Proteomic profiling of the interface between the stomach wall and the pancreas in dystrophinopathy


Submitted: 20 January 2021
Accepted: 3 February 2021
Published: 17 February 2021
Abstract Views: 1521
PDF: 628
HTML: 3
Publisher's note
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.

Authors

  • Paul Dowling Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland.
  • Stephen Gargan Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland.
  • Margit Zweyer Department of Neonatology and Paediatric Intensive Care, Children’s Hospital, University of Bonn, Bonn, Germany.
  • Hemmen Sabir Department of Neonatology and Paediatric Intensive Care, Children’s Hospital, University of Bonn, Bonn, Germany.
  • Michael Henry National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland.
  • Paula Meleady National Institute for Cellular Biotechnology, Dublin City University, Dublin 9, Ireland.
  • Dieter Swandulla Institute of Physiology II, University of Bonn, Bonn, Germany.
  • Kay Ohlendieck Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare, Ireland.

The neuromuscular disorder Duchenne muscular dystrophy is a multi-systemic disease that is caused by a primary abnormality in the X-chromosomal Dmd gene. Although progressive skeletal muscle wasting and cardio-respiratory complications are the most serious symptoms that are directly linked to the almost complete loss of the membrane cytoskeletal protein dystrophin, dystrophic patients also suffer from gastrointestinal dysfunction. In order to determine whether proteome-wide changes potentially occur in the gastrointestinal system due to dystrophin deficiency, total tissue extracts from the interface between the stomach wall and the pancreas of the mdx-4cv model of dystrophinopathy were analysed by mass spectrometry. Following the proteomic establishment of both smooth muscle markers of the gastrointestinal system and key enzymes of the pancreas, core members of the dystrophin-glycoprotein complex, including dystrophin, dystroglycans, sarcoglycans, dystrobrevins and syntrophins were identified in this tissue preparation. Comparative proteomics revealed a drastic reduction in dystrophin, sarcoglycan, dystroglycan, laminin, titin and filamin suggesting loss of cytoskeletal integrity in mdx-4cv smooth muscles. A concomitant increase in various mitochondrial enzymes is indicative of metabolic disturbances. These findings agree with abnormal gastrointestinal function in dystrophinopathy.


Mercuri E, Bönnemann CG, Muntoni F. Muscular dystrophies. Lancet. 2019;394(10213):2025-2038. DOI: https://doi.org/10.1016/S0140-6736(19)32910-1

Guiraud S, Aartsma-Rus A, Vieira NM, Davies KE, van Ommen GJ, Kunkel LM. The Pathogenesis and Therapy of Muscular Dystrophies. Annu Rev Genomics Hum Genet. 2015;16:281-308. DOI: https://doi.org/10.1146/annurev-genom-090314-025003

Bladen CL, Salgado D, Monges S, Foncuberta ME, Kekou K, Kosma K, Dawkins H, Lamont L, Roy AJ, Chamova T, Guergueltcheva V, Chan S, Korngut L, Campbell C, Dai Y, Wang J, Barišić N, Brabec P, Lahdetie J, Walter MC, Schreiber-Katz O, Karcagi V, Garami M, Viswanathan V, Bayat F, Buccella F, Kimura E, Koeks Z, van den Bergen JC, Rodrigues M, Roxburgh R, Lusakowska A, Kostera-Pruszczyk A, Zimowski J, Santos R, Neagu E, Artemieva S, Rasic VM, Vojinovic D, Posada M, Bloetzer C, Jeannet PY, Joncourt F, Díaz-Manera J, Gallardo E, Karaduman AA, Topaloğlu H, El Sherif R, Stringer A, Shatillo AV, Martin AS, Peay HL, Bellgard MI, Kirschner J, Flanigan KM, Straub V, Bushby K, Verschuuren J, Aartsma-Rus A, Béroud C, Lochmüller H. The TREAT-NMD DMD Global Database: analysis of more than 7,000 Duchenne muscular dystrophy mutations. Hum Mutat. 2015 Apr;36(4):395-402. DOI: https://doi.org/10.1002/humu.22758

Goemans N, Buyse G. Current treatment and management of dystrophinopathies. Curr Treat Options Neurol. 2014 May;16(5):287. DOI: https://doi.org/10.1007/s11940-014-0287-4

Birnkrant DJ, Bushby K, Bann CM, Apkon SD, Blackwell A, Brumbaugh D, Case LE, Clemens PR, Hadjiyannakis S, Pandya S, Street N, Tomezsko J, Wagner KR, Ward LM, Weber DR; DMD Care Considerations Working Group. Diagnosis and management of Duchenne muscular dystrophy, part 1: diagnosis, and neuromuscular, rehabilitation, endocrine, and gastrointestinal and nutritional management. Lancet Neurol. 2018;17(3):251-267. DOI: https://doi.org/10.1016/S1474-4422(18)30024-3

Birnkrant DJ, Bushby K, Bann CM, Alman BA, Apkon SD, Blackwell A, Case LE, Cripe L, Hadjiyannakis S, Olson AK, Sheehan DW, Bolen J, Weber DR, Ward LM; DMD Care Considerations Working Group. Diagnosis and management of Duchenne muscular dystrophy, part 2: respiratory, cardiac, bone health, and orthopaedic management. Lancet Neurol. 2018 Apr;17(4):347-361. DOI: https://doi.org/10.1016/S1474-4422(18)30025-5

Birnkrant DJ, Bushby K, Bann CM, Apkon SD, Blackwell A, Colvin MK, Cripe L, Herron AR, Kennedy A, Kinnett K, Naprawa J, Noritz G, Poysky J, Street N, Trout CJ, Weber DR, Ward LM; DMD Care Considerations Working Group. Diagnosis and management of Duchenne muscular dystrophy, part 3: primary care, emergency management, psychosocial care, and transitions of care across the lifespan. Lancet Neurol. 2018 May;17(5):445-455. DOI: https://doi.org/10.1016/S1474-4422(18)30026-7

Messina S, Vita GL. Clinical management of Duchenne muscular dystrophy: the state of the art. Neurol Sci. 2018 Nov;39(11):1837-1845. DOI: https://doi.org/10.1007/s10072-018-3555-3

Tidball JG, Welc SS, Wehling-Henricks M. Immunobiology of Inherited Muscular Dystrophies. Compr Physiol. 2018 Sep 14;8(4):1313-1356. DOI: https://doi.org/10.1002/cphy.c170052

Hoffman EP , Hudecki MS, Rosenberg P A, Pollina CM, Kunkel LM. Cell and fiber-type distribution of dystrophin. Neuron. 1988 Jul;1(5):411-20. DOI: https://doi.org/10.1016/0896-6273(88)90191-2

Lionarons JM, Hoogland G, Hendriksen RGF, Faber CG, Hellebrekers DMJ, Van Koeveringe GA, Schipper S, Vles JSH. Dystrophin is expressed in smooth muscle and afferent nerve fibers in the rat urinary bladder. Muscle Nerve. 2019 Aug;60(2): 202- 210. DOI: https://doi.org/10.1002/mus.26518

Barohn RJ, Levine EJ, Olson JO, Mendell JR. Gastric hypomotility in Duchenne's muscular dystrophy. N Engl J Med. 1988 Jul 7;319(1):15-8. DOI: https://doi.org/10.1056/NEJM198807073190103

Jaffe KM, McDonald CM, Ingman E, Haas J. Symptoms of upper gastrointestinal dysfunction in Duchenne muscular dystrophy: case-control study. Arch Phys Med Rehabil. 1990 Sep;71(10):742-4.

Borrelli O, Salvia G, Mancini V, Santoro L, Tagliente F, Romeo EF, Cucchiara S. Evolution of gastric electrical features and gastric emptying in children with Duchenne and Becker muscular dystrophy. Am J Gastroenterol. 2005 Mar;100(3):695-702. DOI: https://doi.org/10.1111/j.1572-0241.2005.41303.x

Lo Cascio CM, Goetze O, Latshang TD, Bluemel S, Frauenfelder T, Bloch KE. Gastrointestinal Dysfunction in Patients with Duchenne Muscular Dystrophy. PLoS One. 2016 Oct 13;11(10):e01 63779. DOI: https://doi.org/10.1371/journal.pone.0163779

Mulè F, Amato A, Serio R. Gastric emptying, small intestinal transit and fecal output in dystrophic (mdx) mice. J Physiol Sci. 2010 Jan;60(1):75-9. DOI: https://doi.org/10.1007/s12576-009-0060-8

Singh K, Randhwa G, Salloum FN, Grider JR, Murthy KS. Decreased smooth muscle function, peristaltic activity, and gastrointestinal transit in dystrophic (mdx) mice. Neurogastroenterol Motil. 2020 Aug 12;e13968. DOI: https://doi.org/10.1111/nmo.13968

Halayko AJ, Stelmack GL. The association of caveolae, actin, and the dystrophin-glycoprotein complex: a role in smooth muscle phenotype and function? Can J Physiol Pharmacol. 2005 Oct;83(10):877-91. DOI: https://doi.org/10.1139/y05-107

Sharma P, Ghavami S, Stelmack GL, McNeill KD, Mutawe MM, Klonisch T, Unruh H, Halayko AJ. beta-Dystroglycan binds caveolin-1 in smooth muscle: a functional role in caveolae distribution and Ca2+ release. J Cell Sci. 2010 Sep 15;123(Pt 18):3061-70. DOI: https://doi.org/10.1242/jcs.066712

Sharma P, Jha A, Stelmack GL, Detillieux K, Basu S, Klonisch T, Unruh H, Halayko AJ. Characterization of the dystrophin-glycoprotein complex in airway smooth muscle: role of delta-sarcoglycan in airway responsiveness. Can J Physiol Pharmacol. 2015;93(3):195-202. DOI: https://doi.org/10.1139/cjpp-2014-0389

Murphy S, Zweyer M, Raucamp M, Henry M, Meleady P, Swandulla D, Ohlendieck K. Proteomic profiling of the mouse diaphragm and refined mass spectrometric analysis of the dystrophic phenotype. J Muscle Res Cell Motil. 2019 Mar;40(1):9-28. DOI: https://doi.org/10.1007/s10974-019-09507-z

22.Gargan S, Dowling P, Zweyer M, Swandulla D, Ohlendieck K. Identification of marker proteins of muscular dystrophy in the urine proteome from the mdx-4cv model of dystrophinopathy. Mol Omics. 2020 Jun 1;16(3):268-278. DOI: https://doi.org/10.1039/C9MO00182D

23. Murphy S, Dowling P, Zweyer M, Henry M, Meleady P, Mundegar RR, Swandulla D, Ohlendieck K. Proteomic profiling of mdx-4cv serum reveals highly elevated levels of the inflammation-induced plasma marker haptoglobin in muscular dystrophy. Int J Mol Med. 2017;39(6):1357-1370. DOI: https://doi.org/10.3892/ijmm.2017.2952

Dowling P, Gargan S, Zweyer M, Henry M, Meleady P , Swandulla D, Ohlendieck K. Proteome-wide Changes in the mdx-4cv Spleen due to Pathophysiological Cross Talk with Dystrophin- Deficient Skeletal Muscle. iScience. 2020 Aug 26;23(9):101500. DOI: https://doi.org/10.1016/j.isci.2020.101500

Dowling P, Gargan S, Zweyer M, Henry M, Meleady P, Swandulla D, Ohlendieck K. Protocol for the Bottom-Up Proteomic Analysis of Mouse Spleen. STAR Protocol. 2020 Dec 3;1(3):100196. DOI: https://doi.org/10.1016/j.xpro.2020.100196

Wilhelm M, Schlegl J, Hahne H, Gholami AM, Lieberenz M, Savitski MM, Ziegler E, Butzmann L, Gessulat S, Marx H, Mathieson T, Lemeer S, Schnatbaum K, Reimer U, Wenschuh H, Mollenhauer M, Slotta-Huspenina J, Boese JH, Bantscheff M, Gerstmair A, Faerber F, Kuster B. Mass- spectrometry-based draft of the human proteome. Nature. 2014 May 29;509(7502):582-7. DOI: https://doi.org/10.1038/nature13319

Kim MS, Pinto SM, Getnet D, Nirujogi RS, Manda SS, Chaerkady R, Madugundu AK, Kelkar DS, Isserlin R, Jain S, Thomas JK, Muthusamy B, Leal- Rojas P , Kumar P , Sahasrabuddhe NA, Balakrishnan L, Advani J, George B, Renuse S, Selvan LD, Patil AH, Nanjappa V, Radhakrishnan A, Prasad S, Subbannayya T, Raju R, Kumar M, Sreenivasamurthy SK, Marimuthu A, Sathe GJ, Chavan S, Datta KK, Subbannayya Y, Sahu A, Yelamanchi SD, Jayaram S, Rajagopalan P, Sharma J, Murthy KR, Syed N, Goel R, Khan AA, Ahmad S, Dey G, Mudgal K, Chatterjee A, Huang TC, Zhong J, Wu X, Shaw PG, Freed D, Zahari MS, Mukherjee KK, Shankar S, Mahadevan A, Lam H, Mitchell CJ, Shankar SK, Satishchandra P, Schroeder JT, Sirdeshmukh R, Maitra A, Leach SD, Drake CG, Halushka MK, Prasad TS, Hruban RH, Kerr CL, Bader GD, Iacobuzio-Donahue CA, Gowda H, Pandey A. A draft map of the human proteome. Nature. 2014 May 29;509(7502):575-81. DOI: https://doi.org/10.1038/nature13302

Uhlén M, Fagerberg L, Hallström BM, Lindskog C, Oksvold P, Mardinoglu A, Sivertsson Å, Kampf C, Sjöstedt E, Asplund A, Olsson I, Edlund K, Lundberg E, Navani S, Szigyarto CA, Odeberg J, Djureinovic D, Takanen JO, Hober S, Alm T, Edqvist PH, Berling H, Tegel H, Mulder J, Rockberg J, Nilsson P, Schwenk JM, Hamsten M, von Feilitzen K, Forsberg M, Persson L, Johansson F, Zwahlen M, von Heijne G, Nielsen J, Pontén F. Proteomics. Tissue-based map of the human proteome. Science. 2015 Jan 23;347(6220): 1260419. DOI: https://doi.org/10.1126/science.1260419

Murphy S, Zweyer M, Henry M, Meleady P, Mundegar RR, Swandulla D, Ohlendieck K. Proteomic profiling of liver tissue from the mdx-4cv mouse model of Duchenne muscular dystrophy. Clin Proteomics. 2018 Oct 29;15:34. DOI: https://doi.org/10.1186/s12014-018-9212-2

Dowling P, Zweyer M, Raucamp M, Henry M, Meleady P, Swandulla D, Ohlendieck K. Proteomic and cell biological profiling of the renal phenotype of the mdx-4cv mouse model of Duchenne muscular dystrophy. Eur J Cell Biol. 2020;99(1):151059. DOI: https://doi.org/10.1016/j.ejcb.2019.151059

Dowling, P., Gargan, S., Zweyer, M., Sabir, H., Henry, M., Meleady, P., Swandulla, D., & Ohlendieck, K. (2021). Proteomic profiling of the interface between the stomach wall and the pancreas in dystrophinopathy. European Journal of Translational Myology, 31(1). https://doi.org/10.4081/ejtm.2021.9627

Downloads

Download data is not yet available.

Citations