1
|
Alvarez-Artime A, Garcia-Soler B, Gonzalez-Menendez P, Fernandez-Vega S, Cernuda-Cernuda R, Hevia D, Mayo JC, Sainz RM. Castration promotes the browning of the prostate tumor microenvironment. Cell Commun Signal 2023; 21:267. [PMID: 37770940 PMCID: PMC10536697 DOI: 10.1186/s12964-023-01294-y] [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: 06/27/2023] [Accepted: 08/23/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Adipose tissue has gained attention due to its potential paracrine role. Periprostatic adipose tissue surrounds the prostate and the prostatic urethra, and it is an essential player in prostate cancer progression. Since obesity is directly related to human tumor progression, and adipose tissue depots are one of the significant components of the tumor microenvironment, the molecular mediators of the communication between adipocytes and epithelial cells are in the spotlight. Although periprostatic white adipose tissue contributes to prostate cancer progression, brown adipose tissue (BAT), which has beneficial effects in metabolic pathologies, has been scarcely investigated concerning cancer progression. Given that adipose tissue is a target of androgen signaling, the actual role of androgen removal on the periprostatic adipose tissue was the aim of this work. METHODS Surgical castration of the transgenic adenocarcinoma of the mouse prostate (TRAMP) was employed. By histology examination and software analysis, WAT and BAT tissue was quantified. 3T3-like adipocytes were used to study the role of Casodex® in modifying adipocyte differentiation and to investigate the function of the secretome of adipocytes on the proliferation of androgen-dependent and independent prostate cancer cells. Finally, the role of cell communication was assayed by TRAMP-C1 xenograft implanted in the presence of 3T3-like adipocytes. RESULTS Androgen removal increases brown/beige adipose tissue in the fat immediately surrounding the prostate glands of TRAMP mice, concomitant with an adjustment of the metabolism. Castration increases body temperature, respiratory exchange rate, and energy expenditure. Also, in vitro, it is described that blocking androgen signaling by Casodex® increases the uncoupling protein 1 (UCP1) marker in 3T3-like adipocytes. Finally, the effect of brown/beige adipocyte secretome was studied on the proliferation of prostate cancer cells in vivo and in vitro. The secretome of brown/beige adipocytes reduces the proliferation of prostate cancer cells mediated partly by the secretion of extracellular vesicles. CONCLUSIONS Consequently, we concluded that hampering androgen signaling plays a crucial role in the browning of the periprostatic adipose tissue. Also, the presence of brown adipocytes exhibits the opposite effect to that of white adipocytes in vitro regulating processes that govern the mechanisms of cell proliferation of prostate cancer cells. And finally, promoting the browning of adipose tissue in the periprostatic adipose tissue might be a way to handle prostate cancer cell progression. Video Abstract.
Collapse
Affiliation(s)
- Alejandro Alvarez-Artime
- Departamento de Morfologia y Biologia Celular, Facultad de Medicina, University of Oviedo, Julian Claveria 6, 33006, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario, 33011, Oviedo, Spain
| | - Belen Garcia-Soler
- Departamento de Morfologia y Biologia Celular, Facultad de Medicina, University of Oviedo, Julian Claveria 6, 33006, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario, 33011, Oviedo, Spain
| | - Pedro Gonzalez-Menendez
- Departamento de Morfologia y Biologia Celular, Facultad de Medicina, University of Oviedo, Julian Claveria 6, 33006, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario, 33011, Oviedo, Spain
| | - Sheila Fernandez-Vega
- Departamento de Morfologia y Biologia Celular, Facultad de Medicina, University of Oviedo, Julian Claveria 6, 33006, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario, 33011, Oviedo, Spain
| | - Rafael Cernuda-Cernuda
- Departamento de Morfologia y Biologia Celular, Facultad de Medicina, University of Oviedo, Julian Claveria 6, 33006, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario, 33011, Oviedo, Spain
| | - David Hevia
- Departamento de Morfologia y Biologia Celular, Facultad de Medicina, University of Oviedo, Julian Claveria 6, 33006, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario, 33011, Oviedo, Spain
| | - Juan C Mayo
- Departamento de Morfologia y Biologia Celular, Facultad de Medicina, University of Oviedo, Julian Claveria 6, 33006, Oviedo, Spain.
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006, Oviedo, Spain.
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario, 33011, Oviedo, Spain.
| | - Rosa M Sainz
- Departamento de Morfologia y Biologia Celular, Facultad de Medicina, University of Oviedo, Julian Claveria 6, 33006, Oviedo, Spain.
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), 33006, Oviedo, Spain.
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Avda. Hospital Universitario, 33011, Oviedo, Spain.
| |
Collapse
|
2
|
Gordon BS, Burns PK, Laskin GR, Dunlap KR, Boykin JR, Rossetti ML, Fukuda DH, Steiner JL. SIRT1 induction in the skeletal muscle of male mice partially preserves limb muscle mass but not contractile force in response to androgen deprivation. J Physiol 2023; 601:3885-3903. [PMID: 37531448 DOI: 10.1113/jp284869] [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: 04/11/2023] [Accepted: 07/13/2023] [Indexed: 08/04/2023] Open
Abstract
In males, the factors that decrease limb muscle mass and strength in response to androgen deprivation are largely unknown. Sirtuin1 (SIRT1) protein levels are lower in the limb muscle of male mice subjected to androgen deprivation. The present study aimed to assess whether SIRT1 induction preserved limb muscle mass and force production in response to androgen deprivation. Physically mature male mice containing an inducible muscle-specific SIRT1 transgene were subjected to a sham or castration surgery and compared to sham and castrated male mice where the SIRT1 transgene was not induced. SIRT1 induction partially preserved whole-body lean mass, tibialis anterior (TA) mass and triceps surae muscle mass in response to castration. Further analysis of the TA muscle showed that muscle-specific SIRT1 induction partially preserved limb muscle soluble protein content and fibre cross-sectional area. Unilateral AAV9-mediated SIRT1 induction in the TA muscle showed that SIRT1 partially preserved mass by acting directly in the muscle. Despite those positive outcomes to limb muscle morphology, muscle-specific SIRT1 induction did not preserve the force generating capacity of the TA or triceps surae muscles. Interestingly, SIRT1 induction in females did not alter limb muscle mass or limb muscle strength even though females have naturally low androgen levels. SIRT1 also did not alter the androgen-mediated increase in limb muscle mass or strength in females. In all, these data suggest that decreases in SIRT1 protein in the limb muscle of males may partially contribute to the loss of limb muscle mass in response to androgen deprivation. KEY POINTS: SIRT1 induction in skeletal muscle of male mice subjected to androgen deprivation partially preserved limb muscle mass and fibre cross-sectional area. SIRT1 induction in skeletal muscle of male mice subjected to androgen deprivation did not prevent preserve limb muscle force generating capacity. SIRT1 induction in skeletal muscle of females did not alter baseline limb muscle mass, nor did it affect the androgen-mediated increase in limb muscle mass.
Collapse
Affiliation(s)
- Bradley S Gordon
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL, USA
- Institute of Sports Sciences and Medicine, Florida State University, Tallahassee, FL, USA
| | - Patrick K Burns
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL, USA
| | - Grant R Laskin
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL, USA
| | - Kirsten R Dunlap
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL, USA
| | - Jake R Boykin
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL, USA
| | - Michael L Rossetti
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL, USA
| | - David H Fukuda
- School of Kinesiology and Physical Therapy, University of Central Florida, Orlando, FL, USA
| | - Jennifer L Steiner
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, FL, USA
- Institute of Sports Sciences and Medicine, Florida State University, Tallahassee, FL, USA
| |
Collapse
|
3
|
Abstract
Myostatin (GDF-8) was discovered 25 years ago as a new transforming growth factor-β family member that acts as a master regulator of skeletal muscle mass. Myostatin is made by skeletal myofibers, circulates in the blood, and acts back on myofibers to limit growth. Myostatin appears to have all of the salient properties of a chalone, which is a term proposed over a half century ago to describe hypothetical circulating, tissue-specific growth inhibitors that control tissue size. The elucidation of the molecular, cellular, and physiological mechanisms underlying myostatin activity suggests that myostatin functions as a negative feedback regulator of muscle mass and raises the question as to whether this type of chalone mechanism is unique to skeletal muscle or whether it also operates in other tissues.
Collapse
Affiliation(s)
- Se-Jin Lee
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, Connecticut, USA.,The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA;
| |
Collapse
|
4
|
Abati E, Manini A, Comi GP, Corti S. Inhibition of myostatin and related signaling pathways for the treatment of muscle atrophy in motor neuron diseases. Cell Mol Life Sci 2022; 79:374. [PMID: 35727341 PMCID: PMC9213329 DOI: 10.1007/s00018-022-04408-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/16/2022] [Accepted: 06/01/2022] [Indexed: 11/26/2022]
Abstract
Myostatin is a negative regulator of skeletal muscle growth secreted by skeletal myocytes. In the past years, myostatin inhibition sparked interest among the scientific community for its potential to enhance muscle growth and to reduce, or even prevent, muscle atrophy. These characteristics make it a promising target for the treatment of muscle atrophy in motor neuron diseases, namely, amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA), which are rare neurological diseases, whereby the degeneration of motor neurons leads to progressive muscle loss and paralysis. These diseases carry a huge burden of morbidity and mortality but, despite this unfavorable scenario, several therapeutic advancements have been made in the past years. Indeed, a number of different curative therapies for SMA have been approved, leading to a revolution in the life expectancy and outcomes of SMA patients. Similarly, tofersen, an antisense oligonucleotide, is now undergoing clinical trial phase for use in ALS patients carrying the SOD1 mutation. However, these therapies are not able to completely halt or reverse progression of muscle damage. Recently, a trial evaluating apitegromab, a myostatin inhibitor, in SMA patients was started, following positive results from preclinical studies. In this context, myostatin inhibition could represent a useful strategy to tackle motor symptoms in these patients. The aim of this review is to describe the myostatin pathway and its role in motor neuron diseases, and to summarize and critically discuss preclinical and clinical studies of myostatin inhibitors in SMA and ALS. Then, we will highlight promises and pitfalls related to the use of myostatin inhibitors in the human setting, to aid the scientific community in the development of future clinical trials.
Collapse
Affiliation(s)
- Elena Abati
- Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Neuroscience Section, Neurology Unit, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
- Neurology Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Arianna Manini
- Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Neuroscience Section, Neurology Unit, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Giacomo Pietro Comi
- Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Neuroscience Section, Neurology Unit, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
- Neurology Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefania Corti
- Department of Pathophysiology and Transplantation (DEPT), Dino Ferrari Centre, Neuroscience Section, Neurology Unit, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico, University of Milan, Milan, Italy.
- Neurology Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
5
|
Omosule CL, Joseph D, Weiler B, Gremminger VL, Silvey S, Jeong Y, Rafique A, Krueger P, Kleiner S, Phillips CL. Combinatorial Inhibition of Myostatin and Activin A Improves Femoral Bone Properties in the G610C Mouse Model of Osteogenesis Imperfecta. J Bone Miner Res 2022; 37:938-953. [PMID: 35195284 PMCID: PMC10041862 DOI: 10.1002/jbmr.4529] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 01/23/2022] [Accepted: 02/11/2022] [Indexed: 01/28/2023]
Abstract
Osteogenesis imperfecta (OI) is a collagen-related bone disorder characterized by fragile osteopenic bone and muscle weakness. We have previously shown that the soluble activin receptor type IIB decoy (sActRIIB) molecule increases muscle mass and improves bone strength in the mild to moderate G610C mouse model of OI. The sActRIIB molecule binds multiple transforming growth factor-β (TGF-β) ligands, including myostatin and activin A. Here, we investigate the musculoskeletal effects of inhibiting activin A alone, myostatin alone, or both myostatin and activin A in wild-type (Wt) and heterozygous G610C (+/G610C) mice using specific monoclonal antibodies. Male and female Wt and +/G610C mice were treated twice weekly with intraperitoneal injections of monoclonal control antibody (Ctrl-Ab, Regn1945), anti-activin A antibody (ActA-Ab, Regn2476), anti-myostatin antibody (Mstn-Ab, Regn647), or both ActA-Ab and Mstn-Ab (Combo, Regn2476, and Regn647) from 5 to 16 weeks of age. Prior to euthanasia, whole body composition, metabolism and muscle force generation assessments were performed. Post euthanasia, hindlimb muscles were evaluated for mass, and femurs were evaluated for changes in microarchitecture and biomechanical strength using micro-computed tomography (μCT) and three-point bend analyses. ActA-Ab treatment minimally impacted the +/G610C musculoskeleton, and was detrimental to bone strength in male +/G610C mice. Mstn-Ab treatment, as previously reported, resulted in substantial increases in hindlimb muscle weights and overall body weights in Wt and male +/G610C mice, but had minimal skeletal impact in +/G610C mice. Conversely, the Combo treatment outperformed ActA-Ab alone or Mstn-Ab alone, consistently increasing hindlimb muscle and body weights regardless of sex or genotype and improving bone microarchitecture and strength in both male and female +/G610C and Wt mice. Combinatorial inhibition of activin A and myostatin more potently increased muscle mass and bone microarchitecture and strength than either antibody alone, recapturing most of the observed benefits of sActRIIB treatment in +/G610C mice. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
| | - Dominique Joseph
- Department of Biochemistry, University of Missouri, Columbia, MO, USA
| | - Brooke Weiler
- Department of Biochemistry, University of Missouri, Columbia, MO, USA
| | | | - Spencer Silvey
- Department of Biochemistry, University of Missouri, Columbia, MO, USA
| | - Youngjae Jeong
- Department of Biochemistry, University of Missouri, Columbia, MO, USA
| | | | | | | | - Charlotte L Phillips
- Department of Biochemistry, University of Missouri, Columbia, MO, USA.,Department of Child Health, University of Missouri, Columbia, MO, USA
| |
Collapse
|
6
|
Puolakkainen T, Rummukainen P, Pihala-Nieminen V, Ritvos O, Savontaus E, Kiviranta R. Treatment with Soluble Activin Type IIB Receptor Ameliorates Ovariectomy-Induced Bone Loss and Fat Gain in Mice. Calcif Tissue Int 2022; 110:504-517. [PMID: 35024891 PMCID: PMC8927044 DOI: 10.1007/s00223-021-00934-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 11/25/2021] [Indexed: 11/23/2022]
Abstract
INTRODUCTION In postmenopausal osteoporosis, hormonal changes lead to increased bone turnover and metabolic alterations including increased fat mass and insulin resistance. Activin type IIB receptors bind several growth factors of the TGF-β superfamily and have been demonstrated to increase muscle and bone mass. We hypothesized that ActRIIB-Fc treatment could improve bone and muscle mass, inhibit fat accumulation, and restore metabolic alterations in an ovariectomy (OVX) model of postmenopausal osteoporosis. MATERIALS AND METHODS Female C57Bl/6 N mice were subjected to SHAM or OVX procedures and received intraperitoneal injections of either PBS or ActRIIB-Fc (5 mg/kg) once weekly for 7 weeks. Glucose and insulin tolerance tests (GTT and ITT, respectively) were performed at 7 and 8 weeks, respectively. Bone samples were analyzed with micro-computed tomography imaging, histomorphometry, and quantitative RT-PCR. RESULTS Bone mass decreased in OVX PBS mice compared to the SHAM PBS group but ActRIIB-Fc was able to prevent these changes as shown by µCT and histological analyses. This was due to decreased osteoclast numbers and function demonstrated by histomorphometric and qRT-PCR analyses. OVX induced adipocyte hypertrophy that was rescued by ActRIIB-Fc, which also decreased systemic adipose tissue accumulation. OVX itself did not affect glucose levels in GTT but ActRIIB-Fc treatment resulted in impaired glucose clearance in both SHAM and OVX groups. OVX induced mild insulin resistance in ITT but ActRIIB-Fc treatment did not affect this. CONCLUSION Our results reinforce the potency of ActRIIB-Fc as a bone-enhancing agent but also bring new insight into the metabolic effects of ActRIIB-Fc in normal and OVX mice.
Collapse
Affiliation(s)
- Tero Puolakkainen
- Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - Petri Rummukainen
- Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - Vappu Pihala-Nieminen
- Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland
| | - Olli Ritvos
- Department of Physiology, University of Helsinki, Helsinki, Finland
| | - Eriika Savontaus
- Clinical Pharmacology, Turku University Hospital, Turku, Finland
- Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Riku Kiviranta
- Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, 20520, Turku, Finland.
- Department of Endocrinology, Division of Medicine, University of Turku and Turku University Hospital, Turku, Finland.
- Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland.
| |
Collapse
|
7
|
Rodgers BD, Ward CW. Myostatin/Activin Receptor Ligands in Muscle and the Development Status of Attenuating Drugs. Endocr Rev 2022; 43:329-365. [PMID: 34520530 PMCID: PMC8905337 DOI: 10.1210/endrev/bnab030] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Indexed: 02/07/2023]
Abstract
Muscle wasting disease indications are among the most debilitating and often deadly noncommunicable disease states. As a comorbidity, muscle wasting is associated with different neuromuscular diseases and myopathies, cancer, heart failure, chronic pulmonary and renal diseases, peripheral neuropathies, inflammatory disorders, and, of course, musculoskeletal injuries. Current treatment strategies are relatively ineffective and can at best only limit the rate of muscle degeneration. This includes nutritional supplementation and appetite stimulants as well as immunosuppressants capable of exacerbating muscle loss. Arguably, the most promising treatments in development attempt to disrupt myostatin and activin receptor signaling because these circulating factors are potent inhibitors of muscle growth and regulators of muscle progenitor cell differentiation. Indeed, several studies demonstrated the clinical potential of "inhibiting the inhibitors," increasing muscle cell protein synthesis, decreasing degradation, enhancing mitochondrial biogenesis, and preserving muscle function. Such changes can prevent muscle wasting in various disease animal models yet many drugs targeting this pathway failed during clinical trials, some from serious treatment-related adverse events and off-target interactions. More often, however, failures resulted from the inability to improve muscle function despite preserving muscle mass. Drugs still in development include antibodies and gene therapeutics, all with different targets and thus, safety, efficacy, and proposed use profiles. Each is unique in design and, if successful, could revolutionize the treatment of both acute and chronic muscle wasting. They could also be used in combination with other developing therapeutics for related muscle pathologies or even metabolic diseases.
Collapse
Affiliation(s)
| | - Christopher W Ward
- Department of Orthopedics and Center for Biomedical Engineering and Technology (BioMET), University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
8
|
Hughes L, Hackney KJ, Patterson SD. Optimization of Exercise Countermeasures to Spaceflight Using Blood Flow Restriction. Aerosp Med Hum Perform 2022; 93:32-45. [PMID: 35063054 DOI: 10.3357/amhp.5855.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION: During spaceflight missions, astronauts work in an extreme environment with several hazards to physical health and performance. Exposure to microgravity results in remarkable deconditioning of several physiological systems, leading to impaired physical condition and human performance, posing a major risk to overall mission success and crew safety. Physical exercise is the cornerstone of strategies to mitigate physical deconditioning during spaceflight. Decades of research have enabled development of more optimal exercise strategies and equipment onboard the International Space Station. However, the effects of microgravity cannot be completely ameliorated with current exercise countermeasures. Moreover, future spaceflight missions deeper into space require a new generation of spacecraft, which will place yet more constraints on the use of exercise by limiting the amount, size, and weight of exercise equipment and the time available for exercise. Space agencies are exploring ways to optimize exercise countermeasures for spaceflight, specifically exercise strategies that are more efficient, require less equipment, and are less time-consuming. Blood flow restriction exercise is a low intensity exercise strategy that requires minimal equipment and can elicit positive training benefits across multiple physiological systems. This method of exercise training has potential as a strategy to optimize exercise countermeasures during spaceflight and reconditioning in terrestrial and partial gravity environments. The possible applications of blood flow restriction exercise during spaceflight are discussed herein.Hughes L, Hackney KJ, Patterson SD. Optimization of exercise countermeasures to spaceflight using blood flow restriction. Aerosp Med Hum Perform. 2021; 93(1):32-45.
Collapse
|
9
|
Functionally diverse heteromeric traps for ligands of the transforming growth factor-β superfamily. Sci Rep 2021; 11:18341. [PMID: 34526551 PMCID: PMC8443706 DOI: 10.1038/s41598-021-97203-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/18/2021] [Indexed: 01/19/2023] Open
Abstract
Ligands of the transforming growth factor-β (TGF-β) superfamily are important targets for therapeutic intervention but present challenges because they signal combinatorially and exhibit overlapping activities in vivo. To obtain agents capable of sequestering multiple TGF-β superfamily ligands with novel selectivity, we generated soluble, heterodimeric ligand traps by pairing the extracellular domain (ECD) of the native activin receptor type IIB (ActRIIB) alternately with the ECDs of native type I receptors activin receptor-like kinase 4 (ALK4), ALK7, or ALK3. Systematic analysis of these heterodimeric constructs by surface plasmon resonance, and comparison with their homodimeric counterparts, revealed that each type I receptor partner confers a distinct ligand-binding profile to the heterodimeric construct. Additional characterization in cell-based reporter gene assays confirmed that the heterodimeric constructs possessed different profiles of signaling inhibition in vitro, which translated into altered patterns of pharmacological activity when constructs were administered systemically to wild-type mice. Our results detail a versatile platform for the modular recombination of naturally occurring receptor domains, giving rise to inhibitory ligand traps that could aid in defining the physiological roles of TGF-β ligand sets or be directed therapeutically to human diseases arising from dysregulated TGF-β superfamily signaling.
Collapse
|
10
|
Local versus systemic control of bone and skeletal muscle mass by components of the transforming growth factor-β signaling pathway. Proc Natl Acad Sci U S A 2021; 118:2111401118. [PMID: 34385332 PMCID: PMC8379946 DOI: 10.1073/pnas.2111401118] [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] [Indexed: 12/20/2022] Open
Abstract
Skeletal muscle and bone homeostasis are regulated by members of the myostatin/GDF-11/activin branch of the transforming growth factor-β superfamily, which share many regulatory components, including inhibitory extracellular binding proteins and receptors that mediate signaling. Here, we present the results of genetic studies demonstrating a critical role for the binding protein follistatin (FST) in regulating both skeletal muscle and bone. Using an allelic series corresponding to varying expression levels of endogenous Fst, we show that FST acts in an exquisitely dose-dependent manner to regulate both muscle mass and bone density. Moreover, by employing a genetic strategy to target Fst expression only in the posterior (caudal) region of the animal, we show that the effects of Fst loss are mostly restricted to the posterior region, implying that locally produced FST plays a much more important role than circulating FST with respect to regulation of muscle and bone. Finally, we show that targeting receptors for these ligands specifically in osteoblasts leads to dramatic increases in bone mass, with trabecular bone volume fraction being increased by 12- to 13-fold and bone mineral density being increased by 8- to 9-fold in humeri, femurs, and lumbar vertebrae. These findings demonstrate that bone, like muscle, has an enormous inherent capacity for growth that is normally kept in check by this signaling system and suggest that the extent to which this regulatory mechanism may be used throughout the body to regulate tissue mass may be more significant than previously appreciated.
Collapse
|
11
|
Lee SJ. Targeting the myostatin signaling pathway to treat muscle loss and metabolic dysfunction. J Clin Invest 2021; 131:148372. [PMID: 33938454 DOI: 10.1172/jci148372] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Since the discovery of myostatin (MSTN; also known as GDF-8) as a critical regulator of skeletal muscle mass in 1997, there has been an extensive effort directed at understanding the cellular and physiological mechanisms underlying MSTN activity, with the long-term goal of developing strategies and agents capable of blocking MSTN signaling to treat patients with muscle loss. Considerable progress has been made in elucidating key components of this regulatory system, and in parallel with this effort has been the development of numerous biologics that have been tested in clinical trials for a wide range of indications, including muscular dystrophy, sporadic inclusion body myositis, spinal muscular atrophy, cachexia, muscle loss due to aging or following falls, obesity, and type 2 diabetes. Here, I review what is known about the MSTN regulatory system and the current state of efforts to target this pathway for clinical applications.
Collapse
Affiliation(s)
- Se-Jin Lee
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA.,University of Connecticut School of Medicine, Department of Genetics and Genome Sciences, Farmington, Connecticut, USA
| |
Collapse
|
12
|
Lodberg A. Principles of the activin receptor signaling pathway and its inhibition. Cytokine Growth Factor Rev 2021; 60:1-17. [PMID: 33933900 DOI: 10.1016/j.cytogfr.2021.04.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 01/19/2023]
Abstract
This review captures the anabolic and stimulatory effects observed with inhibition of the transforming growth factor β superfamily in muscle, blood, and bone. New medicinal substances that rectify activin, myostatin, and growth differentiation factor 11 signaling give hope to the many whose lives are affected by deterioration of these tissues. The review first covers the origin, structure, and common pathway of activins, myostatin, and growth differentiation factor 11 along with the pharmacodynamics of the new class of molecules designed to oppose the activin receptor signaling pathway. Current terminology surrounding this new class of molecules is inconsistent and does not infer functionality. Adopting inhibitors of the activin receptor signaling pathway (IASPs) as a generic term is proposed because it encapsulates the molecular mechanisms along the pathway trajectory. To conclude, a pragmatic classification of IASPs is presented that integrates functionality and side effects based on the data available from animals and humans. This provides researchers and clinicians with a tool to tailor IASPs therapy according to the need of projects or patients and with respect to side effects.
Collapse
Affiliation(s)
- Andreas Lodberg
- Department of Biomedicine, Aarhus University, Department of Respiratory Diseases and Allergy, Aarhus University Hospital, Wilhelm Meyers Allé, DK-8000, Aarhus, Denmark.
| |
Collapse
|
13
|
Meier D, Lodberg A, Gvozdenovic A, Pellegrini G, Neklyudova O, Born W, Fuchs B, Eijken M, M. Botter S. Inhibition of the activin receptor signaling pathway: A novel intervention against osteosarcoma. Cancer Med 2021; 10:286-296. [PMID: 33179858 PMCID: PMC7826474 DOI: 10.1002/cam4.3581] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 01/02/2023] Open
Abstract
Osteosarcoma is a cancer of pathological bone remodeling with high mortality and severe comorbidity. New therapies are urgently needed. Activin A, a member of the transforming growth factor β (TGFβ) superfamily, has been suggested to stimulate proliferation and invasion of osteosarcoma cells in vitro, thus representing a potential therapeutic target. In this study, inhibition of the activin receptor signaling pathway was explored as a therapy for osteosarcoma. In a murine intratibial osteosarcoma xenograft model, two types of inhibitors were tested: (a) a soluble activin type IIA decoy receptor (ActRIIA-mFc), or (b) a modified variant of follistatin (FSTΔHBS -hFc), either alone or in combination with a bisphosphonate. Both inhibitors reduced primary tumor development by nearly 50% compared to vehicle treatment. When ActRIIA-mFc was combined with bisphosphonate, the effect on tumor size became even more pronounced (78% reduction vs. vehicle). Moreover, FSTΔHBS -hFc increased body weight in the face of tumor progression (14% increase vs. vehicle), and ActRIIA-mFc reduced the number of lung metastases when combined with bisphosphonate. The present study demonstrates a novel approach to treating osteosarcoma and encourages further investigation of inhibition of the activin receptor signaling pathway as an intervention against the disease.
Collapse
Affiliation(s)
- Daniela Meier
- Department of OrthopedicsBalgrist University HospitalZurichSwitzerland
| | - Andreas Lodberg
- Department of BiomedicineAarhus UniversityAarhusDenmark
- Department of Pulmonary MedicineAarhus University HospitalAarhusDenmark
| | - Ana Gvozdenovic
- Department of OrthopedicsBalgrist University HospitalZurichSwitzerland
| | - Giovanni Pellegrini
- Laboratory for Animal Model PathologyInstitute of Veterinary Pathology, University of ZurichZurichSwitzerland
| | - Olga Neklyudova
- Department of OrthopedicsBalgrist University HospitalZurichSwitzerland
| | - Walter Born
- Department of OrthopedicsBalgrist University HospitalZurichSwitzerland
| | - Bruno Fuchs
- Department of OrthopedicsBalgrist University HospitalZurichSwitzerland
| | - Marco Eijken
- Department of Renal MedicineAarhus University HospitalAarhusDenmark
- Department of Clinical ImmunologyAarhus University HospitalAarhusDenmark
| | - Sander M. Botter
- Department of OrthopedicsBalgrist University HospitalZurichSwitzerland
| |
Collapse
|
14
|
Functional redundancy of type I and type II receptors in the regulation of skeletal muscle growth by myostatin and activin A. Proc Natl Acad Sci U S A 2020; 117:30907-30917. [PMID: 33219121 PMCID: PMC7733802 DOI: 10.1073/pnas.2019263117] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Myostatin (MSTN) is a transforming growth factor-β (TGF-β) family member that normally acts to limit muscle growth. The function of MSTN is partially redundant with that of another TGF-β family member, activin A. MSTN and activin A are capable of signaling through a complex of type II and type I receptors. Here, we investigated the roles of two type II receptors (ACVR2 and ACVR2B) and two type I receptors (ALK4 and ALK5) in the regulation of muscle mass by these ligands by genetically targeting these receptors either alone or in combination specifically in myofibers in mice. We show that targeting signaling in myofibers is sufficient to cause significant increases in muscle mass, showing that myofibers are the direct target for signaling by these ligands in the regulation of muscle growth. Moreover, we show that there is functional redundancy between the two type II receptors as well as between the two type I receptors and that all four type II/type I receptor combinations are utilized in vivo. Targeting signaling specifically in myofibers also led to reductions in overall body fat content and improved glucose metabolism in mice fed either regular chow or a high-fat diet, demonstrating that these metabolic effects are the result of enhanced muscling. We observed no effect, however, on either bone density or muscle regeneration in mice in which signaling was targeted in myofibers. The latter finding implies that MSTN likely signals to other cells, such as satellite cells, in addition to myofibers to regulate muscle homeostasis.
Collapse
|
15
|
Zhuang Z, John JV, Liao H, Luo J, Rubery P, Mesfin A, Boda SK, Xie J, Zhang X. Periosteum Mimetic Coating on Structural Bone Allografts via Electrospray Deposition Enhances Repair and Reconstruction of Segmental Defects. ACS Biomater Sci Eng 2020; 6:6241-6252. [PMID: 33449646 DOI: 10.1021/acsbiomaterials.0c00421] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Structural bone allograft transplantation remains one of the common strategies for repair and reconstruction of large bone defects. Due to the loss of periosteum that covers the outer surface of the cortical bone, the healing and incorporation of allografts is extremely slow and limited. To enhance the biological performance of allografts, herein, we report a novel and simple approach for engineering a periosteum mimetic coating on the surface of structural bone allografts via polymer-mediated electrospray deposition. This approach enables the coating on allografts with precisely controlled composition and thickness. In addition, the periosteum mimetic coating can be tailored to achieve desired drug release profiles by making use of an appropriate biodegradable polymer or polymer blend. The efficacy study in a murine segmental femoral bone defect model demonstrates that the allograft coating composed of poly(lactic-co-glycolic acid) and bone morphogenetic protein-2 mimicking peptide significantly improves allograft healing as evidenced by decreased fibrotic tissue formation, increased periosteal bone formation, and enhanced osseointegration. Taken together, this study provides a platform technology for engineering a periosteum mimetic coating which can greatly promote bone allograft healing. This technology could eventually result in an off-the-shelf and multifunctional structural bone allograft for highly effective repair and reconstruction of large segmental bone defects. The technology can also be used to ameliorate the performance of other medical implants by modifying their surfaces.
Collapse
Affiliation(s)
- Zhou Zhuang
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14621, United States
| | - Johnson V John
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska, Omaha, Nebraska 68198, United States
| | - Haofu Liao
- Department of Computer Science, University of Rochester, Rochester, New York 14627, United States
| | - Jiebo Luo
- Department of Computer Science, University of Rochester, Rochester, New York 14627, United States
| | - Paul Rubery
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642, United States
| | - Addisu Mesfin
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642, United States
| | - Sunil Kumar Boda
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska, Omaha, Nebraska 68198, United States
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary & Dick Holland Regenerative Medicine Program, University of Nebraska, Omaha, Nebraska 68198, United States
| | - Xinping Zhang
- Center for Musculoskeletal Research, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642, United States
| |
Collapse
|
16
|
Lee SJ, Lehar A, Meir JU, Koch C, Morgan A, Warren LE, Rydzik R, Youngstrom DW, Chandok H, George J, Gogain J, Michaud M, Stoklasek TA, Liu Y, Germain-Lee EL. Targeting myostatin/activin A protects against skeletal muscle and bone loss during spaceflight. Proc Natl Acad Sci U S A 2020; 117:23942-23951. [PMID: 32900939 PMCID: PMC7519220 DOI: 10.1073/pnas.2014716117] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Among the physiological consequences of extended spaceflight are loss of skeletal muscle and bone mass. One signaling pathway that plays an important role in maintaining muscle and bone homeostasis is that regulated by the secreted signaling proteins, myostatin (MSTN) and activin A. Here, we used both genetic and pharmacological approaches to investigate the effect of targeting MSTN/activin A signaling in mice that were sent to the International Space Station. Wild type mice lost significant muscle and bone mass during the 33 d spent in microgravity. Muscle weights of Mstn-/- mice, which are about twice those of wild type mice, were largely maintained during spaceflight. Systemic inhibition of MSTN/activin A signaling using a soluble form of the activin type IIB receptor (ACVR2B), which can bind each of these ligands, led to dramatic increases in both muscle and bone mass, with effects being comparable in ground and flight mice. Exposure to microgravity and treatment with the soluble receptor each led to alterations in numerous signaling pathways, which were reflected in changes in levels of key signaling components in the blood as well as their RNA expression levels in muscle and bone. These findings have implications for therapeutic strategies to combat the concomitant muscle and bone loss occurring in people afflicted with disuse atrophy on Earth as well as in astronauts in space, especially during prolonged missions.
Collapse
Affiliation(s)
- Se-Jin Lee
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032;
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Adam Lehar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032
| | - Jessica U Meir
- The National Aeronautics and Space Administration, NASA Johnson Space Center, Houston, TX 77058
| | - Christina Koch
- The National Aeronautics and Space Administration, NASA Johnson Space Center, Houston, TX 77058
| | - Andrew Morgan
- The National Aeronautics and Space Administration, NASA Johnson Space Center, Houston, TX 77058
| | - Lara E Warren
- Center for the Advancement of Science in Space, Houston, TX 77058
| | - Renata Rydzik
- Department of Orthopaedic Surgery, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Daniel W Youngstrom
- Department of Orthopaedic Surgery, University of Connecticut School of Medicine, Farmington, CT 06030
| | | | - Joshy George
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032
| | | | - Michael Michaud
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032
| | | | - Yewei Liu
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032
| | - Emily L Germain-Lee
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT 06030
- Connecticut Children's Center for Rare Bone Disorders, Farmington, CT 06032
| |
Collapse
|
17
|
Invernizzi M, de Sire A, Carda S, Venetis K, Renò F, Cisari C, Fusco N. Bone Muscle Crosstalk in Spinal Cord Injuries: Pathophysiology and Implications for Patients' Quality of Life. Curr Osteoporos Rep 2020; 18:422-431. [PMID: 32519284 DOI: 10.1007/s11914-020-00601-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The goal of this review is to provide a comprehensive overview of (i) bone and muscle tissue modifications pathophysiology in spinal cord injury (SCI), (ii) experimental data on the physiopathological mechanisms underpinning these modifications and their similarities with the aging process, and (iii) potential clinical implications in the management of the disabling sequelae of SCI. RECENT FINDINGS Several studies attempted to describe the biology underpinning the links between bone and muscle tissues in the setting of highly disabling conditions, such as osteoporosis, sarcopenia, and neurodegenerative disorders, although these bidirectional connections remain still unclear. SCI could be considered an in vivo paradigmatic model of the bone muscle interactions in unloading conditions that might be expanded in the field of neurodegenerative disorders or cancer studies. Future studies should take into consideration the newer insights into bone muscle crosstalk in order to develop multitargeted and therapeutic interventions.
Collapse
Affiliation(s)
- Marco Invernizzi
- Physical and Rehabilitative Medicine, Department of Health Sciences, University of Eastern Piedmont, Novara, Italy.
| | - Alessandro de Sire
- Physical and Rehabilitative Medicine, Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
- Rehabilitation Unit, "Mons. L. Novarese" Hospital, Moncrivello, Vercelli, Italy
| | - Stefano Carda
- Neuropsychology and Neurorehabilitation Service, Department of Clinical Neuroscience, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Konstantinos Venetis
- Ph.D. Program in Translational Medicine, University of Milan, Milan, Italy
- Division of Pathology, IRCCS European Institute of Oncology (IEO), Milan, Italy
| | - Filippo Renò
- Innovative Research Laboratory for Wound Healing, Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
| | - Carlo Cisari
- Physical and Rehabilitative Medicine, Department of Health Sciences, University of Eastern Piedmont, Novara, Italy
- Physical Medicine and Rehabilitation Unit, University Hospital "Maggiore della Carità", Novara, Italy
| | - Nicola Fusco
- Division of Pathology, IRCCS European Institute of Oncology (IEO), Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
18
|
Farhang-Sardroodi S, Wilkie KP. Mathematical Model of Muscle Wasting in Cancer Cachexia. J Clin Med 2020; 9:jcm9072029. [PMID: 32605273 PMCID: PMC7409297 DOI: 10.3390/jcm9072029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/17/2020] [Accepted: 06/19/2020] [Indexed: 12/12/2022] Open
Abstract
Cancer cachexia is a debilitating condition characterized by an extreme loss of skeletal muscle mass, which negatively impacts patients' quality of life, reduces their ability to sustain anti-cancer therapies, and increases the risk of mortality. Recent discoveries have identified the myostatin/activin A/ActRIIB pathway as critical to muscle wasting by inducing satellite cell quiescence and increasing muscle-specific ubiquitin ligases responsible for atrophy. Remarkably, pharmacological blockade of the ActRIIB pathway has been shown to reverse muscle wasting and prolong the survival time of tumor-bearing animals. To explore the implications of this signaling pathway and potential therapeutic targets in cachexia, we construct a novel mathematical model of muscle tissue subjected to tumor-derived cachectic factors. The model formulation tracks the intercellular interactions between cancer cell, satellite cell, and muscle cell populations. The model is parameterized by fitting to colon-26 mouse model data, and the analysis provides insight into tissue growth in healthy, cancerous, and post-cachexia treatment conditions. Model predictions suggest that cachexia fundamentally alters muscle tissue health, as measured by the stem cell ratio, and this is only partially recovered by anti-cachexia treatment. Our mathematical findings suggest that after blocking the myostatin/activin A pathway, partial recovery of cancer-induced muscle loss requires the activation and proliferation of the satellite cell compartment with a functional differentiation program.
Collapse
|
19
|
Pan C, Jaiswal Agrawal N, Zulia Y, Singh S, Sha K, Mohler JL, Eng KH, Chakkalakal JV, Krolewski JJ, Nastiuk KL. Prostate tumor-derived GDF11 accelerates androgen deprivation therapy-induced sarcopenia. JCI Insight 2020; 5:127018. [PMID: 32078585 PMCID: PMC7213789 DOI: 10.1172/jci.insight.127018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 02/19/2020] [Indexed: 12/31/2022] Open
Abstract
Most prostate cancers depend on androgens for growth, and therefore, the mainstay treatment for advanced, recurrent, or metastatic prostate cancer is androgen deprivation therapy (ADT). A prominent side effect in patients receiving ADT is an obese frailty syndrome that includes fat gain and sarcopenia, defined as the loss of muscle function accompanied by reduced muscle mass or quality. Mice bearing Pten-deficient prostate cancers were examined to gain mechanistic insight into ADT-induced sarcopenic obesity. Castration induced fat gain as well as skeletal muscle mass and strength loss. Catabolic TGF-β family myokine protein levels were increased immediately prior to strength loss, and pan-myokine blockade using a soluble receptor (ActRIIB-Fc) completely reversed the castration-induced sarcopenia. The onset of castration-induced strength and muscle mass loss, as well as the increase in catabolic TGF-β family myokine protein levels, were coordinately accelerated in tumor-bearing mice relative to tumor-free mice. Notably, growth differentiation factor 11 (GDF11) increased in muscle after castration only in tumor-bearing mice, but not in tumor‑free mice. An early surge of GDF11 in prostate tumor tissue and in the circulation suggests that endocrine GDF11 signaling from tumor to muscle is a major driver of the accelerated ADT-induced sarcopenic phenotype. In tumor-bearing mice, GDF11 blockade largely prevented castration-induced strength loss but did not preserve muscle mass, which confirms a primary role for GDF11 in muscle function and suggests an additional role for the other catabolic myokines.
Collapse
Affiliation(s)
| | | | | | | | - Kai Sha
- Department of Cancer Genetics and Genomics
| | | | - Kevin H Eng
- Department of Cancer Genetics and Genomics.,Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Joe V Chakkalakal
- Department of Pharmacology & Physiology and.,Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, New York, USA
| | | | - Kent L Nastiuk
- Department of Cancer Genetics and Genomics.,Department of Urology, and
| |
Collapse
|
20
|
Long KK, O’Shea KM, Khairallah RJ, Howell K, Paushkin S, Chen KS, Cote SM, Webster MT, Stains JP, Treece E, Buckler A, Donovan A. Specific inhibition of myostatin activation is beneficial in mouse models of SMA therapy. Hum Mol Genet 2019; 28:1076-1089. [PMID: 30481286 PMCID: PMC6423420 DOI: 10.1093/hmg/ddy382] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/29/2018] [Accepted: 10/31/2018] [Indexed: 12/22/2022] Open
Abstract
Spinal muscular atrophy (SMA) is a neuromuscular disease characterized by loss of α-motor neurons, leading to profound skeletal muscle atrophy. Patients also suffer from decreased bone mineral density and increased fracture risk. The majority of treatments for SMA, approved or in clinic trials, focus on addressing the underlying cause of disease, insufficient production of full-length SMN protein. While restoration of SMN has resulted in improvements in functional measures, significant deficits remain in both mice and SMA patients following treatment. Motor function in SMA patients may be additionally improved by targeting skeletal muscle to reduce atrophy and improve muscle strength. Inhibition of myostatin, a negative regulator of muscle mass, offers a promising approach to increase muscle function in SMA patients. Here we demonstrate that muSRK-015P, a monoclonal antibody which specifically inhibits myostatin activation, effectively increases muscle mass and function in two variants of the pharmacological mouse model of SMA in which pharmacologic restoration of SMN has taken place either 1 or 24 days after birth to reflect early or later therapeutic intervention. Additionally, muSRK-015P treatment improves the cortical and trabecular bone phenotypes in these mice. These data indicate that preventing myostatin activation has therapeutic potential in addressing muscle and bone deficiencies in SMA patients. An optimized variant of SRK-015P, SRK-015, is currently in clinical development for treatment of SMA.
Collapse
Affiliation(s)
| | | | | | - Kelly Howell
- SMA Foundation, 888 7th Avenue #400, New York, NY
| | | | - Karen S Chen
- SMA Foundation, 888 7th Avenue #400, New York, NY
| | - Shaun M Cote
- Scholar Rock Inc., 620 Memorial Drive, Cambridge, MA
| | | | - Joseph P Stains
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Erin Treece
- Scholar Rock Inc., 620 Memorial Drive, Cambridge, MA
| | - Alan Buckler
- Scholar Rock Inc., 620 Memorial Drive, Cambridge, MA
| | | |
Collapse
|
21
|
Lodberg A, van der Eerden BCJ, Boers-Sijmons B, Thomsen JS, Brüel A, van Leeuwen JPTM, Eijken M. A follistatin-based molecule increases muscle and bone mass without affecting the red blood cell count in mice. FASEB J 2019; 33:6001-6010. [PMID: 30759349 DOI: 10.1096/fj.201801969rr] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Inhibitors of the activin receptor signaling pathway (IASPs) have become candidate therapeutics for sarcopenia and bone remodeling disorders because of their ability to increase muscle and bone mass. However, IASPs utilizing activin type IIA and IIB receptors are also potent stimulators of erythropoiesis, a feature that may restrict their usage to anemic patients because of increased risk of venous thromboembolism. Based on the endogenous TGF-β superfamily antagonist follistatin (FST), a molecule in the IASP class, FSTΔHBS-mFc, was generated and tested in both ovariectomized and naive BALB/c and C57BL/6 mice. In ovariectomized mice, FSTΔHBS-mFc therapy dose-dependently increased cancellous bone mass up to 42% and improved bone microstructural indices. For the highest dosage of FSTΔHBS-mFc (30 mg/kg, 2 times/wk), the increase in cancellous bone mass was similar to that observed with parathyroid hormone therapy (1-34, 80 µg/kg, 5 times/wk). Musculus quadriceps femoris mass dose-dependently increased up to 21% in ovariectomized mice. In both ovariectomized and naive mice, FSTΔHBS-mFc therapy did not influence red blood cell count or hematocrit or hemoglobin levels. If the results are reproduced, a human FSTΔHBS-mFc version could be applicable in patients with musculoskeletal conditions irrespective of hematocrit status.-Lodberg, A., van der Eerden, B. C. J., Boers-Sijmons, B., Thomsen, J. S., Brüel, A., van Leeuwen, J. P. T. M., Eijken, M. A follistatin-based molecule increases muscle and bone mass without affecting the red blood cell count in mice.
Collapse
Affiliation(s)
- Andreas Lodberg
- Department of Pulmonary Medicine, Aarhus University Hospital, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Bianca Boers-Sijmons
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Annemarie Brüel
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Marco Eijken
- Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
22
|
Age-related bone loss and sarcopenia in men. Maturitas 2019; 122:51-56. [PMID: 30797530 DOI: 10.1016/j.maturitas.2019.01.006] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 01/20/2019] [Accepted: 01/22/2019] [Indexed: 12/20/2022]
Abstract
Bone and muscle are required for mobility but they also have endocrine and metabolic functions. In ageing as well as in many chronic diseases, bone loss and muscle atrophy occur simultaneously, leading to concomitant osteoporosis and sarcopenia. This occurs in both genders but compared with postmenopausal women, men appear to be better protected against age-related bone and muscle decay. Sex steroids (both androgens like testosterone and oestrogens like estradiol) are mainly responsible for musculoskeletal sexual dimorphism. They stimulate peak bone and muscle mass accretion during puberty and midlife, and prevent subsequent loss in ageing men but not post-menopausal women. Still, recent studies have highlighted the importance of intrinsic ageing mechanisms such as cellular senescence and oxidative stress in both genders. Sarcopenia may predispose to dysmobility, frailty, falls and fractures, but whether so-called osteosarcopenia qualifies as a distinct entity remains debated. Although randomized clinical trials in male osteoporosis are smaller and therefore underpowered for some outcomes like hip fractures, the available evidence suggests that the clinical diagnostic and therapeutic approach to male osteoporosis is largely similar to that in postmenopausal women. There is a clear unmet medical need for effective and safe anabolic drugs to rebuild the ageing skeleton, muscle, and preferably both tissues simultaneously. The Wnt/sclerostin and myostatin/activin receptor signalling pathways appear particularly promising in this regard. In this narrative review, we aim to provide an overview of our current understanding of the pathophysiology and treatment of male osteoporosis and sarcopenia, and interactions between these two diseases.
Collapse
|
23
|
Abstract
Bone morphogenetic proteins (BMPs) constitute the largest subdivision of the transforming growth factor-β family of ligands. BMPs exhibit widespread utility and pleiotropic, context-dependent effects, and the strength and duration of BMP pathway signaling is tightly regulated at numerous levels via mechanisms operating both inside and outside the cell. Defects in the BMP pathway or its regulation underlie multiple human diseases of different organ systems. Yet much remains to be discovered about the BMP pathway in its original context, i.e., the skeleton. In this review, we provide a comprehensive overview of the intricacies of the BMP pathway and its inhibitors in bone development, homeostasis, and disease. We frame the content of the review around major unanswered questions for which incomplete evidence is available. First, we consider the gene regulatory network downstream of BMP signaling in osteoblastogenesis. Next, we examine why some BMP ligands are more osteogenic than others and what factors limit BMP signaling during osteoblastogenesis. Then we consider whether specific BMP pathway components are required for normal skeletal development, and if the pathway exerts endogenous effects in the aging skeleton. Finally, we propose two major areas of need of future study by the field: greater resolution of the gene regulatory network downstream of BMP signaling in the skeleton, and an expanded repertoire of reagents to reliably and specifically inhibit individual BMP pathway components.
Collapse
Affiliation(s)
- Jonathan W Lowery
- Division of Biomedical Science, Marian University College of Osteopathic Medicine , Indianapolis, Indiana ; and Department of Developmental Biology, Harvard School of Dental Medicine , Boston, Massachusetts
| | - Vicki Rosen
- Division of Biomedical Science, Marian University College of Osteopathic Medicine , Indianapolis, Indiana ; and Department of Developmental Biology, Harvard School of Dental Medicine , Boston, Massachusetts
| |
Collapse
|
24
|
Jeong Y, Daghlas SA, Yixia X, Hulbert MA, Pfeiffer FM, Dallas MR, Omosule CL, Pearsall RS, Dallas SL, Phillips CL. Skeletal Response to Soluble Activin Receptor Type IIB in Mouse Models of Osteogenesis Imperfecta. J Bone Miner Res 2018; 33:1760-1772. [PMID: 29813187 PMCID: PMC6400483 DOI: 10.1002/jbmr.3473] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 05/20/2018] [Accepted: 05/29/2018] [Indexed: 01/21/2023]
Abstract
Osteogenesis imperfecta (OI) is a heritable connective tissue disorder primarily due to mutations in the type I collagen genes (COL1A1 and COL1A2), leading to compromised biomechanical integrity in type I collagen-containing tissues such as bone. Bone is inherently mechanosensitive and thus responds and adapts to external stimuli, such as muscle mass and contractile strength, to alter its mass and shape. Myostatin, a member of the TGF-β superfamily, signals through activin receptor type IIB to negatively regulate muscle fiber growth. Because of the positive impact of myostatin deficiency on bone mass, we utilized a soluble activin receptor type IIB-mFc (sActRIIB-mFc) fusion protein in two molecularly distinct OI mouse models (G610C and oim) and evaluated their bone properties. Wild-type (WT), +/G610C, and oim/oim mice were treated from 2 to 4 months of age with either vehicle (Tris-buffered saline) or sActRIIB-mFc (10 mg/kg). Femurs of sActRIIB-mFc-treated mice exhibited increased trabecular bone volume regardless of genotype, whereas the cortical bone microarchitecture and biomechanical strength were only improved in WT and +/G610C mice. Dynamic histomorphometric analyses suggest the improved cortical bone geometry and biomechanical integrity reflect an anabolic effect due to increased mineral apposition and bone formation rates, whereas static histomorphometric analyses supported sActRIIB-mFc treatment also having an anti-catabolic impact with decreased osteoclast number per bone surface on trabecular bone regardless of sex and genotype. Together, our data suggest that sActRIIB-mFc may provide a new therapeutic direction to improve both bone and muscle properties in OI. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Youngjae Jeong
- Department of Biochemistry, University of Missouri, Columbia, Missouri, 65211
| | - Salah A. Daghlas
- Department of Biochemistry, University of Missouri, Columbia, Missouri, 65211
| | - Xie Yixia
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, Missouri, 64108
| | - Molly A Hulbert
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, Missouri, 64108
| | - Ferris M. Pfeiffer
- Department of Orthopaedic Surgery and Bioengineering, University of Missouri, Columbia, MO, 65211
| | - Mark R. Dallas
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, Missouri, 64108
| | | | | | - Sarah L. Dallas
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, Missouri, 64108
| | - Charlotte L. Phillips
- Department of Biochemistry, University of Missouri, Columbia, Missouri, 65211
- Department of Child Health, University of Missouri, Columbia, Missouri, 65211
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Osteogenesis imperfecta (OI) is a hereditary connective tissue disorder of skeletal fragility and more recently muscle weakness. This review highlights our current knowledge of the impact of compromised OI muscle function on muscle-bone interactions and skeletal strength in OI. RECENT FINDINGS The ramifications of inherent muscle weakness in OI muscle-bone interactions are just beginning to be elucidated. Studies in patients and in OI mouse models implicate altered mechanosensing, energy metabolism, mitochondrial dysfunction, and paracrine/endocrine crosstalk in the pathogenesis of OI. Compromised muscle-bone unit impacts mechanosensing and the ability of OI muscle and bone to respond to physiotherapeutic and pharmacologic treatment strategies. Muscle and bone are both compromised in OI, making it essential to understand the mechanisms responsible for both impaired muscle and bone functions and their interdependence, as this will expand and drive new physiotherapeutic and pharmacological approaches to treat OI and other musculoskeletal disorders.
Collapse
Affiliation(s)
- Charlotte L Phillips
- Department of Biochemistry, University of Missouri, 117 Schweitzer Hall, Columbia, MO, 65211, USA.
- Department of Child Health, University of Missouri, Columbia, MO, 65211, USA.
| | - Youngjae Jeong
- Department of Biochemistry, University of Missouri, 117 Schweitzer Hall, Columbia, MO, 65211, USA
| |
Collapse
|
26
|
Formicola L, Pannérec A, Correra RM, Gayraud-Morel B, Ollitrault D, Besson V, Tajbakhsh S, Lachey J, Seehra JS, Marazzi G, Sassoon DA. Inhibition of the Activin Receptor Type-2B Pathway Restores Regenerative Capacity in Satellite Cell-Depleted Skeletal Muscle. Front Physiol 2018; 9:515. [PMID: 29881353 PMCID: PMC5978452 DOI: 10.3389/fphys.2018.00515] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 04/20/2018] [Indexed: 12/11/2022] Open
Abstract
Degenerative myopathies typically display a decline in satellite cells coupled with a replacement of muscle fibers by fat and fibrosis. During this pathological remodeling, satellite cells are present at lower numbers and do not display a proper regenerative function. Whether a decline in satellite cells directly contributes to disease progression or is a secondary result is unknown. In order to dissect these processes, we used a genetic model to reduce the satellite cell population by ~70–80% which leads to a nearly complete loss of regenerative potential. We observe that while no overt tissue damage is observed following satellite cell depletion, muscle fibers atrophy accompanied by changes in the stem cell niche cellular composition. Treatment of these mice with an Activin receptor type-2B (AcvR2B) pathway blocker reverses muscle fiber atrophy as expected, but also restores regenerative potential of the remaining satellite cells. These findings demonstrate that in addition to controlling fiber size, the AcvR2B pathway acts to regulate the muscle stem cell niche providing a more favorable environment for muscle regeneration.
Collapse
Affiliation(s)
- Luigi Formicola
- UMR S 1166 French National Institute of Health and Medical Research, France and the Institute of Cardiometabolism and Nutrition, Stem Cells and Regenerative Medicine, University of Pierre and Marie Curie Paris VI, Paris, France
| | - Alice Pannérec
- UMR S 1166 French National Institute of Health and Medical Research, France and the Institute of Cardiometabolism and Nutrition, Stem Cells and Regenerative Medicine, University of Pierre and Marie Curie Paris VI, Paris, France
| | - Rosa Maria Correra
- UMR S 1166 French National Institute of Health and Medical Research, France and the Institute of Cardiometabolism and Nutrition, Stem Cells and Regenerative Medicine, University of Pierre and Marie Curie Paris VI, Paris, France
| | - Barbara Gayraud-Morel
- Centre National de la Recherche Scientifique URA 2578, Institut Pasteur, Stem Cells and Development, Paris, France
| | - David Ollitrault
- UMR S 1166 French National Institute of Health and Medical Research, France and the Institute of Cardiometabolism and Nutrition, Stem Cells and Regenerative Medicine, University of Pierre and Marie Curie Paris VI, Paris, France
| | - Vanessa Besson
- UMR S 1166 French National Institute of Health and Medical Research, France and the Institute of Cardiometabolism and Nutrition, Stem Cells and Regenerative Medicine, University of Pierre and Marie Curie Paris VI, Paris, France
| | - Shahragim Tajbakhsh
- Centre National de la Recherche Scientifique URA 2578, Institut Pasteur, Stem Cells and Development, Paris, France
| | - Jennifer Lachey
- Acceleron Pharma, Cambridge, MA, United States.,Ember Therapeutics, Watertown, MA, United States
| | - Jasbir S Seehra
- Acceleron Pharma, Cambridge, MA, United States.,Ember Therapeutics, Watertown, MA, United States
| | - Giovanna Marazzi
- UMR S 1166 French National Institute of Health and Medical Research, France and the Institute of Cardiometabolism and Nutrition, Stem Cells and Regenerative Medicine, University of Pierre and Marie Curie Paris VI, Paris, France
| | - David A Sassoon
- UMR S 1166 French National Institute of Health and Medical Research, France and the Institute of Cardiometabolism and Nutrition, Stem Cells and Regenerative Medicine, University of Pierre and Marie Curie Paris VI, Paris, France
| |
Collapse
|
27
|
Lodberg A, Eijken M, van der Eerden BCJ, Okkels MW, Thomsen JS, Brüel A. A soluble activin type IIA receptor mitigates the loss of femoral neck bone strength and cancellous bone mass in a mouse model of disuse osteopenia. Bone 2018; 110:326-334. [PMID: 29499419 DOI: 10.1016/j.bone.2018.02.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 02/21/2018] [Accepted: 02/26/2018] [Indexed: 10/17/2022]
Abstract
Disuse causes a rapid and substantial bone loss distinct in its pathophysiology from the bone loss associated with cancers, age, and menopause. While inhibitors of the activin-receptor signaling pathway (IASPs) have been shown to prevent ovariectomy- and cancer-induced bone loss, their application in a model of disuse osteopenia remains to be tested. Here, we show that a soluble activin type IIA receptor (ActRIIA-mFc) increases diaphyseal bone strength and cancellous bone mass, and mitigates the loss of femoral neck bone strength in the Botulinum Toxin A (BTX)-model of disuse osteopenia in female C57BL/6J mice. We show that ActRIIA-mFc treatment preferentially stimulates a dual-effect (anabolic-antiresorptive) on the periosteal envelope of diaphyseal bone, demonstrating in detail the effects of ActRIIA-mFc on cortical bone. These observations constitute a previously undescribed feature of IASPs that mediates at least part of their ability to mitigate detrimental effects of unloading on bone tissue. The study findings support the application of IASPs as a strategy to combat bone loss during disuse.
Collapse
Affiliation(s)
- Andreas Lodberg
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Aarhus University Hospital, Aarhus, Denmark.
| | - Marco Eijken
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark; Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark.
| | | | | | | | - Annemarie Brüel
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
28
|
Durán J, Estrada M. Commentary: Blockade of activin type II receptors with a dual anti-ActRIIA/IIB antibody is critical to promote maximal skeletal muscle hypertrophy. Front Pharmacol 2018; 9:381. [PMID: 29726548 PMCID: PMC5917040 DOI: 10.3389/fphar.2018.00381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 04/03/2018] [Indexed: 11/26/2022] Open
Affiliation(s)
- Javier Durán
- Program of Physiology and Biophysics, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Manuel Estrada
- Program of Physiology and Biophysics, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
29
|
Electrical impedance myography as a biomarker of myostatin inhibition with ActRIIB-mFc: a study in wild-type mice. Future Sci OA 2018; 4:FSO308. [PMID: 30057785 PMCID: PMC6060391 DOI: 10.4155/fsoa-2018-0002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 03/15/2018] [Indexed: 12/14/2022] Open
Abstract
Aim: We sought to determine the sensitivity of electrical impedance myography (EIM) to myofiber hypertrophy induced by treatment with various doses of ActRIIB-mFc, an inhibitor of myostatin signaling. Methods: Wild-type C57BL/6 J mice (n = 40, male) were treated with three different doses of ActRIIB-mFc (i.e., RAP-031) or vehicle twice weekly for 5 weeks. End point assessments included gastrocnemius EIM, force measurements, muscle mass and myofiber size quantification. Results: ActRIIB-mFc increased body mass, muscle mass and myofiber size across all doses. Alterations in EIM 50 kHz phase and center frequency (fc) were also present, with trends in a dose-dependent fashion. Significant correlations between EIM parameters and myofiber/functional data were identified. Conclusion: EIM outcomes can serve as effective biomarkers of myostatin signaling inhibition, demonstrating a dose sensitivity and correlation to standard assessments. We were interested in studying the sensitivity of a technique, called electrical impedance myography (EIM), to noninvasively assess the size of muscle fibers. In this technique a minute electrical current is used to probe the tissue. To do so, we gave a drug (ActRIIB-mFc) to mice that enlarges muscle fibers at three different doses. We were able to show that the EIM technique was able to detect this differential effect and functional changes induced by the drug correlated to the EIM data. This work suggests that EIM will be useful as a noninvasive marker muscle health.
Collapse
|
30
|
Kokabu S, Rosen V. BMP3 expression by osteoblast lineage cells is regulated by canonical Wnt signaling. FEBS Open Bio 2017; 8:168-176. [PMID: 29435407 PMCID: PMC5794463 DOI: 10.1002/2211-5463.12347] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 10/28/2017] [Accepted: 11/02/2017] [Indexed: 12/19/2022] Open
Abstract
Bone morphogenetic protein (BMP) and canonical Wnt (cWnt) signaling factors are both known to regulate bone mass, fracture risk, fracture repair, and osteoblastogenesis. BMP3 is the most abundant BMP and negatively regulates osteoblastogenesis and bone mass. Thus, identifying the mechanism by which BMP3 acts to depress bone formation may allow for the development of new therapeutics useful in the treatment for osteopenia and osteoporosis. Here, we report that cWnt signaling stimulates BMP3 expression in osteoblast (OB) lineage cells. The expression of BMP3 increases with OB differentiation. Treatment of cells with various cWnt proteins stimulated BMP3 expression. Mice with enhanced cWnt signaling had high expression levels of BMP3. Our data suggest that reduction in BMP3 levels may contribute beneficially to the positive effect of cWnt agonists on bone mass.
Collapse
Affiliation(s)
- Shoichiro Kokabu
- Department of Developmental Biology Harvard School of Dental Medicine Boston MA USA.,Division of Molecular Signaling and Biochemistry Department of Health Promotion Kyushu Dental University Kitakyushu Japan.,Department of Oral and Maxillofacial Surgery Faculty of Medicine Saitama Medical University Moroyama-machiIruma-gun Japan
| | - Vicki Rosen
- Department of Developmental Biology Harvard School of Dental Medicine Boston MA USA
| |
Collapse
|
31
|
Hudnall AM, Arthur JW, Lowery JW. Clinical Relevance and Mechanisms of Antagonism Between the BMP and Activin/TGF-β Signaling Pathways. J Osteopath Med 2017; 116:452-61. [PMID: 27367950 DOI: 10.7556/jaoa.2016.089] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The transforming growth factor β (TGF-β) superfamily is a large group of signaling molecules that participate in embryogenesis, organogenesis, and tissue homeostasis. These molecules are present in all animal genomes. Dysfunction in the regulation or activity of this superfamily's components underlies numerous human diseases and developmental defects. There are 2 distinct arms downstream of the TGF-β superfamily ligands-the bone morphogenetic protein (BMP) and activin/TGF-β signaling pathways-and these 2 responses can oppose one another's effects, most notably in disease states. However, studies have commonly focused on a single arm of the TGF-β superfamily, and the antagonism between these pathways is unknown in most physiologic and pathologic contexts. In this review, the authors summarize the clinically relevant scenarios in which the BMP and activin/TGF-β pathways reportedly oppose one another and identify several molecular mechanisms proposed to mediate this interaction. Particular attention is paid to experimental findings that may be informative to human pathology to highlight potential therapeutic approaches for future investigation.
Collapse
|
32
|
Cardozo CP, Graham ZA. Muscle-bone interactions: movement in the field of mechano-humoral coupling of muscle and bone. Ann N Y Acad Sci 2017; 1402:10-17. [PMID: 28763828 DOI: 10.1111/nyas.13411] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 05/15/2017] [Indexed: 12/12/2022]
Abstract
Cyclical, mechanical loading of bone by skeletal muscle is widely recognized as a critical determinant of bone structure and mass. A growing body of evidence indicates that substances released from skeletal muscle into the bloodstream also regulate bone mass and metabolism. In this commentary, we discuss the status of research in the area of humoral regulation of bone mass by the skeletal muscle secretome, with an emphasis on the roles of myostatin, irisin, interleukin-6, and exosomes. The interplay between muscle, bone, and other modulators of bone mass, including circadian rhythm and sympathetic tone, is also discussed.
Collapse
Affiliation(s)
- Christopher P Cardozo
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center, Bronx, New York.,Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Pharmacologic Science, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Zachary A Graham
- National Center for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center, Bronx, New York.,Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
33
|
Puolakkainen T, Rummukainen P, Lehto J, Ritvos O, Hiltunen A, Säämänen AM, Kiviranta R. Soluble activin type IIB receptor improves fracture healing in a closed tibial fracture mouse model. PLoS One 2017; 12:e0180593. [PMID: 28704409 PMCID: PMC5509431 DOI: 10.1371/journal.pone.0180593] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 06/16/2017] [Indexed: 12/26/2022] Open
Abstract
Fractures still present a significant burden to patients due to pain and periods of unproductivity. Numerous growth factors have been identified to regulate bone remodeling. However, to date, only the bone morphogenetic proteins (BMPs) are used to enhance fracture healing in clinical settings. Activins are pleiotropic growth factors belonging to the TGF-β superfamily. We and others have recently shown that treatment with recombinant fusion proteins of activin receptors greatly increases bone mass in different animal models by trapping activins and other ligands thus inhibiting their signaling pathways. However, their effects on fracture healing are less known. Twelve-week old male C57Bl mice were subjected to a standardized, closed tibial fracture model. Animals were divided into control and treatment groups and were administered either PBS control or a soluble activin type IIB receptor (ActRIIB-Fc) intraperitoneally once a week for a duration of two or four weeks. There were no significant differences between the groups at two weeks but we observed a significant increase in callus mineralization in ActRIIB-Fc-treated animals by microcomputed tomography imaging at four weeks. Bone volume per tissue volume was 60%, trabecular number 55% and bone mineral density 60% higher in the 4-week calluses of the ActRIIB-Fc-treated mice (p<0.05 in all). Biomechanical strength of 4-week calluses was also significantly improved by ActRIIB-Fc treatment as stiffness increased by 64% and maximum force by 45% (p<0.05) compared to the PBS-injected controls. These results demonstrate that ActRIIB-Fc treatment significantly improves healing of closed long bone fractures. Our findings support the previous reports of activin receptors increasing bone mass but also demonstrate a novel approach for using ActRIIB-Fc to enhance fracture healing.
Collapse
Affiliation(s)
| | | | - Jemina Lehto
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Olli Ritvos
- Department of Physiology, University of Helsinki, Helsinki, Finland
| | | | | | - Riku Kiviranta
- Institute of Biomedicine, University of Turku, Turku, Finland.,Division of Endocrinology, Turku University Hospital, Turku, Finland
| |
Collapse
|
34
|
Goh BC, Singhal V, Herrera AJ, Tomlinson RE, Kim S, Faugere MC, Germain-Lee EL, Clemens TL, Lee SJ, DiGirolamo DJ. Activin receptor type 2A (ACVR2A) functions directly in osteoblasts as a negative regulator of bone mass. J Biol Chem 2017; 292:13809-13822. [PMID: 28659341 DOI: 10.1074/jbc.m117.782128] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 06/27/2017] [Indexed: 02/06/2023] Open
Abstract
Bone and skeletal muscle mass are highly correlated in mammals, suggesting the existence of common anabolic signaling networks that coordinate the development of these two anatomically adjacent tissues. The activin signaling pathway is an attractive candidate to fulfill such a role. Here, we generated mice with conditional deletion of activin receptor (ACVR) type 2A, ACVR2B, or both, in osteoblasts, to determine the contribution of activin receptor signaling in regulating bone mass. Immunohistochemistry localized ACVR2A and ACVR2B to osteoblasts and osteocytes. Primary osteoblasts expressed activin signaling components, including ACVR2A, ACVR2B, and ACVR1B (ALK4) and demonstrated increased levels of phosphorylated Smad2/3 upon exposure to activin ligands. Osteoblasts lacking ACVR2B did not show significant changes in vitro However, osteoblasts deficient in ACVR2A exhibited enhanced differentiation indicated by alkaline phosphatase activity, mineral deposition, and transcriptional expression of osterix, osteocalcin, and dentin matrix acidic phosphoprotein 1. To investigate activin signaling in osteoblasts in vivo, we analyzed the skeletal phenotypes of mice lacking these receptors in osteoblasts and osteocytes (osteocalcin-Cre). Similar to the lack of effect in vitro, ACVR2B-deficient mice demonstrated no significant change in any bone parameter. By contrast, mice lacking ACVR2A had significantly increased femoral trabecular bone volume at 6 weeks of age. Moreover, mutant mice lacking both ACVR2A and ACVR2B demonstrated sustained increases in trabecular bone volume, similar to those in ACVR2A single mutants, at 6 and 12 weeks of age. Taken together, these results indicate that activin receptor signaling, predominantly through ACVR2A, directly and negatively regulates bone mass in osteoblasts.
Collapse
Affiliation(s)
- Brian C Goh
- From the Departments of Orthopaedic Surgery and
| | | | | | | | - Soohyun Kim
- From the Departments of Orthopaedic Surgery and
| | - Marie-Claude Faugere
- the Albert B. Chandler Medical Center, University of Kentucky, Lexington, Kentucky 40536
| | - Emily L Germain-Lee
- the Department of Pediatrics, University of Connecticut School of Medicine/UConn Health, Farmington, Connecticut 06030.,the Division of Pediatric Endocrinology, Connecticut Children's Medical Center, Hartford, Connecticut 06106, and
| | - Thomas L Clemens
- From the Departments of Orthopaedic Surgery and.,the Veterans Administration Medical Center, Baltimore, Maryland 21201
| | - Se-Jin Lee
- Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287
| | | |
Collapse
|
35
|
Guo W, Pencina KM, O'Connell K, Montano M, Peng L, Westmoreland S, Glowacki J, Bhasin S. Administration of an activin receptor IIB ligand trap protects male juvenile rhesus macaques from simian immunodeficiency virus-associated bone loss. Bone 2017; 97:209-215. [PMID: 28132908 PMCID: PMC5985824 DOI: 10.1016/j.bone.2017.01.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 01/07/2017] [Accepted: 01/25/2017] [Indexed: 11/30/2022]
Abstract
UNLABELLED HIV-infected individuals are at an increased risk of osteoporosis despite effective viral suppression. Observations that myostatin null mice have increased bone mass led us to hypothesize that simian immunodeficiency virus (SIV)-associated bone loss may be attenuated by blocking myostatin/TGFβ signaling. In this proof-of-concept study, pair-housed juvenile male rhesus macaques were inoculated with SIVmac239. Four weeks later, animals were treated with vehicle or Fc-conjugated soluble activin receptor IIB (ActR2B·Fc, iv. 10mg∗kg-1∗week-1) - an antagonist of myostatin and related members of TGFβ superfamily. Limb and trunk bone mineral content (BMC) and density (BMD) using dual-energy X-Ray absorptiometry, circulating markers of bone growth and turnover, and serum testosterone levels were measured at baseline and during the 12-week intervention period. The increase in BMC was significantly greater in the ActRIIB.Fc-treated group (+8g) than in the placebo group (-4g) (p<0.05). BMD also increased significantly more in the ActRIIB.Fc-treated macaques (+0.03g/cm2) than in the placebo-treated animals (+0g/cm2) (p<0.005). Serum osteocalcin was about two-fold higher in the ActRIIB.Fc-treated group than in the placebo group (p<0.05), but serum C-terminal telopeptide and testosterone levels did not differ significantly between groups. The expression levels of TNFalpha (p<0.05), GADD45 (p<0.005), and sclerostin (p<0.038) in the bone-marrow were significantly lower in the ActRIIB.Fc-treated group than in the placebo group. CONCLUSION The administration of ActRIIB.FC in SIV-infected juvenile macaques significantly increases BMC and BMD in association with reduced expression levels of markers of bone marrow inflammation.
Collapse
Affiliation(s)
- Wen Guo
- Research Program in Men's Health: Aging and Metabolism, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States.
| | - Karol M Pencina
- Research Program in Men's Health: Aging and Metabolism, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Karyn O'Connell
- Department of Comparative Pathology, New England Primate Research Center, One Pine Hill Drive, PO Box 9102, Southborough, MA 01772-9102, United States
| | - Monty Montano
- Research Program in Men's Health: Aging and Metabolism, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Liming Peng
- Research Program in Men's Health: Aging and Metabolism, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Susan Westmoreland
- Department of Comparative Pathology, New England Primate Research Center, One Pine Hill Drive, PO Box 9102, Southborough, MA 01772-9102, United States
| | - Julie Glowacki
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Shalender Bhasin
- Research Program in Men's Health: Aging and Metabolism, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| |
Collapse
|
36
|
Puolakkainen T, Ma H, Kainulainen H, Pasternack A, Rantalainen T, Ritvos O, Heikinheimo K, Hulmi JJ, Kiviranta R. Treatment with soluble activin type IIB-receptor improves bone mass and strength in a mouse model of Duchenne muscular dystrophy. BMC Musculoskelet Disord 2017; 18:20. [PMID: 28103859 PMCID: PMC5244551 DOI: 10.1186/s12891-016-1366-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 12/14/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Inhibition of activin/myostatin pathway has emerged as a novel approach to increase muscle mass and bone strength. Duchenne muscular dystrophy (DMD) is a neuromuscular disorder that leads to progressive muscle degeneration and also high incidence of fractures. The aim of our study was to test whether inhibition of activin receptor IIB ligands with or without exercise could improve bone strength in the mdx mouse model for DMD. METHODS Thirty-two mdx mice were divided to running and non-running groups and to receive either PBS control or soluble activin type IIB-receptor (ActRIIB-Fc) once weekly for 7 weeks. RESULTS Treatment of mdx mice with ActRIIB-Fc resulted in significantly increased body and muscle weights in both sedentary and exercising mice. Femoral μCT analysis showed increased bone volume and trabecular number (BV/TV +80%, Tb.N +70%, P < 0.05) in both ActRIIB-Fc treated groups. Running also resulted in increased bone volume and trabecular number in PBS-treated mice. However, there was no significant difference in trabecular bone structure or volumetric bone mineral density between the ActRIIB-Fc and ActRIIB-Fc-R indicating that running did not further improve bone structure in ActRIIB-Fc-treated mice. ActRIIB-Fc increased bone mass also in vertebrae (BV/TV +20%, Tb.N +30%, P < 0.05) but the effects were more modest. The number of osteoclasts was decreased in histological analysis and the expression of several osteoblast marker genes was increased in ActRIIB-Fc treated mice suggesting decreased bone resorption and increased bone formation in these mice. Increased bone mass in femurs translated into enhanced bone strength in biomechanical testing as the maximum force and stiffness were significantly elevated in ActRIIB-Fc-treated mice. CONCLUSIONS Our results indicate that treatment of mdx mice with the soluble ActRIIB-Fc results in a robust increase in bone mass, without any additive effect by voluntary running. Thus ActRIIB-Fc could be an attractive option in the treatment of musculoskeletal disorders.
Collapse
Affiliation(s)
- Tero Puolakkainen
- Department of Medical Biochemistry and Genetics, University of Turku, Kiinamyllynkatu 10, FI-20520, Turku, Finland
| | - Hongqian Ma
- Department of Biology of Physical Activity, University of Jyväskylä, Jyväskylä, Finland.,Institute of Dentistry, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Heikki Kainulainen
- Department of Biology of Physical Activity, University of Jyväskylä, Jyväskylä, Finland
| | - Arja Pasternack
- Department of Physiology, University of Helsinki, Helsinki, Finland
| | - Timo Rantalainen
- Centre for Physical Activity and Nutrition Research, Deakin University, Melbourne, Australia
| | - Olli Ritvos
- Department of Physiology, University of Helsinki, Helsinki, Finland
| | - Kristiina Heikinheimo
- Department of Oral and Maxillofacial Surgery, Institute of Dentistry, University of Turku, Turku, Finland.,Department of Oral Diagnostic Sciences, Institute of Dentistry, University of Eastern Finland, Kuopio, Finland.,Kuopio University Hospital, Kuopio, Finland
| | - Juha J Hulmi
- Department of Biology of Physical Activity, University of Jyväskylä, Jyväskylä, Finland.,Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Riku Kiviranta
- Department of Medical Biochemistry and Genetics, University of Turku, Kiinamyllynkatu 10, FI-20520, Turku, Finland. .,Department of Endocrinology, Turku University Hospital, Turku, Finland.
| |
Collapse
|
37
|
Pan C, Singh S, Sahasrabudhe DM, Chakkalakal JV, Krolewski JJ, Nastiuk KL. TGFβ Superfamily Members Mediate Androgen Deprivation Therapy-Induced Obese Frailty in Male Mice. Endocrinology 2016; 157:4461-4472. [PMID: 27611336 PMCID: PMC5414572 DOI: 10.1210/en.2016-1580] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
First line treatment for recurrent and metastatic prostate cancer is androgen deprivation therapy (ADT). Use of ADT has been increasing in frequency and duration, such that side effects increasingly impact patient quality of life. One of the most significant side effects of ADT is sarcopenia, which leads to a loss of skeletal muscle mass and function, resulting in a clinical disability syndrome known as obese frailty. Using aged mice, we developed a mouse model of ADT-induced sarcopenia that closely resembles the phenotype seen in patients, including loss of skeletal muscle strength, reduced lean muscle mass, and increased adipose tissue. Sarcopenia onset occurred about 6 weeks after castration and was blocked by a soluble receptor (ActRIIB-Fc) that binds multiple TGFβ superfamily members, including myostatin, growth differentiation factor 11, activin A, activin B, and activin AB. Analysis of ligand expression in both gastrocnemius and triceps brachii muscles demonstrates that each of these proteins is induced in response to ADT, in 1 of 3 temporal patterns. Specifically, activin A and activin AB levels increase and decline before onset of strength loss at 6 weeks after castration, and myostatin levels increase coincident with the onset of strength loss and then decline. In contrast, activin B and growth differentiation factor 11 levels increase after the onset of strength loss, 8-10 weeks after castration. The observed patterns of ligand induction may represent differential contributions to the development and/or maintenance of sarcopenia. We hypothesize that some or all of these ligands are targets for therapy to ameliorate ADT-induced sarcopenia in prostate cancer patients.
Collapse
Affiliation(s)
- Chunliu Pan
- Department of Cancer Genetics (C.P., S.S., J.J.K., K.L.N.) and Center for Personalized Medicine (J.J.K.), Roswell Park Cancer Institute; Buffalo, New York 14263; and James P. Wilmot Cancer Center and Department of Medicine (D.M.S.), Department of Orthopedics and Center for Musculoskeletal Research (J.V.C.), and Department of Pathology and Laboratory Medicine (K.L.N.), University of Rochester, School of Medicine and Dentistry, Rochester, New York 14642
| | - Shalini Singh
- Department of Cancer Genetics (C.P., S.S., J.J.K., K.L.N.) and Center for Personalized Medicine (J.J.K.), Roswell Park Cancer Institute; Buffalo, New York 14263; and James P. Wilmot Cancer Center and Department of Medicine (D.M.S.), Department of Orthopedics and Center for Musculoskeletal Research (J.V.C.), and Department of Pathology and Laboratory Medicine (K.L.N.), University of Rochester, School of Medicine and Dentistry, Rochester, New York 14642
| | - Deepak M Sahasrabudhe
- Department of Cancer Genetics (C.P., S.S., J.J.K., K.L.N.) and Center for Personalized Medicine (J.J.K.), Roswell Park Cancer Institute; Buffalo, New York 14263; and James P. Wilmot Cancer Center and Department of Medicine (D.M.S.), Department of Orthopedics and Center for Musculoskeletal Research (J.V.C.), and Department of Pathology and Laboratory Medicine (K.L.N.), University of Rochester, School of Medicine and Dentistry, Rochester, New York 14642
| | - Joe V Chakkalakal
- Department of Cancer Genetics (C.P., S.S., J.J.K., K.L.N.) and Center for Personalized Medicine (J.J.K.), Roswell Park Cancer Institute; Buffalo, New York 14263; and James P. Wilmot Cancer Center and Department of Medicine (D.M.S.), Department of Orthopedics and Center for Musculoskeletal Research (J.V.C.), and Department of Pathology and Laboratory Medicine (K.L.N.), University of Rochester, School of Medicine and Dentistry, Rochester, New York 14642
| | - John J Krolewski
- Department of Cancer Genetics (C.P., S.S., J.J.K., K.L.N.) and Center for Personalized Medicine (J.J.K.), Roswell Park Cancer Institute; Buffalo, New York 14263; and James P. Wilmot Cancer Center and Department of Medicine (D.M.S.), Department of Orthopedics and Center for Musculoskeletal Research (J.V.C.), and Department of Pathology and Laboratory Medicine (K.L.N.), University of Rochester, School of Medicine and Dentistry, Rochester, New York 14642
| | - Kent L Nastiuk
- Department of Cancer Genetics (C.P., S.S., J.J.K., K.L.N.) and Center for Personalized Medicine (J.J.K.), Roswell Park Cancer Institute; Buffalo, New York 14263; and James P. Wilmot Cancer Center and Department of Medicine (D.M.S.), Department of Orthopedics and Center for Musculoskeletal Research (J.V.C.), and Department of Pathology and Laboratory Medicine (K.L.N.), University of Rochester, School of Medicine and Dentistry, Rochester, New York 14642
| |
Collapse
|
38
|
Laurent MR, Dubois V, Claessens F, Verschueren SMP, Vanderschueren D, Gielen E, Jardí F. Muscle-bone interactions: From experimental models to the clinic? A critical update. Mol Cell Endocrinol 2016; 432:14-36. [PMID: 26506009 DOI: 10.1016/j.mce.2015.10.017] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 10/13/2015] [Accepted: 10/20/2015] [Indexed: 02/06/2023]
Abstract
Bone is a biomechanical tissue shaped by forces from muscles and gravitation. Simultaneous bone and muscle decay and dysfunction (osteosarcopenia or sarco-osteoporosis) is seen in ageing, numerous clinical situations including after stroke or paralysis, in neuromuscular dystrophies, glucocorticoid excess, or in association with vitamin D, growth hormone/insulin like growth factor or sex steroid deficiency, as well as in spaceflight. Physical exercise may be beneficial in these situations, but further work is still needed to translate acceptable and effective biomechanical interventions like vibration therapy from animal models to humans. Novel antiresorptive and anabolic therapies are emerging for osteoporosis as well as drugs for sarcopenia, cancer cachexia or muscle wasting disorders, including antibodies against myostatin or activin receptor type IIA and IIB (e.g. bimagrumab). Ideally, increasing muscle mass would increase muscle strength and restore bone loss from disuse. However, the classical view that muscle is unidirectionally dominant over bone via mechanical loading is overly simplistic. Indeed, recent studies indicate a role for neuronal regulation of not only muscle but also bone metabolism, bone signaling pathways like receptor activator of nuclear factor kappa-B ligand (RANKL) implicated in muscle biology, myokines affecting bone and possible bone-to-muscle communication. Moreover, pharmacological strategies inducing isolated myocyte hypertrophy may not translate into increased muscle power because tendons, connective tissue, neurons and energy metabolism need to adapt as well. We aim here to critically review key musculoskeletal molecular pathways involved in mechanoregulation and their effect on the bone-muscle unit as a whole, as well as preclinical and emerging clinical evidence regarding the effects of sarcopenia therapies on osteoporosis and vice versa.
Collapse
Affiliation(s)
- Michaël R Laurent
- Gerontology and Geriatrics, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium; Centre for Metabolic Bone Diseases, University Hospitals Leuven, 3000 Leuven, Belgium.
| | - Vanessa Dubois
- Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Frank Claessens
- Laboratory of Molecular Endocrinology, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Sabine M P Verschueren
- Research Group for Musculoskeletal Rehabilitation, Department of Rehabilitation Science, KU Leuven, 3000 Leuven, Belgium
| | - Dirk Vanderschueren
- Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Evelien Gielen
- Gerontology and Geriatrics, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium; Centre for Metabolic Bone Diseases, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Ferran Jardí
- Clinical and Experimental Endocrinology, Department of Clinical and Experimental Medicine, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
39
|
Castration induced browning in subcutaneous white adipose tissue in male mice. Biochem Biophys Res Commun 2016; 478:1746-50. [PMID: 27608598 DOI: 10.1016/j.bbrc.2016.09.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 09/03/2016] [Indexed: 11/23/2022]
Abstract
We demonstrated that castration enhanced the expression of uncoupling protein 1 (Ucp1), a thermogenic protein, in brown adipose tissue (BAT) and subcutaneous (sc) white adipose tissue (WAT) in male mice. Castration of male mice increased body temperature and reduced body weight gain compared with those of sham-operated mice. BAT Ucp1 mRNA expression in castrated male mice was significantly higher than that in sham-operated mice. Histologically, cells with multilocular fat droplets were observed in the castrated inguinal scWAT. Immunohistochemical staining revealed that these cells positively reacted with the anti-Ucp1 antibody. The Ucp1-positive area near the inguinal lymph node in the castrated WAT was extensive compared with that of the sham-operated WAT. Castration-induced Ucp1 up-regulation in scWAT was suppressed by high-fat diet feeding. These findings suggest that thermogenesis by BAT activation and scWAT browning contribute to castration-induced inhibition of body weight gain. However, considering that the effect of castration was blunted by high-fat diet consumption, thermogenesis stimulation in response to castration is inhibited by chronic over-nutrition.
Collapse
|
40
|
Graham ZA, Collier L, Peng Y, Saéz JC, Bauman WA, Qin W, Cardozo CP. A Soluble Activin Receptor IIB Fails to Prevent Muscle Atrophy in a Mouse Model of Spinal Cord Injury. J Neurotrauma 2016; 33:1128-35. [PMID: 26529111 DOI: 10.1089/neu.2015.4058] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Myostatin (MST) is a potent regulator of muscle growth and size. Spinal cord injury (SCI) results in marked atrophy of muscle below the level of injury. Currently, there is no effective pharmaceutical treatment available to prevent sublesional muscle atrophy post-SCI. To determine whether inhibition of MST with a soluble activin IIB receptor (RAP-031) prevents sublesional SCI-induced muscle atrophy, mice were randomly assigned to the following groups: Sham-SCI; SCI+Vehicle group (SCI-VEH); and SCI+RAP-031 (SCI-RAP-031). SCI was induced by complete transection at thoracic level 10. Animals were euthanized at 56 days post-surgery. RAP-031 reduced, but did not prevent, body weight loss post-SCI. RAP-031 increased total lean tissue mass compared to SCI-VEH (14.8%). RAP-031 increased forelimb muscle mass post-SCI by 38% and 19% for biceps and triceps, respectively (p < 0.001). There were no differences in hindlimb muscle weights between the RAP-031 and SCI-VEH groups. In the gastrocnemius, messenger RNA (mRNA) expression was elevated for interleukin (IL)-6 (8-fold), IL-1β (3-fold), and tumor necrosis factor alpha (8-fold) in the SCI-VEH, compared to the Sham group. Muscle RING finger protein 1 mRNA was 2-fold greater in the RAP-031 group, compared to Sham-SCI. RAP-031 did not influence cytokine expression. Bone mineral density of the distal femur and proximal tibia were decreased post-SCI (-26% and -28%, respectively) and were not altered by RAP-031. In conclusion, MST inhibition increased supralesional muscle mass, but did not prevent sublesional muscle or bone loss, or the inflammation in paralyzed muscle.
Collapse
Affiliation(s)
- Zachary A Graham
- 1 National Center of Excellence for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center , Bronx, New York.,5 Department of Medicine, Icahn School of Medicine at Mount Sinai , New York, New York
| | - Lauren Collier
- 1 National Center of Excellence for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center , Bronx, New York
| | - Yuanzhen Peng
- 1 National Center of Excellence for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center , Bronx, New York
| | - Juan C Saéz
- 3 Department of Physiology, Pontificia Universidad Católica , Santiago, Chile .,4 Centro Interdisciplinario de Neurociencias de Valparaiso , Valparaiso, Chile
| | - William A Bauman
- 1 National Center of Excellence for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center , Bronx, New York.,2 Medical Service, James J. Peters VA Medical Center , Bronx, New York.,5 Department of Medicine, Icahn School of Medicine at Mount Sinai , New York, New York.,6 Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai , New York, New York
| | - Weiping Qin
- 1 National Center of Excellence for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center , Bronx, New York.,5 Department of Medicine, Icahn School of Medicine at Mount Sinai , New York, New York
| | - Christopher P Cardozo
- 1 National Center of Excellence for the Medical Consequences of Spinal Cord Injury, James J. Peters VA Medical Center , Bronx, New York.,2 Medical Service, James J. Peters VA Medical Center , Bronx, New York.,5 Department of Medicine, Icahn School of Medicine at Mount Sinai , New York, New York.,6 Department of Rehabilitation Medicine, Icahn School of Medicine at Mount Sinai , New York, New York.,7 Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai , New York, New York
| |
Collapse
|
41
|
The soluble form of BMPRIB is a novel therapeutic candidate for treating bone related disorders. Sci Rep 2016; 6:18849. [PMID: 26732094 PMCID: PMC4702141 DOI: 10.1038/srep18849] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 11/27/2015] [Indexed: 11/08/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are multi-functional growth factors that belong to the TGF-beta superfamily. Recently, several soluble BMP receptors, such as ActRIIA-Fc, ActRIIB-Fc, and ALK1-Fc, are undergoing clinical trials. Both BMPRIA and BMPRIB are type I BMP receptors, and while BMPRIA-Fc has been reported to have bone-increasing properties, there have been no investigations concerning the biological functions of BMPRIB-Fc. Therefore, comparing the effects of BMPRIA-Fc and BMPRIB-Fc in vivo should be helpful in revealing the differences in biological function between BMPRIA and BMPRIB, and would also aid in the evaluation of BMPRIB-Fc as a therapeutic agent. Here, we produced Tg chimeras in which BMPRIA-Fc and BMPRIB-Fc proteins circulated at high concentrations (36.8–121.4 μg/mL). Both Tg chimeras showed a significant increase of bone volume and strength. Using histological analysis, adenoma of the glandular stomach was observed only in BMPRIA-Fc chimeras suggesting the tumorigenic activity of this protein. Administration of recombinant BMPRIB-Fc protein to normal mice also increased bone volumes. Finally, treatment with BMPRIB-Fc decreased the area of osteolytic regions in a mouse model of breast cancer metastasis. In conclusion, our data suggest that BMPRIB-Fc can be used for the treatment of bone-related disorders with a lower risk than BMPRIA-Fc.
Collapse
|
42
|
Aoki A, Fujitani K, Takagi K, Kimura T, Nagase H, Nakanishi T. Male Hypogonadism Causes Obesity Associated with Impairment of Hepatic Gluconeogenesis in Mice. Biol Pharm Bull 2016; 39:587-92. [DOI: 10.1248/bpb.b15-00942] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Akira Aoki
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University
| | - Kohei Fujitani
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University
| | - Kohei Takagi
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University
| | - Tomoki Kimura
- Laboratory of Toxicology, Faculty of Pharmaceutical Sciences, Setsunan University
| | - Hisamitsu Nagase
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University
| | - Tsuyoshi Nakanishi
- Laboratory of Hygienic Chemistry and Molecular Toxicology, Gifu Pharmaceutical University
| |
Collapse
|
43
|
Kokabu S, Tsuchiya-Hirata S, Fukushima H, Sugiyama G, Lowery JW, Katagiri T, Jimi E. Inhibition of bone morphogenetic protein-induced osteoblast differentiation. J Oral Biosci 2015. [DOI: 10.1016/j.job.2015.05.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
44
|
Kalinkovich A, Livshits G. Sarcopenia--The search for emerging biomarkers. Ageing Res Rev 2015; 22:58-71. [PMID: 25962896 DOI: 10.1016/j.arr.2015.05.001] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 05/06/2015] [Accepted: 05/06/2015] [Indexed: 12/12/2022]
Abstract
Sarcopenia, an age-related decline in skeletal muscle mass and function, dramatically affects the life quality of elder people. In view of increasing life expectancy, sarcopenia renders a heavy burden on the health care system. However, although there is a consensus that sarcopenia is a multifactorial syndrome, its etiology, underlying mechanisms, and even definition remain poorly delineated, thus, preventing development of a precise treatment strategy. The main aim of our review is to critically analyze potential sarcopenia biomarkers in light of the molecular mechanisms of their involvement in sarcopenia pathogenesis. Normal muscle mass and function maintenance are proposed to be dependent on the dynamic balance between the positive regulators of muscle growth such as bone morphogenetic proteins (BMPs), brain-derived neurotrophic factor (BDNF), follistatin (FST) and irisin, and negative regulators including TGFβ, myostatin, activins A and B, and growth and differentiation factor-15 (GDF-15). We hypothesize that the shift in this balance to muscle growth inhibitors, along with increased expression of the C- terminal agrin fragment (CAF) associated with age-dependent neuromuscular junction (NMJ) dysfunction, as well as skeletal muscle-specific troponin T (sTnT), a key component of contractile machinery, is a main mechanism underlying sarcopenia pathogenesis. Thus, this review proposes and emphasizes that these molecules are the emerging sarcopenia biomarkers.
Collapse
|
45
|
Girgis CM. Integrated therapies for osteoporosis and sarcopenia: from signaling pathways to clinical trials. Calcif Tissue Int 2015; 96:243-55. [PMID: 25633430 DOI: 10.1007/s00223-015-9956-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 01/15/2015] [Indexed: 12/15/2022]
Abstract
Sarcopenia and osteoporosis are two sides of the same coin. They represent different aspects of the same age-related process of musculoskeletal atrophy and together culminate in falls, fractures, deconditioning, and increased mortality in older individuals. However, the current therapeutic approach to the prevention of minimal trauma fracture is unilateral and focuses solely on bone. In theory, an integrated approach that recognizes the interaction between muscle and bone could break the vicious cycle of their combined involution and more effectively minimize falls/fractures. In this review, signaling pathways and cross-talk mechanisms that integrate bone/muscle, and the emergence of novel therapies that exploit these pathways to target osteoporosis/sarcopenia will be discussed. In broad terms, these agents act on nuclear receptors (e.g., VDR, AR) or transmembrane receptors (e.g., activins, GH/IGF-1) expressed in muscle and bone, and seek to alter biologic responses to musculoskeletal aging, loading, and injury. Challenges in the development of these dual bone-muscle therapies, early clinical trials examining their safety/efficacy, and novel targets that hold promise in the reversal of musculoskeletal aging will be discussed.
Collapse
Affiliation(s)
- Christian M Girgis
- Westmead Millennium Institute for Medical Research, 176 Hawkesbury Rd, Westmead, NSW, Australia,
| |
Collapse
|
46
|
DiGirolamo DJ, Singhal V, Chang X, Lee SJ, Germain-Lee EL. Administration of soluble activin receptor 2B increases bone and muscle mass in a mouse model of osteogenesis imperfecta. Bone Res 2015; 3:14042. [PMID: 26161291 PMCID: PMC4472144 DOI: 10.1038/boneres.2014.42] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 11/01/2014] [Accepted: 11/03/2014] [Indexed: 11/09/2022] Open
Abstract
Osteogenesis imperfecta (OI) comprises a group of heritable connective tissue disorders generally defined by recurrent fractures, low bone mass, short stature and skeletal fragility. Beyond the skeletal complications of OI, many patients also report intolerance to physical activity, fatigue and muscle weakness. Indeed, recent studies have demonstrated that skeletal muscle is also negatively affected by OI, both directly and indirectly. Given the well-established interdependence of bone and skeletal muscle in both physiology and pathophysiology and the observations of skeletal muscle pathology in patients with OI, we investigated the therapeutic potential of simultaneous anabolic targeting of both bone and skeletal muscle using a soluble activin receptor 2B (ACVR2B) in a mouse model of type III OI (oim). Treatment of 12-week-old oim mice with ACVR2B for 4 weeks resulted in significant increases in both bone and muscle that were similar to those observed in healthy, wild-type littermates. This proof of concept study provides encouraging evidence for a holistic approach to treating the deleterious consequences of OI in the musculoskeletal system.
Collapse
Affiliation(s)
- Douglas J DiGirolamo
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine , Baltimore, MD, USA
| | - Vandana Singhal
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine , Baltimore, MD, USA
| | - Xiaoli Chang
- Bone & Osteogenesis Imperfecta Department, Kennedy Krieger Institute , Baltimore, MD, USA ; Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine , Baltimore, MD, USA
| | - Se-Jin Lee
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine , Baltimore, MD, USA
| | - Emily L Germain-Lee
- Bone & Osteogenesis Imperfecta Department, Kennedy Krieger Institute , Baltimore, MD, USA ; Department of Pediatrics, Johns Hopkins University School of Medicine , Baltimore, MD, USA
| |
Collapse
|
47
|
Lowery JW, Intini G, Gamer L, Lotinun S, Salazar VS, Ote S, Cox K, Baron R, Rosen V. Loss of BMPR2 leads to high bone mass due to increased osteoblast activity. J Cell Sci 2015; 128:1308-15. [PMID: 25663702 DOI: 10.1242/jcs.156737] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Imbalances in the ratio of bone morphogenetic protein (BMP) versus activin and TGFβ signaling are increasingly associated with human diseases yet the mechanisms mediating this relationship remain unclear. The type 2 receptors ACVR2A and ACVR2B bind BMPs and activins but the type 2 receptor BMPR2 only binds BMPs, suggesting that type 2 receptor utilization might play a role in mediating the interaction of these pathways. We tested this hypothesis in the mouse skeleton, where bone mass is reciprocally regulated by BMP signaling and activin and TGFβ signaling. We found that deleting Bmpr2 in mouse skeletal progenitor cells (Bmpr2-cKO mice) selectively impaired activin signaling but had no effect on BMP signaling, resulting in an increased bone formation rate and high bone mass. Additionally, activin sequestration had no effect on bone mass in Bmpr2-cKO mice but increased bone mass in wild-type mice. Our findings suggest a novel model whereby BMPR2 availability alleviates receptor-level competition between BMPs and activins and where utilization of ACVR2A and ACVR2B by BMPs comes at the expense of activins. As BMP and activin pathway modulation are of current therapeutic interest, our findings provide important mechanistic insight into the relationship between these pathways in human health.
Collapse
Affiliation(s)
- Jonathan W Lowery
- Department of Biomedical Science, Marian University College of Osteopathic Medicine, Indianapolis, IN 46222, USA Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Giuseppe Intini
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Laura Gamer
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Sutada Lotinun
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, USA Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Valerie S Salazar
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Satoshi Ote
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Karen Cox
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Roland Baron
- Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Vicki Rosen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| |
Collapse
|
48
|
Wang Q, Guo T, Portas J, McPherron AC. A soluble activin receptor type IIB does not improve blood glucose in streptozotocin-treated mice. Int J Biol Sci 2015; 11:199-208. [PMID: 25561902 PMCID: PMC4279095 DOI: 10.7150/ijbs.10430] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 11/05/2014] [Indexed: 12/18/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM), or insulin dependent DM, is accompanied by decreased muscle mass. The growth factor myostatin (MSTN) is a negative regulator of muscle growth, and a loss of MSTN signaling has been shown to increase muscle mass and prevent the development of obesity, insulin resistance and lipodystrophic diabetes in mice. The effects of MSTN inhibition in a T1DM model on muscle mass and blood glucose are unknown. We asked whether MSTN inhibition would increase muscle mass and decrease hyperglycemia in mice treated with streptozotocin (STZ) to destroy pancreatic beta cells. After diabetes developed, mice were treated with a soluble MSTN/activin receptor fused to Fc (ACVR2B:Fc). ACVR2B:Fc increased body weight and muscle mass compared to vehicle treated mice. Unexpectedly, ACVR2B:Fc reproducibly exacerbated hyperglycemia within approximately one week of administration. ACVR2B:Fc treatment also elevated serum levels of the glucocorticoid corticosterone. These results suggest that although MSTN/activin inhibitors increased muscle mass, they may be counterproductive in improving health in patients with T1DM.
Collapse
Affiliation(s)
- Qian Wang
- 1. Current Addresses: Pathology Department, Stony Brook University Medical Center, Stony Brook, New York, USA
| | - Tingqing Guo
- 2. Novo Nordisk Research Centre China, Changping District, Beijing, China
| | - Jennifer Portas
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | | |
Collapse
|
49
|
Abstract
Musculoskeletal diseases are highly prevalent with staggering annual health care costs across the globe. The combined wasting of muscle (sarcopenia) and bone (osteoporosis)-both in normal aging and pathologic states-can lead to vastly compounded risk for fracture in patients. Until now, our therapeutic approach to the prevention of such fractures has focused solely on bone, but our increasing understanding of the interconnected biology of muscle and bone has begun to shift our treatment paradigm for musculoskeletal disease. Targeting pathways that centrally regulate both bone and muscle (eg, GH/IGF-1, sex steroids, etc.) and newly emerging pathways that might facilitate communication between these 2 tissues (eg, activin/myostatin) might allow a greater therapeutic benefit and/or previously unanticipated means by which to treat these frail patients and prevent fracture. In this review, we will discuss a number of therapies currently under development that aim to treat musculoskeletal disease in precisely such a holistic fashion.
Collapse
Affiliation(s)
- Christian M Girgis
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney NSW, Australia,
| | | | | |
Collapse
|
50
|
Yamawaki K, Ueda S, Okada T, Oshima T, Kakitani M, Kato T, Tomizuka K. Adult-specific systemic over-expression reveals novel in vivo effects of the soluble forms of ActRIIA, ActRIIB and BMPRII. PLoS One 2013; 8:e78076. [PMID: 24205096 PMCID: PMC3804470 DOI: 10.1371/journal.pone.0078076] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 09/16/2013] [Indexed: 12/21/2022] Open
Abstract
Bone morphogenetic proteins (BMPs)/growth differentiation factors (GDFs), which belong to the TGF-beta superfamily, are pleiotropic factors that play a role in regulating the embryonic development and postnatal homeostasis of various organs and tissues by controlling cellular differentiation, proliferation and apoptosis. Conventional transgenic and knockout (KO) mouse approaches have provided only limited information regarding the in vivo functions of BMP signaling in adult animals due to the effects on prenatal development and the difficulty in manipulating multiligand signals simultaneously. We recently produced transgenic chimeric mice(Tg chimeras) in which the soluble IgG1-Fc fusion protein of three BMP type II receptors (ActRIIA, ActRIIB, BMPRII) was highly circulated (281-709 μg/ml), specifically in adult mouse blood. Since each BMP receptor can bind to multiple BMP ligands, these Tg chimeras should be useful to investigate the effects of trapping multiple BMP ligands. Remarkably, some phenotypes were unexpected based on previous studies, such as KO mouse analyses, presumably representing the effects of the multiple ligand trapping. These phenotypes included increased red blood cells (RBCs) and decreased viability in adults. In a further study, we focused on the phenotype of increased RBCs and found that extramedullary hematopoiesis in the spleen, not in the bone marrow, was increased using histological and flow cytometric analyses. Although it remains to be elucidated whether the transgene products affect the tissues directly or indirectly, our data provide novel and important insight into the biological functions of the soluble IgG1-Fc fusion protein of three BMP type II receptors in adults, and our approach should have broad applications to research on other ligand receptor families and studies involving mouse models.
Collapse
Affiliation(s)
- Kengo Yamawaki
- Biologics Research Laboratories, Kyowa Hakko Kirin Co., Ltd, Machida-shi, Tokyo, Japan
- * E-mail: (KY); (KT)
| | - Shinobu Ueda
- Comprehensive Research Organization, Institute for Innovation Design, Waseda University, Shinjuku-ku, Tokyo, Japan
| | - Tsutomu Okada
- Biologics Research Laboratories, Kyowa Hakko Kirin Co., Ltd, Machida-shi, Tokyo, Japan
| | - Takeshi Oshima
- Biologics Research Laboratories, Kyowa Hakko Kirin Co., Ltd, Machida-shi, Tokyo, Japan
| | - Makoto Kakitani
- Biologics Research Laboratories, Kyowa Hakko Kirin Co., Ltd, Machida-shi, Tokyo, Japan
| | - Takashi Kato
- Department of Biology, School of Education, Waseda University, Shinjuku-ku, Tokyo, Japan
| | - Kazuma Tomizuka
- Kyowa Hakko Kirin California, Inc., La Jolla, San Diego, California, United States of America
- * E-mail: (KY); (KT)
| |
Collapse
|