1
|
Pignolo RJ, Chandra A. Insights into age-related osteoporosis from senescence-based preclinical models and human accelerated aging paradigms. Mech Ageing Dev 2025; 224:112025. [PMID: 39805505 PMCID: PMC11938943 DOI: 10.1016/j.mad.2025.112025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/17/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
Preclinical models of age-related osteoporosis have been developed based on the accumulation and clearance of senescent cells. The former include animal models based on telomere dysfunction and focal radiation; the latter based on genetic and pharmacological targeting (i.e., removal) of senescent cells. The weight of evidence using these models suggests that cellular senescence plays a key role in the pathophysiology of aging-onset bone loss with the senescence-associated secretory phenotype (SASP) mediating local and systemic deleterious effects on the skeleton. Mitochondrial dysfunction has also been implicated in senescence and age-related comorbidities, including osteoporosis, and knock-in mutations in the mtDNA polymerase gamma (Polg) gene in mice may recapitulate similar respiratory chain complex defects in aged individual with osteoporosis. This and other contributions to senile osteoporosis may also be identified by the careful evaluation of non-genetic paradigms of human accelerated aging. Premature aging syndromes, especially those with a prominent bone loss phenotype, include clinical scenarios of skeletal unloading, premature ovarian failure and survival from childhood cancers. These non-hereditary progeroid syndromes implicate the involvement of lineage switching to an adipogenic fate, inhibition of Wnt signaling, increased osteoclastogenesis and activation frequency of osteoclasts, as well as the substantial burden of senescent cell accumulation.
Collapse
Affiliation(s)
- Robert J Pignolo
- Department of Medicine, Divisions of Geriatric Medicine and Gerontology, Endocrinology, and Hospital Internal Medicine, the Department of Physiology and Biomedical Engineering, and the Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States.
| | - Abhishek Chandra
- Department of Medicine, Divisions of Geriatric Medicine and Gerontology, the Department of Physiology and Biomedical Engineering, and the Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
2
|
Welc SS, Brotto M, White KE, Bonewald LF. Aging: A struggle for beneficial to overcome negative factors made by muscle and bone. Mech Ageing Dev 2025; 224:112039. [PMID: 39952614 PMCID: PMC11893237 DOI: 10.1016/j.mad.2025.112039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/15/2024] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
Musculoskeletal health is strongly influenced by regulatory interactions of bone and muscle. Recent discoveries have identified a number of key mechanisms through which soluble factors released during exercise by bone exert positive effects on muscle and by muscle on bone. Although exercise can delay the negative effects of aging, these beneficial effects are diminished with aging. The limited response of aged muscle and bone tissue to exercise are accompanied by a failure in bone and muscle communication. Here, we propose that exercise induced beneficial factors must battle changes in circulating endocrine and inflammatory factors that occur with aging. Furthermore, sedentary behavior results in the release of negative factors impacting the ability of bone and muscle to respond to physical activity especially with aging. In this review we report on exercise responsive factors and evidence of modification occurring with aging.
Collapse
Affiliation(s)
- Steven S Welc
- Department of Anatomy, Cell Biology, & Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN 46202, USA.
| | - Marco Brotto
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas-Arlington, Arlington, TX 76019, USA.
| | - Kenneth E White
- Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Department of Molecular and Medical Genetics, School of Medicine, Indiana University, Indianapolis, IN 46202, USA.
| | - Lynda F Bonewald
- Department of Anatomy, Cell Biology, & Physiology, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN 46202, USA.
| |
Collapse
|
3
|
Ali MM, Nookaew I, Resende-Coelho A, Marques-Carvalho A, Warren A, Fu Q, Kim HN, O’Brien CA, Almeida M. Mechanisms of mitochondrial reactive oxygen species action in bone mesenchymal cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.24.643319. [PMID: 40196660 PMCID: PMC11974693 DOI: 10.1101/2025.03.24.643319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Mitochondrial (mt)ROS, insufficient NAD+, and cellular senescence all contribute to the decrease in bone formation with aging. ROS can cause senescence and decrease NAD+, but it remains unknown whether these mechanisms mediate the effects of ROS in vivo. Here, we generated mice lacking the mitochondrial antioxidant enzyme Sod2 in osteoblast lineage cells targeted by Osx1-Cre and showed that Sod2ΔOsx1 mice had low bone mass. Osteoblastic cells from these mice had impaired mitochondrial respiration and attenuated NAD+ levels. Administration of an NAD+ precursor improved mitochondrial function in vitro but failed to rescue the low bone mass of Sod2ΔOsx1 mice. Single-cell RNA-sequencing of bone mesenchymal cells indicated that ROS had no significant effects on markers of senescence but disrupted parathyroid hormone signaling, iron metabolism, and proteostasis. Our data supports the rationale that treatment combinations aimed at decreasing mtROS and senescent cells and increasing NAD+ should confer additive effects in delaying age-associated osteoporosis.
Collapse
Affiliation(s)
- Md Mohsin Ali
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Intawat Nookaew
- Department of Biomedical Informatics; University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Center for Musculoskeletal Disease Research; University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Ana Resende-Coelho
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Adriana Marques-Carvalho
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Aaron Warren
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Qiang Fu
- Center for Musculoskeletal Disease Research; University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Ha-Neui Kim
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Center for Musculoskeletal Disease Research; University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Charles A O’Brien
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Orthopedic Surgery; University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Center for Musculoskeletal Disease Research; University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Maria Almeida
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Orthopedic Surgery; University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Center for Musculoskeletal Disease Research; University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Lead contact
| |
Collapse
|
4
|
Resende-Coelho A, Ali MM, James A, Warren A, Gatrell L, Kadhim I, Fu Q, Xiong J, Onal M, Almeida M. Mitochondrial oxidative stress or decreased autophagy in osteoblast lineage cells is not sufficient to mimic the deleterious effects of aging on bone mechanoresponsiveness. Aging (Albany NY) 2025; 17:610-629. [PMID: 40105873 PMCID: PMC11984430 DOI: 10.18632/aging.206213] [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] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 01/23/2025] [Indexed: 03/20/2025]
Abstract
Exercise-induced mechanical load stimulates bone cells, including osteocytes, to promote bone formation. The bone response to loading is less effective with aging, but the cellular and molecular mechanisms responsible for the impaired mechanoresponsiveness remain unclear. Excessive mitochondrial reactive oxygen species (mtROS) and deficient autophagy are common aging mechanisms implicated in decreased bone formation in old mice. Here, we confirmed that the osteogenic effects of tibia compressive loading are lower in old versus young female mice. We also examined whether an increase in mtROS or decreased autophagy in osteoblast-lineage cells of adult female mice could mimic the deleterious effects of aging. To this end, we loaded mice lacking the antioxidant enzyme superoxide dismutase 2 (Sod2) or autophagy-related 7 (Atg7) in cells targeted by Osterix1 (Osx1)-Cre. Osteocytes in Atg7ΔOsx1 exhibited altered morphology and decreased osteocyte dendrite projections. Two weeks of loading increased cortical bone mass and bone formation rate at both periosteal and endosteal surfaces of Osx1-Cre control mice. Nonetheless, in both Atg7ΔOsx1 and Sod2ΔOsx1 mice the response to loading was identical to that observed in control mice, indicating that compromised Atg7-dependent autophagy or excessive mtROS are not sufficient to impair the bone response to tibial compressive loading. Thus, alternative mechanisms of aging might be responsible for the decreased response of the aged skeleton to mechanical stimuli. These findings also suggest that an intact osteocyte dendrite network is not required for the osteogenic response in this model of bone loading.
Collapse
Affiliation(s)
- Ana Resende-Coelho
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Md Mohsin Ali
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Alicen James
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Aaron Warren
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Landon Gatrell
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Ilham Kadhim
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Qiang Fu
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jinhu Xiong
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Melda Onal
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Maria Almeida
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
5
|
Zhang M, Liu S, Chen Y, Chen Y, He J, Xia Y, Yang Y. Matrix Gla protein suppresses osteoblast senescence and promotes osteogenic differentiation by the PI3K-AKT signaling pathway. Exp Cell Res 2025; 444:114329. [PMID: 39536932 DOI: 10.1016/j.yexcr.2024.114329] [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: 09/22/2024] [Revised: 11/05/2024] [Accepted: 11/10/2024] [Indexed: 11/16/2024]
Abstract
Age-related bone loss in mice is associated with senescent cell accumulation and reduced bone formation by osteoblasts. Matrix Gla protein (MGP), secreted by osteoblasts, is pivotal in regulating the bone extracellular matrix mineralization. Previous research has demonstrated that Mgp null mice exhibit osteopenia and fractures, and ultimately die prematurely. To elucidate the mechanisms underlying MGP's role of MGP in bone metabolism, we generated osteoblast-specific Mgp knockout (Mgp cKO) mice by crossing Mgpfl/fl mice with Bglap-Cre mice. The study revealed that in 3-month-old Mgp cKO male mice, trabecular bone volume decreased, and the senescence marker protein p21 increased. Primary osteoblasts from Mgp cKO mice exhibited markers of DNA damage and senescence, such as increased γH2AX foci, p21, and senescence-associated β-galactosidase staining, as well as attenuated cellular proliferation and osteogenic differentiation abilities. In addition, bone marrow stromal cells' colony formation and spontaneous osteogenic ability were impaired in Mgp cKO mice, whereas osteoclastogenesis was enhanced. In vitro treatment with recombinant human MGP promotes osteogenesis in osteoblasts derived from Mgp cKO mice via the PI3K-AKT signaling pathway. Thus, our results suggest that MGP is protective by suppressing osteoblast senescence, offering new insights into potential therapeutic strategies for age-related osteoporosis.
Collapse
Affiliation(s)
- Min Zhang
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, 330006, Nanchang, Jiangxi, China; Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, 330006, China
| | - Sha Liu
- Department of Endocrinology, The Eighth Affiliated Hospital, Sun Yat-sen University, 518000, Shenzhen, China
| | - Yulin Chen
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, 330006, Nanchang, Jiangxi, China; Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, 330006, China
| | - Yifa Chen
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, 330006, Nanchang, Jiangxi, China; Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, 330006, China
| | - Jiaojiao He
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, 330006, Nanchang, Jiangxi, China; Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, 330006, China
| | - Yuting Xia
- Department of General Practice, Jingzhou Central Hospital, 434000, Jingzhou, Hubei, China
| | - Ya Yang
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, 330006, Nanchang, Jiangxi, China; Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, 330006, China.
| |
Collapse
|
6
|
Schwartzman JD, McCall M, Ghattas Y, Pugazhendhi AS, Wei F, Ngo C, Ruiz J, Seal S, Coathup MJ. Multifunctional scaffolds for bone repair following age-related biological decline: Promising prospects for smart biomaterial-driven technologies. Biomaterials 2024; 311:122683. [PMID: 38954959 DOI: 10.1016/j.biomaterials.2024.122683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/09/2024] [Accepted: 06/23/2024] [Indexed: 07/04/2024]
Abstract
The repair of large bone defects due to trauma, disease, and infection can be exceptionally challenging in the elderly. Despite best clinical practice, bone regeneration within contemporary, surgically implanted synthetic scaffolds is often problematic, inconsistent, and insufficient where additional osteobiological support is required to restore bone. Emergent smart multifunctional biomaterials may drive important and dynamic cellular crosstalk that directly targets, signals, stimulates, and promotes an innate bone repair response following age-related biological decline and when in the presence of disease or infection. However, their role remains largely undetermined. By highlighting their mechanism/s and mode/s of action, this review spotlights smart technologies that favorably align in their conceivable ability to directly target and enhance bone repair and thus are highly promising for future discovery for use in the elderly. The four degrees of interactive scaffold smartness are presented, with a focus on bioactive, bioresponsive, and the yet-to-be-developed autonomous scaffold activity. Further, cell- and biomolecular-assisted approaches were excluded, allowing for contemporary examination of the capabilities, demands, vision, and future requisites of next-generation biomaterial-induced technologies only. Data strongly supports that smart scaffolds hold significant promise in the promotion of bone repair in patients with a reduced osteobiological response. Importantly, many techniques have yet to be tested in preclinical models of aging. Thus, greater clarity on their proficiency to counteract the many unresolved challenges within the scope of aging bone is highly warranted and is arguably the next frontier in the field. This review demonstrates that the use of multifunctional smart synthetic scaffolds with an engineered strategy to circumvent the biological insufficiencies associated with aging bone is a viable route for achieving next-generation therapeutic success in the elderly population.
Collapse
Affiliation(s)
| | - Max McCall
- College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Yasmine Ghattas
- College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Abinaya Sindu Pugazhendhi
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA
| | - Fei Wei
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA
| | - Christopher Ngo
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA
| | - Jonathan Ruiz
- College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Sudipta Seal
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA; Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC), Materials Science and Engineering, College of Medicine, University of Central Florida, USA, Orlando, FL
| | - Melanie J Coathup
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA.
| |
Collapse
|
7
|
You X, Yan J, Herzog J, Nobakhti S, Campbell R, Hoke A, Hammamieh R, Sartor RB, Shefelbine S, Kacena MA, Chakraborty N, Charles JF. Bone loss with aging is independent of gut microbiome in mice. Bone Res 2024; 12:65. [PMID: 39523344 PMCID: PMC11551211 DOI: 10.1038/s41413-024-00366-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 08/19/2024] [Accepted: 08/27/2024] [Indexed: 11/16/2024] Open
Abstract
Emerging evidence suggests a significant role of gut microbiome in bone health. Aging is well recognized as a crucial factor influencing the gut microbiome. In this study, we investigated whether age-dependent microbial change contributes to age-related bone loss in CB6F1 mice. The bone phenotype of 24-month-old germ-free (GF) mice was indistinguishable compared to their littermates colonized by fecal transplant at 1-month-old. Moreover, bone loss from 3 to 24-month-old was comparable between GF and specific pathogen-free (SPF) mice. Thus, GF mice were not protected from age-related bone loss. 16S rRNA gene sequencing of fecal samples from 3-month and 24-month-old SPF males indicated an age-dependent microbial shift with an alteration in energy and nutrient metabolism potential. An integrative analysis of 16S predicted metagenome function and LC-MS fecal metabolome revealed an enrichment of protein and amino acid biosynthesis pathways in aged mice. Microbial S-adenosyl methionine metabolism was increased in the aged mice, which has previously been associated with the host aging process. Collectively, aging caused microbial taxonomic and functional alteration in mice. To demonstrate the functional importance of young and old microbiome to bone, we colonized GF mice with fecal microbiome from 3-month or 24-month-old SPF donor mice for 1 and 8 months. The effect of microbial colonization on bone phenotypes was independent of the microbiome donors' age. In conclusion, our study indicates age-related bone loss occurs independent of gut microbiome.
Collapse
Affiliation(s)
- Xiaomeng You
- Department of Orthopaedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| | - Jing Yan
- Department of Orthopaedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jeremy Herzog
- National Gnotobiotic Rodent Resource Center, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Sabah Nobakhti
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Ross Campbell
- The Geneva Foundation, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Allison Hoke
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
- ORISE, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Rasha Hammamieh
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - R Balfour Sartor
- National Gnotobiotic Rodent Resource Center, Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Sandra Shefelbine
- Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, 46202, USA
| | - Nabarun Chakraborty
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD, 20910, USA
| | - Julia F Charles
- Department of Orthopaedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Division of Rheumatology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
8
|
Rakhymzhan A, Fiedler AF, Günther R, Domingue SR, Wooldridge L, Leben R, Cao Y, Bias A, Roodselaar J, Köhler R, Ulbricht C, Heidelin J, Andresen V, Beckers I, Haibel A, Duda G, Hauser AE, Niesner RA. Optimized intravital three-photon imaging of intact mouse tibia links plasma cell motility to functional states. iScience 2024; 27:110985. [PMID: 39391739 PMCID: PMC11466647 DOI: 10.1016/j.isci.2024.110985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 06/29/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024] Open
Abstract
Intravital deep bone marrow imaging is crucial to studying cellular dynamics and functions but remains challenging, and minimally invasive methods are needed. We employed a high pulse-energy 1650 nm laser to perform three-photon microscopy in vivo, reaching ≈400 μm depth in intact mouse tibia. Repetition rates of 3 and 4 MHz allowed us to analyze motility patterns of fast and rare cells within unperturbed marrow and to identify a bi-modal migratory behavior for plasma cells. Third harmonic generation (THG) was identified as a label-free marker for cellular organelles, particularly endoplasmic reticulum, indicating protein synthesis capacity. We found a strong THG signal, suggesting high antibody secretion, in one-third of plasma cells while the rest showed low signals. We discovered an inverse relationship between migratory behavior and THG signal, linking motility to functional plasma cell states. This method may enhance our understanding of marrow microenvironment effects on cellular functions.
Collapse
Affiliation(s)
- Asylkhan Rakhymzhan
- German Rheumatism Research Center – a Leibniz Institute, Biophysical Analytics, Berlin, Germany
- German Rheumatism Research Center – a Leibniz Institute, Immune Dynamics, Berlin, Germany
- Charité – Universitätsmedizin, Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Clinics for Rheumatology and Clinical Immunology, Berlin, Germany
| | - Alexander F. Fiedler
- German Rheumatism Research Center – a Leibniz Institute, Biophysical Analytics, Berlin, Germany
- Freie Universität Berlin, Dynamic and Functional in vivo Imaging, Berlin, Germany
| | - Robert Günther
- German Rheumatism Research Center – a Leibniz Institute, Biophysical Analytics, Berlin, Germany
- German Rheumatism Research Center – a Leibniz Institute, Immune Dynamics, Berlin, Germany
| | | | | | - Ruth Leben
- German Rheumatism Research Center – a Leibniz Institute, Biophysical Analytics, Berlin, Germany
- Freie Universität Berlin, Dynamic and Functional in vivo Imaging, Berlin, Germany
| | - Yu Cao
- German Rheumatism Research Center – a Leibniz Institute, Immune Dynamics, Berlin, Germany
- Charité – Universitätsmedizin, Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Clinics for Rheumatology and Clinical Immunology, Berlin, Germany
| | - Anne Bias
- German Rheumatism Research Center – a Leibniz Institute, Biophysical Analytics, Berlin, Germany
- Berlin University of Applied Sciences and Technology, Berlin, Germany
| | - Jay Roodselaar
- German Rheumatism Research Center – a Leibniz Institute, Immune Dynamics, Berlin, Germany
- Charité – Universitätsmedizin, Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Clinics for Rheumatology and Clinical Immunology, Berlin, Germany
| | - Ralf Köhler
- German Rheumatism Research Center – a Leibniz Institute, Biophysical Analytics, Berlin, Germany
- German Rheumatism Research Center – a Leibniz Institute, Immune Dynamics, Berlin, Germany
| | - Carolin Ulbricht
- German Rheumatism Research Center – a Leibniz Institute, Immune Dynamics, Berlin, Germany
- Charité – Universitätsmedizin, Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Clinics for Rheumatology and Clinical Immunology, Berlin, Germany
| | | | | | - Ingeborg Beckers
- Berlin University of Applied Sciences and Technology, Berlin, Germany
| | - Astrid Haibel
- Berlin University of Applied Sciences and Technology, Berlin, Germany
| | - Georg Duda
- Charité – Universitätsmedizin, Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Julius Wolff Institute, Berlin, Germany
| | - Anja E. Hauser
- German Rheumatism Research Center – a Leibniz Institute, Immune Dynamics, Berlin, Germany
- Charité – Universitätsmedizin, Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Clinics for Rheumatology and Clinical Immunology, Berlin, Germany
| | - Raluca A. Niesner
- German Rheumatism Research Center – a Leibniz Institute, Biophysical Analytics, Berlin, Germany
- Freie Universität Berlin, Dynamic and Functional in vivo Imaging, Berlin, Germany
| |
Collapse
|
9
|
Richardson KK, Adam GO, Ling W, Warren A, Marques-Carvalho A, Thostenson JD, Krager K, Aykin-Burns N, Byrum SD, Almeida M, Kim HN. Mitochondrial protein deacetylation by SIRT3 in osteoclasts promotes bone resorption with aging in female mice. Mol Metab 2024; 88:102012. [PMID: 39154858 PMCID: PMC11399565 DOI: 10.1016/j.molmet.2024.102012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 08/12/2024] [Accepted: 08/12/2024] [Indexed: 08/20/2024] Open
Abstract
OBJECTIVES The mitochondrial deacetylase sirtuin-3 (SIRT3) is necessary for the increased bone resorption and enhanced function of mitochondria in osteoclasts that occur with advancing age; how SIRT3 drives bone resorption remains elusive. METHODS To determine the role of SIRT3 in osteoclast mitochondria, we used mice with conditional loss of Sirt3 in osteoclast lineage and mice with germline deletion of either Sirt3 or its known target Pink1. RESULTS SIRT3 stimulates mitochondrial quality in osteoclasts in a PINK1-independent manner, promoting mitochondrial activity and osteoclast maturation and function, thereby contributing to bone loss in female but not male mice. Quantitative analyses of global proteomes and acetylomes revealed that deletion of Sirt3 dramatically increased acetylation of osteoclast mitochondrial proteins, particularly ATPase inhibitory factor 1 (ATPIF1), an essential protein for mitophagy. Inhibition of mitophagy via mdivi-1 recapitulated the effect of deletion of Sirt3 or Atpif1 in osteoclast formation and mitochondrial function. CONCLUSIONS Decreasing mitophagic flux in osteoclasts may be a promising pharmacotherapeutic approach to treat osteoporosis in older adults.
Collapse
Affiliation(s)
- Kimberly K Richardson
- Center for Musculoskeletal Disease Research, USA; Division of Endocrinology, Department of Internal Medicine, USA
| | - Gareeballah Osman Adam
- Center for Musculoskeletal Disease Research, USA; Division of Endocrinology, Department of Internal Medicine, USA
| | - Wen Ling
- Center for Musculoskeletal Disease Research, USA; Division of Endocrinology, Department of Internal Medicine, USA
| | - Aaron Warren
- Center for Musculoskeletal Disease Research, USA; Division of Endocrinology, Department of Internal Medicine, USA
| | - Adriana Marques-Carvalho
- Center for Musculoskeletal Disease Research, USA; Division of Endocrinology, Department of Internal Medicine, USA
| | - Jeff D Thostenson
- Center for Musculoskeletal Disease Research, USA; Department of Biostatistics, USA
| | - Kimberly Krager
- Division of Radiation Health, Department of Pharmaceutical Sciences, USA
| | - Nukhet Aykin-Burns
- Division of Radiation Health, Department of Pharmaceutical Sciences, USA
| | - Stephanie D Byrum
- Department of Biochemistry and Molecular Biology, USA; Arkansas Children's Research Institute, Little Rock, AR, USA
| | - Maria Almeida
- Center for Musculoskeletal Disease Research, USA; Division of Endocrinology, Department of Internal Medicine, USA; Department of Orthopedic Surgery, University of Arkansas for Medical Sciences, USA
| | - Ha-Neui Kim
- Center for Musculoskeletal Disease Research, USA; Division of Endocrinology, Department of Internal Medicine, USA.
| |
Collapse
|
10
|
Yin Y, Chen G, Yang C, Wang J, Peng J, Huang X, Tang Q, Chen L. Osteocyte ferroptosis induced by ATF3/TFR1 contributes to cortical bone loss during ageing. Cell Prolif 2024; 57:e13657. [PMID: 38764128 PMCID: PMC11471391 DOI: 10.1111/cpr.13657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/20/2024] [Accepted: 05/03/2024] [Indexed: 05/21/2024] Open
Abstract
Cortical bone loss is intricately associated with ageing and coincides with iron accumulation. The precise role of ferroptosis, characterized by iron overload and lipid peroxidation, in senescent osteocytes remains elusive. We found that ferroptosis was a crucial mode of osteocyte death in cortical bone during ageing. Using a single-cell transcriptome analysis, we identified activating transcription factor 3 (ATF3) as a critical driver of osteocyte ferroptosis. Elevated ATF3 expression in senescent osteocytes promotes iron uptake by upregulating transferrin receptor 1 while simultaneously inhibiting solute carrier family 7-member 11-mediated cystine import. This process leads to an iron overload and lipid peroxidation, culminating in ferroptosis. Importantly, ATF3 inhibition in aged mice effectively alleviated ferroptosis in the cortical bone and mitigated cortical bone mass loss. Taken together, our findings establish a pivotal role of ferroptosis in cortical bone loss in older adults, providing promising prevention and treatment strategies for osteoporosis and fractures.
Collapse
Affiliation(s)
- Ying Yin
- Department of Stomatology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Guang‐Jin Chen
- Department of Stomatology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Chen Yang
- Department of Stomatology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Jia‐Jia Wang
- Department of Stomatology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Jin‐Feng Peng
- Department of Stomatology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Xiao‐Fei Huang
- Department of Stomatology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Qing‐Ming Tang
- Department of Stomatology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Li‐Li Chen
- Department of Stomatology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| |
Collapse
|
11
|
Jiang C, Ruan Y, Li J, Huang J, Xiao M, Xu H. Tissue expression and promoter activity analysis of the porcine TNFSF11 gene. Theriogenology 2024; 226:277-285. [PMID: 38954996 DOI: 10.1016/j.theriogenology.2024.06.018] [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: 04/17/2024] [Revised: 06/20/2024] [Accepted: 06/20/2024] [Indexed: 07/04/2024]
Abstract
Tumour necrosis factor (TNF) superfamily member 11 (TNFSF11), also known as RANKL, plays a crucial role in regulating several physiological and pathological activities. Additionally, it is a vital factor in bone physiology, and the sex hormone progesterone regulates the expansion of stem cells and the proliferation of mammary epithelial cells. It is essential for animal growth and reproductive physiological processes. This study aimed to evaluate the tissue-specific expression characteristics and promoter activity of the TNFSF11 gene in pigs. As a result, the study examined the presence of TNFSF11 expression in the tissues of Xiangsu pigs at 0.6 and 12 months of age. Moreover, the core promoter region of TNFSF11 was also identified by utilizing a combination of bioinformatic prediction and dual-luciferase activity tests. Finally, the effect of transcription factors on the transcriptional activity of the core promoter region was determined using site-directed mutagenesis. TNFSF11 was uniformly expressed in all tissues; however, its expression in muscles was comparatively low. The core promoter region of TNFSF11 was located in the -555 to -1 region. The prediction of the transcription start site of TNFSF11 gene-2000 ∼ + 500bp showed that there was a CpG site in 17 ∼ + 487bp. Analysis of mutations in the transcription factor binding sites revealed that mutations in the Stat5b, Myog, Trl, and EN1 binding sites had significant effects on the transcriptional activity of the TNFSF11 gene, particularly following the EN1 binding site mutation (P < 0.001). This study provides insights into both the tissue-specific expression patterns of TNFSF11 in the tissues of Xiangsu pigs and the potential regulatory effects of transcription factors on its promoter activity. These results may be helpful for future research aimed at clarifying the expression and role of the porcine TNFSF11 gene.
Collapse
Affiliation(s)
- Chuanmei Jiang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang, China; College of Animal Science, Guizhou University, Guiyang, China
| | - Yong Ruan
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang, China; College of Animal Science, Guizhou University, Guiyang, China
| | - Jifeng Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang, China; College of Animal Science, Guizhou University, Guiyang, China
| | - Jiajin Huang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang, China; College of Animal Science, Guizhou University, Guiyang, China
| | - Meimei Xiao
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang, China; College of Animal Science, Guizhou University, Guiyang, China
| | - Houqiang Xu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang, China; Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, Guizhou University, Guiyang, China; College of Animal Science, Guizhou University, Guiyang, China.
| |
Collapse
|
12
|
Kuroda Y, Yoda M, Kawaai K, Tatenuma M, Mizoguchi T, Ito S, Kasahara M, Wu Y, Takano H, Momose A, Matsuo K. Developing long bones respond to surrounding tissues by trans-pairing of periosteal osteoclasts and endocortical osteoblasts. Development 2024; 151:dev202194. [PMID: 39119717 PMCID: PMC11423808 DOI: 10.1242/dev.202194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 08/01/2024] [Indexed: 08/10/2024]
Abstract
Developing long bones alter their shape while maintaining uniform cortical thickness via coordinated activity of bone-forming osteoblasts and bone-resorbing osteoclasts at periosteal and endosteal surfaces, a process we designate trans-pairing. Two types of trans-pairing shift cortical bone in opposite orientations: peri-forming trans-pairing (peri-t-p) increases bone marrow space and endo-forming trans-pairing (endo-t-p) decreases it, via paired activity of bone resorption and formation across the cortex. Here, we focused on endo-t-p in growing bones. Analysis of endo-t-p activity in the cortex of mouse fibulae revealed osteoclasts under the periosteum compressed by muscles, and expression of RANKL in periosteal cells of the cambium layer. Furthermore, mature osteoblasts were localized on the endosteum, while preosteoblasts were at the periosteum and within cortical canals. X-ray tomographic microscopy revealed the presence of cortical canals more closely associated with endo- than with peri-t-p. Sciatic nerve transection followed by muscle atrophy and unloading induced circumferential endo-t-p with concomitant spread of cortical canals. Such canals likely supply the endosteum with preosteoblasts from the periosteum under endo-t-p, allowing bone shape to change in response to mechanical stress or nerve injury.
Collapse
Affiliation(s)
- Yukiko Kuroda
- Laboratory of Cell and Tissue Biology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Masaki Yoda
- Laboratory of Cell and Tissue Biology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Katsuhiro Kawaai
- Laboratory of Cell and Tissue Biology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | - Motoharu Tatenuma
- Laboratory of Cell and Tissue Biology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| | | | - Shinichirou Ito
- Department of Pharmacology, Tokyo Dental College, Tokyo 101-0061, Japan
| | - Masataka Kasahara
- Department of Pharmacology, Tokyo Dental College, Tokyo 101-0061, Japan
| | - Yanlin Wu
- Institute of Multidisciplinary Research for Advanced Materials (IMRAM), Tohoku University, Katahira 2-1-1, Aoba, Sendai Miyagi 980-8577, Japan
| | - Hidekazu Takano
- Institute of Multidisciplinary Research for Advanced Materials (IMRAM), Tohoku University, Katahira 2-1-1, Aoba, Sendai Miyagi 980-8577, Japan
| | - Atsushi Momose
- Institute of Multidisciplinary Research for Advanced Materials (IMRAM), Tohoku University, Katahira 2-1-1, Aoba, Sendai Miyagi 980-8577, Japan
- JASRI/SPring-8, 1-1-1 Kouto, Sayo-cho, Hyogo 679-5198, Japan
| | - Koichi Matsuo
- Laboratory of Cell and Tissue Biology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku, Tokyo 160-8582, Japan
| |
Collapse
|
13
|
Ding Z, Ma G, Zhou B, Cheng S, Tang W, Han Y, Chen L, Pang W, Chen Y, Yang D, Cao H. Targeting miR-29 mitigates skeletal senescence and bolsters therapeutic potential of mesenchymal stromal cells. Cell Rep Med 2024; 5:101665. [PMID: 39168101 PMCID: PMC11384963 DOI: 10.1016/j.xcrm.2024.101665] [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: 03/05/2024] [Revised: 06/07/2024] [Accepted: 07/08/2024] [Indexed: 08/23/2024]
Abstract
Mesenchymal stromal cell (MSC) senescence is a key factor in skeletal aging, affecting the potential of MSC applications. Identifying targets to prevent MSC and skeletal senescence is crucial. Here, we report increased miR-29 expression in bone tissues of aged mice, osteoporotic patients, and senescent MSCs. Genetic overexpression of miR-29 in Prx1-positive MSCs significantly accelerates skeletal senescence, reducing cortical bone thickness and trabecular bone mass, while increasing femur cross-sectional area, bone marrow adiposity, p53, and senescence-associated secretory phenotype (SASP) levels. Mechanistically, miR-29 promotes senescence by upregulating p53 via targeting Kindlin-2 mRNA. miR-29 knockdown in BMSCs impedes skeletal senescence, enhances bone mass, and accelerates calvarial defect regeneration, also reducing lipopolysaccharide (LPS)-induced organ injuries and mortality. Thus, our findings underscore miR-29 as a promising therapeutic target for senescence-related skeletal diseases and acute inflammation-induced organ damage.
Collapse
Affiliation(s)
- Zhen Ding
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Guixing Ma
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China.
| | - Bo Zhou
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Siyuan Cheng
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Wanze Tang
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yingying Han
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Litong Chen
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Wei Pang
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yangshan Chen
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Dazhi Yang
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China
| | - Huiling Cao
- Department of Biochemistry, School of Medicine, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
14
|
Chen W, Zheng H, Liao Q, Zeng S, Bai R, Shi J, Jiang Y, Wang T, Jia H, Liang W, Du W, Chen H. Zhuang-Gu-Fang promotes osteoblast differentiation via myoblasts and myoblast-derived exosomal miRNAs:miR-5100, miR-126a-3p, miR-450b-5p, and miR-669a-5p. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155718. [PMID: 38795694 DOI: 10.1016/j.phymed.2024.155718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 04/15/2024] [Accepted: 05/05/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND Senile osteoporosis (SOP) is an age-related systemic metabolic bone disorder. Previous studies have proved that Zhuang-Gu-Fang (ZGF) modulates myokines, stimulates osteogenic differentiation, and mitigates osteoporosis. OBJECTIVE To elucidate the mechanism by which ZGF promotes osteogenic differentiation via myoblast and myoblast exosomal microRNAs (miRNAs) and investigate its potential implications in senile osteoporosis. METHODS Characterization of ZGF and ZGF serum using UHPLC-MS/MS. An alkaline phosphatase (ALP) activity assay and staining techniques were employed to corroborate the impacts of ZGF on the osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) via myoblasts. Subsequently, exosomes derived from myoblasts were isolated through ultracentrifugation. The effects of ZGF on the BMSCs' osteogenic differentiation were substantiated through ALP activity, alizarin red staining, and a quantitative real-time polymerase reaction system (qRT-PCR). Selected miRNAs were identified via high-throughput sequencing and subjected to differential expression analysis, and subsequently validated through qRT-PCR. The senescence-accelerated (SAMP6) mice were selected as the SOP models. qRT-PCR analyses were further conducted to confirm the expression levels of these selected miRNAs in the muscle and bone tissues of the SAMP6 mice, and the protein expression of osteogenesis-related transcription factors OCN and Osterix in its bone tissue was evaluated by immunofluorescence staining analysis (IF). RESULTS ZGF may enhance the osteogenic differentiation of BMSCs through myoblasts and myoblast-derived exosomes. High-throughput sequencing, differential expression analysis, and subsequent qRT-PCR validation identified four miRNAs that stood out due to their significant differential expression: miR-5100, miR-142a-3p, miR-126a-3p, miR-450b-5p and miR-669a-5p. Moreover, the mice experiment corroborated these findings, which revealed that ZGF not only up-regulated the expression of miR-5100, miR-450b-5p and miR-126a-3p in muscle and bone tissues but also concurrently down-regulated the expression of miR-669a-5p in these tissues. IF staining analysis indicated that ZGF can significantly increase the protein expression of the osteogenic transcription factors OCN and Osterix in the bone tissue of mice with SOP. CONCLUSIONS ZGF can promote osteogenic differentiation of osteoblasts, regulate bone metabolism, and thereby delay the process of SOP. Perhaps, its mechanism is to upregulate myoblast-derived exosomes miR-5100, miR-126a-3p, and miR-450b-5p or downregulate miR-669a-5p. This study reports for the first time that myoblast exosomes miR-669a-5p and miR-450b-5p are novel targets for the regulation of osteoblastic differentiation and the treatment of SOP.
Collapse
Affiliation(s)
- Wenhui Chen
- School of Graduate, Guangxi University of Chinese Medicine, Nanning 530001, China; Department of Endocrinology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, 89-9 Dongge Road, Nanning 530023, China.
| | - Hongxiang Zheng
- School of Graduate, Guangxi University of Chinese Medicine, Nanning 530001, China
| | - Qiulan Liao
- School of Graduate, Guangxi University of Chinese Medicine, Nanning 530001, China
| | - Shiqi Zeng
- School of Graduate, Guangxi University of Chinese Medicine, Nanning 530001, China
| | - Rui Bai
- School of Graduate, Guangxi University of Chinese Medicine, Nanning 530001, China; Faculty of Chinese Medicine Science, Guangxi University of Chinese Medicine, Nanning 530222, China
| | - Jun Shi
- School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning 530007, China
| | - Yunxia Jiang
- Department of Endocrinology, The First Affiliated Hospital of Guangxi University of Chinese Medicine, 89-9 Dongge Road, Nanning 530023, China
| | - Ting Wang
- School of Graduate, Guangxi University of Chinese Medicine, Nanning 530001, China
| | - Hongyang Jia
- School of Graduate, Guangxi University of Chinese Medicine, Nanning 530001, China
| | - Wei Liang
- School of Graduate, Guangxi University of Chinese Medicine, Nanning 530001, China
| | - Wei Du
- School of Graduate, Guangxi University of Chinese Medicine, Nanning 530001, China
| | - Haiqing Chen
- School of Graduate, Guangxi University of Chinese Medicine, Nanning 530001, China
| |
Collapse
|
15
|
Koh NYY, Miszkiewicz JJ, Fac ML, Wee NKY, Sims NA. Preclinical Rodent Models for Human Bone Disease, Including a Focus on Cortical Bone. Endocr Rev 2024; 45:493-520. [PMID: 38315213 PMCID: PMC11244217 DOI: 10.1210/endrev/bnae004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 11/22/2023] [Accepted: 02/02/2024] [Indexed: 02/07/2024]
Abstract
Preclinical models (typically ovariectomized rats and genetically altered mice) have underpinned much of what we know about skeletal biology. They have been pivotal for developing therapies for osteoporosis and monogenic skeletal conditions, including osteogenesis imperfecta, achondroplasia, hypophosphatasia, and craniodysplasias. Further therapeutic advances, particularly to improve cortical strength, require improved understanding and more rigorous use and reporting. We describe here how trabecular and cortical bone structure develop, are maintained, and degenerate with aging in mice, rats, and humans, and how cortical bone structure is changed in some preclinical models of endocrine conditions (eg, postmenopausal osteoporosis, chronic kidney disease, hyperparathyroidism, diabetes). We provide examples of preclinical models used to identify and test current therapies for osteoporosis, and discuss common concerns raised when comparing rodent preclinical models to the human skeleton. We focus especially on cortical bone, because it differs between small and larger mammals in its organizational structure. We discuss mechanisms common to mouse and human controlling cortical bone strength and structure, including recent examples revealing genetic contributors to cortical porosity and osteocyte network configurations during growth, maturity, and aging. We conclude with guidelines for clear reporting on mouse models with a goal for better consistency in the use and interpretation of these models.
Collapse
Affiliation(s)
- Natalie Y Y Koh
- Bone Cell Biology & Disease Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Justyna J Miszkiewicz
- School of Social Science, The University of Queensland, Brisbane, QLD 4072, Australia
- Vertebrate Evolution Development and Ecology, Naturalis Biodiversity Center, 2333 CR Leiden, The Netherlands
| | - Mary Louise Fac
- Bone Cell Biology & Disease Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Natalie K Y Wee
- Bone Cell Biology & Disease Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Natalie A Sims
- Bone Cell Biology & Disease Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC 3065, Australia
| |
Collapse
|
16
|
Zhang L, Wang Z, Zhang Y, Ji R, Li Z, Zou J, Gao B. Regulatory cellular and molecular networks in the bone microenvironment during aging. LIFE MEDICINE 2024; 3:lnae019. [PMID: 39871887 PMCID: PMC11749081 DOI: 10.1093/lifemedi/lnae019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 04/30/2024] [Indexed: 01/29/2025]
Abstract
Age-induced abnormalities in bone metabolism disrupt the equilibrium between bone resorption and formation. This largely stems from disturbances in bone homeostasis, in which signaling pathways exert a significant regulatory influence. Aging compromises the functionality of the bone marrow mesenchymal stem cells (BMSCs), ultimately resulting in tissue dysfunction and pathological aging. Age-related bone degradation primarily manifests as reduced bone formation and the increased accumulation of bone marrow fat. Cellular senescence diminishes bone cell vitality, thereby disrupting the balance of bone remodeling. Intensive osteoclast differentiation leads to the generation of more osteoclasts and increased bone resorption. This review provides insight into the impact of aging on bone, encompassing bone cell states during the aging process and bone signaling pathway transformations. It primarily delves into aging-related signaling pathways, such as the bone morphogenetic protein/Smad, Wnt/β-catenin, osteoprotegerin/receptor activator of NF-κB ligand/receptor activator of NF-κB, connexin43/miR21, and nuclear factor erythroid 2-related factor 2/antioxidant response element pathways, seeking to enhance our comprehension of crucial bone cells and their secretory phenotypes during aging. Furthermore, the precise molecular regulatory mechanisms underlying the interactions between bone signaling pathways and aging are investigated.
Collapse
Affiliation(s)
- Lingli Zhang
- School of Athletic Performance, Shanghai University of Sport, Shanghai 200438, China
| | - Zhikun Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Yuan Zhang
- School of Athletic Performance, Shanghai University of Sport, Shanghai 200438, China
| | - Rui Ji
- Department of Orthopedic Surgery, Xijing Hospital, Airforce Medical University, Xi'an 710032, China
| | - Zhiben Li
- Department of Orthopedic Surgery, Xijing Hospital, Airforce Medical University, Xi'an 710032, China
| | - Jun Zou
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Bo Gao
- Department of Orthopedic Surgery, Xijing Hospital, Airforce Medical University, Xi'an 710032, China
| |
Collapse
|
17
|
Cai Y, Han Z, Cheng H, Li H, Wang K, Chen J, Liu ZX, Xie Y, Lin Y, Zhou S, Wang S, Zhou X, Jin S. The impact of ageing mechanisms on musculoskeletal system diseases in the elderly. Front Immunol 2024; 15:1405621. [PMID: 38774874 PMCID: PMC11106385 DOI: 10.3389/fimmu.2024.1405621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 04/22/2024] [Indexed: 05/24/2024] Open
Abstract
Ageing is an inevitable process that affects various tissues and organs of the human body, leading to a series of physiological and pathological changes. Mechanisms such as telomere depletion, stem cell depletion, macrophage dysfunction, and cellular senescence gradually manifest in the body, significantly increasing the incidence of diseases in elderly individuals. These mechanisms interact with each other, profoundly impacting the quality of life of older adults. As the ageing population continues to grow, the burden on the public health system is expected to intensify. Globally, the prevalence of musculoskeletal system diseases in elderly individuals is increasing, resulting in reduced limb mobility and prolonged suffering. This review aims to elucidate the mechanisms of ageing and their interplay while exploring their impact on diseases such as osteoarthritis, osteoporosis, and sarcopenia. By delving into the mechanisms of ageing, further research can be conducted to prevent and mitigate its effects, with the ultimate goal of alleviating the suffering of elderly patients in the future.
Collapse
Affiliation(s)
- Yijin Cai
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhongyu Han
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong Cheng
- School of Automation Engineering, University of Electronic Science and Technology, Chengdu, China
| | - Hongpeng Li
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ke Wang
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jia Chen
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhi-Xiang Liu
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yulong Xie
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yumeng Lin
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shuwei Zhou
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Siyu Wang
- Department of Gastroenterology, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Xiao Zhou
- Second Clinical Medical College, Heilongjiang University of Chinese Medicine, Heilongjiang, China
| | - Song Jin
- Department of Rehabilitation, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
18
|
Wang H, Yuan T, Wang Y, Liu C, Li D, Li Z, Sun S. Osteoclasts and osteoarthritis: Novel intervention targets and therapeutic potentials during aging. Aging Cell 2024; 23:e14092. [PMID: 38287696 PMCID: PMC11019147 DOI: 10.1111/acel.14092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/28/2023] [Accepted: 01/02/2024] [Indexed: 01/31/2024] Open
Abstract
Osteoarthritis (OA), a chronic degenerative joint disease, is highly prevalent among the aging population, and often leads to joint pain, disability, and a diminished quality of life. Although considerable research has been conducted, the precise molecular mechanisms propelling OA pathogenesis continue to be elusive, thereby impeding the development of effective therapeutics. Notably, recent studies have revealed subchondral bone lesions precede cartilage degeneration in the early stage of OA. This development is marked by escalated osteoclast-mediated bone resorption, subsequent imbalances in bone metabolism, accelerated bone turnover, and a decrease in bone volume, thereby contributing significantly to the pathological changes. While the role of aging hallmarks in OA has been extensively elucidated from the perspective of chondrocytes, their connection with osteoclasts is not yet fully understood. There is compelling evidence to suggest that age-related abnormalities such as epigenetic alterations, proteostasis network disruption, cellular senescence, and mitochondrial dysfunction, can stimulate osteoclast activity. This review intends to systematically discuss how aging hallmarks contribute to OA pathogenesis, placing particular emphasis on the age-induced shifts in osteoclast activity. It also aims to stimulate future studies probing into the pathological mechanisms and therapeutic approaches targeting osteoclasts in OA during aging.
Collapse
Affiliation(s)
- Haojue Wang
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanShandongChina
| | - Tao Yuan
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanShandongChina
| | - Yi Wang
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Changxing Liu
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanShandongChina
| | - Dengju Li
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Ziqing Li
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Shui Sun
- Department of Joint Surgery, Shandong Provincial Hospital, Cheeloo College of MedicineShandong UniversityJinanShandongChina
- Department of Joint SurgeryShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongChina
- Orthopaedic Research Laboratory, Medical Science and Technology Innovation CenterShandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| |
Collapse
|
19
|
Hendrixson JA, James A, Akel NS, Laster DJ, Crawford JA, Berryhill SB, Onal M. Loss of chaperone-mediated autophagy does not alter age-related bone loss in male mice. FASEB Bioadv 2024; 6:73-84. [PMID: 38463697 PMCID: PMC10918985 DOI: 10.1096/fba.2023-00133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/18/2024] [Accepted: 02/06/2024] [Indexed: 03/12/2024] Open
Abstract
Chaperone-mediated autophagy (CMA) is a lysosome-dependent degradation pathway that eliminates proteins that are damaged, partially unfolded, or targeted for selective proteome remodeling. CMA contributes to several cellular processes, including stress response and proteostasis. Age-associated increase in cellular stressors and decrease in CMA contribute to pathologies associated with aging in various tissues. CMA contributes to bone homeostasis in young mice. An age-associated reduction in CMA was reported in osteoblast lineage cells; however, whether declining CMA contributes to skeletal aging is unknown. Herein we show that cellular stressors stimulate CMA in UAMS-32 osteoblastic cells. Moreover, the knockdown of an essential component of the CMA pathway, LAMP2A, sensitizes osteoblasts to cell death caused by DNA damage, ER stress, and oxidative stress. As elevations in these stressors are thought to contribute to age-related bone loss, we hypothesized that declining CMA contributes to the age-associated decline in bone formation by sensitizing osteoblast lineage cells to elevated stressors. To test this, we aged male CMA-deficient mice and controls up to 24 months of age and examined age-associated changes in bone mass and architecture. We showed that lack of CMA did not alter age-associated decline in bone mineral density as measured by dual x-ray absorptiometry (DXA). Moreover, microCT analysis performed at 24 months of age showed that vertebral cancellous bone volume, cortical thickness, and porosity of CMA-deficient and control mice were similar. Taken together, these results suggest that reduction of CMA does not contribute to age-related bone loss.
Collapse
Affiliation(s)
- James A. Hendrixson
- Department of Physiology and Cell BiologyUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Alicen James
- Department of Physiology and Cell BiologyUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Nisreen S. Akel
- Department of Physiology and Cell BiologyUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Dominique J. Laster
- Department of Physiology and Cell BiologyUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Julie A. Crawford
- Center for Musculoskeletal Disease Research (CMDR)University of Arkansas for Medical SciencesLittle RockArkansasUSA
- Division of EndocrinologyUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Stuart B. Berryhill
- Center for Musculoskeletal Disease Research (CMDR)University of Arkansas for Medical SciencesLittle RockArkansasUSA
- Division of EndocrinologyUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Melda Onal
- Department of Physiology and Cell BiologyUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| |
Collapse
|
20
|
Fan H, Qiao Z, Li J, Shang G, Shang C, Chen S, Leng Z, Su H, Kou H, Liu H. Recent advances in senescence-associated secretory phenotype and osteoporosis. Heliyon 2024; 10:e25538. [PMID: 38375248 PMCID: PMC10875379 DOI: 10.1016/j.heliyon.2024.e25538] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 02/21/2024] Open
Abstract
The worldwide elderly population is on the rise, and aging is a major osteoporosis risk factor. Senescent cells accumulation can have a detrimental effect the body as we age. The senescence-associated secretory phenotype (SASP), an essential cellular senescence hallmark, is an important mechanism connecting cellular senescence to osteoporosis. This review describes in detail the characteristics of SASPs and their regulatory agencies, and shed fresh light on how SASPs from different senescent cells contribute to osteoporosis development. Furthermore, we summarized various innovative therapy techniques that target SASPs to lower the burden of osteoporosis in the elderly and discussed the potential challenges of SASPs-based therapy for osteoporosis as a new clinical trial.
Collapse
Affiliation(s)
- Haonan Fan
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Zhi Qiao
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Jitian Li
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital)/Henan Institute of Orthopedic and Traumatology, Luoyang 471000, China
| | - Guowei Shang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Chunfeng Shang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Songfeng Chen
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Zikuan Leng
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Huifang Su
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Hongwei Kou
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Hongjian Liu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| |
Collapse
|
21
|
Mertens TF, Liebheit AT, Ehl J, Köhler R, Rakhymzhan A, Woehler A, Katthän L, Ebel G, Liublin W, Kasapi A, Triantafyllopoulou A, Schulz TJ, Niesner RA, Hauser AE. MarShie: a clearing protocol for 3D analysis of single cells throughout the bone marrow at subcellular resolution. Nat Commun 2024; 15:1764. [PMID: 38409121 PMCID: PMC10897183 DOI: 10.1038/s41467-024-45827-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 02/01/2024] [Indexed: 02/28/2024] Open
Abstract
Analyzing immune cell interactions in the bone marrow is vital for understanding hematopoiesis and bone homeostasis. Three-dimensional analysis of the complete, intact bone marrow within the cortex of whole long bones remains a challenge, especially at subcellular resolution. We present a method that stabilizes the marrow and provides subcellular resolution of fluorescent signals throughout the murine femur, enabling identification and spatial characterization of hematopoietic and stromal cell subsets. By combining a pre-processing algorithm for stripe artifact removal with a machine-learning approach, we demonstrate reliable cell segmentation down to the deepest bone marrow regions. This reveals age-related changes in the marrow. It highlights the interaction between CX3CR1+ cells and the vascular system in homeostasis, in contrast to other myeloid cell types, and reveals their spatial characteristics after injury. The broad applicability of this method will contribute to a better understanding of bone marrow biology.
Collapse
Affiliation(s)
- Till Fabian Mertens
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
- Immune Dynamics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
| | - Alina Tabea Liebheit
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
- Immune Dynamics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
- Institute of Chemistry and Biochemistry, Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Johanna Ehl
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
- Immune Dynamics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
| | - Ralf Köhler
- Immune Dynamics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
| | - Asylkhan Rakhymzhan
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
- Biophysical Analytics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
| | - Andrew Woehler
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, 10115, Berlin, Germany
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, 20147, USA
| | - Lukas Katthän
- Miltenyi Biotec B.V. and Co. Bertha-von-Suttner-Straße 5, 37085, Göttingen, Germany
| | - Gernot Ebel
- Miltenyi Biotec B.V. and Co. Bertha-von-Suttner-Straße 5, 37085, Göttingen, Germany
| | - Wjatscheslaw Liublin
- Biophysical Analytics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
| | - Ana Kasapi
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
- Innate Immunity in Rheumatic Diseases, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
| | - Antigoni Triantafyllopoulou
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
- Innate Immunity in Rheumatic Diseases, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
| | - Tim Julius Schulz
- Department of Adipocyte Development and Nutrition, German Institute of Human Nutrition (DIfE) Potsdam-Rehbruecke, 14558, Nuthetal, Germany
- German Center for Diabetes Research (DZD), 85764, Munich-Neuherberg, Germany
| | - Raluca Aura Niesner
- Biophysical Analytics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
- Dynamic and Functional in vivo Imaging, Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Anja Erika Hauser
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany.
- Immune Dynamics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
22
|
Bi J, Zhang C, Lu C, Mo C, Zeng J, Yao M, Jia B, Liu Z, Yuan P, Xu S. Age-related bone diseases: Role of inflammaging. J Autoimmun 2024; 143:103169. [PMID: 38340675 DOI: 10.1016/j.jaut.2024.103169] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/03/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024]
Abstract
Bone aging is characterized by an imbalance in the physiological and pathological processes of osteogenesis, osteoclastogenesis, adipogenesis, and chondrogenesis, resulting in exacerbated bone loss and the development of age-related bone diseases, including osteoporosis, osteoarthritis, rheumatoid arthritis, and periodontitis. Inflammaging, a novel concept in the field of aging research, pertains to the persistent and gradual escalation of pro-inflammatory reactions during the aging process. This phenomenon is distinguished by its low intensity, systemic nature, absence of symptoms, and potential for management. The mechanisms by which inflammaging contribute to age-related chronic diseases, particularly in the context of age-related bone diseases, remain unclear. The precise manner in which systemic inflammation induces bone aging and consequently contributes to the development of age-related bone diseases has yet to be fully elucidated. This article primarily examines the mechanisms underlying inflammaging and its association with age-related bone diseases, to elucidate the potential mechanisms of inflammaging in age-related bone diseases and offer insights for developing preventive and therapeutic strategies for such conditions.
Collapse
Affiliation(s)
- Jiaming Bi
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Caimei Zhang
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Caihong Lu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Chuzi Mo
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiawei Zeng
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Mingyan Yao
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, China; Department of Endocrinology, Baoding No.1 Central Hospital, Baoding, China
| | - Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhongjun Liu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Peiyan Yuan
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Shuaimei Xu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
23
|
Liu LL, Liu ZR, Cao LJ, Wang J, Huang SM, Hu SG, Yang YZ, Li DS, Cao WW, Zeng QB, Huang S, Wu Q, Xiao JH, Liu WY, Xiao YS. Iron accumulation induced by hepcidin1 knockout accelerates the progression of aging osteoporosis. J Orthop Surg Res 2024; 19:59. [PMID: 38216929 PMCID: PMC10785403 DOI: 10.1186/s13018-024-04535-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 01/04/2024] [Indexed: 01/14/2024] Open
Abstract
OBJECTIVE Iron accumulation is associated with osteoporosis. This study aims to explore the effect of chronic iron accumulation induced by hepcidin1 deficiency on aging osteoporosis. METHODS Iron accumulation in hepcidin1 knockout aging mice was assessed by atomic absorption spectroscopy and Perl's staining. Bone microarchitecture was observed using Micro-CT. Hepcidin, ferritin, oxidative stress, and markers of bone turnover in serum were detected by enzyme-linked immunosorbent assay. Bone formation and resorption markers were measured by real-time quantitative PCR. Cell aging was induced by D-galactose treatment. CCK-8, flow cytometry, EdU assays, and Alizarin red staining were performed to reveal the role of hepcidin1 knockout in cell model. Iron Colorimetric Assay Kit and western blot were applied to detect iron and ferritin levels in cells, respectively. RESULTS In hepcidin1-knockout mice, the ferritin and iron contents in liver and tibia were significantly increased. Iron accumulation induced by hepcidin1 knockout caused a phenotype of low bone mass and deteriorated bone microarchitecture. Osteogenic marker was decreased and osteoclast marker was increased in mice, accompanied by increased oxidative stress level. The mRNA expression levels of osteoclast differentiation markers (RANKL, Mmp9, OPG, Trap, and CTSK) were up-regulated, while bone formation markers (OCN, ALP, Runx2, SP7, and Col-1) were down-regulated in model group, compared to wild type mice. In vitro, hepcidin1 knockdown inhibited proliferation and osteogenic differentiation, while promoted apoptosis, with increased levels of iron and ferritin. CONCLUSION Iron accumulation induced by hepcidin1 deficiency aggravates the progression of aging osteoporosis via inhibiting osteogenesis and promoting osteoclast genesis.
Collapse
Affiliation(s)
- Lu-Lin Liu
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, No. 128, Jinling Road, Ganzhou, 341000, Jiangxi, China
- Ganzhou Key Laboratory of Osteoporosis Research, No. 23, Qingnian Road, Ganzhou, 341000, Jiangxi, China
| | - Zhong-Rui Liu
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, No. 128, Jinling Road, Ganzhou, 341000, Jiangxi, China
- Ganzhou Key Laboratory of Osteoporosis Research, No. 23, Qingnian Road, Ganzhou, 341000, Jiangxi, China
| | - Lu-Jun Cao
- Department of Orthopedics, The People's Hospital of Ningdu County, No. 109, Zhongshan South Road, Ningdu County, Ganzhou, 342800, Jiangxi, China
| | - Jun Wang
- Department of Orthopedics, The People's Hospital of Ningdu County, No. 109, Zhongshan South Road, Ningdu County, Ganzhou, 342800, Jiangxi, China
| | - San-Ming Huang
- Department of Orthopedics, The People's Hospital of Ningdu County, No. 109, Zhongshan South Road, Ningdu County, Ganzhou, 342800, Jiangxi, China
| | - Shui-Gen Hu
- Department of Orthopedics, The People's Hospital of Ningdu County, No. 109, Zhongshan South Road, Ningdu County, Ganzhou, 342800, Jiangxi, China
| | - Yi-Zhong Yang
- Department of Orthopedics, The People's Hospital of Ningdu County, No. 109, Zhongshan South Road, Ningdu County, Ganzhou, 342800, Jiangxi, China
| | - Dong-Sheng Li
- Department of Orthopedics, The People's Hospital of Ningdu County, No. 109, Zhongshan South Road, Ningdu County, Ganzhou, 342800, Jiangxi, China
| | - Wei-Wei Cao
- Department of Orthopedics, The People's Hospital of Ningdu County, No. 109, Zhongshan South Road, Ningdu County, Ganzhou, 342800, Jiangxi, China
| | - Qing-Bao Zeng
- Department of Orthopedics, The People's Hospital of Ningdu County, No. 109, Zhongshan South Road, Ningdu County, Ganzhou, 342800, Jiangxi, China
| | - Sheng Huang
- Department of Orthopedics, The People's Hospital of Ningdu County, No. 109, Zhongshan South Road, Ningdu County, Ganzhou, 342800, Jiangxi, China
| | - Qiong Wu
- Department of Orthopedics, The People's Hospital of Ningdu County, No. 109, Zhongshan South Road, Ningdu County, Ganzhou, 342800, Jiangxi, China
| | - Jian-Hua Xiao
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, No. 128, Jinling Road, Ganzhou, 341000, Jiangxi, China
- Ganzhou Key Laboratory of Osteoporosis Research, No. 23, Qingnian Road, Ganzhou, 341000, Jiangxi, China
| | - Wu-Yang Liu
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, No. 128, Jinling Road, Ganzhou, 341000, Jiangxi, China
- Ganzhou Key Laboratory of Osteoporosis Research, No. 23, Qingnian Road, Ganzhou, 341000, Jiangxi, China
| | - Yao-Sheng Xiao
- Department of Orthopedics, The First Affiliated Hospital of Gannan Medical University, No. 128, Jinling Road, Ganzhou, 341000, Jiangxi, China.
- Ganzhou Key Laboratory of Osteoporosis Research, No. 23, Qingnian Road, Ganzhou, 341000, Jiangxi, China.
| |
Collapse
|
24
|
Kim YK, Kameo Y, Tanaka S, Adachi T. Aging effects on osteoclast progenitor dynamics affect variability in bone turnover via feedback regulation. JBMR Plus 2024; 8:ziad003. [PMID: 38690125 PMCID: PMC11059999 DOI: 10.1093/jbmrpl/ziad003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/31/2023] [Accepted: 11/17/2023] [Indexed: 05/02/2024] Open
Abstract
Bone turnover markers (BTMs) are commonly used in osteoporosis treatment as indicators of cell activities of bone-resorbing osteoclasts and bone-forming osteoblasts. The wide variability in their values due to multiple factors, such as aging and diseases, makes it difficult for physicians to utilize them for clinical decision-making. The progenitors of osteoclasts and osteoblasts are indispensable for a comprehensive interpretation of the variability in BTM values because these upstream progenitors strongly regulate the downstream cell activities of bone turnover. However, understanding the complex interactions among the multiple populations of bone cells is challenging. In this study, we aimed to gain a fundamental understanding of the mechanism by which the progenitor dynamics affect the variability in bone turnover through in silico experiments by exploring the cell dynamics with aging effects on osteoporosis. Negative feedback control driven by the consumptive loss of progenitors prevents rapid bone loss due to excessive bone turnover, and through feedback regulation, aging effects on osteoclast differentiation and osteoclast progenitor proliferation cause variability in the osteoclast and osteoblast activity balance and its temporal transition. By expressing the variability in the bone turnover status, our model describes the individualities of patients based on their clinical backgrounds. Therefore, our model could play a powerful role in assisting tailored treatment and has the potential to resolve the various health problems associated with osteoporosis worldwide.
Collapse
Affiliation(s)
- Young Kwan Kim
- Laboratory of Biomechanics, Department of Biosystems Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Yoshitaka Kameo
- Laboratory of Biomechanics, Department of Biosystems Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
- Department of Engineering Science and Mechanics, Shibaura Institute of Technology, Tokyo 135-8548, Japan
| | - Sakae Tanaka
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - Taiji Adachi
- Laboratory of Biomechanics, Department of Biosystems Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
25
|
Nguyen DK, Vanden-Bossche A, Laroche N, Thomas M, Linossier MT, Peyroche S, Farlay D, Follet H, Laquerrière P, Lafage-Proust MH, Thomas T, Vico L, Marotte H, Rousseau M. Dietary supplementation with nacre reduces cortical bone loss in aged female mice. Exp Gerontol 2023; 184:112337. [PMID: 38006949 DOI: 10.1016/j.exger.2023.112337] [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: 05/04/2023] [Revised: 08/17/2023] [Accepted: 11/22/2023] [Indexed: 11/27/2023]
Abstract
Aging is associated with detrimental bone loss leading to fragility fractures in both men and women. Notably, a majority of bone loss with aging is cortical, as well as a large number of fractures are non-vertebral and at the non-hip sites. Nacre is a product of mollusks composed of calcium carbonate embedded in organic components. As our previous study demonstrated the protective effect of nacre supplementation on trabecular bone loss in ovariectomized rats, we sought to evaluate the effect of dietary nacre on bone loss related to aging in female mice which do not suffer true menopause as observed in women. The current study compared the effect of a 90-day long nacre-supplemented diet to that of Standard or CaCO3 diets on both bone mass and strength in 16-month-old C57BL/6 female mice. Multiple approaches were performed to assess the microarchitecture and mechanical properties of long bones, analyze trabecular histomorphometry, and measure bone cell-related gene expressions, and bone turnover markers. In the cortex, dietary nacre improved cortical bone strength in line with lower expression levels of genes reflecting osteoclasts activity compared to Standard or CaCO3 diets (p < 0.05). In the trabeculae, nacre-fed mice were characterized by a bone remodeling process more active than the other groups as shown by greater histomorphometric parameters and osteoblast-related gene expressions (p < 0.05). But these differences were not exhibited at the level of the trabecular microarchitecture at this age. Collectively, these data suggest that dietary nacre should be a potential candidate for reducing aging-associated cortical bone loss in the elderly.
Collapse
Affiliation(s)
- Dung Kim Nguyen
- Université Jean Monnet Saint-Étienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023 Saint-Étienne, France.
| | - Arnaud Vanden-Bossche
- Université Jean Monnet Saint-Étienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023 Saint-Étienne, France
| | - Norbert Laroche
- Université Jean Monnet Saint-Étienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023 Saint-Étienne, France
| | - Mireille Thomas
- Université Jean Monnet Saint-Étienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023 Saint-Étienne, France
| | - Marie-Thérèse Linossier
- Université Jean Monnet Saint-Étienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023 Saint-Étienne, France
| | - Sylvie Peyroche
- Université Jean Monnet Saint-Étienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023 Saint-Étienne, France
| | - Delphine Farlay
- INSERM, LYOS UMR 1033, Université Claude Bernard Lyon 1, 69008 Lyon, France
| | - Hélène Follet
- INSERM, LYOS UMR 1033, Université Claude Bernard Lyon 1, 69008 Lyon, France
| | - Patrice Laquerrière
- Université de Strasbourg, Strasbourg, France; CNRS, Institut Pluridisciplinaire Hubert Curien UMR 7178, Strasbourg, France
| | - Marie-Hélène Lafage-Proust
- Université Jean Monnet Saint-Étienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023 Saint-Étienne, France; Université Jean Monnet Saint-Étienne, Department of Rheumatology, CHU Saint-Etienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023, F-42055 Saint-Étienne, France
| | - Thierry Thomas
- Université Jean Monnet Saint-Étienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023 Saint-Étienne, France; Université Jean Monnet Saint-Étienne, Department of Rheumatology, CHU Saint-Etienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023, F-42055 Saint-Étienne, France
| | - Laurence Vico
- Université Jean Monnet Saint-Étienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023 Saint-Étienne, France
| | - Hubert Marotte
- Université Jean Monnet Saint-Étienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023 Saint-Étienne, France; Université Jean Monnet Saint-Étienne, Department of Rheumatology, CHU Saint-Etienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023, F-42055 Saint-Étienne, France
| | - Marthe Rousseau
- Université Jean Monnet Saint-Étienne, INSERM, Mines Saint Etienne, SAINBIOSE U1059, F-42023 Saint-Étienne, France; UMR5510 MATEIS, CNRS, Lyon University, INSA-Lyon, Lyon, France
| |
Collapse
|
26
|
Venkatesh VS, Nie T, Zajac JD, Grossmann M, Davey RA. The Utility of Preclinical Models in Understanding the Bone Health of Transgender Individuals Undergoing Gender-Affirming Hormone Therapy. Curr Osteoporos Rep 2023; 21:825-841. [PMID: 37707757 PMCID: PMC10724092 DOI: 10.1007/s11914-023-00818-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/16/2023] [Indexed: 09/15/2023]
Abstract
PURPOSE OF REVIEW To summarise the evidence regarding the effects of gender-affirming hormone therapy (GAHT) on bone health in transgender people, to identify key knowledge gaps and how these gaps can be addressed using preclinical rodent models. RECENT FINDINGS Sex hormones play a critical role in bone physiology, yet there is a paucity of research regarding the effects of GAHT on bone microstructure and fracture risk in transgender individuals. The controlled clinical studies required to yield fracture data are unethical to conduct making clinically translatable preclinical research of the utmost importance. Novel genetic and surgical preclinical models have yielded significant mechanistic insight into the roles of sex steroids on skeletal integrity. Preclinical models of GAHT have the potential inform clinical approaches to preserve skeletal integrity and prevent fractures in transgender people undergoing GAHT. This review highlights the key considerations required to ensure the information gained from preclinical models of GAHT are informative.
Collapse
Affiliation(s)
- Varun S Venkatesh
- Department of Medicine, Austin Health, The University of Melbourne, Heidelberg, Victoria, 3084, Australia
| | - Tian Nie
- Department of Medicine, Austin Health, The University of Melbourne, Heidelberg, Victoria, 3084, Australia
| | - Jeffrey D Zajac
- Department of Medicine, Austin Health, The University of Melbourne, Heidelberg, Victoria, 3084, Australia
- Department of Endocrinology, Austin Health, Heidelberg, Victoria, 3084, Australia
| | - Mathis Grossmann
- Department of Medicine, Austin Health, The University of Melbourne, Heidelberg, Victoria, 3084, Australia
- Department of Endocrinology, Austin Health, Heidelberg, Victoria, 3084, Australia
| | - Rachel A Davey
- Department of Medicine, Austin Health, The University of Melbourne, Heidelberg, Victoria, 3084, Australia.
| |
Collapse
|
27
|
Zhang L, Guan Q, Wang Z, Feng J, Zou J, Gao B. Consequences of Aging on Bone. Aging Dis 2023; 15:2417-2452. [PMID: 38029404 PMCID: PMC11567267 DOI: 10.14336/ad.2023.1115] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/16/2023] [Indexed: 12/01/2023] Open
Abstract
With the aging of the global population, the incidence of musculoskeletal diseases has been increasing, seriously affecting people's health. As people age, the microenvironment within skeleton favors bone resorption and inhibits bone formation, accompanied by bone marrow fat accumulation and multiple cellular senescence. Specifically, skeletal stem/stromal cells (SSCs) during aging tend to undergo adipogenesis rather than osteogenesis. Meanwhile, osteoblasts, as well as osteocytes, showed increased apoptosis, decreased quantity, and multiple functional limitations including impaired mechanical sensing, intercellular modulation, and exosome secretion. Also, the bone resorption function of macrophage-lineage cells (including osteoclasts and preosteoclasts) was significantly enhanced, as well as impaired vascularization and innervation. In this study, we systematically reviewed the effect of aging on bone and the within microenvironment (including skeletal cells as well as their intracellular structure variations, vascular structures, innervation, marrow fat distribution, and lymphatic system) caused by aging, and mechanisms of osteoimmune regulation of the bone environment in the aging state, and the causal relationship with multiple musculoskeletal diseases in addition with their potential therapeutic strategy.
Collapse
Affiliation(s)
- Lingli Zhang
- College of Athletic Performance, Shanghai University of Sport, Shanghai, China
| | - Qiao Guan
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Zhikun Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Jie Feng
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Jun Zou
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Bo Gao
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
| |
Collapse
|
28
|
Laster DJ, Akel NS, Hendrixson JA, James A, Crawford JA, Fu Q, Berryhill SB, Thostenson JD, Nookaew I, O’Brien CA, Onal M. CRISPR interference provides increased cell type-specificity compared to the Cre-loxP system. iScience 2023; 26:107428. [PMID: 37575184 PMCID: PMC10415806 DOI: 10.1016/j.isci.2023.107428] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 06/12/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023] Open
Abstract
Cre-mediated recombination is frequently used for cell type-specific loss of function (LOF) studies. A major limitation of this system is recombination in unwanted cell types. CRISPR interference (CRISPRi) has been used effectively for global LOF in mice. However, cell type-specific CRISPRi, independent of recombination-based systems, has not been reported. To test the feasibility of cell type-specific CRISPRi, we produced two novel knock-in mouse models that achieve gene suppression when used together: one expressing dCas9::KRAB under the control of a cell type-specific promoter and the other expressing a single guide RNA from a safe harbor locus. We then compared the phenotypes of mice in which the same gene was targeted by either CRISPRi or the Cre-loxP system, with cell specificity conferred by Dmp1 regulatory elements in both cases. We demonstrate that CRISPRi is effective for cell type-specific LOF and that it provides improved cell type-specificity compared to the Cre-loxP system.
Collapse
Affiliation(s)
- Dominique J. Laster
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Nisreen S. Akel
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - James A. Hendrixson
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Alicen James
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Julie A. Crawford
- Center for Musculoskeletal Disease Research (CMDR), University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Division of Endocrinology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Qiang Fu
- Center for Musculoskeletal Disease Research (CMDR), University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Division of Endocrinology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Stuart B. Berryhill
- Center for Musculoskeletal Disease Research (CMDR), University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Division of Endocrinology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jeff D. Thostenson
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Intawat Nookaew
- Center for Musculoskeletal Disease Research (CMDR), University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Charles A. O’Brien
- Center for Musculoskeletal Disease Research (CMDR), University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Division of Endocrinology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Melda Onal
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Center for Musculoskeletal Disease Research (CMDR), University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
29
|
Kitase Y, Prideaux M. Regulation of the Osteocyte Secretome with Aging and Disease. Calcif Tissue Int 2023; 113:48-67. [PMID: 37148298 DOI: 10.1007/s00223-023-01089-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/21/2023] [Indexed: 05/08/2023]
Abstract
As the most numerous and long-lived of all bone cells, osteocytes have essential functions in regulating skeletal health. Through the lacunar-canalicular system, secreted proteins from osteocytes can reach cells throughout the bone. Furthermore, the intimate connectivity between the lacunar-canalicular system and the bone vasculature allows for the transport of osteocyte-secreted factors into the circulation to reach the entire body. Local and endocrine osteocyte signaling regulates physiological processes such as bone remodeling, bone mechanoadaptation, and mineral homeostasis. However, these processes are disrupted by impaired osteocyte function induced by aging and disease. Dysfunctional osteocyte signaling is now associated with the pathogenesis of many disorders, including chronic kidney disease, cancer, diabetes mellitus, and periodontitis. In this review, we focus on the targeting of bone and extraskeletal tissues by the osteocyte secretome. In particular, we highlight the secreted osteocyte proteins, which are known to be dysregulated during aging and disease, and their roles during disease progression. We also discuss how therapeutic or genetic targeting of osteocyte-secreted proteins can improve both skeletal and systemic health.
Collapse
Affiliation(s)
- Yukiko Kitase
- Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
| | - Matthew Prideaux
- Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.
| |
Collapse
|
30
|
Lad SE. Absence of secondary osteons in femora of aged rats: Implications of lifespan on Haversian remodeling in mammals. J Morphol 2023; 284:e21600. [PMID: 37313764 DOI: 10.1002/jmor.21600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 06/15/2023]
Abstract
Bone is a dynamic tissue capable of adapting to its loading environment, allowing the skeleton to remain structurally sound throughout life. One way adaptation occurs in mammals is via Haversian remodeling: the site-specific, coupled resorption and formation of cortical bone that results in secondary osteons. Remodeling occurs at a baseline rate in most mammals, but it also occurs in relation to strain by repairing deleterious microdamage. Yet, not all animals with bony skeletons remodel. Among mammals, there is inconsistent or absent evidence for Haversian remodeling among monotremes, insectivores, chiropterans, cingulates, and rodents. Three possible explanations for this disparity are discussed: the capacity for Haversian remodeling, body size as a constraint, and age and lifespan as constraints. It is generally accepted, although not thoroughly documented, that rats (a common model used in bone research) do not typically exhibit Haversian remodeling. The present aim is to more specifically test the hypothesis that rats of advanced age do remodel intracortically because of the longer lifespan over which baseline remodeling could occur. Most published histological descriptions of rat bone only include young (3-6 months) rats. Excluding aged rats possibly overlooks a transition from modeling (i.e., bone growth) to Haversian remodeling as the primary mode of bone adaptation. Here, midshaft and distal femora (typical sites for remodeling in other mammals) of 24-month-old rats were examined for presence of secondary osteons. None were found, suggesting that Haversian remodeling does not occur in rats under normal physiological conditions at any age. A likely explanation is that modeling of cortical bone continues throughout most of the short rat lifespan, negating the stimulus for Haversian remodeling. Thorough sampling of key rodent taxa of varying body sizes and lifespans is key to elucidating the reasons why (i.e., body size, age/lifespan, phylogenetic factors) Haversian remodeling might not occur in all mammals.
Collapse
Affiliation(s)
- Susan E Lad
- Department of Exercise Science, High Point University, High Point, North Carolina, USA
- Department of Physical Therapy, High Point University, High Point, North Carolina, USA
| |
Collapse
|
31
|
Abstract
Senescence is a complex cell state characterized by stable cell cycle arrest and a unique secretory pattern known as the senescence-associated secretory phenotype (SASP). The SASP factors, which are heterogeneous and tissue specific, normally include chemokines, cytokines, growth factors, adhesion molecules, and lipid components that can lead to multiple age-associated disorders by eliciting local and systemic consequences. The skeleton is a highly dynamic organ that changes constantly in shape and composition. Senescent cells in bone and bone marrow produce diverse SASP factors that induce alterations of the skeleton through paracrine effects. Herein, we refer to bone cell-associated SASP as "bone-SASP." In this review, we describe current knowledge of cellular senescence and SASP, focusing on the role of senescent cells in mediating bone pathologies during natural aging and premature aging syndromes. We also summarize the role of cellular senescence and the bone-SASP in glucocorticoids-induced bone damage. In addition, we discuss the role of bone-SASP in the development of osteoarthritis, highlighting the mechanisms by which bone-SASP drives subchondral bone changes in metabolic syndrome-associated osteoarthritis.
Collapse
Affiliation(s)
- Ching-Lien Fang
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Bin Liu
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Mei Wan
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Ross Building, Room 209, 720 Rutland Avenue, Baltimore, MD, 21205, USA.
| |
Collapse
|
32
|
Yang Q, Zou Y, Wei X, Ye P, Wu Y, Ai H, Zhang Z, Tan J, Zhou J, Yang Y, Dai Q, Dou C, Luo F. PTP1B knockdown alleviates BMSCs senescence via activating AMPK-mediated mitophagy and promotes osteogenesis in senile osteoporosis. Biochim Biophys Acta Mol Basis Dis 2023:166795. [PMID: 37385514 DOI: 10.1016/j.bbadis.2023.166795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 06/09/2023] [Accepted: 06/21/2023] [Indexed: 07/01/2023]
Abstract
The senescence of bone marrow mesenchymal stem cells (BMSCs) is the basis of senile osteoporosis (SOP). Targeting BMSCs senescence is of paramount importance for developing anti-osteoporotic strategy. In this study, we found that protein tyrosine phosphatase 1B (PTP1B), an enzyme responsible for tyrosine dephosphorylation, was significantly upregulated in BMSCs and femurs with advancing chronological age. Therefore, the potential role of PTP1B in BMSCs senescence and senile osteoporosis was studied. Firstly, significantly upregulated PTP1B expression along with impaired osteogenic differentiation capacity was observed in D-galactose (D-gal)-induced BMSCs and naturally-aged BMSCs. Furthermore, PTP1B silencing could effectively alleviate senescence, improve mitochondrial dysfunction, and restore osteogenic differentiation in aged BMSCs, which was attributable to enhanced mitophagy mediated by PKM2/AMPK pathway. In addition, hydroxychloroquine (HCQ), an autophagy inhibitor, significantly reversed the protective effects from PTP1B knockdown. In SOP animal model, transplantation of LVsh-PTP1B-transfected D-gal-induced BMSCs harvested double protective effects, including increased bone formation and reduced osteoclastogenesis. Similarly, HCQ treatment remarkably suppressed osteogenesis of LVsh-PTP1B-transfected D-gal-induced BMSCs in vivo. Taken together, our data demonstrated that PTP1B silencing protects against BMSCs senescence and mitigates SOP via activating AMPK-mediated mitophagy. Targeting PTP1B may represent a promising interventional strategy to attenuate SOP.
Collapse
Affiliation(s)
- QianKun Yang
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - YuChi Zou
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - XiaoYu Wei
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Peng Ye
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - YuTong Wu
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - HongBo Ai
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zhao Zhang
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China; Orthopedics Department, The General Hospital of Western Theater Command PLA, Chengdu 610083, Sichuan Province, China
| | - JiuLin Tan
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jiangling Zhou
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - YuSheng Yang
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - QiJie Dai
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ce Dou
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Fei Luo
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
33
|
Marahleh A, Kitaura H, Ohori F, Noguchi T, Mizoguchi I. The osteocyte and its osteoclastogenic potential. Front Endocrinol (Lausanne) 2023; 14:1121727. [PMID: 37293482 PMCID: PMC10244721 DOI: 10.3389/fendo.2023.1121727] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 04/07/2023] [Indexed: 06/10/2023] Open
Abstract
The skeleton is an organ of dual functionality; on the one hand, it provides protection and structural competence. On the other hand, it participates extensively in coordinating homeostasis globally given that it is a mineral and hormonal reservoir. Bone is the only tissue in the body that goes through strategically consistent bouts of bone resorption to ensure its integrity and organismal survival in a temporally and spatially coordinated process, known as bone remodeling. Bone remodeling is directly enacted by three skeletal cell types, osteoclasts, osteoblasts, and osteocytes; these cells represent the acting force in a basic multicellular unit and ensure bone health maintenance. The osteocyte is an excellent mechanosensory cell and has been positioned as the choreographer of bone remodeling. It is, therefore, not surprising that a holistic grasp of the osteocyte entity in the bone is warranted. This review discusses osteocytogenesis and associated molecular and morphological changes and describes the osteocytic lacunocanalicular network (LCN) and its organization. We highlight new knowledge obtained from transcriptomic analyses of osteocytes and discuss the regulatory role of osteocytes in promoting osteoclastogenesis with an emphasis on the case of osteoclastogenesis in anosteocytic bones. We arrive at the conclusion that osteocytes exhibit several redundant means through which osteoclast formation can be initiated. However, whether osteocytes are true "orchestrators of bone remodeling" cannot be verified from the animal models used to study osteocyte biology in vivo. Results from studying osteocyte biology using current animal models should come with the caveat that these models are not osteocyte-specific, and conclusions from these studies should be interpreted cautiously.
Collapse
Affiliation(s)
- Aseel Marahleh
- Frontier Research Institute for Interdisciplinary Sciences (FRIS), Tohoku University, Sendai, Japan
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Hideki Kitaura
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Fumitoshi Ohori
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Takahiro Noguchi
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Itaru Mizoguchi
- Division of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| |
Collapse
|
34
|
Rizzo MG, Best TM, Huard J, Philippon M, Hornicek F, Duan Z, Griswold AJ, Kaplan LD, Hare JM, Kouroupis D. Therapeutic Perspectives for Inflammation and Senescence in Osteoarthritis Using Mesenchymal Stem Cells, Mesenchymal Stem Cell-Derived Extracellular Vesicles and Senolytic Agents. Cells 2023; 12:1421. [PMID: 37408255 PMCID: PMC10217382 DOI: 10.3390/cells12101421] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/08/2023] [Accepted: 05/13/2023] [Indexed: 07/07/2023] Open
Abstract
Osteoarthritis (OA) is the most common cause of disability worldwide among the elderly. Alarmingly, the incidence of OA in individuals less than 40 years of age is rising, likely due to the increase in obesity and post-traumatic osteoarthritis (PTOA). In recent years, due to a better understanding of the underlying pathophysiology of OA, several potential therapeutic approaches targeting specific molecular pathways have been identified. In particular, the role of inflammation and the immune system has been increasingly recognized as important in a variety of musculoskeletal diseases, including OA. Similarly, higher levels of host cellular senescence, characterized by cessation of cell division and the secretion of a senescence-associated secretory phenotype (SASP) within the local tissue microenvironments, have also been linked to OA and its progression. New advances in the field, including stem cell therapies and senolytics, are emerging with the goal of slowing disease progression. Mesenchymal stem/stromal cells (MSCs) are a subset of multipotent adult stem cells that have demonstrated the potential to modulate unchecked inflammation, reverse fibrosis, attenuate pain, and potentially treat patients with OA. Numerous studies have demonstrated the potential of MSC extracellular vesicles (EVs) as cell-free treatments that comply with FDA regulations. EVs, including exosomes and microvesicles, are released by numerous cell types and are increasingly recognized as playing a critical role in cell-cell communication in age-related diseases, including OA. Treatment strategies for OA are being developed that target senescent cells and the paracrine and autocrine secretions of SASP. This article highlights the encouraging potential for MSC or MSC-derived products alone or in combination with senolytics to control patient symptoms and potentially mitigate the progression of OA. We will also explore the application of genomic principles to the study of OA and the potential for the discovery of OA phenotypes that can motivate more precise patient-driven treatments.
Collapse
Affiliation(s)
- Michael G. Rizzo
- Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami Miller School of Medicine, Miami, FL 33146, USA; (M.G.R.); (T.M.B.)
| | - Thomas M. Best
- Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami Miller School of Medicine, Miami, FL 33146, USA; (M.G.R.); (T.M.B.)
| | - Johnny Huard
- Center for Regenerative and Personalized Medicine (CRPM), Steadman Philippon Research Institute, Vail, CO 81657, USA (M.P.)
| | - Marc Philippon
- Center for Regenerative and Personalized Medicine (CRPM), Steadman Philippon Research Institute, Vail, CO 81657, USA (M.P.)
| | - Francis Hornicek
- Department of Orthopedics, Sarcoma Biology Laboratory, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.H.); (Z.D.)
| | - Zhenfeng Duan
- Department of Orthopedics, Sarcoma Biology Laboratory, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.H.); (Z.D.)
| | - Anthony J. Griswold
- John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Lee D. Kaplan
- Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami Miller School of Medicine, Miami, FL 33146, USA; (M.G.R.); (T.M.B.)
| | - Joshua M. Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33101, USA
| | - Dimitrios Kouroupis
- Department of Orthopedics, UHealth Sports Medicine Institute, University of Miami Miller School of Medicine, Miami, FL 33146, USA; (M.G.R.); (T.M.B.)
- Diabetes Research Institute, Cell Transplant Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
35
|
Li X, Zhang C, Bowman HH, Stambough JB, Stronach BM, Mears SC, Barnes LC, Ambrogini E, Xiong J. Piezo1 opposes age-associated cortical bone loss. Aging Cell 2023:e13846. [PMID: 37147884 DOI: 10.1111/acel.13846] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 05/07/2023] Open
Abstract
As we age, our bones undergo a process of loss, often accompanied by muscle weakness and reduced physical activity. This is exacerbated by decreased responsiveness to mechanical stimulation in aged skeleton, leading to the hypothesis that decreased mechanical stimulation plays an important role in age-related bone loss. Piezo1, a mechanosensitive ion channel, is critical for bone homeostasis and mechanotransduction. Here, we observed a decrease in Piezo1 expression with age in both murine and human cortical bone. Furthermore, loss of Piezo1 in osteoblasts and osteocytes resulted in an increase in age-associated cortical bone loss compared to control mice. The loss of cortical bone was due to an expansion of the endosteal perimeter resulting from increased endocortical resorption. In addition, expression of Tnfrsf11b, encoding anti-osteoclastogenic protein OPG, decreases with Piezo1 in vitro and in vivo in bone cells, suggesting that Piezo1 suppresses osteoclast formation by promoting Tnfrsf11b expression. Our results highlight the importance of Piezo1-mediated mechanical signaling in protecting against age-associated cortical bone loss by inhibiting bone resorption in mice.
Collapse
Affiliation(s)
- Xuehua Li
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Connie Zhang
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Hayden H Bowman
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Jeffrey B Stambough
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Benjamin M Stronach
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Simon C Mears
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Lowry C Barnes
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Elena Ambrogini
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Division of Endocrinology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Jinhu Xiong
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
36
|
Martiniakova M, Kovacova V, Mondockova V, Svik K, Londzin P, Folwarczna J, Soltesova Prnova M, Stefek M, Omelka R. The Effects of Prolonged Treatment with Cemtirestat on Bone Parameters Reflecting Bone Quality in Non-Diabetic and Streptozotocin-Induced Diabetic Rats. Pharmaceuticals (Basel) 2023; 16:ph16040628. [PMID: 37111385 PMCID: PMC10145951 DOI: 10.3390/ph16040628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/16/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023] Open
Abstract
Cemtirestat, a bifunctional drug acting as an aldose reductase inhibitor with antioxidant ability, is considered a promising candidate for the treatment of diabetic neuropathy. Our study firstly examined the effects of prolonged cemtirestat treatment on bone parameters reflecting bone quality in non-diabetic rats and rats with streptozotocin (STZ)-induced diabetes. Experimental animals were assigned to four groups: non-diabetic rats, non-diabetic rats treated with cemtirestat, diabetic rats, and diabetic rats treated with cemtirestat. Higher levels of plasma glucose, triglycerides, cholesterol, glycated hemoglobin, magnesium, reduced femoral weight and length, bone mineral density and content, parameters characterizing trabecular bone mass and microarchitecture, cortical microarchitecture and geometry, and bone mechanical properties were determined in STZ-induced diabetic versus non-diabetic rats. Treatment with cemtirestat did not affect all aforementioned parameters in non-diabetic animals, suggesting that this drug is safe. In diabetic rats, cemtirestat supplementation reduced plasma triglyceride levels, increased the Haversian canal area and slightly, but insignificantly, improved bone mineral content. Nevertheless, the insufficient effect of cemtirestat treatment on diabetic bone disease does not support its use in the therapy of this complication of type 1 diabetes mellitus.
Collapse
Affiliation(s)
- Monika Martiniakova
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 01 Nitra, Slovakia
| | - Veronika Kovacova
- Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 01 Nitra, Slovakia
| | - Vladimira Mondockova
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 01 Nitra, Slovakia
| | - Karol Svik
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia
| | - Piotr Londzin
- Department of Pharmacology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland
| | - Joanna Folwarczna
- Department of Pharmacology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 41-200 Sosnowiec, Poland
| | - Marta Soltesova Prnova
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia
- Faculty of Informatics and Information Technologies, Slovak University of Technology in Bratislava, 842 16 Bratislava, Slovakia
| | - Milan Stefek
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia
| | - Radoslav Omelka
- Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 01 Nitra, Slovakia
| |
Collapse
|
37
|
Farr JN, Saul D, Doolittle ML, Kaur J, Rowsey JL, Vos SJ, Froemming MN, Lagnado AB, Zhu Y, Weivoda M, Ikeno Y, Pignolo RJ, Niedernhofer LJ, Robbins PD, Jurk D, Passos JF, LeBrasseur NK, Tchkonia T, Kirkland JL, Monroe DG, Khosla S. Local senolysis in aged mice only partially replicates the benefits of systemic senolysis. J Clin Invest 2023; 133:e162519. [PMID: 36809340 PMCID: PMC10104901 DOI: 10.1172/jci162519] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 02/16/2023] [Indexed: 02/23/2023] Open
Abstract
Clearance of senescent cells (SnCs) can prevent several age-related pathologies, including bone loss. However, the local versus systemic roles of SnCs in mediating tissue dysfunction remain unclear. Thus, we developed a mouse model (p16-LOX-ATTAC) that allowed for inducible SnC elimination (senolysis) in a cell-specific manner and compared the effects of local versus systemic senolysis during aging using bone as a prototype tissue. Specific removal of Sn osteocytes prevented age-related bone loss at the spine, but not the femur, by improving bone formation without affecting osteoclasts or marrow adipocytes. By contrast, systemic senolysis prevented bone loss at the spine and femur and not only improved bone formation, but also reduced osteoclast and marrow adipocyte numbers. Transplantation of SnCs into the peritoneal cavity of young mice caused bone loss and also induced senescence in distant host osteocytes. Collectively, our findings provide proof-of-concept evidence that local senolysis has health benefits in the context of aging, but, importantly, that local senolysis only partially replicates the benefits of systemic senolysis. Furthermore, we establish that SnCs, through their senescence-associated secretory phenotype (SASP), lead to senescence in distant cells. Therefore, our study indicates that optimizing senolytic drugs may require systemic instead of local SnC targeting to extend healthy aging.
Collapse
Affiliation(s)
- Joshua N. Farr
- Robert and Arlene Kogod Center on Aging
- Division of Endocrinology
- Department of Physiology and Biomedical Engineering, and
| | - Dominik Saul
- Robert and Arlene Kogod Center on Aging
- Division of Endocrinology
| | | | - Japneet Kaur
- Robert and Arlene Kogod Center on Aging
- Division of Endocrinology
| | | | - Stephanie J. Vos
- Robert and Arlene Kogod Center on Aging
- Division of Endocrinology
| | | | - Anthony B. Lagnado
- Robert and Arlene Kogod Center on Aging
- Department of Physiology and Biomedical Engineering, and
| | - Yi Zhu
- Robert and Arlene Kogod Center on Aging
- Department of Physiology and Biomedical Engineering, and
| | - Megan Weivoda
- Department of Hematology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Yuji Ikeno
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Robert J. Pignolo
- Robert and Arlene Kogod Center on Aging
- Department of Physiology and Biomedical Engineering, and
- Department of Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Laura J. Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Paul D. Robbins
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Diana Jurk
- Robert and Arlene Kogod Center on Aging
- Department of Physiology and Biomedical Engineering, and
| | - João F. Passos
- Robert and Arlene Kogod Center on Aging
- Department of Physiology and Biomedical Engineering, and
| | - Nathan K. LeBrasseur
- Robert and Arlene Kogod Center on Aging
- Department of Physiology and Biomedical Engineering, and
- Department of Physical Medicine and Rehabilitation, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | | | | | - David G. Monroe
- Robert and Arlene Kogod Center on Aging
- Division of Endocrinology
| | - Sundeep Khosla
- Robert and Arlene Kogod Center on Aging
- Division of Endocrinology
- Department of Physiology and Biomedical Engineering, and
| |
Collapse
|
38
|
Matsubayashi S, Ito S, Araya J, Kuwano K. Drugs against metabolic diseases as potential senotherapeutics for aging-related respiratory diseases. Front Endocrinol (Lausanne) 2023; 14:1079626. [PMID: 37077349 PMCID: PMC10106576 DOI: 10.3389/fendo.2023.1079626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 03/22/2023] [Indexed: 04/05/2023] Open
Abstract
Recent advances in aging research have provided novel insights for the development of senotherapy, which utilizes cellular senescence as a therapeutic target. Cellular senescence is involved in the pathogenesis of various chronic diseases, including metabolic and respiratory diseases. Senotherapy is a potential therapeutic strategy for aging-related pathologies. Senotherapy can be classified into senolytics (induce cell death in senescent cells) and senomorphics (ameliorate the adverse effects of senescent cells represented by the senescence-associated secretory phenotype). Although the precise mechanism has not been elucidated, various drugs against metabolic diseases may function as senotherapeutics, which has piqued the interest of the scientific community. Cellular senescence is involved in the pathogenesis of chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF), which are aging-related respiratory diseases. Large-scale observational studies have reported that several drugs, such as metformin and statins, may ameliorate the progression of COPD and IPF. Recent studies have reported that drugs against metabolic diseases may exert a pharmacological effect on aging-related respiratory diseases that can be different from their original effect on metabolic diseases. However, high non-physiological concentrations are needed to determine the efficacy of these drugs under experimental conditions. Inhalation therapy may increase the local concentration of drugs in the lungs without exerting systemic adverse effects. Thus, the clinical application of drugs against metabolic diseases, especially through an inhalation treatment modality, can be a novel therapeutic approach for aging-related respiratory diseases. This review summarizes and discusses accumulating evidence on the mechanisms of aging, as well as on cellular senescence and senotherapeutics, including drugs against metabolic diseases. We propose a developmental strategy for a senotherapeutic approach for aging-related respiratory diseases with a special focus on COPD and IPF.
Collapse
|
39
|
Abstract
Changes in bone architecture and metabolism with aging increase the likelihood of osteoporosis and fracture. Age-onset osteoporosis is multifactorial, with contributory extrinsic and intrinsic factors including certain medical problems, specific prescription drugs, estrogen loss, secondary hyperparathyroidism, microenvironmental and cellular alterations in bone tissue, and mechanical unloading or immobilization. At the histological level, there are changes in trabecular and cortical bone as well as marrow cellularity, lineage switching of mesenchymal stem cells to an adipogenic fate, inadequate transduction of signals during skeletal loading, and predisposition toward senescent cell accumulation with production of a senescence-associated secretory phenotype. Cumulatively, these changes result in bone remodeling abnormalities that over time cause net bone loss typically seen in older adults. Age-related osteoporosis is a geriatric syndrome due to the multiple etiologies that converge upon the skeleton to produce the ultimate phenotypic changes that manifest as bone fragility. Bone tissue is dynamic but with tendencies toward poor osteoblastic bone formation and relative osteoclastic bone resorption with aging. Interactions with other aging physiologic systems, such as muscle, may also confer detrimental effects on the aging skeleton. Conversely, individuals who maintain their BMD experience a lower risk of fractures, disability, and mortality, suggesting that this phenotype may be a marker of successful aging. © 2023 American Physiological Society. Compr Physiol 13:4355-4386, 2023.
Collapse
Affiliation(s)
- Robert J Pignolo
- Department of Medicine, Divisions of Geriatric Medicine and Gerontology, Endocrinology, and Hospital Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA.,The Department of Physiology and Biomedical Engineering, and the Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
40
|
Abstract
Osteoclasts are multinucleated cells with the unique ability to resorb bone matrix. Excessive production or activation of osteoclasts leads to skeletal pathologies that affect a significant portion of the population. Although therapies that effectively target osteoclasts have been developed, they are associated with sometimes severe side effects, and a fuller understanding of osteoclast biology may lead to more specific treatments. Along those lines, a rich body of work has defined essential signaling pathways required for osteoclast formation, function, and survival. Nonetheless, recent studies have cast new light on long-held views regarding the origin of these cells during development and homeostasis, their life span, and the cellular sources of factors that drive their production and activity during homeostasis and disease. In this review, we discuss these new findings in the context of existing work and highlight areas of ongoing and future investigation.
Collapse
Affiliation(s)
- Deborah J Veis
- Division of Bone and Mineral Diseases, Musculoskeletal Research Center; and Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA; .,Shriners Hospitals for Children, St. Louis, Missouri, USA
| | - Charles A O'Brien
- Center for Musculoskeletal Disease Research, Division of Endocrinology, and Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Central Arkansas Veterans Healthcare System, Little Rock, Arkansas, USA
| |
Collapse
|
41
|
Long-Term Cola Intake Does Not Cause Evident Pathological Alterations in the Femoral Bone Microstructure: An Animal Study in Adult Mice. Nutrients 2023; 15:nu15030583. [PMID: 36771291 PMCID: PMC9920312 DOI: 10.3390/nu15030583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
Short-term animal experiments and association studies in humans have shown that cola intake may have a detrimental impact on bone mineral density (BMD); however, other bone parameters have not been investigated. This study examined the effects of long-term cola consumption on the femoral bone microstructure using adult mice (n = 32) as an animal model, which were divided into water and cola groups depending on whether they received water or cola along with a standard rodent diet for 6 months. Micro-computed tomography revealed that cola intake did not significantly affect all measured parameters characterizing trabecular bone mass and microarchitecture, as well as cortical microarchitecture and geometry in both sexes, although a slight deterioration of these parameters was noted. Cola consumption also resulted in a slightly, statistically insignificant worsening of bone mechanical properties. In contrast to female mice, males receiving cola had a lower area of primary osteons' vascular canals. Nevertheless, long-term cola intake did not cause evident pathological alterations in the femur of adult mice, possibly due to a balanced diet and no restriction of physical activity. Therefore, the adverse effects of cola consumption on BMD, the only bone parameter studied so far, may be caused by other risk and lifestyle factors.
Collapse
|
42
|
Abstract
Bone is a living organ that exhibits active metabolic processes, presenting constant bone formation and resorption. The bone cells that maintain local homeostasis are osteoblasts, osteoclasts, osteocytes and bone marrow stem cells, their progenitor cells. Osteoblasts are the main cells that govern bone formation, osteoclasts are involved in bone resorption, and osteocytes, the most abundant bone cells, also participate in bone remodeling. All these cells have active metabolic activities, are interconnected and influence each other, having both autocrine and paracrine effects. Ageing is associated with multiple and complex bone metabolic changes, some of which are currently incompletely elucidated. Ageing causes important functional changes in bone metabolism, influencing all resident cells, including the mineralization process of the extracellular matrix. With advancing age, a decrease in bone mass, the appearance of specific changes in the local microarchitecture, a reduction in mineralized components and in load-bearing capacity, as well as the appearance of an abnormal response to different humoral molecules have been observed. The present review points out the most important data regarding the formation, activation, functioning, and interconnection of these bone cells, as well as data on the metabolic changes that occur due to ageing.
Collapse
Affiliation(s)
- Anca Cardoneanu
- Department of Rheumatology, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania
- Clinical Rehabilitation Hospital, 1st Rheumatology Clinic, Iasi, Romania
| | - Ciprian Rezus
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania
- IIIrd Medical Clinic, "Saint Spiridon" Clinic Emergency County Hospital, Iasi, Romania
| | - Bogdan Ionel Tamba
- Advanced Research and Development Center for Experimental Medicine (CEMEX), "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania.
| | - Elena Rezus
- Department of Rheumatology, "Grigore T. Popa" University of Medicine and Pharmacy, Iasi, Romania
- Clinical Rehabilitation Hospital, 1st Rheumatology Clinic, Iasi, Romania
| |
Collapse
|
43
|
Chang X, Xu S, Zhang H. Regulation of bone health through physical exercise: Mechanisms and types. Front Endocrinol (Lausanne) 2022; 13:1029475. [PMID: 36568096 PMCID: PMC9768366 DOI: 10.3389/fendo.2022.1029475] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 11/15/2022] [Indexed: 12/12/2022] Open
Abstract
Osteoporosis, characterized by bone mineral density reduction, bone mass loss, increased bone fragility, and propensity to fractures, is a common disease in older individuals and one of the most serious health problems worldwide. The imbalance between osteoblasts and osteoclasts results in the predominance of bone resorption and decreased bone formation. In recent years, it has been found that regular and proper exercise not only helps prevent the occurrence of osteoporosis but also adds benefits to osteoporosis therapy; accordingly, bone homeostasis is closely associated with mechanical stress and the intricate crosstalk between osteoblasts and osteoclasts. In this review, we summarize the mechanisms of exercise on osteoporosis and provide new proposals for the prevention and treatment of osteoporosis.
Collapse
Affiliation(s)
- Xinyu Chang
- Department of Traumatic Orthopedics, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Sheng Xu
- National Key Laboratory of Medical Immunology and Institute of Immunology, Institute of Immunology, Naval Medical University, Shanghai, China
| | - Hao Zhang
- Department of Traumatic Orthopedics, the First Affiliated Hospital of Naval Medical University, Naval Medical University, Shanghai, China
| |
Collapse
|
44
|
Choi RB, Hoggatt AM, Horan DJ, Rogers EZ, Hong JM, Robling AG. Targeting Sclerostin and Dkk1 at Optimized Proportions of Low-Dose Antibody Achieves Similar Skeletal Benefits to Higher-Dose Sclerostin Targeting in the Mature Adult and Aged Skeleton. Aging Dis 2022; 13:1891-1900. [PMID: 36465166 PMCID: PMC9662273 DOI: 10.14336/ad.2022.0315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/15/2022] [Indexed: 11/18/2022] Open
Abstract
Age-associated low bone mass disease is a growing problem in the US. Development of osteoanabolic therapies for treating skeletal fragility has lagged behind anti-catabolic therapies, but several bone-building molecules are clinically available. We reported previously that antibody-based neutralization of the Lrp5/Lrp6 inhibitor Dkk1 has minimal effects on bone gain, but can potentiate the already potent osteoanabolic effects of sclerostin inhibition (another Lrp5/Lrp6 inhibitor highly expressed by osteocytes). In this communication, we test whether an optimized ratio of sclerostin and Dkk1 antibodies (Scl-mAb and Dkk1-mAb, respectively), administered at low doses, can maintain the same bone-building effects as higher dose Scl-mAb, in adult (6 months of age) and aged (20 months of age) wild-type mice. A 3:1 dose of Scl-mAb:Dkk1-mAb at 12.5 mg/kg was equally efficacious as 25 mg/kg of Scl-mAb in both age groups, using radiographic (DXA, µCT), biomechanical, (3-point bending tests), and histological (fluorochrome-based bone formation parameters) outcome measures. For some bone properties, including trabecular thickness and bone mineral density in the spine, and endocortical bone formation rates in the femur, the 3:1 treatment was associated with significantly improved skeletal properties compared to twice the dose of Scl-mAb. Cortical porosity in aged mice was also reduced by both Scl-mAb and low-dose 3:1 treatment. Overall, both treatments were efficacious in the mature adult (6 mo.) and aged (20 mo.) skeletons, suggesting Wnt targeting is a viable strategy for improving skeletal fragility in the very old. Further, the data suggest that low dose of combination therapy can be at least equally efficacious as higher doses of Scl-mAb monotherapy.
Collapse
Affiliation(s)
- Roy B. Choi
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - April M. Hoggatt
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Daniel J. Horan
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Emily Z. Rogers
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Jung Min Hong
- Division of Biomedical and Applied Sciences, Indiana University School of Dentistry, Indianapolis, IN, USA.
| | - Alexander G. Robling
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.,Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, IN, USA.,Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, USA.,Indiana Center for Musculoskeletal Health, Indianapolis, IN, USA.,Correspondence should be addressed to: Dr. Alexander G. Robling, Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA. .
| |
Collapse
|
45
|
Safwan-Zaiter H, Wagner N, Wagner KD. P16INK4A-More Than a Senescence Marker. Life (Basel) 2022; 12:1332. [PMID: 36143369 PMCID: PMC9501954 DOI: 10.3390/life12091332] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Aging is a biological feature that is characterized by gradual degeneration of function in cells, tissues, organs, or an intact organism due to the accumulation of environmental factors and stresses with time. Several factors have been attributed to aging such as oxidative stress and augmented production or exposure to reactive oxygen species, inflammatory cytokines production, telomere shortening, DNA damage, and, importantly, the deposit of senescent cells. These are irreversibly mitotically inactive, yet metabolically active cells. The reason underlying their senescence lies within the extrinsic and the intrinsic arms. The extrinsic arm is mainly characterized by the expression and the secretory profile known as the senescence-associated secretory phenotype (SASP). The intrinsic arm results from the impact of several genes meant to regulate the cell cycle, such as tumor suppressor genes. P16INK4A is a tumor suppressor and cell cycle regulator that has been linked to aging and senescence. Extensive research has revealed that p16 expression is significantly increased in senescent cells, as well as during natural aging or age-related pathologies. Based on this fact, p16 is considered as a specific biomarker for detecting senescent cells and aging. Other studies have found that p16 is not only a senescence marker, but also a protein with many functions outside of senescence and aging. In this paper, we discuss and shed light on several studies that show the different functions of p16 and provide insights in its role in several biological processes besides senescence and aging.
Collapse
Affiliation(s)
| | - Nicole Wagner
- CNRS, INSERM, iBV, Université Côte d’Azur, 06107 Nice, France
| | | |
Collapse
|
46
|
Wang T, Huang S, He C. Senescent cells: A therapeutic target for osteoporosis. Cell Prolif 2022; 55:e13323. [DOI: 10.1111/cpr.13323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Tiantian Wang
- Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital Sichuan University Chengdu Sichuan China
- Institute of Rehabilitation Medicine, West China Hospital Sichuan University Chengdu Sichuan China
| | - Shishu Huang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital and West China School of Medicine Sichuan University Chengdu Sichuan China
| | - Chengqi He
- Department of Rehabilitation Medicine, Key Laboratory of Rehabilitation Medicine, West China Hospital Sichuan University Chengdu Sichuan China
- Institute of Rehabilitation Medicine, West China Hospital Sichuan University Chengdu Sichuan China
| |
Collapse
|
47
|
Yamada T, Fukasawa K, Horie T, Kadota T, Lyu J, Tokumura K, Ochiai S, Iwahashi S, Suzuki A, Park G, Ueda R, Yamamoto M, Kitao T, Shirahase H, Ochi H, Sato S, Iezaki T, Hinoi E. The role of CDK8 in mesenchymal stem cells in controlling osteoclastogenesis and bone homeostasis. Stem Cell Reports 2022; 17:1576-1588. [PMID: 35777359 PMCID: PMC9287674 DOI: 10.1016/j.stemcr.2022.06.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 01/02/2023] Open
Abstract
Bone marrow mesenchymal stem cells (MSCs) are critical regulators of postnatal bone homeostasis. Osteoporosis is characterized by bone volume and strength deterioration, partly due to MSC dysfunction. Cyclin-dependent kinase 8 (CDK8) belongs to the transcription-related CDK family. Here, CDK8 in MSCs was identified as important for bone homeostasis. CDK8 level was increased in aged MSCs along with the association with aging-related signals. Mouse genetic studies revealed that CDK8 in MSCs plays a crucial role in bone resorption and homeostasis. Mechanistically, CDK8 in MSCs extrinsically controls osteoclastogenesis through the signal transducer and transcription 1 (STAT1)-receptor activator of the nuclear factor κ Β ligand (RANKL) axis. Moreover, aged MSCs have high osteoclastogenesis-supporting activity, partly through a CDK8-dependent manner. Finally, pharmacological inhibition of CDK8 effectively repressed MSC-dependent osteoclastogenesis and prevented ovariectomy-induced osteoclastic activation and bone loss. These findings highlight that the CDK8-STAT1-RANKL axis in MSCs could play a crucial role in bone resorption and homeostasis. Aging increases CDK8 expression level in MSCs and their progeny CDK8 in MSCs plays a crucial role in bone resorption and homeostasis CDK8 in MSCs extrinsically controls osteoclastogenesis through STAT1/RANKL axis CDK8 inhibitor prevents ovariectomy-induced osteoclastic activation and bone loss
Collapse
Affiliation(s)
- Takanori Yamada
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Kazuya Fukasawa
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Tetsuhiro Horie
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Takuya Kadota
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan; Drug Discovery Research Department, Kyoto Pharmaceutical Industries, Kyoto, Japan
| | - Jiajun Lyu
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Kazuya Tokumura
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Shinsuke Ochiai
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Sayuki Iwahashi
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Akane Suzuki
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Gyujin Park
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Rie Ueda
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Megumi Yamamoto
- Drug Discovery Research Department, Kyoto Pharmaceutical Industries, Kyoto, Japan
| | - Tatsuya Kitao
- Drug Discovery Research Department, Kyoto Pharmaceutical Industries, Kyoto, Japan
| | - Hiroaki Shirahase
- Drug Discovery Research Department, Kyoto Pharmaceutical Industries, Kyoto, Japan
| | - Hiroki Ochi
- Department of Physiology and Cell Biology, Tokyo Medical and Dental University, Graduate School, Tokyo 113-8510, Japan
| | - Shingo Sato
- Center for Innovative Cancer Treatment, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Takashi Iezaki
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Eiichi Hinoi
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu 501-1196, Japan; United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu 501-1196, Japan.
| |
Collapse
|
48
|
Goff E, Cohen A, Shane E, Recker RR, Kuhn G, Müller R. Large-scale osteocyte lacunar morphological analysis of transiliac bone in normal and osteoporotic premenopausal women. Bone 2022; 160:116424. [PMID: 35460961 DOI: 10.1016/j.bone.2022.116424] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 11/29/2022]
Abstract
Bone's ability to adapt is governed by the network of embedded osteocytes, which inhabit individual pores called lacunae. The morphology of these lacunae and their resident osteocytes are known to change with age and diseases such as postmenopausal osteoporosis. However, it is unclear whether alterations in lacunar morphology are present in younger populations with osteoporosis. To investigate this, we implemented a previously validated methodology to image and quantify the three-dimensional morphometries of lacunae on a large scale with ultra-high-resolution micro-computed tomography (microCT) in transiliac bone biopsies from three groups of premenopausal women: control n = 39; idiopathic osteoporosis (IOP) n = 45; idiopathic low BMD (ILBMD) n = 19. Lacunar morphometric parameters were measured in both trabecular and cortical bone such as lacunar density (Lc.N/BV), lacunar volume (Lc.V), and lacunar sphericity (Lc.Sr). These were then compared against each other and also with previously measured tissue morphometries such as bone volume density (BV/TV), trabecular separation (Tb.Sp), trabecular number (Tb.N), and others. We detected no differences in lacunar morphology between the IOP, ILBMD and healthy premenopausal women. In contrast, we did find significant differences between lacunar morphologies including Lc.N/BV, Lc. V, and Lc. Sr in cortical and trabecular regions within all three groups (p < 0.001), which was consistent with our previous findings on a subgroup of the healthy group. Furthermore, we discovered strong correlations between Lc. Sr from trabecular regions with the measured BV/TV (R = -0.90, p < 0.05). The findings and comprehensive lacunar dataset we present here will be a crucial foundation for future investigations of the relationship between osteocyte lacunar morphology and disease.
Collapse
Affiliation(s)
- Elliott Goff
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Adi Cohen
- Department of Medicine, Columbia University Vagelos College of Physicians & Surgeons, New York, NY, USA
| | - Elizabeth Shane
- Department of Medicine, Columbia University Vagelos College of Physicians & Surgeons, New York, NY, USA
| | - Robert R Recker
- Department of Medicine, Creighton University Medical Center, Omaha, NE, USA
| | - Gisela Kuhn
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
49
|
Mmp13 deletion in mesenchymal cells increases bone mass and may attenuate the cortical bone loss caused by estrogen deficiency. Sci Rep 2022; 12:10257. [PMID: 35715555 PMCID: PMC9205908 DOI: 10.1038/s41598-022-14470-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 05/17/2022] [Indexed: 12/30/2022] Open
Abstract
The protective effect of estrogens against cortical bone loss is mediated via direct actions on mesenchymal cells, but functional evidence for the mediators of these effects has only recently begun to emerge. We report that the matrix metalloproteinase 13 (MMP13) is the highest up-regulated gene in mesenchymal cells from mice lacking the estrogen receptor alpha (ERα). In sham-operated female mice with conditional Mmp13 deletion in Prrx1 expressing cells (Mmp13ΔPrrx1), the femur and tibia length was lower as compared to control littermates (Mmp13f./f). Additionally, in the sham-operated female Mmp13ΔPrrx1 mice cortical thickness and trabecular bone volume in the femur and tibia were higher and osteoclast number at the endocortical surfaces was lower, whereas bone formation rate was unaffected. Notably, the decrease of cortical thickness caused by ovariectomy (OVX) in the femur and tibia of Mmp13f./f mice was attenuated in the Mmp13ΔPrrx1 mice; but the decrease of trabecular bone caused by OVX was not affected. These results reveal that mesenchymal cell-derived MMP13 may regulate osteoclast number and/or activity, bone resorption, and bone mass. And increased production of mesenchymal cell-derived factors may be important mediators of the adverse effect of estrogen deficiency on cortical, but not trabecular, bone.
Collapse
|
50
|
BMP9 reduces age-related bone loss in mice by inhibiting osteoblast senescence through Smad1-Stat1-P21 axis. Cell Death Dis 2022; 8:254. [PMID: 35523787 PMCID: PMC9076651 DOI: 10.1038/s41420-022-01048-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 04/22/2022] [Accepted: 04/27/2022] [Indexed: 11/27/2022]
Abstract
Age-related osteoporosis is characterized by the accumulation of senescent osteoblastic cells in bone microenvironment and significantly reduced osteogenic differentiation. Clearing of the senescent cells is helpful to improve bone formation in aged mice. Bone morphogenetic protein 9 (BMP9), a multifunctional protein produced and secreted by liver, was reported to improve osteoporosis caused by estrogen withdrawal. However, the mechanism of BMP9 has not been fully elucidated, and its effect on senile osteoporosis has not been reported. This study reveals that BMP9 significantly increases bone mass and improves bone biomechanical properties in aged mice. Furthermore, BMP9 reduces expression of senescent genes in bone microenvironment, accompanied by decreased senescence-associated secretory phenotypes (SASPs) such as Ccl5, Mmp9, Hmgb1, Nfkb1, and Vcam1. In vitro, Bmp9 treatment inhibits osteoblast senescence through activating Smad1, which suppresses the transcriptional activity of Stat1, thereby inhibits P21 expression and SASPs production. Furthermore, inhibiting the Smad1 signal in vivo can reverse the inhibitory effect of BMP9 on Stat1 and downstream senescent genes, which eliminates the protection of BMP9 on age-related osteoporosis. These findings highlight the critical role of BMP9 on reducing age-related bone loss by inhibiting osteoblast senescence through Smad1-Stat1-P21 axis. BMP9 inhibits cellular senescence by activation of Smad1, which suppresses the transcription of Stat1, resulting in decreased P21 expression and SASPs production in osteoblast. The anti-aging effect of BMP9 is benefit to improving age-related osteoporosis.![]()
Collapse
|