1
|
Falvino A, Gasperini B, Cariati I, Bonanni R, Chiavoghilefu A, Gasbarra E, Botta A, Tancredi V, Tarantino U. Cellular Senescence: The Driving Force of Musculoskeletal Diseases. Biomedicines 2024; 12:1948. [PMID: 39335461 PMCID: PMC11429507 DOI: 10.3390/biomedicines12091948] [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: 07/23/2024] [Revised: 08/20/2024] [Accepted: 08/24/2024] [Indexed: 09/30/2024] Open
Abstract
The aging of the world population is closely associated with an increased prevalence of musculoskeletal disorders, such as osteoporosis, sarcopenia, and osteoarthritis, due to common genetic, endocrine, and mechanical risk factors. These conditions are characterized by degeneration of bone, muscle, and cartilage tissue, resulting in an increased risk of fractures and reduced mobility. Importantly, a crucial role in the pathophysiology of these diseases has been proposed for cellular senescence, a state of irreversible cell cycle arrest induced by factors such as DNA damage, telomere shortening, and mitochondrial dysfunction. In addition, senescent cells secrete pro-inflammatory molecules, called senescence-associated secretory phenotype (SASP), which can alter tissue homeostasis and promote disease progression. Undoubtedly, targeting senescent cells and their secretory profiles could promote the development of integrated strategies, including regular exercise and a balanced diet or the use of senolytics and senomorphs, to improve the quality of life of the aging population. Therefore, our review aimed to highlight the role of cellular senescence in age-related musculoskeletal diseases, summarizing the main underlying mechanisms and potential anti-senescence strategies for the treatment of osteoporosis, sarcopenia, and osteoarthritis.
Collapse
Affiliation(s)
- Angela Falvino
- Department of Biomedicine and Prevention, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Beatrice Gasperini
- Department of Biomedicine and Prevention, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Ida Cariati
- Department of Systems Medicine, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Roberto Bonanni
- Department of Biomedicine and Prevention, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Angela Chiavoghilefu
- Department of Orthopaedics and Traumatology, "Policlinico Tor Vergata" Foundation, Viale Oxford 81, 00133 Rome, Italy
| | - Elena Gasbarra
- Department of Orthopaedics and Traumatology, "Policlinico Tor Vergata" Foundation, Viale Oxford 81, 00133 Rome, Italy
| | - Annalisa Botta
- Department of Biomedicine and Prevention, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Virginia Tancredi
- Department of Systems Medicine, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
- Centre of Space Bio-Medicine, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| | - Umberto Tarantino
- Department of Orthopaedics and Traumatology, "Policlinico Tor Vergata" Foundation, Viale Oxford 81, 00133 Rome, Italy
- Centre of Space Bio-Medicine, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
- Department of Clinical Sciences and Translational Medicine, "Tor Vergata" University of Rome, Via Montpellier 1, 00133 Rome, Italy
| |
Collapse
|
2
|
Guan L, Eisenmenger A, Crasta KC, Sandalova E, Maier AB. Therapeutic effect of dietary ingredients on cellular senescence in animals and humans: A systematic review. Ageing Res Rev 2024; 95:102238. [PMID: 38382678 DOI: 10.1016/j.arr.2024.102238] [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: 10/12/2023] [Revised: 01/12/2024] [Accepted: 02/16/2024] [Indexed: 02/23/2024]
Abstract
BACKGROUND Cellular senescence has been regarded as a therapeutic target for ageing and age-related diseases. Several senotherapeutic agents have been proposed, including compounds derived from natural products which hold the translational potential to promote healthy ageing. This systematic review examined the association of dietary ingredients with cellular senescence in animals and humans, with an intent to identify dietary ingredients with senotherapeutic potential. METHODS This systematic review was registered at PROSPERO International prospective register of systematic reviews (Reg #: CRD42022338885). The databases PubMed and Embase were systematically searched for key terms related to cellular senescence, senescence markers, diets, nutrients and bioactive compounds. Intervention and observational studies on human and animals investigating the effects of dietary ingredients via oral administration on cellular senescence load were included. The SYRCLE's risk of bias tool and Cochrane risk of bias tool v2.0 were used to assess the risk of bias for animal and human studies respectively. RESULTS Out of 5707 identified articles, 83 articles consisting of 78 animal studies and 5 human studies aimed to reduce cellular senescence load using dietary ingredients. In animal studies, the most-frequently used senescence model was normative ageing (26 studies), followed by D-galactose-induced models (17 studies). Resveratrol (8 studies), vitamin E (4 studies) and soy protein isolate (3 studies) showed positive effects on reducing the level of senescence markers such as p53, p21, p16 and senescence-associated ß-galactosidase in various tissues of physiological systems. In three out of five human studies, ginsenoside Rg1 had no positive effect on reducing senescence in muscle tissues after exercise. The risk of bias for both animal and human studies was largely unclear. CONCLUSION Resveratrol, vitamin E and soy protein isolate are promising senotherapeutics studied in animal models. Studies testing dietary ingredients with senotherapeutic potential in humans are limited and translation is highly warranted.
Collapse
Affiliation(s)
- Lihuan Guan
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore
| | - Anna Eisenmenger
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore
| | - Karen C Crasta
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore; Department of Physiology, National University of Singapore, Singapore; NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Agency for Science, Technology & Research (A⁎STAR), Institute of Molecular and Cell Biology (IMCB), Singapore
| | - Elena Sandalova
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore
| | - Andrea B Maier
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore; Department of Human Movement Sciences, @AgeAmsterdam, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, the Netherlands.
| |
Collapse
|
3
|
Studzińska-Sroka E, Paczkowska-Walendowska M, Erdem C, Paluszczak J, Kleszcz R, Hoszman-Kulisz M, Cielecka-Piontek J. Anti-Aging Properties of Chitosan-Based Hydrogels Rich in Bilberry Fruit Extract. Antioxidants (Basel) 2024; 13:105. [PMID: 38247529 PMCID: PMC10812676 DOI: 10.3390/antiox13010105] [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: 12/14/2023] [Revised: 12/30/2023] [Accepted: 01/10/2024] [Indexed: 01/23/2024] Open
Abstract
Photoaging is a process related to an increased level of reactive oxygen species (ROS). Polyphenols can scavenge free radicals in the body, which can delay skin aging. Therefore, our work aimed to prepare a biologically active extract from dry fruits of Vaccinium myrtillus or Vaccinium corymbosum and use it for the preparation of hydrogels for topical application. Therefore, eight different extracts (using V. myrtillus and V. corymbosum and different extraction mixtures: methanol, methanol-water 1:1, water, acetone-water 1:1) were prepared and their phytochemical (total polyphenolic content, total flavonoid content, total anthocyanin content) and biological properties (antioxidant, anti-hyaluronidase, and anti-tyrosinase activity) were assessed. Cytotoxicity towards HaCaT keratinocytes was also determined. Based on the results, the acetone-water extract from V. myrtillus was selected for further study. Using the Design of Experiments approach, chitosan-based hydrogels with bilberry fruit extract were prepared. The content of extract and chitosan were selected as independent factors. The activity of hydrogels depended on the extract content; however, the enzyme-inhibiting (anti-hyaluronidase and anti-tyrosinase) activity resulted from the presence of both the extract and chitosan. Increased concentration of chitosan in the hydrogel base led to increased viscosity of the hydrogel and, consequently, a slower release of active compounds. To get optimal hydrogel characteristics, 1% extract and 2.5% MMW chitosan were utilized. The research suggests the validity of using bilberry fruit extracts in topical preparations with anti-aging properties.
Collapse
Affiliation(s)
- Elżbieta Studzińska-Sroka
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3 Str, 60-806 Poznań, Poland; (E.S.-S.); (M.H.-K.); (J.C.-P.)
| | - Magdalena Paczkowska-Walendowska
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3 Str, 60-806 Poznań, Poland; (E.S.-S.); (M.H.-K.); (J.C.-P.)
| | - Cansu Erdem
- Department Pharmaceutical Chemistry, Ege Üniversitesi, 35040 İzmir, Turkey;
| | - Jarosław Paluszczak
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3 Str, 60-806 Poznań, Poland; (J.P.); (R.K.)
| | - Robert Kleszcz
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3 Str, 60-806 Poznań, Poland; (J.P.); (R.K.)
| | - Marta Hoszman-Kulisz
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3 Str, 60-806 Poznań, Poland; (E.S.-S.); (M.H.-K.); (J.C.-P.)
| | - Judyta Cielecka-Piontek
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3 Str, 60-806 Poznań, Poland; (E.S.-S.); (M.H.-K.); (J.C.-P.)
| |
Collapse
|
4
|
Zhu C, Ding H, Shi L, Zhang S, Tong X, Huang M, Liu L, Guan X, Zou J, Yuan Y, Chen X. Exercise improved bone health in aging mice: a role of SIRT1 in regulating autophagy and osteogenic differentiation of BMSCs. Front Endocrinol (Lausanne) 2023; 14:1156637. [PMID: 37476496 PMCID: PMC10355118 DOI: 10.3389/fendo.2023.1156637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 06/07/2023] [Indexed: 07/22/2023] Open
Abstract
Introduction This study was designed to investigate the effect of running exercise on improving bone health in aging mice and explore the role of the SIRT1 in regulating autophagy and osteogenic differentiation of Bone marrow Mesenchymal Stem Cells (BMSCs). Methods Twelve-month-old male C57BL/6J mice were used in this study as the aging model and were assigned to treadmill running exercise for eight weeks. Non-exercise male C57BL/6J mice of the same old were used as aging control and five-month-old mice were used as young controls. BMSCs were isolated from mice and subjected to mechanical stretching stimulation in vitro. Results The results showed that aging mice had lower bone mass, bone mineral density (BMD), and autophagy than young mice, while running exercise improved BMD and bone mass as well as upregulated autophagy in bone cells. Mechanical loading increased osteogenic differentiation and autophagy in BMSCs, and knockdown of SIRT1 in BMSCs demonstrated that SIRT1-regulated autophagy involved the mechanical loading activation of osteogenic differentiation. Conclusion Taken together, this study revealed that exercise improved bone health during aging by activating bone formation, which can be attributed to osteogenic differentiation of BMSCs through the activation of SIRT1-mediated autophagy. The mechanisms underlying this effect may involve mechanical loading.
Collapse
Affiliation(s)
- Chengyu Zhu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
- School of Sports Science, Wenzhou Medical University, Wenzhou, China
| | - Haili Ding
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Liang Shi
- Department of Gynaecology and Obstetrics, Xinchang People’s Hospital, Shaoxing, China
| | - Shihua Zhang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Xiaoyang Tong
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Mei Huang
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Lifei Liu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
- Department of Rehabilitation, The People’s Hospital of Liaoning Province, Shenyang, China
| | - Xiaotian Guan
- Institute of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Jun Zou
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Yu Yuan
- School of Exercise and Health, Guangzhou Sport University, Guangzhou, China
| | - Xi Chen
- School of Sports Science, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
5
|
Effect of Hydrogen Oxide-Induced Oxidative Stress on Bone Formation in the Early Embryonic Development Stage of Chicken. Biomolecules 2023; 13:biom13010154. [PMID: 36671539 PMCID: PMC9855391 DOI: 10.3390/biom13010154] [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: 12/08/2022] [Revised: 01/02/2023] [Accepted: 01/09/2023] [Indexed: 01/15/2023] Open
Abstract
The current study aimed to monitor the impact of H2O2-induced oxidative stress on avian bone formation during the early stage of embryonic development. Fertilized Cobb broiler eggs were divided into five treatment groups and micro-injected with varying concentrations of H2O2, i.e., control (PBS; 0 nM), 10 nM, 30 nM, 100 nM, and 300 nM, on embryonic day 3, with continued incubation thereafter. The treatment concentrations were selected based on the level of lipid peroxidation and the survival rate of embryo. Embryos were collected at 6 h, 24 h, 48 h, and 72 h post-injection. The mRNA expression levels of apoptotic markers, antioxidant enzymes, and early bone formation gene markers were measured. The results showed that the microinjection of H2O2 altered the expression pattern of antioxidant enzymes' mRNA during early embryogenesis and decreased the expression of COL1A2 and COL2A1 at 6 h and 24 h post-injection. Decreased expression of BMP, BGLAP, and RUNX2 was observed 48 h post-injection. Additionally, a shorter embryo length was observed in the 100 nM and 300 nM H2O2 treatment groups 72 h post-injection. In conclusion, H2O2-induced oxidative stress suppressed the expression of bone formation gene markers, with chronic effects on avian embryonic development.
Collapse
|
6
|
Naude, MTech (Hom) DF. Chronic Sub-Clinical Systemic Metabolic Acidosis - A Review with Implications for Clinical Practice. J Evid Based Integr Med 2022; 27:2515690X221142352. [PMID: 36448194 PMCID: PMC9716591 DOI: 10.1177/2515690x221142352] [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] [Indexed: 12/03/2022] Open
Abstract
When arterial serum pH remains near the lower pH limit of 7.35 for protracted periods of time, a low-grade, sub-clinical form of acidosis results, referred to in this review as chronic, sub-clinical, systemic metabolic acidosis (CSSMA). This narrative review explores the scientific basis for CSSMA, its consequences for health, and potential therapeutic interventions. The major etiology of CSSMA is the shift away from the ancestral, alkaline diet which was rich in fruit and vegetables, toward the contemporary, acidogenic 'Westernized' diet characterized by higher animal protein consumption and lack of base forming minerals. Urine pH is reduced with high dietary acid load and may be a convenient marker of CSSMA. Evidence suggests further that CSSMA negatively influences cortisol levels potentially contributing significantly to the pathophysiology thereof. Both CSSMA and high dietary acid load are associated with the risk and prognosis of various chronic diseases. Clinical trials show that CSSMA can be addressed successfully through alkalizing the diet by increasing fruit and vegetable intake and/or supplementing with alkaline minerals. This review confirms the existence of a significant body of evidence regarding this low-grade form of acidosis as well as evidence to support its diverse negative implications for health, and concludes that CSSMA is a condition warranting further research.
Collapse
Affiliation(s)
- David Francis Naude, MTech (Hom)
- Irma Schutte Foundation, Drummond, South Africa,David Francis Naude, Irma Schutte Foundation, 42 Protea Hill Rd, Drummond, KwaZulu Natal, 3626, South Africa. Postal address: P.O Box 8, Hillcrest, KwaZulu Natal, 3650, South Africa.
| |
Collapse
|
7
|
Chen JR, Lazarenko OP, Blackburn ML, Chen JF, Randolph CE, Zabaleta J, Schroder K, Pedersen KB, Ronis MJJ. Nox4 expression in osteo-progenitors controls bone development in mice during early life. Commun Biol 2022; 5:583. [PMID: 35701603 PMCID: PMC9198054 DOI: 10.1038/s42003-022-03544-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/01/2022] [Indexed: 11/09/2022] Open
Abstract
Tightly regulated and cell-specific NADPH-oxidases (Nox) represent one of the major sources of reactive oxygen species (ROS) signaling molecules that are involved in tissue development and stem cell self-renewal. We have characterized the role of Nox4 in osteo-progenitors during postnatal bone development. Nox4 expression in bone and ROS generation were increased during early osteoblast differentiation and bone development. Stromal osteoblastic cell self-renewal, proliferation and ROS production were significantly lower in samples from whole-body Nox4 knockout mice (Nox4-/-) and conditional knockout (CKO) mice with depletion of Nox4 in the limb bud mesenchyme compared with those from control mice (Nox4fl/fl), but they were reversed after 9 passages. In both sexes, bone volume, trabecular number and bone mineral density were significantly lower in 3-week old CKO and Nox4-/- mice compared with Nox4fl/fl controls. This was reflected in serum levels of bone formation markers alkaline phosphatase (ALP) and procollagen 1 intact N-terminal propeptide (P1NP). However, under-developed bone formation in 3-week old CKO and Nox4-/- mice quickly caught up to levels of control mice by 6-week of age, remained no different at 13-week of age, and was reversed in 32-week old male mice. Osteoclastogenesis showed no differences among groups, however, CTX1 reflecting osteoclast activity was significantly higher in 3-week old male CKO and Nox4-/- mice compared with control mice, and significantly lower in 32-week old Nox4-/- mice compared with control mice. These data suggest that Nox4 expression and ROS signaling in bone and osteoblastic cells coordinately play an important role in osteoblast differentiation, proliferation and maturation.
Collapse
Affiliation(s)
- Jin-Ran Chen
- Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA. .,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA.
| | - Oxana P. Lazarenko
- grid.508987.bArkansas Children’s Nutrition Center, Little Rock, AR 72202 USA ,grid.241054.60000 0004 4687 1637Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202 USA
| | - Michael L. Blackburn
- grid.508987.bArkansas Children’s Nutrition Center, Little Rock, AR 72202 USA ,grid.241054.60000 0004 4687 1637Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202 USA
| | - Jennifer F. Chen
- grid.411017.20000 0001 2151 0999Undergraduate Pre-Medical Program, University of Arkansas at Fayetteville, Fayetteville, AR 72701 USA
| | - Christopher E. Randolph
- grid.488749.eCenter for Translational Pediatric Research, Arkansas Children’s Research Institute, Little Rock, AR 72202 USA
| | - Jovanny Zabaleta
- grid.279863.10000 0000 8954 1233Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112 USA
| | - Katrin Schroder
- grid.7839.50000 0004 1936 9721Institute of Physiology I, Goethe-University, Frankfurt, Germany
| | - Kim B. Pedersen
- grid.279863.10000 0000 8954 1233Department of Interdisciplinary Oncology (DIO), Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, Louisiana Cancer Research Center, New Orleans, LA 70112 USA
| | - Martin J. J. Ronis
- grid.279863.10000 0000 8954 1233Department of Interdisciplinary Oncology (DIO), Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, Louisiana Cancer Research Center, New Orleans, LA 70112 USA
| |
Collapse
|
8
|
Role of Histone Deacetylases in Monocyte Function in Health and Chronic Inflammatory Diseases. Rev Physiol Biochem Pharmacol 2021; 180:1-47. [PMID: 33974124 DOI: 10.1007/112_2021_59] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Histone deacetylases (HDACs) are a family of 18 members that participate in the epigenetic regulation of gene expression. In addition to histones, some HDACs also deacetylate transcription factors and specific cytoplasmic proteins.Monocytes, as part of the innate immune system, maintain tissue homeostasis and help fight infections and cancer. In these cells, HDACs are involved in multiple processes including proliferation, migration, differentiation, inflammatory response, infections, and tumorigenesis. Here, a systematic description of the role that most HDACs play in these functions is reviewed. Specifically, some HDACs induce a pro-inflammatory response and play major roles in host defense. Conversely, other HDACs reprogram monocytes and macrophages towards an immunosuppressive phenotype. The right balance between both types helps monocytes to respond correctly to the different physiological/pathological stimuli. However, aberrant expressions or activities of specific HDACs are associated with autoimmune diseases along with other chronic inflammatory diseases, infections, or cancer.This paper critically reviews the interesting and extensive knowledge regarding the role of some HDACs in these pathologies. It also shows that as yet, very little progress has been made toward the goal of finding effective HDAC-targeted therapies. However, given their obvious potential, we conclude that it is worth the effort to develop monocyte-specific drugs that selectively target HDAC subtypes with the aim of finding effective treatments for diseases in which our innate immune system is involved.
Collapse
|
9
|
Sato AY, Pellegrini GG, Cregor M, McAndrews K, Choi RB, Maiz M, Johnson O, McCabe LD, McCabe GP, Ferruzzi MG, Lila MA, Peacock M, Burr DB, Nakatsu CH, Weaver CM, Bellido T. Skeletal Protection and Promotion of Microbiome Diversity by Dietary Boosting of the Endogenous Antioxidant Response. J Bone Miner Res 2021; 36:768-778. [PMID: 33316081 DOI: 10.1002/jbmr.4231] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 12/09/2020] [Accepted: 12/09/2020] [Indexed: 01/28/2023]
Abstract
There is an unmet need for interventions with better compliance that prevent the adverse effects of sex steroid deficiency on the musculoskeletal system. We identified a blueberry cultivar (Montgomerym [Mont]) that added to the diet protects female mice from musculoskeletal loss and body weight changes induced by ovariectomy. Mont, but not other blueberries, increased the endogenous antioxidant response by bypassing the traditional antioxidant transcription factor Nrf2 and without activating estrogen receptor canonical signaling. Remarkably, Mont did not protect the male skeleton from androgen-induced bone loss. Moreover, Mont increased the variety of bacterial communities in the gut microbiome (α-diversity) more in female than in male mice; shifted the phylogenetic relatedness of bacterial communities (β-diversity) further in females than males; and increased the prevalence of the taxon Ruminococcus1 in females but not males. Therefore, this nonpharmacologic intervention (i) protects from estrogen but not androgen deficiency; (ii) preserves bone, skeletal muscle, and body composition; (iii) elicits antioxidant defense responses independently of classical antioxidant/estrogenic signaling; and (iv) increases gut microbiome diversity toward a healthier signature. These findings highlight the impact of nutrition on musculoskeletal and gut microbiome homeostasis and support the precision medicine principle of tailoring dietary interventions to patient individualities, like sex. © 2020 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Amy Y Sato
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Gretel G Pellegrini
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Meloney Cregor
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kevin McAndrews
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Roy B Choi
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Maria Maiz
- Department of Nutrition, Purdue University, West Lafayette, IN, USA
| | - Olivia Johnson
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Linda D McCabe
- Department of Statistics, Purdue University, West Lafayette, IN, USA
| | - George P McCabe
- Department of Statistics, Purdue University, West Lafayette, IN, USA
| | - Mario G Ferruzzi
- Department of Food Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, USA
| | - Mary A Lila
- Department of Food Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, USA
| | - Munro Peacock
- Department of Medicine, Division of Endocrinology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - David B Burr
- Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cindy H Nakatsu
- Department of Agronomy, Purdue University, West Lafayette, IN, USA
| | - Connie M Weaver
- Department of Nutrition, Purdue University, West Lafayette, IN, USA
| | - Teresita Bellido
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Department of Anatomy, Cell Biology, and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Medicine, Division of Endocrinology, Indiana University School of Medicine, Indianapolis, IN, USA.,Central Arkansas Veterans Healthcare System, Little Rock, AR, USA.,Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, IN, USA
| |
Collapse
|
10
|
Wang JS, Yoon SH, Wein MN. Role of histone deacetylases in bone development and skeletal disorders. Bone 2021; 143:115606. [PMID: 32829038 PMCID: PMC7770092 DOI: 10.1016/j.bone.2020.115606] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/11/2020] [Accepted: 08/15/2020] [Indexed: 02/08/2023]
Abstract
Bone cells must constantly respond to hormonal and mechanical cues to change gene expression programs. Of the myriad of epigenomic mechanisms used by cells to dynamically alter cell type-specific gene expression, histone acetylation and deacetylation has received intense focus over the past two decades. Histone deacetylases (HDACs) represent a large family of proteins with a conserved deacetylase domain first described to deacetylate lysine residues on histone tails. It is now appreciated that multiple classes of HDACs exist, some of which are clearly misnamed in that acetylated lysine residues on histone tails is not the major function of their deacetylase domain. Here, we will review the roles of proteins bearing deacetylase domains in bone cells, focusing on current genetic evidence for each individual HDAC gene. While class I HDACs are nuclear proteins whose primary role is to deacetylate histones, class IIa and class III HDACs serve other important cellular functions. Detailed knowledge of the roles of individual HDACs in bone development and remodeling will set the stage for future efforts to specifically target individual HDAC family members in the treatment of skeletal diseases such as osteoporosis.
Collapse
Affiliation(s)
- Jialiang S Wang
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sung-Hee Yoon
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marc N Wein
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
11
|
Li P, Mao WW, Zhang S, Zhang L, Chen ZR, Lu ZD. Sodium hydrosulfide alleviates dexamethasone-induced cell senescence and dysfunction through targeting the miR-22/sirt1 pathway in osteoblastic MC3T3-E1 cells. Exp Ther Med 2021; 21:238. [PMID: 33603846 PMCID: PMC7851607 DOI: 10.3892/etm.2021.9669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 09/15/2020] [Indexed: 01/30/2023] Open
Abstract
Glucocorticoid-induced osteoporosis is characterized by osteoblastic cell and microarchitecture dysfunction, as well as a loss of bone mass. Cell senescence contributes to the pathological process of osteoporosis and sodium hydrosulfide (NaHS) regulates the potent protective effects through delaying cell senescence. The aim of the present study was to investigate whether senescence could contribute to dexamethasone (Dex)-induced osteoblast impairment and to examine the effect of NaHS on Dex-induced cell senescence and damage. It was found that the levels of the senescence-associated markers, p53 and p21, were markedly increased in osteoblasts exposed to Dex. A p53 inhibitor reversed Dex-induced osteoblast injury, a process that was mitigated by NaHS administration through alleviating osteoblastic cell senescence. MicroRNA (miR)-22 blocked the impact of NaHS on Dex-induced osteoblast damage and senescence through targeting the regulation of Sirtuin 1 (sirt1) expression, as shown by the decreased cell viability and alkaline phosphatase activity, as well as an increased expression of p53 and p21. It was revealed that the sirt1 gene was the target of miR-22 in osteoblastic MC3T3-E1 cells through combining the results of dual luciferase reporter assays and reverse transcription-quantitative PCR, as well as western blot analyses. Silencing of sirt1 abolished the protective effect of NaHS against Dex-associated osteoblast senescence and injury. Taken together, the present study showed that NaHS prevents Dex-induced cell senescence and damage through targeting the miR-22/sirt1 pathway in osteoblastic MC3T3-E1 cells.
Collapse
Affiliation(s)
- Peng Li
- Department of Orthopedics, General Hospital of Ningxia Medical University, Xingqing, Yinchuan, Ningxia 750004, P.R. China
| | - Wei-Wei Mao
- Clinical Skill Center of Yinchuan First People's Hospital, Yinchuan, Ningxia 750001, P.R. China
| | - Shuai Zhang
- Department of Orthopedics, General Hospital of Ningxia Medical University, Xingqing, Yinchuan, Ningxia 750004, P.R. China
| | - Liang Zhang
- Department of Orthopedics, General Hospital of Ningxia Medical University, Xingqing, Yinchuan, Ningxia 750004, P.R. China
| | - Zhi-Rong Chen
- Department of Orthopedics, General Hospital of Ningxia Medical University, Xingqing, Yinchuan, Ningxia 750004, P.R. China
| | - Zhi-Dong Lu
- Department of Orthopedics, General Hospital of Ningxia Medical University, Xingqing, Yinchuan, Ningxia 750004, P.R. China
| |
Collapse
|
12
|
Chen Y, Zhou F, Liu H, Li J, Che H, Shen J, Luo E. SIRT1, a promising regulator of bone homeostasis. Life Sci 2021; 269:119041. [PMID: 33453243 DOI: 10.1016/j.lfs.2021.119041] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/29/2020] [Accepted: 01/06/2021] [Indexed: 12/16/2022]
Abstract
Sirtuin 1 (SIRT1), a nicotinamide adenine dinucleotide-dependent deacetylase, epigenetically regulates various cell metabolisms, including inflammation, tumorigenesis, and bone metabolism. Many clinical studies have found the potential of SIRT1 in predicting and treating bone-related disorders, such as osteoporosis and osteonecrosis, suggesting that SIRT1 might be a regulator of bone homeostasis. In order to identify the mechanisms that underlie the pivotal role of SIRT1 in bone homeostasis, many studies revealed that SIRT1 could maintain the balance between bone formation and absorption via regulating the ratio of osteoblasts to osteoclasts. SIRT1 controls the differentiation of mesenchymal stem cells (MSCs) and bone marrow-derived macrophages, increasing osteogenesis and reducing osteoclastogenesis. Besides, SIRT1 can enhance bone-forming cells' viability, including MSCs and osteoblasts under adverse conditions by resisting senescence, suppressing apoptosis, and promoting autophagy in favor of osteogenesis. Furthermore, the effect on bone vasculature homeostasis enables SIRT1 to become a valuable strategy for ischemic osteonecrosis and senile osteoporosis. The review systemically discusses SIRT1 pathways and the critical role in bone homeostasis and assesses whether SIRT1 is a potential target for manipulation and therapy, to lay a solid foundation for further researches in the future.
Collapse
Affiliation(s)
- Ye Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Feng Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Hanghang Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China; Maine Medical Center Research Institute, Maine Medical Center, Scarborough, ME 04074, USA
| | - Jiaxuan Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Huiling Che
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jiaqi Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - En Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
13
|
Chen JR, Zhao H, Wankhade UD, Chintapalli SV, Li C, Gai D, Shankar K, Zhan F, Lazarenko OP. GPR109A mediates the effects of hippuric acid on regulating osteoclastogenesis and bone resorption in mice. Commun Biol 2021; 4:53. [PMID: 33420329 PMCID: PMC7794563 DOI: 10.1038/s42003-020-01564-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 11/26/2020] [Indexed: 12/26/2022] Open
Abstract
The G protein-coupled receptor 109 A (GPR109A) is robustly expressed in osteoclastic precursor macrophages. Previous studies suggested that GPR109A mediates effects of diet-derived phenolic acids such as hippuric acid (HA) and 3-(3-hydroxyphenyl) propionic acid (3-3-PPA) on promoting bone formation. However, the role of GPR109A in metabolic bone homeostasis and osteoclast differentiation has not been investigated. Using densitometric, bone histologic and molecular signaling analytic methods, we uncovered that bone mass and strength were significantly higher in tibia and spine of standard rodent diet weaned 4-week-old and 6-month-old GPR109A gene deletion (GPR109A-/-) mice, compared to their wild type controls. Osteoclast numbers in bone and in ex vivo bone marrow cell cultures were significantly decreased in GPR109A-/- mice compared to wild type controls. In accordance with these data, CTX-1 in bone marrow plasma and gene expression of bone resorption markers (TNFα, TRAP, Cathepsin K) were significantly decreased in GPR109A-/- mice, while on the other hand, P1NP was increased in serum from both male and female GPR109A-/- mice compared to their respective controls. GPR109A deletion led to suppressed Wnt/β-catenin signaling in osteoclast precursors to inhibit osteoclast differentiation and activity. Indeed, HA and 3-3-PPA substantially inhibited RANKL-induced GPR109A expression and Wnt/β-catenin signaling in osteoclast precursors and osteoclast differentiation. Resultantly, HA significantly inhibited bone resorption and increased bone mass in wild type mice, but had no additional effects on bone in GPR109A-/- mice compared with their respective untreated control mice. These results suggest an important role for GPR109A during osteoclast differentiation and bone resorption mediating effects of HA and 3-3-PPA on inhibiting bone resorption during skeletal development.
Collapse
Affiliation(s)
- Jin-Ran Chen
- Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA.
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA.
| | - Haijun Zhao
- Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA
| | - Umesh D Wankhade
- Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA
| | - Sree V Chintapalli
- Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA
| | - Can Li
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA
| | - Dongzheng Gai
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA
| | - Kartik Shankar
- Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Fenghuang Zhan
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA
| | - Oxana P Lazarenko
- Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA
| |
Collapse
|
14
|
Chen J, Lazarenko OP, Zhao H, Wankhade UD, Pedersen K, Watt J, Ronis MJJ. Nox4 Expression Is Not Required for OVX-Induced Osteoblast Senescence and Bone Loss in Mice. JBMR Plus 2020; 4:e10376. [PMID: 32803108 PMCID: PMC7422714 DOI: 10.1002/jbm4.10376] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/04/2020] [Accepted: 05/09/2020] [Indexed: 12/12/2022] Open
Abstract
Estrogen deficiency and aging play critical roles in the pathophysiology of bone as a result of increased oxidative stress. It has been suggested that prevention of NADPH oxidase- (Nox-) dependent accumulation of ROS may be an approach to potentially minimize bone loss caused by these conditions. Using ovariectomized (OVX) and Nox4 gene-deletion mouse models, we investigated the role of Nox4 in OVX-induced bone loss and osteoblast senescence signaling. Six-month-old WT C57Bl6 mice were allocated to a sham control group, OVX, and OVX plus E2 treatment group for 8 weeks. Decreased bone mass including BMD and BMC were found in the OVX group compared with the sham control (p < 0.05); E2 treatment completely reversed OVX-induced bone loss. Interestingly, the prevention of OVX-induced bone loss by E2 was associated with the elimination of increased senescence signaling in bone osteoblastic cells from the OVX group. E2 blunted OVX-induced p53 and p21 overexpression, but not p16 and Nox4 in bone. In addition, 8- and 11-month-old Nox4 KO female mice were OVX for 8 weeks. Significant bone loss and increased bone osteoblastic cell senescence signaling occurred not only in Nox4 KO OVX mice compared with sham-operated animals, but also in 11-month-old Nox4 KO sham mice compared with 8-month-old Nox4 KO sham mice (p < 0.05). These data suggest that Nox4-mediated ROS in bone osteoblastic cells may be dispensable for sex steroid deficiency-induced bone loss and senescence. © 2020 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Jin‐Ran Chen
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockARUSA
- Arkansas Children's Nutrition CenterLittle RockARUSA
| | - Oxana P Lazarenko
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockARUSA
- Arkansas Children's Nutrition CenterLittle RockARUSA
| | - Haijun Zhao
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockARUSA
- Arkansas Children's Nutrition CenterLittle RockARUSA
| | - Umesh D Wankhade
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockARUSA
- Arkansas Children's Nutrition CenterLittle RockARUSA
| | - Kim Pedersen
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLAUSA
| | - James Watt
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLAUSA
| | - Martin J J Ronis
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLAUSA
| |
Collapse
|
15
|
Chen JR, Wankhade UD, Alund AW, Blackburn ML, Shankar K, Lazarenko OP. 3-(3-Hydroxyphenyl)-Propionic Acid (PPA) Suppresses Osteoblastic Cell Senescence to Promote Bone Accretion in Mice. JBMR Plus 2019; 3:e10201. [PMID: 31667457 PMCID: PMC6808226 DOI: 10.1002/jbm4.10201] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 04/18/2019] [Accepted: 04/23/2019] [Indexed: 12/24/2022] Open
Abstract
Phenolic acids (PAs) are metabolites derived from polyphenolic compounds found in fruits and vegetables resulting from the actions of gut bacteria. Previously, we reported that the levels of seven individual PAs were found to be at least 10 times higher in the serum of rats fed a blueberry (BB)‐containing diet compared to those fed a control diet. We have characterized the effects of one such BB‐associated serum PA, 3‐(3‐hydroxyphenyl)‐propionic acid (PPA), on senescence signaling and promotion of mesenchymal stem cell differentiation toward osteoblasts, while suppressing adipogenesis in the stem cells. To better understand the mechanistic actions of PPA on bone formation in vivo, we administered four doses of PPA (0.1, 0.5, 1, and 5 mg/kg/day; daily i.p.) to 1‐month‐old female C57BL6/J mice for 30 days. We did not observe significant effects of PPA on cortical bone; however, there were significantly higher bone volume and trabecular thickness and increased osteoblastic cell number, but decreased osteoclastic cell number in PPA‐treated groups compared to controls. These morphological and cellular outcomes of bone were reflected in changes of bone formation markers in serum and bone marrow plasma. PPA treatment reduced senescence signaling as evaluated by senescence‐associated β‐galactosidase activity, PPARγ, p53, and p21 expression in bone. In conclusion, PPA is capable of altering the mesenchymal stem cell differentiation program and bone cell senescence. This raises the possibility that BB‐rich diets promote bone growth through increasing systemic PAs, a question that merits additional investigation. © 2019 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Jin-Ran Chen
- Arkansas Children's Nutrition Center Little Rock AR USA.,Department of Pediatrics University of Arkansas for Medical Sciences Little Rock AR USA
| | - Umesh D Wankhade
- Arkansas Children's Nutrition Center Little Rock AR USA.,Department of Pediatrics University of Arkansas for Medical Sciences Little Rock AR USA
| | - Alexander W Alund
- Graduate Program in Interdisciplinary Biomedical Sciences University of Arkansas for Medical Sciences Little Rock AR USA
| | - Michael L Blackburn
- Arkansas Children's Nutrition Center Little Rock AR USA.,Department of Pediatrics University of Arkansas for Medical Sciences Little Rock AR USA
| | - Kartik Shankar
- Arkansas Children's Nutrition Center Little Rock AR USA.,Department of Pediatrics University of Arkansas for Medical Sciences Little Rock AR USA
| | - Oxana P Lazarenko
- Arkansas Children's Nutrition Center Little Rock AR USA.,Department of Pediatrics University of Arkansas for Medical Sciences Little Rock AR USA
| |
Collapse
|
16
|
Domazetovic V, Marcucci G, Pierucci F, Bruno G, Di Cesare Mannelli L, Ghelardini C, Brandi ML, Iantomasi T, Meacci E, Vincenzini MT. Blueberry juice protects osteocytes and bone precursor cells against oxidative stress partly through SIRT1. FEBS Open Bio 2019; 9:1082-1096. [PMID: 31006177 PMCID: PMC6551492 DOI: 10.1002/2211-5463.12634] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 03/08/2019] [Accepted: 03/29/2019] [Indexed: 12/17/2022] Open
Abstract
Oxidative stress and abnormal osteocyte apoptosis are often related to dysregulation of bone turnover and chronic bone loss, and so fruit and vegetables with high antioxidant potential may play an important role in the prevention and/or management of osteoporosis. Osteocytes are the main regulators of bone remodelling. For the first time, we demonstrate here that blueberry juice (BJ), obtained from Vaccinium myrtillus, rich in polyphenols, shows antioxidant and antiosteoclastogenic properties in MLO‐Y4 osteocytes. We report that BJ prevents oxidative stress‐induced apoptosis and reverses the increase in receptor activator of nuclear factor κB ligand and sclerostin expression, crucial factors for osteoclast activation and bone resorption. BJ is also able to prevent oxidative stress‐induced cell cytotoxicity in bone marrow mesenchymal stromal cells (MSCs), which are considered to be an important tool for cell therapy in bone disorders. No significant difference in preventing these events was observed between BJ and blueberry dry extract containing equal amounts of total soluble polyphenols. We have also shown that blueberry acts as both an antioxidant and an activator of sirtuin type 1, a class III histone deacetylase involved in cell death regulation and considered a molecular target for blocking bone resorption without affecting osteoclast survival. Overall, these novel data obtained in osteocytes and MSCs may help us clarify the mechanisms by which blueberry counteracts oxidative stress‐induced damage in bone remodelling and osteogenesis at the cellular and molecular level. Our findings are consistent with the reported beneficial effects of blueberry on bone tissue reported in animal studies, which suggest that blueberry may be a useful supplement for the prevention and/or management of osteoporosis and osteogenic process.
Collapse
Affiliation(s)
- Vladana Domazetovic
- Department of Biomedical Experimental and Clinical Sciences 'Mario Serio', University of Florence, Italy
| | - Gemma Marcucci
- Department of Biomedical Experimental and Clinical Sciences 'Mario Serio', University of Florence, Italy
| | - Federica Pierucci
- Department of Biomedical Experimental and Clinical Sciences 'Mario Serio', University of Florence, Italy
| | - Gennaro Bruno
- Department of Health Sciences, University of Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Pharmacology and Toxicology Section, University of Florence, Italy
| | - Maria Luisa Brandi
- Department of Biomedical Experimental and Clinical Sciences 'Mario Serio', University of Florence, Italy
| | - Teresa Iantomasi
- Department of Biomedical Experimental and Clinical Sciences 'Mario Serio', University of Florence, Italy
| | - Elisabetta Meacci
- Department of Biomedical Experimental and Clinical Sciences 'Mario Serio', University of Florence, Italy
| | - Maria Teresa Vincenzini
- Department of Biomedical Experimental and Clinical Sciences 'Mario Serio', University of Florence, Italy
| |
Collapse
|
17
|
Abstract
Intrauterine or early postnatal high-fat diet (HFD) has substantial influences on adult offspring health; however, studies of HFD-induced maternal obesity on regulation of adult offspring bone formation are sparse. Here, we investigated the effects of HFD-induced maternal obesity on both fetal and adult offspring skeletal development. We found that HFD-induced maternal obesity significantly decreased fetal skeletal development, but enhanced fetal osteoblastic cell senescence signaling and significantly increased the expression of inflammatory factors of the senescence-associated secretory phenotype (SASP) in osteo-progenitors. It was found that p300/CBP activation led to H3K27 acetylation to increase the expression of senescence-related genes and PPARγ in embryonic mouse osteogenic calvarial cells from HFD obese dams. These results were recapitulated in human umbilical cord mesenchymal stem cells (UC MSCs) isolated from offspring of pregnant obese and lean mothers following delivery. Regardless of postnatal HFD challenge, adult offspring from HFD obese dams showed significantly suppressed bone formation. Such early involution of bone formation of adult offspring from HFD obese dams may at least in part due to histone acetylation, i.e., epigenetic regulation of genes involved in cell senescence signaling in pre-osteoblasts from prenatal development. These findings indicate fetal pre-osteoblastic cell senescence signaling is epigenetically regulated by maternal obesity to repress bone formation in adult offspring in rodents and suggest that at least some of these effects may also manifest in humans.
Collapse
Affiliation(s)
- Jin-Ran Chen
- Arkansas Children’s Nutrition CenterLittle Rock, Arkansas, USA
- Department of PediatricsUniversity of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Correspondence should be addressed to J-R Chen:
| | - Oxana P Lazarenko
- Arkansas Children’s Nutrition CenterLittle Rock, Arkansas, USA
- Department of PediatricsUniversity of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Haijun Zhao
- Arkansas Children’s Nutrition CenterLittle Rock, Arkansas, USA
- Department of PediatricsUniversity of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Alexander W Alund
- Interdisciplinary Biomedical Sciences University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kartik Shankar
- Arkansas Children’s Nutrition CenterLittle Rock, Arkansas, USA
- Department of PediatricsUniversity of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
18
|
Domazetovic V, Marcucci G, Iantomasi T, Brandi ML, Vincenzini MT. Oxidative stress in bone remodeling: role of antioxidants. ACTA ACUST UNITED AC 2017; 14:209-216. [PMID: 29263736 DOI: 10.11138/ccmbm/2017.14.1.209] [Citation(s) in RCA: 447] [Impact Index Per Article: 63.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
ROS are highly reactive molecules which consist of a number of diverse chemical species, including radical and non-radical oxygen species. Oxidative stress occurs as a result of an overproduction of ROS not balanced by an adequate level of antioxidants. The natural antioxidants are: thiol compounds among which GSH is the most representative, and non-thiol compounds such as polyphenols, vitamins and also various enzymes. Many diseases have been linked to oxidative stress including bone diseases among which one of the most important is the osteoporosis. The redox state changes are also related to the bone remodeling process which allows the continuous bone regeneration through the coordinated action of bone cells: osteoclasts, osteoblasts and osteocytes. Changes in ROS and/or antioxidant systems seem to be involved in the pathogenesis of bone loss. ROS induce the apoptosis of osteoblasts and osteocytes, and this favours osteoclastogenesis and inhibits the mineralization and osteogenesis. Excessive osteocyte apoptosis correlates with oxidative stress causing an imbalance in favor of osteoclastogenesis which leads to increased turnover of bone remodeling and bone loss. Antioxidants either directly or by counteracting the action of oxidants contribute to activate the differentiation of osteoblasts, mineralization process and the reduction of osteoclast activity. In fact, a marked decrease in plasma antioxidants was found in aged or osteoporotic women. Some evidence shows a link among nutrients, antioxidant intake and bone health. Recent data demonstrate the antioxidant properties of various nutrients and their influence on bone metabolism. Polyphenols and anthocyanins are the most abundant antioxidants in the diet, and nutritional approaches to antioxidant strategies, in animals or selected groups of patients with osteoporosis or inflammatory bone diseases, suggest the antioxidant use in anti-resorptive therapies for the treatment and prevention of bone loss.
Collapse
Affiliation(s)
- Vladana Domazetovic
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" (Biochemistry section), University of Florence, Florence, Italy
| | - Gemma Marcucci
- Department of Surgery and Translational Medicine (Endocrinology Section), University of Florence, Florence, Italy
| | - Teresa Iantomasi
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" (Biochemistry section), University of Florence, Florence, Italy
| | - Maria Luisa Brandi
- Department of Surgery and Translational Medicine (Endocrinology Section), University of Florence, Florence, Italy
| | - Maria Teresa Vincenzini
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" (Biochemistry section), University of Florence, Florence, Italy
| |
Collapse
|
19
|
Jiao X, Li B, Zhang Q, Gao N, Zhang X, Meng X. Effect of
in vitro
‐simulated gastrointestinal digestion on the stability and antioxidant activity of blueberry polyphenols and their cellular antioxidant activity towards HepG2 cells. Int J Food Sci Technol 2017. [DOI: 10.1111/ijfs.13516] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Xinyao Jiao
- College of Food Science Shenyang Agricultural University Dongling Road 120 Shenhe District Shenyang China
| | - Bin Li
- College of Food Science Shenyang Agricultural University Dongling Road 120 Shenhe District Shenyang China
| | - Qi Zhang
- College of Food Science Shenyang Agricultural University Dongling Road 120 Shenhe District Shenyang China
| | - Ningxuan Gao
- College of Food Science Shenyang Agricultural University Dongling Road 120 Shenhe District Shenyang China
| | - Xiuyan Zhang
- College of Science Shenyang Agricultural University Dongling Road 120 Shenhe District Shenyang China
| | - Xianjun Meng
- College of Food Science Shenyang Agricultural University Dongling Road 120 Shenhe District Shenyang China
| |
Collapse
|
20
|
Bone-Protective Effects of Dried Plum in Postmenopausal Women: Efficacy and Possible Mechanisms. Nutrients 2017; 9:nu9050496. [PMID: 28505102 PMCID: PMC5452226 DOI: 10.3390/nu9050496] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 05/11/2017] [Accepted: 05/12/2017] [Indexed: 12/12/2022] Open
Abstract
Osteoporosis is an age-related chronic disease characterized by a loss of bone mass and quality, and is associated with an increased risk of fragility fractures. Postmenopausal women are at the greatest risk of developing osteoporosis due to the cessation in ovarian hormone production, which causes accelerated bone loss. As the demographic shifts to a more aged population, a growing number of postmenopausal women will be afflicted with osteoporosis. Certain lifestyle factors, including nutrition and exercise, are known to reduce the risk of developing osteoporosis and therefore play an important role in bone health. In terms of nutrition, accumulating evidence suggests that dried plum (Prunus domestica L.) is potentially an efficacious intervention for preventing and reversing bone mass and structural loss in an ovariectomized rat model of osteoporosis, as well as in osteopenic postmenopausal women. Here, we provide evidence supporting the efficacy of dried plum in preventing and reversing bone loss associated with ovarian hormone deficiency in rodent models and in humans. We end with the results of a recent follow-up study demonstrating that postmenopausal women who previously consumed 100 g dried plum per day during our one-year clinical trial conducted five years earlier retained bone mineral density to a greater extent than those receiving a comparative control. Additionally, we highlight the possible mechanisms of action by which bioactive compounds in dried plum exert bone-protective effects. Overall, the findings of our studies and others strongly suggest that dried plum in its whole form is a promising and efficacious functional food therapy for preventing bone loss in postmenopausal women, with the potential for long-lasting bone-protective effects.
Collapse
|
21
|
Minami A, Ogino M, Nakano N, Ichimura M, Nakanishi A, Murai T, Kitagishi Y, Matsuda S. Roles of oncogenes and tumor-suppressor genes in osteoclastogenesis (Review). Int J Mol Med 2017; 39:261-267. [DOI: 10.3892/ijmm.2017.2847] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 12/28/2016] [Indexed: 11/06/2022] Open
|
22
|
Chen JR, Lazarenko OP, Blackburn ML, Rose S, Frye RE, Badger TM, Andres A, Shankar K. Maternal Obesity Programs Senescence Signaling and Glucose Metabolism in Osteo-Progenitors From Rat and Human. Endocrinology 2016; 157:4172-4183. [PMID: 27653035 DOI: 10.1210/en.2016-1408] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Nutritional status during intrauterine and early postnatal life impacts the risk of chronic diseases, presumably via epigenetic mechanisms. However, evidence on the impact of gestational events on regulation of embryonic bone cell fate is sparse. We investigated the effects of maternal obesity on fetal osteoblast development in both rodents and humans. Female rats were fed control or an obesogenic high-fat diet (HFD) for 12 weeks and mated with male rats fed control diets, and respective maternal diets were continued during pregnancy. Embryonic rat osteogenic calvarial cells (EOCCs) were taken from gestational day 18.5 fetuses from control and HFD dams. EOCCs from HFD obese dams showed increases in p53/p21-mediated cell senescence signaling but decreased glucose metabolism. Decreased aerobic glycolysis in HFD-EOCCs was associated with decreased osteoblastic cell differentiation and proliferation. Umbilical cord human mesenchymal stem cells (MSCs) from 24 pregnant women (12 obese and 12 lean) along with placentas were collected upon delivery. The umbilical cord MSCs of obese mothers displayed less potential toward osteoblastogenesis and more towards adipogenesis. Human MSCs and placenta from obese mothers also exhibited increased cell senescence signaling, whereas MSCs showed decreased glucose metabolism and insulin resistance. Finally, we showed that overexpression of p53 linked increased cell senescence signaling and decreased glucose metabolism in fetal osteo-progenitors from obese rats and humans. These findings suggest programming of fetal preosteoblastic cell senescence signaling and glucose metabolism by maternal obesity.
Collapse
Affiliation(s)
- Jin-Ran Chen
- Arkansas Children's Nutrition Center (J.-R.C., O.P.L., M.L.B., T.M.B., A.A., K.S.), Department of Pediatrics (J.-R.C., O.P.L., M.L.B., R.E.F., T.M.B., A.A., K.S.), University of Arkansas for Medical Sciences, and Arkansas Children's Hospital Research Institute (S.R., R.E.F.), Little Rock, Arkansas 72202
| | - Oxana P Lazarenko
- Arkansas Children's Nutrition Center (J.-R.C., O.P.L., M.L.B., T.M.B., A.A., K.S.), Department of Pediatrics (J.-R.C., O.P.L., M.L.B., R.E.F., T.M.B., A.A., K.S.), University of Arkansas for Medical Sciences, and Arkansas Children's Hospital Research Institute (S.R., R.E.F.), Little Rock, Arkansas 72202
| | - Michael L Blackburn
- Arkansas Children's Nutrition Center (J.-R.C., O.P.L., M.L.B., T.M.B., A.A., K.S.), Department of Pediatrics (J.-R.C., O.P.L., M.L.B., R.E.F., T.M.B., A.A., K.S.), University of Arkansas for Medical Sciences, and Arkansas Children's Hospital Research Institute (S.R., R.E.F.), Little Rock, Arkansas 72202
| | - Shannon Rose
- Arkansas Children's Nutrition Center (J.-R.C., O.P.L., M.L.B., T.M.B., A.A., K.S.), Department of Pediatrics (J.-R.C., O.P.L., M.L.B., R.E.F., T.M.B., A.A., K.S.), University of Arkansas for Medical Sciences, and Arkansas Children's Hospital Research Institute (S.R., R.E.F.), Little Rock, Arkansas 72202
| | - Richard E Frye
- Arkansas Children's Nutrition Center (J.-R.C., O.P.L., M.L.B., T.M.B., A.A., K.S.), Department of Pediatrics (J.-R.C., O.P.L., M.L.B., R.E.F., T.M.B., A.A., K.S.), University of Arkansas for Medical Sciences, and Arkansas Children's Hospital Research Institute (S.R., R.E.F.), Little Rock, Arkansas 72202
| | - Thomas M Badger
- Arkansas Children's Nutrition Center (J.-R.C., O.P.L., M.L.B., T.M.B., A.A., K.S.), Department of Pediatrics (J.-R.C., O.P.L., M.L.B., R.E.F., T.M.B., A.A., K.S.), University of Arkansas for Medical Sciences, and Arkansas Children's Hospital Research Institute (S.R., R.E.F.), Little Rock, Arkansas 72202
| | - Aline Andres
- Arkansas Children's Nutrition Center (J.-R.C., O.P.L., M.L.B., T.M.B., A.A., K.S.), Department of Pediatrics (J.-R.C., O.P.L., M.L.B., R.E.F., T.M.B., A.A., K.S.), University of Arkansas for Medical Sciences, and Arkansas Children's Hospital Research Institute (S.R., R.E.F.), Little Rock, Arkansas 72202
| | - Kartik Shankar
- Arkansas Children's Nutrition Center (J.-R.C., O.P.L., M.L.B., T.M.B., A.A., K.S.), Department of Pediatrics (J.-R.C., O.P.L., M.L.B., R.E.F., T.M.B., A.A., K.S.), University of Arkansas for Medical Sciences, and Arkansas Children's Hospital Research Institute (S.R., R.E.F.), Little Rock, Arkansas 72202
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW The design and the development of functional foods is a key issue for bone health and a scientific challenge as well. As most studies have focused on calcium, and have paid less attention to other nutrients, our knowledge of the influence of nutrition on bone health remains limited. It has been well acknowledged that the human diet contains a wide and complex range of bioactive molecules endowed with interesting protective properties. In this context, and according to their high content in micronutrients, a growing body of evidence has enlightened the high nutritional value of berries. This review addresses the emerging interest in berries for bone health. RECENT FINDINGS Recent studies indicate that berry intakes are relevant to prevent osteopenia in humans. Their bone-sparing effects can be partly explained by their content in phytochemicals and vitamins. Beyond their antioxidant or anti-inflammatory functions, those micronutrients have been shown to modulate enzyme activities, cellular signaling pathways, and gene expression. SUMMARY Berry-enriched foods represent a relevant opportunity in the design of nutritional strategies targeting bone alteration.
Collapse
Affiliation(s)
- Marie-Jeanne Davicco
- aINRA, UMR 1019, Nutrition Humaine, CRNH Auvergne bClermont Université, Université d'Auvergne cEquipe Alimentation, Squelette et Métabolismes, Unité de Nutrition Humaine, UMR 1019, Centre INRA Auvergne Rhone Alpes, Clermont-Ferrand, France
| | | | | |
Collapse
|
24
|
Chen JR, Lazarenko OP, Blackburn ML, Shankar K. Dietary factors during early life program bone formation in female rats. FASEB J 2016; 31:376-387. [PMID: 27733448 DOI: 10.1096/fj.201600703r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 09/28/2016] [Indexed: 01/10/2023]
Abstract
Nutritional status during intrauterine and early postnatal life impacts the risk of chronic diseases; however, evidence for an association between early-life dietary factors and bone health in adults is limited. Soy protein isolate (SPI) may be one such dietary factor that promotes bone accretion during early life with persistent effects into adulthood. In the present study, we fed postnatal day (PND) 24 weanling female rats an SPI diet for 30 d [short-term SPI (ST-SPI)], and on PND 55, we switched SPI diet to control Cas diet until age 6 mo. Rats then underwent either ovariectomy (OVX) or sham surgery and thereafter either continued to be fed an SPI diet or control diet for 1 or 3 wk. We showed significantly increased bone mass in 30-d SPI-fed young rats compared with controls. OVX-induced bone loss was associated with increased osteoblastic cell senescence. On the one hand, both long-term SPI (continuous SPI diet throughout life) and ST-SPI diet only in early life protected against 1 wk post-OVX-associated bone loss. On the other hand, long-term SPI diet diminished the loss of total, trabecular, and cortical bone mineral density, whereas ST-SPI diet only reduced cortical bone mineral density loss 3 wk post-OVX. Persistent and protective effects of SPI diets on OVX-induced bone loss were associated with down-regulation of the caveolin-1/p53-mediated senescence pathway in bone. We recapitulated these results in cell cultures. Reprogramming of cellular senescence signaling by SPI-associated isoflavones in osteoblastic cells may explain the persistent effects of SPI on bone. These results suggest that OVX-induced bone loss, in part, is a result of increased osteoblastic cell senescence, and that ST-SPI diet early in life has modest but persistent programming effects on bone formation to prevent OVX-induced bone loss in adult female rats.-Chen, J.-R., Lazarenko, O. P., Blackburn, M. L., Shankar, K. Dietary factors during early life program bone formation in female rats.
Collapse
Affiliation(s)
- Jin-Ran Chen
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, USA; and .,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Oxana P Lazarenko
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, USA; and.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Michael L Blackburn
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, USA; and.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kartik Shankar
- Arkansas Children's Nutrition Center, Little Rock, Arkansas, USA; and.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
25
|
Chen JR, Lazarenko OP, Blackburn ML, Badger TM, Ronis MJJ. Soy protein isolate inhibits high-fat diet-induced senescence pathways in osteoblasts to maintain bone acquisition in male rats. Endocrinology 2015; 156:475-87. [PMID: 25490147 PMCID: PMC4298323 DOI: 10.1210/en.2014-1427] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Chronic consumption by experimental animals of a typical Western diet high in saturated fats and cholesterol during postnatal life has been demonstrated to impair skeletal development. However, the underlying mechanism by which high-fat, energy-dense diets affect bone-forming cell phenotypes is poorly understood. Here, we show that male weanling rats fed a diet containing 45% fat and 0.5% cholesterol made with casein (HF-Cas) for 6 weeks displayed lower bone mineral density and strength compared with those of AIN-93G-fed dietary controls. Substitution of casein with soy protein isolate (SPI) in the high-fat diet (HF-SPI) prevented these effects. The bone-sparing effects of SPI were associated with prevention of HF-Cas-induced osteoblast senescence pathways through suppression of the p53/p21 signaling pathways. HF-Cas-fed rats had increased caveolin-1 and down-regulated Sirt1, leading to activations of peroxisome proliferator-activated receptor γ (PPARγ) and p53/p21, whereas rats fed HF-SPI suppressed caveolin-1 and activated Sirt1 to deacetylate PPARγ and p53 in bone. Treatment of osteoblastic cells with nonesterified free fatty acid (NEFA) increased cell senescence signaling pathways. Isoflavones significantly blocked activations of senescence-associated β-galactosidase and PPARγ/p53/p21 by NEFA. Finally, replicative senescent osteoblastic cells and bone marrow mesenchymal ST2 cells exhibited behavior similar to that of cells treated with NEFA and in vivo bone cells in rats fed the HF-Cas diet. These results suggest that (1) high concentrations of NEFA occurring with HF intake are mediators of osteoblast cell senescence leading to impairment of bone development and acquisition and (2) the molecular mechanisms underlying the SPI-protective effects involve isoflavone-induced inhibition of osteoblastic cell senescence to prevent HF-induced bone impairments.
Collapse
Affiliation(s)
- Jin-Ran Chen
- Arkansas Children's Nutrition Center (J.-R.C., O.P.L, M.L.B., T.M.B., M.J.J.R.), Little Rock, Arkansas 72202; and Department of Pediatrics (J.-R.C., O.P.L, M.L.B., T.M.B., M.J.J.R.), University of Arkansas for Medical Sciences, Little Rock, Arkansas 72202
| | | | | | | | | |
Collapse
|
26
|
Artsi H, Cohen-Kfir E, Gurt I, Shahar R, Bajayo A, Kalish N, Bellido TM, Gabet Y, Dresner-Pollak R. The Sirtuin1 activator SRT3025 down-regulates sclerostin and rescues ovariectomy-induced bone loss and biomechanical deterioration in female mice. Endocrinology 2014; 155:3508-15. [PMID: 24949665 DOI: 10.1210/en.2014-1334] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Estrogen deficiency leads to rapid bone loss and skeletal fragility. Sclerostin, encoded by the sost gene, and a product of the osteocyte, is a negative regulator of bone formation. Blocking sclerostin increases bone mass and strength in animals and humans. Sirtuin1 (Sirt1), a player in aging and metabolism, regulates bone mass and inhibits sost expression by deacetylating histone 3 at its promoter. We asked whether a Sirt1-activating compound could rescue ovariectomy (OVX)-induced bone loss and biomechanical deterioration in 9-week-old C57BL/6 mice. OVX resulted in a substantial decrease in skeletal Sirt1 expression accompanied by an increase in sclerostin. Oral administration of SRT3025, a Sirt1 activator, at 50 and 100 mg/kg·d for 6 weeks starting 6 weeks after OVX fully reversed the deleterious effects of OVX on vertebral bone mass, microarchitecture, and femoral biomechanical properties. Treatment with SRT3025 decreased bone sclerostin expression and increased cortical periosteal mineralizing surface and serum propeptide of type I procollagen, a bone formation marker. In vitro, in the murine long bone osteocyte-Y4 osteocyte-like cell line SRT3025 down-regulated sclerostin and inactive β-catenin, whereas a reciprocal effect was observed with EX-527, a Sirt1 inhibitor. Sirt1 activation by Sirt1-activating compounds is a potential novel pathway to down-regulate sclerostin and design anabolic therapies for osteoporosis concurrently ameliorating other metabolic and age-associated conditions.
Collapse
Affiliation(s)
- Hanna Artsi
- Endocrinology and Metabolism Service (H.A., E.C.-K., I.G., R.D.-P.), Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; Laboratory of Bone Biomechanics (R.S., N.K.), Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, Rehovot, 76100 Israel; Bone Laboratory (A.B.), The Hebrew University of Jerusalem, Jerusalem, 91120 Israel; Department of Anatomy and Cell Biology (T.M.B.), Division of Endocrinology, Indiana University School of Medicine, Indianapolis, Indiana 46202; and Department of Anatomy and Anthropology (Y.G.), Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978 Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
The normal human chromosome complement consists of 46 chromosomes comprising 22 morphologically different pairs of autosomes and one pair of sex chromosomes. Variations in either chromosome number and/or structure frequently result in significant mental impairment and/or a variety of other clinical problems, among them, altered bone mass and strength. Chromosomal syndromes associated with specific chromosomal abnormalities are classified as either numerical or structural and may involve more than one chromosome. Aneuploidy refers to the presence of an extra copy of a specific chromosome, or trisomy, as seen in Down's syndrome (trisomy 21), or the absence of a single chromosome, or monosomy, as seen in Turner syndrome (a single X chromosome in females: 45, X). Aneuploidies have diverse phenotypic consequences, ranging from severe mental retardation and developmental abnormalities to increased susceptibility to various neoplasms and premature death. In fact, trisomy 21 is the prototypical aneuploidy in humans, is the most common genetic abnormality associated with longevity, and is one of the most widespread genetic causes of intellectual disability. In this review, the impact of trisomy 21 on the bone mass, architecture, skeletal health, and quality of life of people with Down syndrome will be discussed.
Collapse
Affiliation(s)
- Archana Kamalakar
- Department of Physiology & Biophysics, University of Arkansas for Medical Sciences, Little Rock, AR
- Department of Orthopaedic Surgery, Center for Orthopaedic Research, University of Arkansas for Medical Sciences, Little Rock, AR
| | - John R. Harris
- Department of Orthopaedic Surgery, Center for Orthopaedic Research, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Kent D. McKelvey
- Department of Genetics. University of Arkansas for Medical Sciences, Little Rock, AR
| | - Larry J. Suva
- Department of Physiology & Biophysics, University of Arkansas for Medical Sciences, Little Rock, AR
- Department of Orthopaedic Surgery, Center for Orthopaedic Research, University of Arkansas for Medical Sciences, Little Rock, AR
- Corresponding Author
| |
Collapse
|
28
|
Zhang J, Lazarenko OP, Blackburn ML, Badger TM, Ronis MJJ, Chen J. Soy protein isolate down‐regulates caveolin‐1 expression to suppress osteoblastic cell senescence pathways. FASEB J 2014; 28:3134-45. [DOI: 10.1096/fj.13-243659] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Jian Zhang
- Arkansas Children's Nutrition CenterLittle RockArkansasUSA
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Oxana P. Lazarenko
- Arkansas Children's Nutrition CenterLittle RockArkansasUSA
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Michael L. Blackburn
- Arkansas Children's Nutrition CenterLittle RockArkansasUSA
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Thomas M. Badger
- Arkansas Children's Nutrition CenterLittle RockArkansasUSA
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Martin J. J. Ronis
- Arkansas Children's Nutrition CenterLittle RockArkansasUSA
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
- Department of Pharmacology and ToxicologyUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Jin‐Ran Chen
- Arkansas Children's Nutrition CenterLittle RockArkansasUSA
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| |
Collapse
|
29
|
Feeding blueberry diets to young rats dose-dependently inhibits bone resorption through suppression of RANKL in stromal cells. PLoS One 2013; 8:e70438. [PMID: 23936431 PMCID: PMC3735613 DOI: 10.1371/journal.pone.0070438] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 06/18/2013] [Indexed: 11/19/2022] Open
Abstract
Previous studies have demonstrated that weanling rats fed AIN-93G semi-purified diets supplemented with 10% whole blueberry (BB) powder for two weeks beginning on postnatal day 21 (PND21) significantly increased bone formation at PND35. However, the minimal level of dietary BB needed to produce these effects is, as yet, unknown. The current study examined the effects of three different levels of BB diet supplementation (1, 3, and 5%) for 35 days beginning on PND25 on bone quality, and osteoclastic bone resorption in female rats. Peripheral quantitative CT scan (pQCT) of tibia, demonstrated that bone mineral density (BMD) and content (BMC) were dose-dependently increased in BB-fed rats compared to controls (P<0.05). Significantly increased bone mass after feeding 5% BB extracts was also observed in a TEN (total enteral nutrition) rat model in which daily caloric and food intake was precisely controlled. Expression of RANKL (receptor activator of nuclear factor-κB ligand) a protein essential for osteoclast formation was dose-dependently decreased in the femur of BB animals. In addition, expression of PPARγ (peroxisome proliferator-activated receptor γ) which regulates bone marrow adipogenesis was suppressed in BB diet rats compared to non-BB diet controls. Finally, a set of in vitro cell cultures revealed that the inhibitory effect of BB diet rat serum on RANKL expression was more profound in mesenchymal stromal cells compared to its effect on mature osteoblasts, pre-adipocytes and osteocytes. These results suggest that inhibition of bone resorption may contribute to increased bone mass during early development after BB consumption.
Collapse
|