Osteoclast-Mediated Bone Disease: Comparison
Please note this is a comparison between Version 3 by J. Kelly Smith and Version 7 by Vicky Zhou.

Astronauts are at risk of losing 1.0% to 1.5% of their bone mass for every month they spend in space despite their adherence to diets and exercise regimens designed to protect their skeletal systems. This loss is the result of microgravity-related impairment of osteocyte and osteoblast function and the consequent upregulation of osteoclast-mediated bone resorption. This entry describes the ontogeny of osteoclast hematopoietic stem cells and the contributions macrophage colony stimulating factor (M-CSF), receptor activator of nuclear factor kappa-B ligand (RANKL), and the calcineurin pathways make in osteoclast differentiation and provides details of bone formation, the osteoclast cytoskeleton, the immune regulation of osteoclasts, and osteoclast mechanotransduction on Earth, in space, and under conditions of simulated microgravity. The entry discusses the need to better understand how osteoclasts are able to function in zero gravity and reviews current and prospective therapies that may be used to treat osteoclast-mediated bone disease.

Astronauts are at risk of losing 1.0% to 1.5% of their bone mass for every month they spend in space despite their adherence to diets and exercise regimens designed to protect their skeletal systems. This loss is the result of microgravity-related impairment of osteocyte and osteoblast function and the consequent upregulation of osteoclast-mediated bone resorption. This review describes the ontogeny of osteoclast hematopoietic stem cells and the contributions macrophage colony stimulating factor (M-CSF), receptor activator of nuclear factor kappa-B ligand (RANKL), and the calcineurin pathways make in osteoclast differentiation and provides details of bone formation, the osteoclast cytoskeleton, the immune regulation of osteoclasts, and osteoclast mechanotransduction on Earth, in space, and under conditions of simulated microgravity. The article discusses the need to better understand how osteoclasts are able to function in zero gravity and reviews current and prospective therapies that may be used to treat osteoclast-mediated bone disease.

  • osteoclasts
  • spaceflight
  • osteoblasts
  • osteocytes
  • M-CSF
  • RANKL
  • bone
  • microgravity
  • cytokines

1. Introduction

The skeletal system of vertebrates has had millions of years to adapt to the force of gravity on Earth (9.8 m/s

2) and to allow osteocytes and the immune system to balance the activities of osteoblasts and osteoclasts. This osteoimmunological system is complex and involves commonly shared osteoclastogenic factors such as receptor activator of the nuclear factor-kappa B (NF-κB) ligand and macrophage colony stimulating factor as well as cytokines and immune cells that inhibit or enhance osteoclast ontogeny [1]. This adaptation has involved the construction of cytoskeletons supported by actin and intermediate filaments and microtubules [2][3], intracellular adhesion molecules including integrins and cell extension kinases [4][5], plasma membrane and nuclear mechanosensors [6], and thermal convection currents which renew nutrients and remove waste [7].

) and to allow osteocytes and the immune system to balance the activities of osteoblasts and osteoclasts. This osteoimmunological system is complex and involves commonly shared osteoclastogenic factors such as receptor activator of the nuclear factor-kappa B (NF-κB) ligand and macrophage colony stimulating factor as well as cytokines and immune cells that inhibit or enhance osteoclast ontogeny [1]. This adaptation has involved the construction of cytoskeletons supported by actin and intermediate filaments and microtubules [2,3], intracellular adhesion molecules including integrins and cell extension kinases [4,5], plasma membrane and nuclear mechanosensors [6], and thermal convection currents which renew nutrients and remove waste [7].

Man’s venture into the vacuum of space where the force of gravity is one millionth of that on Earth has resulted in adverse effects on the osteoimmunological system, particularly bone homeostasis [8]. Astronauts are not only at risk of progressive bone and cartilage loss while in space but must also face the reality that space-related bone and joint changes may persist for years after their return to Earth despite efforts made to protect their skeletal systems [8][9][10][11][12][13].

Man’s venture into the vacuum of space where the force of gravity is one millionth of that on Earth has resulted in adverse effects on the osteoimmunological system, particularly bone homeostasis [8]. Astronauts are not only at risk of progressive bone and cartilage loss while in space but must also face the reality that space-related bone and joint changes may persist for years after their return to Earth despite efforts made to protect their skeletal systems [8,9,10,11,12,13].

2. Treatment of Osteoclast-Mediated Bone Disease

Unloading of bone on Earth and in the microgravity of space is associated with decreased bone formation and increased bone resorption. The reasons are complex but include the reduction of bone-loading signals normally transduced by shear stresses and hydraulic pressures exerted on osteocytes residing in the lacunar–canalicular network of bone, the increased secretion of sclerostin by pre-apoptotic osteocytes, and microgravity-induced disruption of osteoblast nuclei, cytoskeleton, and intracellular adhesins.

Experiments have been consistent in showing that conditions of microgravity and simulated microgravity decrease the mineral content and cortical and trabecular microstructures of bone, increase osteoclast secretion of sclerostin, decrease osteoblastogenesis and osteoblast secretion of OPG, and increase osteoclast differentiation, fusion, and expression of regulatory and osteoclast-specific genes [14][15][16][17][18][19][20][21][22][23][24][25][26]. However, why, in contrast, to osteocytes and osteoblasts, do osteoclasts with their complex cytoskeletons, remain functional under conditions of bone unloading—both on Earth and in space? How is the integrity of the sealing zone, so essential for bone resorption, maintained under such adverse conditions? What happens to the complex associations of actin and intermediate filaments, septins, and microtubules in osteoclasts subjected to microgravity? Why are M-CSF, RANKL, and calcineurin transcriptional pathways upregulated in osteoclast hematopoietic stems cells but downregulated in osteoblast mesenchymal stem cell precursors in zero gravity? Moreover, is there any relation between osteoclast survival and space-related changes in osteoclast regulation by immune cells and their cytokines? These intriguing questions should provide an ample basis for future research into the amazingly resilient osteoclast, including the development of agents capable of disabling key elements in its cytoskeleton.

Experiments have been consistent in showing that conditions of microgravity and simulated microgravity decrease the mineral content and cortical and trabecular microstructures of bone, increase osteoclast secretion of sclerostin, decrease osteoblastogenesis and osteoblast secretion of OPG, and increase osteoclast differentiation, fusion, and expression of regulatory and osteoclast-specific genes [57,58,59,60,61,62,63,64,65,66,67,68,69]. However, why, in contrast, to osteocytes and osteoblasts, do osteoclasts with their complex cytoskeletons, remain functional under conditions of bone unloading—both on Earth and in space? How is the integrity of the sealing zone, so essential for bone resorption, maintained under such adverse conditions? What happens to the complex associations of actin and intermediate filaments, septins, and microtubules in osteoclasts subjected to microgravity? Why are M-CSF, RANKL, and calcineurin transcriptional pathways upregulated in osteoclast hematopoietic stems cells but downregulated in osteoblast mesenchymal stem cell precursors in zero gravity? Moreover, is there any relation between osteoclast survival and space-related changes in osteoclast regulation by immune cells and their cytokines? These intriguing questions should provide an ample basis for future research into the amazingly resilient osteoclast, including the development of agents capable of disabling key elements in its cytoskeleton.

In addition to bone unloading, a number of physiopathological conditions are characterized by excessive osteoclast activity. These include but are not limited to menopause, juvenile Paget’s disease of bone, inflammatory joint diseases, bone cancers such as multiple myeloma, and glucocorticoid therapy [1][2]. Thus, it is not surprising that many of the studies on bone homeostasis have been motivated by the need to find treatments capable of modifying osteoclast activity without inducing osteopetrosis [8]. I have listed below several potential treatments designed for this purpose.

In addition to bone unloading, a number of physiopathological conditions are characterized by excessive osteoclast activity. These include but are not limited to menopause, juvenile Paget’s disease of bone, inflammatory joint diseases, bone cancers such as multiple myeloma, and glucocorticoid therapy [1,2]. Thus, it is not surprising that many of the studies on bone homeostasis have been motivated by the need to find treatments capable of modifying osteoclast activity without inducing osteopetrosis [8]. I have listed below several potential treatments designed for this purpose.

2.1. Biphosphonates

Bisphosphonates have long been used with success to control osteoclast-mediated bone disease; these agents are incorporated into the bone matrix and are ingested by bone-resorbing osteoclasts, causing their apoptosis. However, biphosphonates inhibit the stimulatory activity of osteoclasts on osteoblast differentiation and, as a consequence, patients on these drugs suffer from a blockade of de novo bone formation [12][27].

Bisphosphonates have long been used with success to control osteoclast-mediated bone disease; these agents are incorporated into the bone matrix and are ingested by bone-resorbing osteoclasts, causing their apoptosis. However, biphosphonates inhibit the stimulatory activity of osteoclasts on osteoblast differentiation and, as a consequence, patients on these drugs suffer from a blockade of de novo bone formation [12,70].

2.2. Anti-RANKL Antibody

A recently developed human monoclonal antibody against RANKL, denosumab, has been shown to have undesirable side effects and, similar to biphosphonates, adversely affects osteoblastogenesis [28].

A recently developed human monoclonal antibody against RANKL, denosumab, has been shown to have undesirable side effects and, similar to biphosphonates, adversely affects osteoblastogenesis [71].

2.3. Cathepsin K Inhibitor

An inhibitor of cathepsin K, odanacatib, was shown to prevent pathological bone loss while preserving bone formation but failed in clinical phase III trials due to increased risk of stroke [29].

An inhibitor of cathepsin K, odanacatib, was shown to prevent pathological bone loss while preserving bone formation but failed in clinical phase III trials due to increased risk of stroke [72].

2.4. Anti-Sclerostin Antibody

Scientists have developed a humanized monoclonal antibody directed against sclerostin (romosozumab), which is approved for the treatment of osteoporosis. Clinical trials have shown that monthly subcutaneous injections of romosozumab are effective in increasing bone formation and density and decreasing bone resorption—results in keeping with the known effects of sclerostin on bone homeostasis [30]. However, there is some concern about the potential cardiotoxicity of romosozumab, prompting the need for further observations [31]. In addition, there is evidence in experimental animals that sclerostin generated in response to TNF-α and IL-1β improves post-traumatic osteoarthritis by inhibiting the activity of proteolytic enzymes involved in cartilage degradation [32]. Because astronauts experience an increased incidence of post-traumatic osteoarthritis involving their knees, ankles, hips, and shoulders, their use of anti-sclerostin antibody during spaceflight may prove to be a double-edge sword. It is important to note that both TNF-α and IL-1β play an important role in the pathophysiology of osteoarthritis [33].

Scientists have developed a humanized monoclonal antibody directed against sclerostin (romosozumab), which is approved for the treatment of osteoporosis. Clinical trials have shown that monthly subcutaneous injections of romosozumab are effective in increasing bone formation and density and decreasing bone resorption—results in keeping with the known effects of sclerostin on bone homeostasis [73]. However, there is some concern about the potential cardiotoxicity of romosozumab, prompting the need for further observations [74]. In addition, there is evidence in experimental animals that sclerostin generated in response to TNF-α and IL-1β improves post-traumatic osteoarthritis by inhibiting the activity of proteolytic enzymes involved in cartilage degradation [75]. Because astronauts experience an increased incidence of post-traumatic osteoarthritis involving their knees, ankles, hips, and shoulders, their use of anti-sclerostin antibody during spaceflight may prove to be a double-edge sword. It is important to note that both TNF-α and IL-1β play an important role in the pathophysiology of osteoarthritis [76].

2.5. Osteoprotegerin

Osteoprotegerin-Fc given subcutaneously to mice flown for 12 days in space produced a sustained suppression of bone resorption and, thus, deserves further study [34].

Osteoprotegerin-Fc given subcutaneously to mice flown for 12 days in space produced a sustained suppression of bone resorption and, thus, deserves further study [77].

2.6. Melatonin

Ikegame and associates reported that melatonin, a well-tolerated and widely available compound, stimulated calcitonin mRNA expression and decreased RANKL mRNA expression in cultured fish scales (a surrogate for bone cultures) during an 11-day space flight aboard the International Space Shuttle. Calcitonin is an osteoclast-inhibiting hormone, and, as previously noted, RANKL binding to RANK is required for osteoclastogenesis. The authors posited that melatonin might prove useful in preventing space-related bone loss and, thus, deserves further evaluation [35].

Ikegame and associates reported that melatonin, a well-tolerated and widely available compound, stimulated calcitonin mRNA expression and decreased RANKL mRNA expression in cultured fish scales (a surrogate for bone cultures) during an 11-day space flight aboard the International Space Shuttle. Calcitonin is an osteoclast-inhibiting hormone, and, as previously noted, RANKL binding to RANK is required for osteoclastogenesis. The authors posited that melatonin might prove useful in preventing space-related bone loss and, thus, deserves further evaluation [78].

2.7. Insulin-Like Growth Factor-1

Insulin-like growth factor (IGF)-1, which plays a major role in all phases of bone and cartilage growth, has been shown to increase rodent humerus periosteal bone formation by 37% during a 10 day Space Shuttle flight [8]. The potential of IGF-1 and other growth factors such as TGF-β and BMP to regulate bone homeostasis in situations of bone unloading merits further investigation.

2.8. Anti-IL-6 neutralizing antibody (IL-6nAb)

IL-6nAb treatment has been shown by He and associates to enhance bone formation in the tibias and femurs of adult mice subjected to simulated microgravity (hindlimb suspension for 4 weeks). They further demonstrated that mIL-6Ab treatment increased osteoprotegerin production and mRNA expression of alkaline phosphatase, osteopontin, and Runx2, and decreased RANKL production in murine pre-osteoblasts cultured under conditions of simulated microgravity. In cultures of murine pre-osteoclasts, simulated microgravity reduced mRNA expression of

cathepsin K and TRAP and reduced numbers of TRAP positive multinucleated osteoclasts.

and reduced numbers of TRAP positive multinucleated osteoclasts [79].

[361]

3. Conclusions

Astronauts are at risk of losing bone mass and damaging joint cartilages despite NASA’s efforts to protect their skeletal system by initiating exercise programs and nutritious diets. Bone loss is the consequence of microgravity-related impairment of osteocyte and osteoblast function and the consequent upregulation of osteoclast-mediated bone resorption. Further research is needed to better understand how osteoclasts are able to function in zero gravity and to develop more effective interventions to prevent osteoclast-mediated bone disease.

References

  1. Okamoto, K.; Nakashima, T.; Shinohara, M.; Negishi-Koga, T.; Komatsu, N.; Terashima, A.; Sawa, S.; Nitta, T.; Takayanagi, H. Osteoimmunology: The conceptual framework unifying the immune and skeletal systems. Physiol. Rev. 2017, 97, 1295–1349.He, B.; Yin, X.; Hao, D.; Zhang, X.; Zhang, Z.; Zhang, K.; Yang, X.; Blockade of IL-6 alleviates bone loss induced by modeled microgravity in mice. Canadian J. Physiol. Pharmacol. 2020, -, -, doi.org/10.1139/cjpp-2019-0632.
  2. Blangy, A.; Bompard, G.; Guerit, D.; Marie, P.; Maurin, J.; Morel, A.; Vives, V. The osteoclast—Current understanding and therapeutic perspectives for osteoporosis. J. Cell Sci. 2020, 133, jcs244798.
  3. Svitkina, T. The actin cytoskeleton and actin-based motility. Cold Spring Harb. Perspect. Biol. 2018, 10, a018267.
  4. LaFlamme, S.E.; Mathew-Steiner, S.; Singh, N.; Colello-Borges, D.; Nieves, B. Integrin and microtubule crosstalk in the regulation of cellular processes. Cell Mol. Life Sci. 2018, 75, 4177–4185.
  5. Martínez, P.T.; Navajas, P.L.; Lietha, D. FAK Structure and regulation by membrane interactions and force in focal adhesions. Biomolecules 2020, 10, 179.
  6. Burke, B. Chain reaction: LINC complexes and nuclear positioning. F1000Research 2019, 8, 136.
  7. Howard, R.; Scheiner, A.; Cunningham, J.; Gatenby, R. Cytoplasmic convection currents and intracellular temperature gradients. PLoS Comput. Biol. 2019, 15, e1007372.
  8. Smith, J.K. Microgravity, bone homeostasis, and insulin-like growth factor-1. Appl. Sci. 2020, 10, 4433.
  9. Iwamoto, J.; Takeda, T.; Sato, Y. Interventions to prevent bone loss in astronauts during space flight. Keio J. Med. 2005, 54, 55–59.
  10. Cavanagh, P.R.; Licata, A.A.; Rice, A.J. Exercise and pharmacological countermeasures for bone loss during long-duration space flight. Gravit. Space Biol. Bull. 2005, 18, 39–58.
  11. Smith, S.M.; McCoy, T.; Gazda, D.; Morgan, J.L.L.; Heer, M.; Zwart, S.R. Space flight calcium: Implications for astronaut health, spaceflight operations, and Earth. Nutrients 2012, 4, 2047–2068.
  12. Leblanc, A.; Matsumoto, T.; Jones, J.; Shapiro, J.; Lang, T.; Shackelford, L.; Smith, S.M.; Evans, H.; Spector, E.; Ploutz-Snyder, R.; et al. Biphosphonates as a supplement to exercise to protect bone during long-duration spaceflight. Osteoporos. Int. 2013, 24, 2105–2114.
  13. NASA—Food for Space Flight. Available online: https:/www.nasa.gov/specials/apollo50thy/index.html (accessed on 12 June 2020).
  14. Vico, L.; Chappard, D.; Palle, S.; Bakulin, A.V.; Novikov, V.E.; Alexandre, C. Trabecular bone remodeling after seven days of weightlessness exposure (BIOCOSMOS 1667). Am. J. Physiol. 1988, 255, R243–R247.
  15. Vico, L.; Chappard, D.; Alexandre, C.; Palle, S.; Minaire, P.; Riffat, G.; Novikov, V.E.; Bakulin, A.V. Effects of weightlessness on bone mass and osteoclast number in pregnant rats after a five-day spaceflight (COSMOS 1514). Bone 1987, 8, 95–103.
  16. Vico, L.; Bourrin, S.; Genty, C.; Palle, S.; Alexandre, C. Histomorphometric analysis of cancellous bone from COSMOS 2044 rats. J. Appl. Physiol. 1985, 75, 2203–2208.
  17. Blaber, E.A.; Dvorochkin, N.; Lee, C.; Alwood, J.S.; Yousuf, R.; Pianetta, P.; Globus, R.K.; Burns, B.P.; Almeida, E.A. Microgravity induces pelvic bone loss through osteoclastic activity, osteocytic osteolysis, and osteoblastic cell cycle inhibition by CDKN1a/p21. PLoS ONE 2013, 8, e61372.
  18. Chatani, M.; Morimoto, H.; Takeyama, K.; Mantoku, A.; Tanigawa, N.; Kubota, K.; Suzuki, H.; Uchida, S.; Tanigaki, F.; Shirakawa, M.; et al. Acute transcriptional up-regulation specific to osteoblasts/osteoclasts in medaka fish immediately after exposure to microgravity. Sci. Rep. 2016, 6, 39545.
  19. Tamma, R.; Colaianni, G.; Camerino, C.; Di Benedetto, A.; Greco, G.; Strippoli, M.; Vergari, R.; Grano, A.; Mancini, L.; Mori, G.; et al. Microgravity during spaceflight directly affects in vitro osteoclastogenesis and bone resorption. FASEB J. 2009, 23, 2549–2554.
  20. Saxena, R.; Pan, G.; Dohm, E.D.; McDonald, J.M. Modeled microgravity and hindlimb unloading sensitize osteoclast precursors to RANKL-mediated osteoclastogenesis. J. Bone Miner. Metab. 2011, 29, 111–122.
  21. Aguirre, J.I.; Plotkin, L.I.; Stewart, S.A.; Weinstein, R.S.; Parfitt, A.M.; Manolagas, S.C.; Bellido, T. Osteocyte apoptosis is induced by weightlessness in mice and precedes osteoclast recruitment and bone loss. J. Bone Miner. Res. 2006, 21, 605–615.
  22. Rucci, N.; Rufo, A.; Alamanou, M.; Teti, A. Modeled microgravity stimulates osteoclastogenesis and bone resorption by increasing osteoblast RANKL/OPG ratio. J. Cell. Biochem. 2007, 100, 464–473.
  23. Sambandam, Y.; Blanchard, J.J.; Daughtridge, G.; Kolb, R.J.; Shanmugarajan, S.; Pandruvada, S.N.M.; Bateman, T.A.; Reddy, S.V. Microarray profile of gene expression during osteoclast differentiation in modelled microgravity. J. Cell. Biochem. 2010, 111, 1179–1187.
  24. Sambandam, Y.; Townsend, T.; Pierce, J.J.; Lipman, C.M.; Haque, A.; Bateman, T.A.; Reddy, S.V. Microgravity control of autophagy modulates osteoclastogenesis. Bone 2014, 61, 125–131.
  25. Sambandam, Y.; Baird, K.L.; Stroebel, M.; Kowai, E.; Balasubramanian, S.; Reddy, S.V. Microgravity induction of TRAIL expression in preosteoclast cells enhances osteoclast differentiation. Sci. Rep. 2016, 6, 25143.
  26. Ethiraj, P.; Link, J.R.; Sinkway, J.M.; Brown, C.D.; Parler, W.A.; Reddy, S.V. Microgravity modulation of syncytin-A expression enhance osteoclast formation. J. Cell. Biochem. 2018, 119, 5695–5703.
  27. Vargas-Franco, J.W.; Castaneda, B.; Rédini, F.; Gómez, D.F.; Heymann, D.; Lézot, F. Paradoxical side effects of biphosphonates on the skeleton: What do we know and what can we do? J. Cell. Physiol. 2018, 233, 5696–5715.
  28. Deeks, E.D. Denosumab: A review in postmenopausal osteoporosis. Drugs Aging 2018, 35, 163–173.
  29. Stone, J.A.; McCrea, J.B.; Witter, R.; Zajic, S.; Stoch, S.A. Clinical and translational pharmacology of the cathepsin K inhibitor odanacatib studied for osteoporosis. Br. J. Clin. Pharmacol. 2019, 85, 1072–1083.
  30. Bandeira, L.; Lewiecki, E.M.; Bilezikian, J.P. Romosozumab for treatment of osteoporosis. Expert Opin. Biol. Ther. 2017, 17, 255–263.
  31. Shakeri, A.; Adanty, C. Romosozumab (sclerostin monoclonal antibody) for the treatment of osteoporosis in postmenopausal women: A review. J. Popul. Ther. Clin. Pharmacol. 2020, 27, e25–e31.
  32. Chang, J.C.; Christiansen, B.A.; Murugesh, D.K.; Sebastian, A.; Hum, N.R.; Collette, N.M.; Hatsell, S.; Economides, A.N.; Blanchette, C.D.; Loots, G.G. SOST/Sclerostin improves posttraumatic osteoarthritis and inhibits MMP2/3 expression after injury. J. Bone Miner. Res. 2018, 33, 1105–1113.
  33. Kapoor, M.; Martel-Pelletier, J.; Lajeunesse, D.; Pelletier, J.-P.; Fahmi, H. Role of proinflammatory cytokines in the pathophysiology of osteoarthritis. Nat. Rev. Rheumatol. 2011, 7, 33–42.
  34. Lloyd, S.A.; Morony, S.E.; Ferguson, V.L.; Simske, S.J.; Stodieck, L.S.; Warmington, K.S.; Livingston, E.W.; Lacey, D.L.; Kostenuik, P.J.; Bateman, T.A. Osteoprotegerin is an effective countermeasure for spaceflight-induced bone loss in mice. Bone 2015, 81, 562–572.
  35. Ikegame, M.; Hattori, A.; Tabata, M.J.; Kitamura, K.; Tabuchi, Y.; Furusawa, Y.; Maruyama, Y.; Yamamoto, T.; Sekiguchi, T.; Matsuoka, R.; et al. Melatonin is a potential drug for the prevention of bone loss during spaceflight. J. Pineal Res. 2019, 67, e1259w.
  36. He, B.; Yin, X.; Hao, D.; Zhang, X.; Zhang, Z.; Zhang, K.; Yang, X.; Blockade of IL-6 alleviates bone loss induced by modeled microgravity in mice. Canadian J. Physiol. Pharmacol. 2020, -, -, doi.org/10.1139/cjpp-2019-0632.
More