - Franz Schaefer
- Division of Pediatric Nephrology, Centre for Pediatric and Adolescent Medicine, University Hospital of Heidelberg, Im Neuenheimer Feld, Heidelberg, Germany
| | - Stefanie Weber
- Pediatric Nephrology, University-Children's Hospital Marburg, Philipps-University Marburg, Germany
| | - Roger Henriksson
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden; Regional Cancer Center Stockholm/Gotland, Stockholm, Sweden
| | - Helen M Stuart
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK; Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Håkan Hedman
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - William G Newman
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK; Manchester Centre for Genomic Medicine, St. Mary's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Adrian S Woolf
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, UK; Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
43
|
Yetman AT, Starr LJ. Newly described recessive MYH11 disorder with clinical overlap of Multisystemic smooth muscle dysfunction and Megacystis microcolon hypoperistalsis syndromes. Am J Med Genet A 2019; 176:1011-1014. [PMID: 29575632 DOI: 10.1002/ajmg.a.38647] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 01/29/2018] [Accepted: 01/29/2018] [Indexed: 12/12/2022]
Abstract
We describe a neonatal patient with fixed dilated pupils and pulmonary, bladder, and bowel dysfunction suspicious for the presence of ACTA2 R179 mediated multisystemic smooth muscle dysfunction syndrome. Whole exome sequencing revealed compound heterozygous mutations in MYH11 after ACTA2 specific testing revealed no abnormalities. The child lived until 18 months of age and represents the only reported case of an MYH11 compound heterozygote with widespread smooth muscle dysfunction.
Collapse
Affiliation(s)
- Anji T Yetman
- Division of Cardiology, Department of Pediatrics, University of Nebraska Medical Center, Omaha, Nebraska
| | - Lois J Starr
- Division of Medical Genetics, Department of Pediatrics, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
44
|
Elrouby A, Kotb MA, Abdelatty M, Waheeb S. Megacystis-Microcolon-intestinal hypoperistalsis syndrome. JOURNAL OF PEDIATRIC SURGERY CASE REPORTS 2019. [DOI: 10.1016/j.epsc.2018.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
45
|
Nakamura H, O'Donnell AM, Puri P. Consanguinity and its relevance for the incidence of megacystis microcolon intestinal hypoperistalsis syndrome (MMIHS): systematic review. Pediatr Surg Int 2019; 35:175-180. [PMID: 30386895 DOI: 10.1007/s00383-018-4390-6] [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] [Accepted: 10/18/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND/PURPOSE Megacystis microcolon intestinal hypoperistalsis syndrome (MMIHS) is a rare congenital and generally fatal cause of functional intestinal obstruction in the newborn. The cause of this syndrome is unknown. Familial occurrence and reports of consanguinity in MMIHS implies that genetic factors may have an important role in the pathogenesis of this syndrome. The aim of the study was to determine the consequence of consanguinity for the incidence of MMIHS. METHODS A literature search was performed using the keywords "megacystis microcolon intestinal hypoperistalsis" for studies published between 1976 and 2018. Retrieved articles, including additional studies from reference lists, were reviewed for consanguinity between parents and recurrence of MMIHS between siblings. Data were extracted for cases where familial MMIHS was present. RESULTS A total of 450 patients with the diagnosis of MMIHS have been reported in the literature. There were 56 (12%) cases in which familial MMIHS was confirmed, 25 families with multiple siblings and 3 families with single affected infant. Of the 25 families with multiple siblings, 22 families had 2 siblings with confirmed MMIHS and 3 families had 3 children each with MMIHS. Consanguinity between parents was confirmed in 30 cases (18 siblings and 12 individual cases). Female-to-male ratio in the 30 patients was 4.4:1. CONCLUSION The occurrence of MMIHS in the offspring of consanguineous parents and recurrence in siblings of healthy parents suggest that MMIHS is an autosomal recessive disorder. Pre-marital and pre-conception counselling of consanguineous populations is recommended to prevent harmful consequences.
Collapse
Affiliation(s)
- Hiroki Nakamura
- National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland
| | - Anne Marie O'Donnell
- National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland
| | - Prem Puri
- National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland. .,School of Medicine and Medical Science, Conway Institute of Biomolecular and Biomedical, Research University College Dublin, Dublin, Ireland.
| |
Collapse
|
46
|
Beaman GM, Woolf AS, Cervellione RM, Keene D, Mushtaq I, Urquhart JE, Stuart HM, Newman WG. 22q11.2 duplications in a UK cohort with bladder exstrophy–epispadias complex. Am J Med Genet A 2019; 179:404-409. [DOI: 10.1002/ajmg.a.61032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/19/2018] [Accepted: 12/07/2018] [Indexed: 11/09/2022]
Affiliation(s)
- Glenda M. Beaman
- Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of Manchester Manchester United Kingdom
- Manchester Centre for Genomic MedicineManchester University NHS Foundation Trust Manchester United Kingdom
| | - Adrian S. Woolf
- Division of Cell Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of Manchester Manchester United Kingdom
- Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust Manchester United Kingdom
| | - Raimondo M. Cervellione
- Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust Manchester United Kingdom
| | - David Keene
- Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust Manchester United Kingdom
| | - Imran Mushtaq
- Department of Paediatric UrologyGreat Ormond Street Hospital for Children NHS Foundation Trust London United Kingdom
| | - Jill E. Urquhart
- Manchester Centre for Genomic MedicineManchester University NHS Foundation Trust Manchester United Kingdom
| | - Helen M. Stuart
- Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of Manchester Manchester United Kingdom
- Manchester Centre for Genomic MedicineManchester University NHS Foundation Trust Manchester United Kingdom
| | - William G. Newman
- Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and HealthUniversity of Manchester Manchester United Kingdom
- Manchester Centre for Genomic MedicineManchester University NHS Foundation Trust Manchester United Kingdom
- Peking University Health Sciences Center Beijing PR China
| |
Collapse
|
47
|
Woolf AS, Lopes FM, Ranjzad P, Roberts NA. Congenital Disorders of the Human Urinary Tract: Recent Insights From Genetic and Molecular Studies. Front Pediatr 2019; 7:136. [PMID: 31032239 PMCID: PMC6470263 DOI: 10.3389/fped.2019.00136] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 03/22/2019] [Indexed: 12/13/2022] Open
Abstract
The urinary tract comprises the renal pelvis, the ureter, the urinary bladder, and the urethra. The tract acts as a functional unit, first propelling urine from the kidney to the bladder, then storing it at low pressure inside the bladder which intermittently and completely voids urine through the urethra. Congenital diseases of these structures can lead to a range of diseases sometimes associated with fetal losses or kidney failure in childhood and later in life. In some of these disorders, parts of the urinary tract are severely malformed. In other cases, the organs appear grossly intact yet they have functional deficits that compromise health. Human studies are beginning to indicate monogenic causes for some of these diseases. Here, the implicated genes can encode smooth muscle, neural or urothelial molecules, or transcription factors that regulate their expression. Furthermore, certain animal models are informative about how such molecules control the development and functional differentiation of the urinary tract. In future, novel therapies, including those based on gene transfer and stem cell technologies, may be used to treat these diseases to complement conventional pharmacological and surgical clinical therapies.
Collapse
Affiliation(s)
- Adrian S Woolf
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom.,Royal Manchester Children's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Filipa M Lopes
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Parisa Ranjzad
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| | - Neil A Roberts
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology Medicine and Health, School of Biological Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
48
|
Ravenscroft G, Pannell S, O'Grady G, Ong R, Ee HC, Faiz F, Marns L, Goel H, Kumarasinghe P, Sollis E, Sivadorai P, Wilson M, Magoffin A, Nightingale S, Freckmann ML, Kirk EP, Sachdev R, Lemberg DA, Delatycki MB, Kamm MA, Basnayake C, Lamont PJ, Amor DJ, Jones K, Schilperoort J, Davis MR, Laing NG. Variants in ACTG2 underlie a substantial number of Australasian patients with primary chronic intestinal pseudo-obstruction. Neurogastroenterol Motil 2018; 30:e13371. [PMID: 29781137 DOI: 10.1111/nmo.13371] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 04/09/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Primary chronic intestinal pseudo-obstruction (CIPO) is a rare, potentially life-threatening disorder characterized by severely impaired gastrointestinal motility. The objective of this study was to examine the contribution of ACTG2, LMOD1, MYH11, and MYLK mutations in an Australasian cohort of patients with a diagnosis of primary CIPO associated with visceral myopathy. METHODS Pediatric and adult patients with primary CIPO and suspected visceral myopathy were recruited from across Australia and New Zealand. Sanger sequencing of the genes encoding enteric gamma-actin (ACTG2) and smooth muscle leiomodin (LMOD1) was performed on DNA from patients, and their relatives, where available. MYH11 and MYLK were screened by next-generation sequencing. KEY RESULTS We identified heterozygous missense variants in ACTG2 in 7 of 17 families (~41%) diagnosed with CIPO and its associated conditions. We also identified a previously unpublished missense mutation (c.443C>T, p.Arg148Leu) in one family. One case presented with megacystis-microcolon-intestinal hypoperistalsis syndrome in utero with subsequent termination of pregnancy at 28 weeks' gestation. All of the substitutions identified occurred at arginine residues. No likely pathogenic variants in LMOD1, MYH11, or MYLK were identified within our cohort. CONCLUSIONS AND INFERENCES ACTG2 mutations represent a significant underlying cause of primary CIPO with visceral myopathy and associated phenotypes in Australasian patients. Thus, ACTG2 sequencing should be considered in cases presenting with hypoperistalsis phenotypes with suspected visceral myopathy. It is likely that variants in other genes encoding enteric smooth muscle contractile proteins will contribute further to the genetic heterogeneity of hypoperistalsis phenotypes.
Collapse
Affiliation(s)
- G Ravenscroft
- Harry Perkins Institute of Medical Research, University of Western Australia, Nedlands, WA, Australia
| | - S Pannell
- Harry Perkins Institute of Medical Research, University of Western Australia, Nedlands, WA, Australia
| | - G O'Grady
- Department of Surgery, University of Auckland, Auckland, New Zealand
| | - R Ong
- Harry Perkins Institute of Medical Research, University of Western Australia, Nedlands, WA, Australia
| | - H C Ee
- Department of Gastroenterology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - F Faiz
- PathWest Diagnostic Genomics, QE II Medical Centre, Nedlands, WA, Australia
| | - L Marns
- PathWest Diagnostic Genomics, QE II Medical Centre, Nedlands, WA, Australia
| | - H Goel
- Hunter Genetics, Waratah, NSW, Australia
| | - P Kumarasinghe
- Faculty of Medicine and Health Sciences, University of Western Australia, Nedlands, WA, Australia
| | - E Sollis
- Harry Perkins Institute of Medical Research, University of Western Australia, Nedlands, WA, Australia
| | - P Sivadorai
- PathWest Diagnostic Genomics, QE II Medical Centre, Nedlands, WA, Australia
| | - M Wilson
- Department of Clinical Genetics, Children's Hospital at Westmead, Sydney, NSW, Australia
| | - A Magoffin
- Department of Gastroenterology, The Children's Hospital at Westmead, Westmead, NSW, Australia
| | - S Nightingale
- Paediatric Gastroenterology, John Hunter Children's Hospital, Newcastle, NSW, Australia
| | - M-L Freckmann
- ACT Genetics, The Canberra Hospital, Woden, ACT, Australia
| | - E P Kirk
- Centre for Clinical Genetics, Sydney Children's Hospital, Randwick, NSW, Australia
| | - R Sachdev
- Centre for Clinical Genetics, Sydney Children's Hospital, Randwick, NSW, Australia
| | - D A Lemberg
- Department of Paediatric Gastroenterology, Sydney Children's Hospital, Women's and Children's Health, University of New South Wales, Randwick, NSW, Australia
| | - M B Delatycki
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Department of Paediatrics, University of Melbourne, Melbourne, Vic., Australia
| | - M A Kamm
- Department of Gastroenterology, St Vincent's Hospital and University of Melbourne, Melbourne, Vic., Australia
| | - C Basnayake
- Department of Gastroenterology, St Vincent's Hospital and University of Melbourne, Melbourne, Vic., Australia
| | - P J Lamont
- Harry Perkins Institute of Medical Research, University of Western Australia, Nedlands, WA, Australia
| | - D J Amor
- Murdoch Children's Research Institute, Department of Paediatrics, University of Melbourne, Melbourne, Vic., Australia
| | - K Jones
- Faculty of Medicine and Health Sciences, University of Western Australia, Nedlands, WA, Australia
| | - J Schilperoort
- Faculty of Medicine and Health Sciences, University of Western Australia, Nedlands, WA, Australia
| | - M R Davis
- PathWest Diagnostic Genomics, QE II Medical Centre, Nedlands, WA, Australia
| | - N G Laing
- Harry Perkins Institute of Medical Research, University of Western Australia, Nedlands, WA, Australia.,PathWest Diagnostic Genomics, QE II Medical Centre, Nedlands, WA, Australia
| |
Collapse
|
49
|
Lu W, Xiao Y, Huang J, Lu L, Tao Y, Yan W, Cao Y, Cai W. Causes and prognosis of chronic intestinal pseudo-obstruction in 48 subjects: A 10-year retrospective case series. Medicine (Baltimore) 2018; 97:e12150. [PMID: 30200110 PMCID: PMC6133590 DOI: 10.1097/md.0000000000012150] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
The aim of the study was to evaluate the prognosis and survival of pediatric subjects with chronic intestinal pseudo-obstruction (CIPO) and investigate the independent risk factors affecting their prognosis.This was a retrospective case series of all pediatric subjects suffering from CIPO and treated at the Pediatric Surgical ward of Xinhua Hospital between January 2006 and January 2016.The overall mortality was 19/48 (39.6%). Because of delayed CIPO diagnosis, many subjects underwent a variety of surgical procedures. The rate of additional surgical procedures was high (35/48, 72.9%), but the number of surgical procedures, parenteral nutrition, and megacystis did not affect mortality. Mycotic infection was significantly associated with mortality, while onset at <1 year and hypoganglionosis showed a tendency to be associated with mortality.Mycotic infection was associated with mortality of children with CIPO. Despite improving treatment approaches, the overall prognosis of CIPO remains poor. The choice of the surgical intervention could be based on standard criteria.
Collapse
Affiliation(s)
- Wei Lu
- Department of Pediatric Surgery, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University
| | - Yongtao Xiao
- Department of Pediatric Surgery, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University
- Shanghai Institute for Pediatric Research
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Jianhu Huang
- Department of Pediatric Surgery, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University
| | - Lina Lu
- Department of Pediatric Surgery, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University
| | - Yiqing Tao
- Department of Pediatric Surgery, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University
| | - Weihui Yan
- Department of Pediatric Surgery, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University
| | - Yi Cao
- Department of Pediatric Surgery, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University
| | - Wei Cai
- Department of Pediatric Surgery, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University
- Shanghai Institute for Pediatric Research
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| |
Collapse
|
50
|
Takasaki C, Yoshihara T, Yawaka Y. Oral findings in a patient with megacystis microcolon intestinal hypoperistalsis syndrome: A case report. PEDIATRIC DENTAL JOURNAL 2018. [DOI: 10.1016/j.pdj.2018.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|