Bonesmashing research behind it and how it came around to what it is and how it could actually be useful Full version

godz

godz

Iron
Joined
Aug 19, 2024
Posts
25
Reputation
10
bone smashing.
German anatomist and surgeon Julius Wolff formulated Wolff's Law. Wolff's Law is based on the observation that you and your bones are not like a Ken doll. Rather than being completely inanimate structures, your bones are constantly undergoing remodeling, with old or damaged bone continuously being resorbed and replaced by newly laid bone. Applying mechanical force or physical stress to your bones increases the rate at which such remodeling occurs and, in the process, results in firmer, thicker bones. Conversely, lacking such force or stress can lead to thinner and weaker bones. That's one of the reasons why physical activity, especially weight-bearing exercises, can help maintain bone strength and prevent bone loss.

AD_4nXcBJhEZWmJKPcOgs0Q4H5cEJOMjK9H4F5zGyuWiPFkQ6PFui3EHSvqR54Te9iOpRm-Zn1zcBcYfqgfpos951pEpZ92MRJ8iMEU5hiU161p2eV2qWSeFlhSN0XS8AJLgdwvHTzqbGEQIeF4IvJL2vIqO-WQ


SHIT, I SHOULDN'T have to say, but I can't be too careful
Bone Smashing Doesn't Work
There are several reasons why bone smashing fails as a facial enhancement technique:
  • It doesn't apply controlled, directional forces. Weightlifting triggers bone growth by applying targeted mechanical stress. Bone smashing is uncontrolled blunt force trauma.
  • It causes microfractures and swelling, not bone growth. The trauma leads to swelling and inflammation, which are not beneficial for remodeling.
  • It can't produce symmetrical, predictable results. Injury patterns are random rather than controlled.
  • It risks severe fractures and breaks. Facial bones are vulnerable to fracture and can require complex surgery for repair.

It Doesn't Follow Wolff's Law​

Wolff's Law requires controlled, cyclic loading along stress lines to thicken bone. Think targeted resistance exercise.
Bone smashing is sporadic, undirected trauma. There's no control over where microfractures and swelling occur. You can't reshape bone predictably like working out muscles.
Smashing may microfracture facial bones, but not along defined lines matching your desired result. Such uncontrolled injuries cause haphazard areas of swelling and thickening.

Other Safer, More Effective Options Exist
You now understand why bone smashing is a dangerous DIY practice that is unlikely to improve your facial structure. But if you're seeking facial enhancement, there are effective options available:
  • Fat grafting involves strategically injecting fat cells to add subtle volume and shape.
  • Facial fillers like Voluma and Radiesse can plump facial contours.
  • Customized facial implants can be inserted surgically to augment structure.
  • Osteoplasty surgically sculpts bone for precision reshaping, e.g., genioplasty.
  • MyEllevate is an innovative, minimally invasive procedure to enhance the neck and jawline.



Are you still interested in bone smashing?
Well DONT, to be blunt, what could work well? Cyclic loading could and is currently used to speed up bone fractures. Well, you would be adequately causing growth at the bones and would get results in ways you could cyclic loading. Well, loading has never been in a specific way, but it is the application of a load at a particular amplitude more than once. The load is increased from a minimum to a maximum value, then decreased back to the minimum, and repeated. This process can be used to study how load changes affect a material's strength. For example, the Comparison between the humerus cross-section in the playing arm and nonplaying arm of a professional tennis player.
AD_4nXd3ZL7pnNmqOL_yu8yO9MPWVdzx7_M75JcsXorARYRvKMRsleAaHSGK1SvZNP0DKsAOoos2H7mVDMXkDy8Up_tn1CGfXlxk059q7FL8PY-BR2r9wkwgAj4vsI9CfDduT3vWRSBUkU5eMh4HoE27ogwz8Tw
.
As seen here, there is growth in the bone by less than a cm, but still, in the jaw, a cm half a cm of more bone means it could make you look a lot different. Here is another example: Radiographs of tibiae from two newborns with reduced tibia loading due to (A) congenital neuromuscular disease and (B) spina-bifida. The reduced mechanical environment of the tibia results in a narrow, underdeveloped diaphysis that remains circular and small rather than triangular and large in cross-section. (A) Reprinted from Rodríguez et al.4 with permission from Springer-Verlag New York Inc. (B) Reprinted from Ralis et al.5 with permission from Cambridge University Press.
AD_4nXf_2iBS3TqnVN2ozhatOa-zhHz7jpWWgkja-IDEhk-z9B70a856HsNlaQutVbybjap2pVyw298yaB0KKKIM9F9PpFUth3kgB6jjw2neis3BOEQuCsYuoQg3TamI1PgEgg1Na4ScNcdQRb0EP2OtTCZjqA

Osteocytes within the bone and lining cells on the bone surface are thought to be the primary mechanosensors in bone tissue. Their abundance and connectivity make them an antenna for detecting mechanical strains. These cells are the most abundant in bone — and outnumber osteoblasts and osteoclasts by over 20 to 1. Osteocytes have long dendritic processes that contact surrounding cells at gap junctions. Deformation of the bone matrix caused by mechanical loads is detected by osteocytes, which in turn send paracrine signals to osteoblasts and osteoclasts. Osteocytes form a functional network that passes intracellular calcium signals from cell to cell through gap junctions or by paracrine-like adenosine triphosphate (ATP).7 Lining cells cover over 90% of the surfaces of adult bone, and they contain gap junction connections to osteocytes and osteoblasts. Several genes have been expressed in osteocytes and bone lining cells in vivo after a mechanical stimulus (. These include the proto-oncogene c-fos (30 min after loading) and insulin-like growth factor I (IGF-I) (6 h after loading). Eight osteocytes provide key chemotactic signals that target osteoclasts to repair focal damage and microcracks.9

With all this knowledge and proof, it's safe to say that cyclic loading would work if we could isolate the location down to the mandible and zygomatic bone. Then, yes, it would work. The main issue is figuring out how to cyclic load those bones, which is a partial reason why I made this thread: so people who might be able to think of ideas that could isolate the loading might be able to help, and just general knowledge.


Take everything here with a grain of salt ALSO AS I SAID THIS IS JUST TO LET PEOPLE KNOW​

  • 1656654183894.jpg
  • 69.6 KB · Views: 0

REFERENCES​

1. Wolff J. The law of bone transformation. A. Hirschwald; Berlin: 1892. [Google Scholar]

2. Murray PDF . Bones – A study of the development and structure of the vertebrate skeleton. Cambridge University Press; Cambridge: 1936. [Google Scholar]

3. von Meyer C. Die architektur der spongiosa. Arch Anat Physiol Wiss Med Reichert DuBois-Reymonds Arch. 1867;34:615–28. [Google Scholar]

4. Rodriguez JI, Palacios J, Garcia-Alix A, Pastor I, Paniagua R. Effects of immobilization on fetal bone development. A morphometric study in newborns with congenital neuromuscular diseases with intrauterine onset. Calcif Tissue Int. 1988;43:335–9. [PubMed] [Google Scholar]

5. Ralis ZA, Ralis HM, Randall M, Watkins G, Blake PD. Changes in shape, ossification and quality of bones in children with spina bifida. Dev Med Child Neurol Suppl. 1976:29–41. [PubMed] [Google Scholar]

6. Jones HH, Priest JD, Hayes WC, Tichenor CC, Nagel DA. Humeral hypertrophy in response to exercise. J Bone Joint Surg Am. 1977;59:204–8. [PubMed] [Google Scholar]

7. Jorgensen NR, Geist ST, Civitelli R, Steinberg TH. ATP- and gap junction-dependent intercellular calcium signaling in osteoblastic cells. J Cell Biol. 1997;139:497–506. [PMC free article] [PubMed] [Google Scholar]

8. Lean JM, Mackay AG, Chow JW, Chambers TJ. Osteocytic expression of mRNA for c-fos and IGF-I: an immediate early gene response to an osteogenic stimulus. Am J Physiol. 1996;270:E937–45. [PubMed] [Google Scholar]

9. Verborgt O, Gibson GJ, Schaffler MB. Loss of osteocyte integrity in association with microdamage and bone remodeling after fatigue in vivo. J Bone Miner Res. 2000;15:60–7. [PubMed] [Google Scholar]

10. Robling AG, Duijvelaar KM, Geevers JV, Ohashi N, Turner CH. Modulation of appositional and longitudinal bone growth in the rat ulna by applied static and dynamic force. Bone. 2001;29:105–13. [PubMed] [Google Scholar]

11. Han Y, Cowin SC, Schaffler MB, Weinbaum S. Mechanotransduction and strain amplification in osteocyte cell processes. Proc Natl Acad Sci U S A. 2004;101:16689–94. [PMC free article] [PubMed] [Google Scholar]

12. Wang Y, McNamara LM, Schaffler MB, Weinbaum S. A model for the role of integrins in flow induced mechanotransduction in osteocytes. Proc Natl Acad Sci U S A. 2007;104:15941–6. [PMC free article] [PubMed] [Google Scholar]

13. Hsieh YF, Turner CH. Effects of loading frequency on mechanically induced bone formation. J Bone Miner Res. 2001;16:918–24. [PubMed] [Google Scholar]

14. Warden SJ, Turner CH. Mechanotransduction in the cortical bone is most efficient at loading frequencies of 5–10 Hz. Bone. 2004;34:261–70. [PubMed] [Google Scholar]

15. Burr DB, Martin RB, Schaffler MB, Radin EL. Bone remodeling in response to in vivo fatigue microdamage. J Biomech. 1985;18:189–200. [PubMed] [Google Scholar]

16. Noble BS, Peet N, Stevens HY, Brabbs A, Mosley JR, Reilly GC, Reeve J, Skerry TM, Lanyon LE. Mechanical loading: biphasic osteocyte survival and targeting of osteoclasts for bone destruction in rat cortical bone. Am J Physiol Cell Physiol. 2003;284:C934–43. [PubMed] [Google Scholar]

17. Plotkin LI, Manolagas SC, Bellido T. Glucocorticoids induce osteocyte apoptosis by blocking focal adhesion kinase-mediated survival. Evidence for inside-out signaling leading to anoikis. J Biol Chem. 2007;282:24120–30. [PubMed] [Google Scholar]

18. Robling AG, Castillo AB, Turner CH. Biomechanical and molecular regulation of bone remodeling. Annu Rev Biomed Eng. 2006;8:455–98. [PubMed] [Google Scholar]

19. Torrance AG, Mosley JR, Suswillo RF, Lanyon LE. Noninvasive loading of the rat ulna in vivo induces a strain-related modeling response uncomplicated by trauma or periostal pressure. Calcif Tissue Int. 1994;54:241–7. [PubMed] [Google Scholar]

20. Robling AG, Hinant FM, Burr DB, Turner CH. Improved bone structure and strength after long-term mechanical loading is greatest if loading is separated into short bouts. J Bone Miner Res. 2002;17:1545–54. [PubMed] [Google Scholar]

21. Mattheck C. Design in nature: learning from trees. Springer; Berlin: 1997. [Google Scholar]

22. Warden SJ, Hurst JA, Sanders MS, Turner CH, Burr DB, Li J. Bone adaptation to a mechanical loading program significantly increases skeletal fatigue resistance. J Bone Miner Res. 2005;20:809–16. [PubMed] [Google Scholar]

23. Gong Y, Slee RB, Fukai N, Rawadi G, Roman-Roman S, Reginato AM, Wang H, Cundy T, Glorieux FH, Lev D, Zacharin M, Oexle K, Marcelino J, Suwairi W, Heeger S, Sabatakos G, Apte S, Adkins WN, Allgrove J, Arslan-Kirchner M, Batch JA, Beighton P, Black GC, Boles RG, Boon LM, Borrone C, Brunner HG, Carle GF, Dallapiccola B, De Paepe A, Floege B, Halfhide ML, Hall B, Hennekam RC, Hirose T, Jans A, Juppner H, Kim CA, Keppler-Noreuil K, Kohlschuetter A, LaCombe D, Lambert M, Lemyre E, Letteboer T, Peltonen L, Ramesar RS, Romanengo M, Somer H, Steichen-Gersdorf E, Steinmann B, Sullivan B, Superti-Furga A, Swoboda W, van den Boogaard MJ, Van Hul W, Vikkula M, Votruba M, Zabel B, Garcia T, Baron R, Olsen BR, Warman ML. LDL receptor-related protein 5 (LRP5) affects bone accrual and eye development. Cell. 2001;107:513–23. [PubMed] [Google Scholar]

24. Sawakami K, Robling AG, Ai M, Pitner ND, Liu D, Warden SJ, Li J, Maye P, Rowe DW, Duncan RL, Warman ML, Turner CH. The Wnt co-receptor LRP5 is essential for skeletal mechanotransduction but not for the anabolic bone response to parathyroid hormone treatment. J Biol Chem. 2006;281:23698–711. [PubMed] [Google Scholar]

25. Kiel DP, Ferrari SL, Cupples LA, Karasik D, Manen D, Imamovic A, Herbert AG, Dupuis J. Genetic variation at the low-density lipoprotein receptor-related protein 5 (LRP5) locus modulates Wnt signaling and the relationship of physical activity with bone mineral density in men. Bone. 2007;40:587–96. [PMC free article] [PubMed] [Google Scholar]

26. Liu C, Li Y, Semenov M, Han C, Baeg GH, Tan Y, Zhang Z, Lin X, He X. Control of beta-catenin phosphorylation/degradation by a dual-kinase mechanism. Cell. 2002;108:837–47. [PubMed] [Google Scholar]

27. Hens JR, Wilson KM, Dann P, Chen X, Horowitz MC, Wysolmerski JJ. TOPGAL mice show that the canonical Wnt signaling pathway is active during bone development and growth and is activated by mechanical loading in vitro. J Bone Miner Res. 2005;20:1103–13. [PubMed] [Google Scholar]

28. Norvell SM, Alvarez M, Bidwell JP, Pavalko FM. Fluid shear stress induces beta-catenin signaling in osteoblasts. Calcif Tissue Int. 2004;75:396–404. [PubMed] [Google Scholar]

29. Armstrong VJ, Muzylak M, Sunters A, Zaman G, Saxon LK, Price JS, Lanyon LE. Wnt/beta-catenin signaling is a component of osteoblastic bone cell early responses to load-bearing and requires estrogen receptor alpha. J Biol Chem. 2007;282:20715–27. [PubMed] [Google Scholar]

30. Robinson JA, Chatterjee-Kishore M, Yaworsky PJ, Cullen DM, Zhao W, Li C, Kharode Y, Sauter L, Babij P, Brown EL, Hill AA, Akhter MP, Johnson ML, Recker RR, Komm BS, Bex FJ. Wnt/beta-catenin signaling is a normal physiological response to mechanical loading in bone. J Biol Chem. 2006;281:31720–8. [PubMed] [Google Scholar]

31. Kim-Weroha NA, Ferris A, Holladay B, Kotha SP, Kamel MA, Johnson ML. In vivo load activated propagation of b-catenin signaling in osteocytes through coordinated downregulation of inhibitors of Lrp5. J Bone Miner Res. 2008;23:S13. [Google Scholar]

32. Balemans W, Ebeling M, Patel N, Van Hul E, Olson P, Dioszegi M, Lacza C, Wuyts W, Van Den Ende J, Willems P, Paes-Alves AF, Hill S, Bueno M, Ramos FJ, Tacconi P, Dikkers FG, Stratakis C, Lindpaintner K, Vickery B, Foernzler D, Van Hul W. Increased bone density in sclerosteosis is due to the deficiency of a novel secreted protein (SOST) Hum Mol Genet. 2001;10:537–43. [PubMed] [Google Scholar]

33. Balemans W, Patel N, Ebeling M, Van Hul E, Wuyts W, Lacza C, Dioszegi M, Dikkers FG, Hildering P, Willems PJ, Verheij JB, Lindpaintner K, Vickery B, Foernzler D, Van Hul W. Identification of a 52 kb deletion downstream of the SOST gene in patients with van Buchem disease. J Med Genet. 2002;39:91–7. [PMC free article] [PubMed] [Google Scholar]

34. Semenov M, Tamai K, He X. SOST is a ligand for LRP5/LRP6 and a Wnt signaling inhibitor. J Biol Chem. 2005;280:26770–5. [PubMed] [Google Scholar]

35. Semenov MV, He X. LRP5 mutations linked to high bone mass diseases cause reduced LRP5 binding and inhibition by SOST. J Biol Chem. 2006;281:38276–84. [PubMed] [Google Scholar]

36. Weidauer SE, Schmieder P, Beerbaum M, Schmitz W, Oschkinat H, Mueller TD. NMR structure of the Wnt modulator protein Sclerostin. Biochem Biophys Res Commun. 2009;380:160–5. [PubMed] [Google Scholar]

37. Moustafa A, Sugiyama T, Saxon LK, Zaman G, Sunters A, Armstrong VJ, Javaheri B, Lanyon LE, Price JS. The mouse fibula as a suitable bone for the study of functional adaptation to mechanical loading. Bone. 2009;44:930–5. [PMC free article] [PubMed] [Google Scholar]

38. Robling AG, Niziolek PJ, Baldridge LA, Condon KW, Allen MR, Alam I, Mantila SM, Gluhak-Heinrich J, Bellido TM, Harris SE, Turner CH. Mechanical stimulation of bone in vivo reduces osteocyte expression of Sost/sclerostin. J Biol Chem. 2008;283:5866–75. [PubMed] [Google Scholar]

39. Duncan RL, Turner CH. Mechanotransduction and the functional response of bone to mechanical strain. Calcif Tissue Int. 1995;57:344–58. [PubMed] [Google Scholar]

40. Li J, Duncan RL, Burr DB, Turner CH. L-type calcium channels mediate mechanically induced bone formation in vivo. J Bone Miner Res. 2002;17:1795–800. [PubMed] [Google Scholar]

41. Reich KM, McAllister TN, Gudi S, Frangos JA. Activation of G proteins mediates flow-induced prostaglandin E2 production in osteoblasts. Endocrinology. 1997;138:1014–8. [PubMed] [Google Scholar]

42. Chachisvilis M, Zhang YL, Frangos JA. G protein-coupled receptors sense fluid shear stress in endothelial cells. Proc Natl Acad Sci U S A. 2006;103:15463–8. [PMC free article] [PubMed] [Google Scholar]

43. Leucht P, Kim JB, Currey JA, Brunski J, Helms JA. FAK-Mediated mechanotransduction in skeletal regeneration. PLoS One. 2007;2:e390. [PMC free article] [PubMed] [Google Scholar]

44. Buckbinder L, Crawford DT, Qi H, Ke HZ, Olson LM, Long KR, Bonnette PC, Baumann AP, Hambor JE, Grasser WA, 3rd, Pan LC, Owen TA, Luzzio MJ, Hulford CA, Gebhard DF, Paralkar VM, Simmons HA, Kath JC, Roberts WG, Smock SL, Guzman-Perez A, Brown TA, Li M. Proline-rich tyrosine kinase 2 regulates osteoprogenitor cells and bone formation, and offers an anabolic treatment approach for osteoporosis. Proc Natl Acad Sci U S A. 2007;104:10619–24. [PMC free article] [PubMed] [Google Scholar]

45. Gil-Henn H, Destaing O, Sims NA, Aoki K, Alles N, Neff L, Sanjay A, Bruzzaniti A, De Camilli P, Baron R, Schlessinger J. Defective microtubule-dependent podosome organization in osteoclasts leads to increased bone density in Pyk2(−/−) mice. J Cell Biol. 2007;178:1053–64. [PMC free article] [PubMed] [Google Scholar]

46. Aguirre JI, Plotkin LI, Stewart SA, Weinstein RS, Parfitt AM, Manolagas SC, Bellido T. Osteocyte apoptosis is induced by weightlessness in mice and precedes osteoclast recruitment and bone loss. J Bone Miner Res. 2006;21:605–15. [PubMed] [Google Scholar]

47. Plotkin LI, Mathov I, Aguirre JI, Parfitt AM, Manolagas SC, Bellido T. Mechanical stimulation prevents osteocyte apoptosis: requirement of integrins, Src kinases, and ERKs. Am J Physiol Cell Physiol. 2005;289:C633–43. [PubMed] [Google Scholar]

48. Hung CT, Allen FD, Pollack SR, Brighton CT. Intracellular Ca2+ stores and extracellular Ca2+ are required in the real-time Ca2+ response of bone cells experiencing fluid flow. J Biomech. 1996;29:1411–7. [PubMed] [Google Scholar]

49. Genetos DC, Geist DJ, Liu D, Donahue HJ, Duncan RL. Fluid shear-induced ATP secretion mediates prostaglandin release in MC3T3-E1 osteoblasts. J Bone Miner Res. 2005;20:41–9. [PMC free article] [PubMed] [Google Scholar]

50. Li J, Liu D, Ke HZ, Duncan RL, Turner CH. The P2X7 nucleotide receptor mediates skeletal mechanotransduction. J Biol Chem. 2005;280:42952–9. [PubMed] [Google Scholar]

51. Forwood MR, Kelly WL, Worth NF. Localisation of prostaglandin endoperoxide H synthase (PGHS)-1 and PGHS-2 in bone following mechanical loading in vivo. Anat Rec. 1998;252:580–6. [PubMed] [Google Scholar]

52. Rawlinson SC, el-Haj AJ, Minter SL, Tavares IA, Bennett A, Lanyon LE. Loading-related increases in prostaglandin production in cores of adult canine cancellous bone in vitro: a role for prostacyclin in adaptive bone remodeling? J Bone Miner Res. 1991;6:1345–51. [PubMed] [Google Scholar]

53. Thorsen K, Kristoffersson AO, Lerner UH, Lorentzon RP. In situ microdialysis in bone tissue. Stimulation of prostaglandin E2 release by weight-bearing mechanical loading. J Clin Invest. 1996;98:2446–9. [PMC free article] [PubMed] [Google Scholar]

54. Chow JW, Chambers TJ. Indomethacin has distinct early and late actions on bone formation induced by mechanical stimulation. Am J Physiol. 1994;267:E287–92. [PubMed] [Google Scholar]

55. Forwood MR. Inducible cyclo-oxygenase (COX-2) mediates the induction of bone formation by mechanical loading in vivo. J Bone Miner Res. 1996;11:1688–93. [PubMed] [Google Scholar]

56. Pead MJ, Lanyon LE. Indomethacin modulation of load-related stimulation of new bone formation in vivo. Calcif Tissue Int. 1989;45:34–40. [PubMed] [Google Scholar]

57. Cullen DM, Peyton AC, Akhter MP, Gong G, Johnson ML. Mechanical loading response in EP2 receptor knockout mice. J Bone Miner Res. 2000;15:S476. [Google Scholar]

58. Gong G, Cullen DM, Peyton AC, Akhter MP, Recker RR. EP1 antagonist inhibition of bone formation induced by mechanical loading. J Bone Miner Res. 2000;15:S477. [Google Scholar]

59. Zaman G, Pitsillides AA, Rawlinson SC, Suswillo RF, Mosley JR, Cheng MZ, Platts LA, Hukkanen M, Polak JM, Lanyon LE. Mechanical strain stimulates nitric oxide production by rapid activation of endothelial nitric oxide synthase in osteocytes. J Bone Miner Res. 1999;14:1123–31. [PubMed] [Google Scholar]

60. Rawlinson SC, Pitsillides AA, Lanyon LE. Involvement of different ion channels in osteoblasts’ and osteocytes’ early responses to mechanical strain. Bone. 1996;19:609–14. [PubMed] [Google Scholar]

61. Watanuki M, Sakai A, Sakata T, Tsurukami H, Miwa M, Uchida Y, Watanabe K, Ikeda K, Nakamura T. Role of inducible nitric oxide synthase in skeletal adaptation to acute increases in mechanical loading. J Bone Miner Res. 2002;17:1015–25. [PubMed] [Google Scholar]

62. Rahnert J, Fan X, Case N, Murphy TC, Grassi F, Sen B, Rubin J. The role of nitric oxide in the mechanical repression of RANKL in bone stromal cells. Bone. 2008;43:48–54. [PMC free article] [PubMed] [Google Scholar]

63. Tan SD, Bakker AD, Semeins CM, Kuijpers-Jagtman AM, Klein-Nulend J. Inhibition of osteocyte apoptosis by fluid flow is mediated by nitric oxide. Biochem Biophys Res Commun. 2008;369:1150–4. [PubMed] [Google Scholar]

64. Tan SD, de Vries TJ, Kuijpers-Jagtman AM, Semeins CM, Everts V, Klein-Nulend J. Osteocytes subjected to fluid flow inhibit osteoclast formation and bone resorption. Bone. 2007;41:745–51. [PubMed] [Google Scholar]

65. Hert J, Pribylova E, Liskova M. Reaction of bone to mechanical stimuli. 3 Microstructure of compact bone of rabbit tibia after intermittent loading. Acta Anat (Basel) 1972;82:218–30. [PubMed] [Google Scholar]

66. Rubin CT, Lanyon LE. Regulation of bone formation by applied dynamic loads. J Bone Joint Surg Am. 1984;66:397–402. [PubMed] [Google Scholar]

67. Turner CH, Forwood MR, Rho JY, Yoshikawa T. Mechanical loading thresholds for lamellar and woven bone formation. J Bone Miner Res. 1994;9:87–97. [PubMed] [Google Scholar]

68. Bouvier M, Hylander WL. Effect of bone strain on cortical bone structure in macaques (Macaca mulatta) J Morphol. 1981;167:1–12. [PubMed] [Google Scholar]

69. Bouvier M, Hylander WL. The effect of dietary consistency on gross and histologic morphology in the craniofacial region of young rats. Am J Anat. 1984;170:117–26. [PubMed] [Google Scholar]

70. Lanyon LE, Goodship AE, Pye CJ, MacFie JH. Mechanically adaptive bone remodelling. J Biomech. 1982;15:141–54. [PubMed] [Google Scholar]

71. Lieberman DE, Pearson OM, Polk JD, Demes B, Crompton AW. Optimization of bone growth and remodeling in response to loading in tapered mammalian limbs. J Exp Biol. 2003;206:3125–38. [PubMed] [Google Scholar]

72. Mori S, Burr DB. Increased intracortical remodeling following fatigue damage. Bone. 1993;14:103–9. [PubMed] [Google Scholar]

73. Hillam RA, Skerry TM. Inhibition of bone resorption and stimulation of formation by mechanical loading of the modeling rat ulna in vivo. J Bone Miner Res. 1995;10:683–9. [PubMed] [Google Scholar]

74. Bacabac RG, Smit TH, Mullender MG, Dijcks SJ, Van Loon JJ, Klein-Nulend J. Nitric oxide production by bone cells is fluid shear stress rate dependent. Biochem Biophys Res Commun. 2004;315:823–9. [PubMed] [Google Scholar]

75. Mullender MG, Dijcks SJ, Bacabac RG, Semeins CM, Van Loon JJ, Klein-Nulend J. Release of nitric oxide, but not prostaglandin E2, by bone cells depends on fluid flow frequency. J Orthop Res. 2006;24:1170–7. [PubMed] [Google Scholar]

76. Fan X, Roy E, Zhu L, Murphy TC, Ackert-Bicknell C, Hart CM, Rosen C, Nanes MS, Rubin J. Nitric oxide regulates receptor activator of nuclear factor-kappaB ligand and osteoprotegerin expression in bone marrow stromal cells. Endocrinology. 2004;145:751–9. [PubMed] [Google Scholar]

77. Turner CH, Owan I, Jacob DS, McClintock R, Peacock M. Effects of nitric oxide synthase inhibitors on bone formation in rats. Bone. 1997;21:487–90. [PubMed] [Google Scholar]

78. Fan X, Rahnert JA, Murphy TC, Nanes MS, Greenfield EM, Rubin J. Response to mechanical strain in an immortalized pre-osteoblast cell is dependent on ERK1/2. J Cell Physiol. 2006;207:454–60. [PubMed] [Google Scholar]

79. Rubin J, Murphy TC, Fan X, Goldschmidt M, Taylor WR. Activation of extracellular signal-regulated kinase is involved in mechanical strain inhibition of RANKL expression in bone stromal cells. J Bone Miner Res. 2002;17:1452–60. [PubMed] [Google Scholar]

80. Gross TS, Akeno N, Clemens TL, Komarova S, Srinivasan S, Weimer DA, Mayorov S. Selected Contribution: Osteocytes upregulate HIF-1alpha in response to acute disuse and oxygen deprivation. J Appl Physiol. 2001;90:2514–9. [PubMed] [Google Scholar]

81. Gross TS, King KA, Rabaia NA, Pathare P, Srinivasan S. Upregulation of osteopontin by osteocytes deprived of mechanical loading or oxygen. J Bone Miner Res. 2005;20:250–6. [PMC free article] [PubMed] [Google Scholar]

82. Basso N, Heersche JN. Effects of hind limb unloading and reloading on nitric oxide synthase expression and apoptosis of osteocytes and chondrocytes. Bone. 2006;39:807–14. [PubMed] [Google Scholar]

83. Verborgt O, Tatton NA, Majeska RJ, Schaffler MB. Spatial distribution of Bax and Bcl-2 in osteocytes after bone fatigue: complementary roles in bone remodeling regulation? J Bone Miner Res. 2002;17:907–14. [PubMed] [Google Scholar]

84. Simpson AE, Stoddart MJ, Davies CM, Jahn K, Furlong PI, Gasser JA, Jones DB, Noble BS, Richards RG. TGFbeta3 and loading increases osteocyte survival in human cancellous bone cultured ex vivo. Cell Biochem Funct. 2009;27:23–9. [PubMed] [Google Scholar]

85. Zaman G, Jessop HL, Muzylak M, De Souza RL, Pitsillides AA, Price JS, Lanyon LL. Osteocytes use estrogen receptor alpha to respond to strain but their ERalpha content is regulated by estrogen. J Bone Miner Res. 2006;21:1297–306. [PubMed] [Google Scholar]

86. Rubin J, Murphy T, Nanes MS, Fan X. Mechanical strain inhibits expression of osteoclast differentiation factor by murine stromal cells. Am J Physiol Cell Physiol. 2000;278:C1126–32. [PubMed] [Google Scholar]

87. Liu J, Zhao Z, Zou L, Li J, Wang F, Li X, Zhang J, Liu Y, Chen S, Zhi M, Wang J. Pressure-Loaded MSCs During early osteodifferentiation promote osteoclastogenesis by increase of RANKL/OPG ratio. Ann Biomed Eng. 2009;37:794–802. [PubMed] [Google Scholar]

88. Mehrotra M, Saegusa M, Wadhwa S, Voznesensky O, Peterson D, Pilbeam C. Fluid flow induces Rankl expression in primary murine calvarial osteoblasts. J Cell Biochem. 2006;98:1271–83. [PubMed] [Google Scholar]

89. Kim CH, You L, Yellowley CE, Jacobs CR. Oscillatory fluid flow-induced shear stress decreases osteoclastogenesis through RANKL and OPG signaling. Bone. 2006;39:1043–7. [PubMed] [Google Scholar]

90. Sanchez C, Gabay O, Salvat C, Henrotin YE, Berenbaum F. Mechanical loading highly increases IL-6 production and decreases OPG expression by osteoblasts. Osteoarthritis Cartilage. 2009;17(4):473–81. [PubMed] [Google Scholar]

91. David V, Laroche N, Boudignon B, Lafage-Proust MH, Alexandre C, Ruegsegger P, Vico L. Noninvasive in vivo monitoring of bone architecture alterations in hindlimb-unloaded female rats using novel three-dimensional microcomputed tomography. J Bone Miner Res. 2003;18:1622–31. [PubMed] [Google Scholar]

92. Tanaka S, Sakai A, Tanaka M, Otomo H, Okimoto N, Sakata T, Nakamura T. Skeletal unloading alleviates the anabolic action of intermittent PTH(1-34) in mouse tibia in association with inhibition of PTH-induced increase in c-fos mRNA in bone marrow cells. J Bone Miner Res. 2004;19:1813–20. [PubMed] [Google Scholar]

93. Martin A, de Vittoris R, David V, Moraes R, Begeot M, Lafage-Proust MH, Alexandre C, Vico L, Thomas T. Leptin modulates both resorption and formation while preventing disuse-induced bone loss in tail-suspended female rats. Endocrinology. 2005;146:3652–9. [PubMed] [Google Scholar]

94. Ichinose Y, Tanaka H, Inoue M, Mochizuki S, Tsuda E, Seino Y. Osteoclastogenesis inhibitory factor/osteoprotegerin reduced bone loss induced by mechanical unloading. Calcif Tissue Int. 2004;75:338–43. [PubMed] [Google Scholar]

95. Bateman TA, Dunstan CR, Ferguson VL, Lacey DL, Ayers RA, Simske SJ. Osteoprotegerin mitigates tail suspension-induced osteopenia. Bone. 2000;26:443–9. [PubMed] [Google Scholar]

96. Umemura Y, Ishiko T, Yamauchi T, Kurono M, Mashiko S. Five jumps per day increase bone mass and breaking force in rats. J Bone Miner Res. 1997;12:1480–5. [PubMed] [Google Scholar]

97. Turner CH. Three rules for bone adaptation to mechanical stimuli. Bone. 1998;23:399–407. [PubMed] [Google Scholar]

98. Forwood MR, Turner CH. The response of rat tibiae to incremental bouts of mechanical loading: a quantum concept for bone formation. Bone. 1994;15:603–9. [PubMed] [Google Scholar]

99. Srinivasan S, Weimer DA, Agans SC, Bain SD, Gross TS. Low-magnitude mechanical loading becomes osteogenic when rest is inserted between each load cycle. J Bone Miner Res.l. 2002;17:1613–20. [PMC free article] [PubMed] [Google Scholar]

100. Robling AG, Burr DB, Turner CH. Recovery periods restore mechanosensitivity to dynamically loaded bone. J Exp Biol. 2001;204:3389–99. [PubMed] [Google Scholar]

101. Robling AG, Hinant FM, Burr DB, Turner CH. Shorter, more frequent mechanical loading sessions enhance bone mass. Med Sci Sports Exerc. 2002;34:196–202. [PubMed] [Google Scholar]

102. Saxon LK, Robling AG, Alam I, Turner CH. Mechanosensitivity of the rat skeleton decreases after a long period of loading, but is improved with time off. Bone. 2005;36:454–64. [PubMed] [Google Scholar]
 
Legally have to include sources cause thats how it be also side note this is not all info but it would've just to much so theres a basic understanding so people can learn and try and help me
 
If I hit my cranium at the sides can it become wider?
 

Similar threads

the MOUSE
Replies
30
Views
4K
highschoolmaxxer
highschoolmaxxer
the MOUSE
Replies
57
Views
13K
Tylermax
Tylermax
D
Replies
36
Views
5K
Deleted member 24712
D

Users who are viewing this thread

  • anka
Back
Top