#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Abnormal Activation of BMP Signaling Causes Myopathy in Null Mice


New strategies for treating congenital muscular dystrophies are needed. Current treatments are limited and aim to prolong ambulation and survival. Since most of the genes responsible for congenital muscular dystrophies are still unknown, elucidation of these genes may provide new insights that can lead to novel treatments. Fibrillin-2 null mice are born with myopathy and contractures and demonstrate accumulation of white fat during the early postnatal period. Both the histological features of myopathy and the accumulation of fat are rescued by inhibiting BMP signaling. Results indicate that FBN2 is a candidate gene for congenital muscular dystrophy and that strategies aimed at inhibition of abnormal BMP signaling may be applicable to muscular dystrophies. Furthermore, results reveal the importance of extracellular control of BMP signaling in skeletal muscle.


Vyšlo v časopise: Abnormal Activation of BMP Signaling Causes Myopathy in Null Mice. PLoS Genet 11(6): e32767. doi:10.1371/journal.pgen.1005340
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1005340

Souhrn

New strategies for treating congenital muscular dystrophies are needed. Current treatments are limited and aim to prolong ambulation and survival. Since most of the genes responsible for congenital muscular dystrophies are still unknown, elucidation of these genes may provide new insights that can lead to novel treatments. Fibrillin-2 null mice are born with myopathy and contractures and demonstrate accumulation of white fat during the early postnatal period. Both the histological features of myopathy and the accumulation of fat are rescued by inhibiting BMP signaling. Results indicate that FBN2 is a candidate gene for congenital muscular dystrophy and that strategies aimed at inhibition of abnormal BMP signaling may be applicable to muscular dystrophies. Furthermore, results reveal the importance of extracellular control of BMP signaling in skeletal muscle.


Zdroje

1. Sakai L. Y., Keene D. R., Engvall E., Fibrillin, a new 350kD glycoprotein, is a component of extracellular microfibrils. J. Cell Biol. 103, 2499–2509 (1986).

2. Reinhardt D. P., Keene D. R., Corson G. M., Pӧschl E., Bächinger H. P., Gambee J. E., Sakai L. Y., Fibrillin-1: organization in microfibrils and structural properties. J. Mol. Biol. 258, 104–116 (1996).

3. Charbonneau N. L., Carlson E. J., Tufa S., Sengle G., Manalo E. C., Carlberg V. M., Ramirez F., Keene D. R., Sakai L. Y., In Vivo Studies of Mutant Fibrillin-1 Microfibrils. J. Biol. Chem. 285, 24943–24955 (2010).

4. Isogai Z., Ono R. N., Ushiro S., Keene D. R., Chen Y., Mazzieri R., Charbonneau N. L., Reinhardt D. P., Rifkin D. B., Sakai L.Y., Latent transforming growth factor beta-binding protein 1 interacts with fibrillin and is a microfibril-associated protein. J. Biol. Chem. 278, 2750–2757 (2003).

5. Gregory K. E., Ono R. N., Charbonneau N. L., Kuo C-L., Keene D. R., Bächinger H. P., Sakai L. Y., The Prodomain of BMP-7 Targets the BMP-7 Complex to the Extracellular Matrix. J. Biol. Chem. 280, 27970–27980 (2005).

6. Sengle G., Charbonneau N. L., Ono R. N., Sasaki T., Alvarez J., Keene D. R., Bächinger H. P., Sakai L. Y., Targeting of Bone Morphogenetic Protein Growth Factor Complexes to fibrillin. J. Biol. Chem. 283, 13874–13888 (2008).

7. Corson G. M., Charbonneau N. L., Keene D. R., Sakai L. Y., Differential expression of fibrillin-3 adds to microfibril variety in human and avian, but not rodent, connective tissues. Genomics 83, 461–472 (2004).

8. Zhang H., Hu W., Ramirez F., Developmental expression of fibrillin genes suggests heterogeneity of extracellular microfibrils. J. Cell Biol. 129, 1165–1176 (1995).

9. Charbonneau N. L., Jordan C. D., Keene D. R., Lee-Arteaga S., Dietz H. C., Rifkin D. B., Ramirez F., Sakai L. Y., Microfibril Structure Masks Fibrillin-2 in Postnatal Tissues. J. Biol. Chem. 285, 20242–20251 (2010).

10. Belleh S., Zhou G., Wang M., Der Kaloustian V. M., Pagon R. A., Godfrey M., Two novel fibrillin-2 mutations in congenital contractural arachnodactyly. Am. J. Med. Genet. 92, 7–12 (2000).

11. Arteaga-Solis E., Gayraud B., Lee S. Y., Shum L., Sakai L., Ramirez F., Regulation of Limb Patterning By Extracellular Microfibrils. J. Cell Biol. 154, 275–281 (2001).

12. Chernousov M. A., Baylor K., Stahl R. C., Stecker M. M., Sakai L. Y., Lee-Arteaga S., Ramirez F., Carey D. J., Fibrillin-2 Is Dispensable for Peripheral Nerve Development, Myelination and Regeneration. Matrix Biol. 29, 357–368 (2010).

13. Miller G., Neilan M., Chia R., Gheryani N., Holt N., Charbit A., Wells S., Tucci V., Lalanne Z., Denny P., Fisher E. M. C., Cheeseman M., Askew G. N., Dear T. N., ENU Mutagenesis Reveals a Novel Phenotype of Reduced Limb Strength in Mice Lacking Fibrillin 2. PLoS ONE 5, e9137 (2010).

14. Neptune E. R., Frischmeyer P. A., Arking D. E., Myers L., Bunton T. E., Gayraud B., Ramirez F., Sakai L. Y., Dietz H. C., Dysregulation of TGFβ Activation Contributes to Pathogenesis in Marfan Syndrome. Nat. Genet. 33, 407–411 (2003).

15. Habashi J. P., Judge D. P., Holm T. M., Cohn R. D., Loeys B. L., Cooper T. K., Meyers L., Klein E. C., Liu G., Calvi C., Podowski M., Neptune E. R., Halushka M. K., Bedja D., Gabrielson K., Rifkin D. B., Carta L., Ramirez F., Huso D. L., Dietz H. C., Losartan, an AT1 Antagonist, Prevents Aortic Aneurysm in a Mouse Model of Marfan Syndrome. Science 312, 117–121 (2006).

16. Cohn R. D., van Erp C., Habashi J. P., Soleimani A. A., Klein E. C., Lisi M. T., Gamradt M., ap Rhys C. M., Holm T. M., Loeys B. L., Ramirez F., Judge D. P., Ward C. W., Dietz H. C., Angiotensin II Type 1 Receptor Blockade Attenuates TGF-beta-induced Failure of Muscle Regeneration in Multiple Myopathic States. Nat. Med. 13, 204–210 (2007).

17. Faivre L., Gorlin R. J., Wirtz M. K., Godfrey M., Dagoneau N., Samples J. R., Le Merrer M., Collod-Beroud G., Boileau C., Munnich A., Cormier-Daire V., In frame fibrillin-1 gene deletion in autosomal dominant Weill-Marchesani syndrome. J. Med. Genet. 40, 34–36 (2003).

18. Sengle G., Tsutsui K., Keene D. R., Tufa S. F., Carlson E. J., Charbonneau N. L., Ono R. N., Sasaki T., Wirtz M. K., Samples J. R., Fessler L. I., Fessler J. H., Sekiguchi K., Hayflick S. J., Sakai L. Y., Microenvironmental reglation by fibrillin-1. PLoS Genet. 8, e1002425 (2012).

19. Le Goff C., Mahaut C., Wang L. W., Allali S., Abhyankar A., Jensen S., Zylberberg L., Collod-Beroud G., Bonnet D., Alanay Y., Brady A. F., Cordier M. P., Devriendt K., Genevieve D., Kiper P.O., Kitoh H., Krakow D., Lynch S. A., Le Merrer M., Megarbane A., Mortier G., Odent S., Polak M., Rohrbach M., Sillence D., Stolte-Dijkstra I., Superti-Furga A., Rimoin D. L., Topouchian V., Unger S., Zabel B., Bole-Feysot C., Nitschke P., Handford P., Casanova J. L., Boileau C., Apte S. S., Munnich A., Cormier-Daire V., Mutations in the TGFβ binding-protein-like domain 5 of FBN1 are responsible for acromicric and geleophysic dysplasias. Am. J. Hum. Genet. 89, 7–14 (2011).

20. Sartori R., Schirwis E., Blaauw B., Bortolanza S., Zhao J., Enzo E., Stantzou A., Mouisel E., Toniolo L., Ferry A., Stricker S., Goldberg A. L., Dupont S., Piccolo S., Amthor H., Sandri M., BMP signaling controls muscle mass. Nat. Genet. 45, 1309–1318 (2013).

21. Winbanks C. E., Chen J. L., Qian H., Liu Y., Bernardo B. C., Beyer C., Watt K. I., Thomson R. E., Connor T., Turner B. J., McMullen J. R., Larsson L., McGee S. L., Harrison C. A., Gregorevic P., The bone morphogenetic protein axis is a positive regulator of skeletal muscle mass. J. Cell Biol. 203, 345–357 (2013).

22. Huang P., Schulz T. J., Beauvais A., Tseng Y. H., Gussoni E., Intramuscular adipogenesis is inhibited by myo-endothelial progenitors with functioning Bmpr1a signaling. Nat. Commun. 5, 4063 (2014).

23. Peat R. A., Smith J. M., Compton A. G., Baker N. L., Pace R. A., Burkin D. J., Kaufman S. J., Lamande S. R., North K. N., Diagnosis and Etiology of Congenital Muscular Dystrophy. Neurology 71, 312–321 (2008).

24. Maddox B. K., Sakai L. Y., Keene D. R., Glanville R. W., Connective Tissue Microfibrils: Isolation and Characterization of Three Large Pepsin-Resistant Domains of Fibrillin. J. Biol. Chem. 264, 21381–21385 (1989).

25. Veugelers M., Bressan M., McDermott D. A., Weremowicz S., Morton C. C., Mabry C. C., Lefaivre J. F., Zunamon A., Destree A., Chaudron J. M., Basson C. T., Mutation of Perinatal Myosin Heavy Chain Associated With a Carney Comlex Variant. N. Engl. J. Med. 351, 460–469 (2004).

26. Katagiri T., Yamaguchi A., Komaki M., Abe E., Takahashi N., Ikeda T., Rosen V., Wozney J. M., Fujisawa-Sehara A., Suda T., Bone Morphogenetic Protein-2 Converts the Differentiation Pathway of C2C12 Myoblasts Into the Osteoblast Lineage. J. Cell Biol. 127, 1755–1766 (1994).

27. Dudley A. T., Lyons K. M., Robertson E. J., A Requirement for Bone Morphogenetic Protein-7 During Development of the Mammalian Kidney and Eye. Genes Dev. 9, 2795–2807 (1995).

28. Zilberberg L., ten Dijke P., Sakai L. Y., Rifkin D. B., A Rapid and Sensitive Bioassay to Measure Bone Morphogenetic Protein Activity. BMC Cell Biol. 8, 41 (2007).

29. Sengle G., Ono R. N., Sasaki T., Sakai L. Y., Prodomains of Transforming Growth Factor-β Superfamily Members Specify Different Functions: Extracellular Matrix Interactions and Growth Factor Bioavailability. J. Biol. Chem. 286, 5087–5099 (2011).

30. Sporn M. B., The early history of TGF-beta, and a brief glimpse of its future. Cytokine Growth Factor Rev. 17, 3–7 (2006).

31. Wakefield L. M., Smith D. M., Broz S., Jackson M., Levinson A. D., Sporn M. B., Recombinant TGF-beta 1 Is Synthesized as a Two-Component Latent Complex that Shares Some Structural Features with the Native Platelet Latent TGF-beta 1 Complex. Growth Factors 1, 203–218 (1989).

32. Lee S. J., McPherron A. C., Regulation of Myostatin Activity and Muscle Growth. Proc. Natl. Acad. Sci. USA 98, 9306–9311 (2001).

33. Ge G., Hopkins D. R., Ho W. B., Greenspan D. S., GDF11 Forms a Bone Morphogenetic Protein 1-activated Latent Complex that Can Modulate Nerve Growth Factor-induced Differentiation of PC12 Cells. Mol Cell Biol. 25, 5846–5858 (2005).

34. Sengle G., Ono R. N., Lyons K. M., Bächinger H. P., Sakai L. Y., A New Model for Growth Factor Activation: Type II Receptors Compete With the Prodomain For BMP-7. J. Mol. Biol. 381, 1025–1039 (2008).

35. Mi L. Z., Brown C. T., Gao Y., Tian Y., Le V. Q., Walz T., Springer T. A., Structure of Bone Morphogenetic Protein 9 Procomplex. Proc. Natl. Acad. Sci. U. S. A. 112, 3710–3715 (2015).

36. Shi M., Zhu J., Wang R., Chen X., Mi L., Walz T., Springer T. A., Latent TGF-β Structure and Activation. Nature 474, 343–349 (2011).

37. Socol M. L., Sabbagha R. E., Elias S., Tamura R. K., Simpson J. L., Dooley S. L., Depp R., Prenatal Diagnosis of Congenital Muscular Dystrophy Producing Arthrogryposis. N. Engl. J. Med. 313, 1230 (1985).

38. Heydemann A., Ceco E., Lim J. E., Hadhazy M., Ryder P., Moran J. L., Beier D. R., Palmer A. A., McNally E. M., Latent TGF-β-Binding Protein 4 Modifies Muscular Dystrophy in Mice. J. Clin. Invest. 119, 3703–3712 (2009).

39. Flanigan K. M., Ceco E., . Lamar K-M, Kaminoh Y., Dunn D. M., Mendell J. R., King W. M., Pestronk A., Florence J. M., Mathews K. D., Finkel R. S., Swoboda K. J., Gappmaier E., Howard M. T., Day J. W., McDonald C., McNally E. M., Weiss R. B., United Dystrophinopathy Project, LTBP4 Genotype Predicts Age of Ambulatory Loss in Duchenne Muscular Dystrophy. Ann. Neurol. 73, 481–488 (2013).

40. Shi S., Hoogaars W. M., de Gorter D. J., van Heiningen S. H., Lin H. Y., Hong C. C., Kemaladewi D. U., Aartsma-Rus A., ten Dijke P., ‘t Hoen P. A., BMP Antagonists Enhance Myogenic Differentiation and Ameliorate the Dystrophic Phenotype in a DMD Mouse Model. Neurobiol. Dis. 41, 353–360 (2011).

41. Tseng Y-H., Kokkotou E., Schulz T. J., Huang T. L., Winnay J. N., Taniguchi C. M., Tran T. T., Suzuki R., Espinoza D. O., Yamamoto Y., Ahrens M. J., Dudley A. T., Norris A. W., Kulkarni R. N., Kahn C. R., New role of bone morphogenetic protein 7 in brown adipogenesis and energy expenditure. Nature 454, 1000–1004 (2008).

42. Schulz T. J., Huang T. L., Tran T. T., Zhang H., Townsend K. L., Shadrach J. L., Cerletti M., McDougall L. E., Giorqadze N., Tchkonia T., Schrier D., Falb D., Kirkland J. L., Wagers A. J., Tseng Y-H., Identification of inducible brown adipocyte progenitors residing in skeletal muscle and white fat. Proc. Natl. Acad. Sci. USA 108, 143–148 (2011).

43. Ono R. N., Sengle G., Charbonneau N. L., Bächinger H. P., Sasaki T., Lee-Arteaga S., Zilberberg L., Rifkin D. B., Ramirez F., Chu M. L., Sakai L. Y., LTBPs and Fibulins Compete for Fibrillin-1 and Exhibit Exquisite Specificities in Binding Sites. J. Biol. Chem. 284, 16872–6881 (2009).

44. Livak K. J., Schmittgen T. D., Analysis of Relative Gene Expression Data Using Real-Time Quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25, 402–408 (2001). 11846609

45. Sengle G., Tufa S. F., Sakai L. Y., Zulliger M. A., Keene D. R., A Correlative Method for Imaging Identical Regions of Samples by Micro-CT, Light Microscopy, and Electron Microscopy: Imaging Adipose Tissue in a Model System. J. Histochem. Cytochem. 61, 263–271 (2013).

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2015 Číslo 6
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#