1
|
Zillinger LS, Liesegang A, Hustedt K, Schnepel N, Sauerwein H, Schmicke M, Schwennen C, Muscher-Banse AS. Influence of N- and/or P-restriction on bone metabolism in young goats. Br J Nutr 2024; 132:874-886. [PMID: 39402760 PMCID: PMC11576093 DOI: 10.1017/s0007114524002150] [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: 02/22/2024] [Revised: 07/31/2024] [Accepted: 08/30/2024] [Indexed: 11/20/2024]
Abstract
Ruminants can recycle nitrogen (N) and phosphorus (P), which are essential for vital body processes. Reduced N- and P-intake in ruminants is desirable for economic and ecologic reasons. Simultaneous modulation of mineral homoeostasis and bone metabolism occurs in young goats. This study aimed to investigate potential effects of dietary N- and/or P-restriction on molecular changes in bone metabolism. The twenty-eight young male goats were fed a control diet, an N-reduced diet, a P-reduced diet or a combined N- and P-reduced diet for 6-8 weeks. The N-restricted goats had lower plasma Ca concentration and higher plasma osteocalcin (OC) and CrossLaps concentrations. The P-restricted goats had reduced plasma inorganic phosphate (Pi) concentrations and increased plasma Ca concentrations. Due to the initiation of a signalling pathway that inhibits the fibroblast growth factor 23 (FGF23) expression, this was lower with P-restriction. Consequently, lower Pi concentrations were the main factor influencing the reduction in FGF23. The changes in mineral homoeostasis associated with P-restriction led to a reduction in OC, bone mineral content and mineral density. Simultaneously, bone resorption potentially increased with P-restriction as indicated by an increased receptor activator of NF-κB ligand/osteoprotegerin (OPG) ratio and an increase in OPG mRNA expression. Additionally, the increased mRNA expression of the calcitonin receptor during P-restriction points to a higher number of osteoclasts. This study demonstrates an impairment of bone remodelling processes in young goats by N- or P-restriction. With P-restriction, bone mineralisation rate was potentially reduced and bone quality impaired, while with N-restriction, bone remodelling increased.
Collapse
Affiliation(s)
- Luisa S Zillinger
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Foundation, Hannover30173, Germany
| | - Annette Liesegang
- Institute of Animal Nutrition and Dietetics, Vetsuisse Faculty Zurich, University of Zurich, Zurich8057, Switzerland
- Center for Applied Biotechnology and Molecular Medicine (CABMM), Zurich, Switzerland
| | - Karin Hustedt
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Foundation, Hannover30173, Germany
| | - Nadine Schnepel
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Foundation, Hannover30173, Germany
| | - Helga Sauerwein
- Institute of Animal Science, Physiology Unit, University of Bonn, Bonn53115, Germany
| | - Marion Schmicke
- Clinic for Diseases of Cattle, University of Veterinary Medicine Hannover Foundation, Hannover30173, Germany
| | - Cornelia Schwennen
- Institute for Animal Nutrition, University of Veterinary Medicine Hannover Foundation, Hannover30173, Germany
| | - Alexandra S Muscher-Banse
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Foundation, Hannover30173, Germany
| |
Collapse
|
2
|
Li S, Yan W, Sun K, Miao J, Liu Z, Xu J, Wang X, Li B, Zhang Q. Norisoboldine, a Natural Alkaloid from Lindera aggregata (Sims) Kosterm, Promotes Osteogenic Differentiation via S6K1 Signaling Pathway and Prevents Bone Loss in OVX Mice. Mol Nutr Food Res 2024; 68:e2400193. [PMID: 38813717 DOI: 10.1002/mnfr.202400193] [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: 03/12/2024] [Revised: 05/06/2024] [Indexed: 05/31/2024]
Abstract
SCOPE Norisoboldine (NOR) is a major isoquinoline alkaloid component in the traditional Chinese herbal plant Lindera aggregata (Sims) Kosterm, with previously reported anti-osteoclast differentiation and antiarthritis properties. However, the roles of NOR on osteoblasts, bone marrow mesenchymal stem cells (BMSCs), and osteoporosis in vivo have never been well established. METHODS AND RESULTS This study investigates the ability of NOR to improve bone formation in vitro and in vivo. Osteoblasts and BMSCs are used to study the effect of NOR on osteogenic and adipogenic differentiation. It finds that NOR promotes osteogenic differentiation of osteoblasts and BMSCs, while inhibiting adipogenic differentiation of BMSCs by reducing the relative expression of peroxisome proliferator-activated receptor γ (Ppar-γ) and adiponectin, C1Q and collagen domain containing (Adipoq). Mechanistic studies show that NOR increases osteoblast differentiation through the mechanistic target of rapamycin kinase (mTOR)/ribosomal protein S6 kinase; polypeptide 1 (S6K1) pathway, and treatment with an mTOR inhibitor rapamycin blocked the NOR-induced increase in mineral accumulation. Finally, the study evaluates the therapeutic potential of NOR in a mouse model of ovariectomy (OVX)-induced bone loss. NOR prevents bone loss in both trabecular and cortical bone by increasing osteoblast number and phospho-S6K1 (p-S6K1) expression in osteoblasts. CONCLUSION NOR effects in enhancing osteoblast-induced bone formation via S6K1 pathway, suggesting the potential of NOR in osteoporosis treatment by increasing bone formation.
Collapse
Affiliation(s)
- Shiming Li
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100193, China
- Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| | - Wenliang Yan
- Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| | - Kainong Sun
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100193, China
- Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| | - Jingyuan Miao
- Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| | - Zichao Liu
- Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| | - Jiayang Xu
- Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| | - Xiaoyu Wang
- Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| | - Bo Li
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100193, China
| | - Qian Zhang
- Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| |
Collapse
|
3
|
Wu M, Wu S, Chen W, Li YP. The roles and regulatory mechanisms of TGF-β and BMP signaling in bone and cartilage development, homeostasis and disease. Cell Res 2024; 34:101-123. [PMID: 38267638 PMCID: PMC10837209 DOI: 10.1038/s41422-023-00918-9] [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: 02/26/2023] [Accepted: 12/15/2023] [Indexed: 01/26/2024] Open
Abstract
Transforming growth factor-βs (TGF-βs) and bone morphometric proteins (BMPs) belong to the TGF-β superfamily and perform essential functions during osteoblast and chondrocyte lineage commitment and differentiation, skeletal development, and homeostasis. TGF-βs and BMPs transduce signals through SMAD-dependent and -independent pathways; specifically, they recruit different receptor heterotetramers and R-Smad complexes, resulting in unique biological readouts. BMPs promote osteogenesis, osteoclastogenesis, and chondrogenesis at all differentiation stages, while TGF-βs play different roles in a stage-dependent manner. BMPs and TGF-β have opposite functions in articular cartilage homeostasis. Moreover, TGF-β has a specific role in maintaining the osteocyte network. The precise activation of BMP and TGF-β signaling requires regulatory machinery at multiple levels, including latency control in the matrix, extracellular antagonists, ubiquitination and phosphorylation in the cytoplasm, nucleus-cytoplasm transportation, and transcriptional co-regulation in the nuclei. This review weaves the background information with the latest advances in the signaling facilitated by TGF-βs and BMPs, and the advanced understanding of their diverse physiological functions and regulations. This review also summarizes the human diseases and mouse models associated with disordered TGF-β and BMP signaling. A more precise understanding of the BMP and TGF-β signaling could facilitate the development of bona fide clinical applications in treating bone and cartilage disorders.
Collapse
Affiliation(s)
- Mengrui Wu
- Department of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Shali Wu
- Department of Cell and Developmental Biology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, LA, USA.
| |
Collapse
|
4
|
Ko FC, Jochum SB, Wilson BM, Adra A, Patel N, Lee H, Wilber S, Shaikh M, Forsyth C, Keshavarzian A, Swanson GR, Sumner DR. Colon epithelial cell-specific Bmal1 deletion impairs bone formation in mice. Bone 2023; 168:116650. [PMID: 36584784 PMCID: PMC9911378 DOI: 10.1016/j.bone.2022.116650] [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: 10/12/2022] [Revised: 12/16/2022] [Accepted: 12/17/2022] [Indexed: 12/29/2022]
Abstract
The circadian clock system regulates multiple metabolic processes, including bone metabolism. Previous studies have demonstrated that both central and peripheral circadian signaling regulate skeletal growth and homeostasis in mice. Disruption in central circadian rhythms has been associated with a decline in bone mineral density in humans and the global and osteoblast-specific disruption of clock genes in bone tissue leads to lower bone mass in mice. Gut physiology is highly sensitive to circadian disruption. Since the gut is also known to affect bone remodeling, we sought to test the hypothesis that circadian signaling disruption in colon epithelial cells affects bone. We therefore assessed structural, functional, and cellular properties of bone in 8 week old Ts4-Cre and Ts4-Cre;Bmal1fl/fl (cBmalKO) mice, where the clock gene Bmal1 is deleted in colon epithelial cells. Axial and appendicular trabecular bone volume was significantly lower in cBmalKO compared to Ts4-Cre 8-week old mice in a sex-dependent fashion, with male but not female mice showing the phenotype. Similarly, the whole bone mechanical properties were deteriorated in cBmalKO male mice. The tissue level mechanisms involved suppressed bone formation with normal resorption, as evidenced by serum markers and dynamic histomorphometry. Our studies demonstrate that colon epithelial cell-specific deletion of Bmal1 leads to failure to acquire trabecular and cortical bone in male mice.
Collapse
Affiliation(s)
- Frank C Ko
- Department of Anatomy& Cell Biology, Rush University Medical Center, Chicago, IL 60612, United States of America; Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612, United States of America; Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, United States of America.
| | - Sarah B Jochum
- Division of Digestive Diseases and Nutrition, Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612, United States of America
| | - Brittany M Wilson
- Department of Anatomy& Cell Biology, Rush University Medical Center, Chicago, IL 60612, United States of America
| | - Amal Adra
- Department of Anatomy& Cell Biology, Rush University Medical Center, Chicago, IL 60612, United States of America
| | - Nikhil Patel
- Department of Anatomy& Cell Biology, Rush University Medical Center, Chicago, IL 60612, United States of America
| | - Hoomin Lee
- Department of Anatomy& Cell Biology, Rush University Medical Center, Chicago, IL 60612, United States of America
| | - Sherry Wilber
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, United States of America
| | - Maliha Shaikh
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, United States of America
| | - Christopher Forsyth
- Department of Anatomy& Cell Biology, Rush University Medical Center, Chicago, IL 60612, United States of America; Division of Digestive Diseases and Nutrition, Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612, United States of America; Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, United States of America
| | - Ali Keshavarzian
- Department of Anatomy& Cell Biology, Rush University Medical Center, Chicago, IL 60612, United States of America; Division of Digestive Diseases and Nutrition, Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612, United States of America; Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, United States of America
| | - Garth R Swanson
- Department of Anatomy& Cell Biology, Rush University Medical Center, Chicago, IL 60612, United States of America; Division of Digestive Diseases and Nutrition, Department of Internal Medicine, Rush University Medical Center, Chicago, IL 60612, United States of America; Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, United States of America
| | - D Rick Sumner
- Department of Anatomy& Cell Biology, Rush University Medical Center, Chicago, IL 60612, United States of America; Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612, United States of America; Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL 60612, United States of America
| |
Collapse
|
5
|
Hussein AI, Carroll D, Bui M, Wolff A, Matheny H, Hogue B, Lybrand K, Cooke M, Bragdon B, Morgan E, Demissie S, Gerstenfeld L. Oxidative metabolism is impaired by phosphate deficiency during fracture healing and is mechanistically related to BMP induced chondrocyte differentiation. Bone Rep 2023. [DOI: 10.1016/j.bonr.2023.101657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
|
6
|
Wang G, Li H, Hou Y. LncRNA MAGI2-AS3 inhibits tumor progression and angiogenesis by regulating ACY1 via interacting with transcription factor HEY1 in clear cell renal cell carcinoma. Cancer Gene Ther 2022; 29:585-596. [PMID: 34002044 DOI: 10.1038/s41417-021-00339-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 03/22/2021] [Accepted: 04/06/2021] [Indexed: 02/02/2023]
Abstract
Clear cell renal cell carcinoma (ccRCC) represents the most common type of RCC in adults, characterized by hyper-vascularization and metastatic relapse. Surgical resection is the main treatment due to poor response of ccRCC to radio-and chemotherapy. However, the high complexity of tumor vasculature in ccRCC has thwarted effects to develop new therapeutic strategies for ccRCC. In this study, we identify the anti-angiogenic activity of MAGI2-AS3 in ccRCC. 86 paired samples of tumor tissues and adjacent no-tumor tissues were collected from ccRCC patients. Dual-luciferase reporter assay, RIP, and ChIP assays were employed to confirm interactions between MAGI2-AS3, transcription factor HEY1, and the ACY1 gene. In other studies, we assayed human ccRCC cells RLC-310 for their viability, migration and invasion using CCK-8 detection and transwell chamber systems. Angiogenesis was evaluated in the Matrigel-based human umbilical vein endothelial cell (HUVEC)-RLC-310 coculture model and immunohistochemical staining for vascular endothelial growth factor (VEGF) and CD31 in tumor tissues collected from a xenograft ccRCC mouse model. MAGI2-AS3 and ACY1 expression was downregulated in ccRCC tissues, and low expression of MAGI2-AS3 was associated with poor patient survival. Overexpression of MAGI2-AS3 could reduce ccRCC cell viability and migration, inhibit vessel-like tube formation of HUVECs in vitro, and repress tumor growth and angiogenesis in vivo. MAGI2-AS3 bound with HEY1 and reduced the HEY1 enrichment at the ACY1 promoter region, thus increasing ACY1 gene transcription. HEY1 knockdown or ACY1 overexpression that resisted MAGI2-AS3 knockdown was found in the in vivo and in vitro settings. The present study demonstrates that MAGI2-AS3 exerts tumor-suppressive, anti-angiogenic activities in ccRCC by modulating the HEY1/ACY1 pathway, thus lending support for conducting further investigations of anti-angiogenesis therapy for ccRCC.
Collapse
Affiliation(s)
- Guanbo Wang
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, People's Republic of China
| | - Hai Li
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, People's Republic of China
| | - Yi Hou
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, People's Republic of China.
| |
Collapse
|
7
|
Rendina-Ruedy E, Smith BJ. Common Dietary Modifications in Preclinical Models to Study Skeletal Health. Front Endocrinol (Lausanne) 2022; 13:932343. [PMID: 35909523 PMCID: PMC9329513 DOI: 10.3389/fendo.2022.932343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 05/27/2022] [Indexed: 12/03/2022] Open
Abstract
Bone is a highly dynamic tissue that undergoes continuous remodeling by bone resorbing osteoclasts and bone forming osteoblasts, a process regulated in large part by osteocytes. Dysregulation of these coupled catabolic and anabolic processes as in the case of menopause, type 2 diabetes mellitus, anorexia nervosa, and chronic kidney disease is known to increase fracture risk. Recent advances in the field of bone cell metabolism and bioenergetics have revealed that maintenance of the skeleton places a high energy demand on these cells involved in bone remodeling. These new insights highlight the reason that bone tissue is the beneficiary of a substantial proportion of cardiac output and post-prandial chylomicron remnants and requires a rich supply of nutrients. Studies designed for the specific purpose of investigating the impact of dietary modifications on bone homeostasis or that alter diet composition and food intake to produce the model can be found throughout the literature; however, confounding dietary factors are often overlooked in some of the preclinical models. This review will examine some of the common pre-clinical models used to study skeletal biology and its pathologies and the subsequent impact of various dietary factors on these model systems. Furthermore, the review will include how inadvertent effects of some of these dietary components can influence bone cell function and study outcomes.
Collapse
Affiliation(s)
- Elizabeth Rendina-Ruedy
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, United States
- *Correspondence: Elizabeth Rendina-Ruedy,
| | - Brenda J. Smith
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
8
|
Frangi G, Guicheteau M, Jacquot F, Pyka G, Kerckhofs G, Feyeux M, Veziers J, Guihard P, Halgand B, Sourice S, Guicheux J, Prieur X, Beck L, Beck-Cormier S. PiT2 deficiency prevents increase of bone marrow adipose tissue during skeletal maturation but not in OVX-induced osteoporosis. Front Endocrinol (Lausanne) 2022; 13:921073. [PMID: 36465661 PMCID: PMC9708882 DOI: 10.3389/fendo.2022.921073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 10/24/2022] [Indexed: 11/17/2022] Open
Abstract
The common cellular origin between bone marrow adipocytes (BMAds) and osteoblasts contributes to the intimate link between bone marrow adipose tissue (BMAT) and skeletal health. An imbalance between the differentiation ability of BMSCs towards one of the two lineages occurs in conditions like aging or osteoporosis, where bone mass is decreased. Recently, we showed that the sodium-phosphate co-transporter PiT2/SLC20A2 is an important determinant for bone mineralization, strength and quality. Since bone mass is reduced in homozygous mutant mice, we investigated in this study whether the BMAT was also affected in PiT2-/- mice by assessing the effect of the absence of PiT2 on BMAT volume between 3 and 16 weeks, as well as in an ovariectomy-induced bone loss model. Here we show that the absence of PiT2 in juveniles leads to an increase in the BMAT that does not originate from an increased adipogenic differentiation of bone marrow stromal cells. We show that although PiT2-/- mice have higher BMAT volume than control PiT2+/+ mice at 3 weeks of age, BMAT volume do not increase from 3 to 16 weeks of age, leading to a lower BMAT volume in 16-week-old PiT2-/- compared to PiT2+/+ mice. In contrast, the absence of PiT2 does not prevent the increase in BMAT volume in a model of ovariectomy-induced bone loss. Our data identify SLC20a2/PiT2 as a novel gene essential for the maintenance of the BMAd pool in adult mice, involving mechanisms of action that remain to be elucidated, but which appear to be independent of the balance between osteoblastic and adipogenic differentiation of BMSCs.
Collapse
Affiliation(s)
- Giulia Frangi
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
| | - Marie Guicheteau
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
| | - Frederic Jacquot
- Nantes Université, CHU Nantes, Inserm, CNRS, CRCI2NA, Nantes, France
| | - Grzegorz Pyka
- Biomechanics lab, Institute of Mechanics, Materials, and Civil Engineering, UC Louvain, Louvain-la-Neuve, Belgium
- Department of Materials Engineering, KU Leuven, Leuven, Belgium
| | - Greet Kerckhofs
- Biomechanics lab, Institute of Mechanics, Materials, and Civil Engineering, UC Louvain, Louvain-la-Neuve, Belgium
- Department of Materials Engineering, KU Leuven, Leuven, Belgium
- IREC, Institute of Experimental and Clinical Research, UC Louvain, Woluwé-Saint-Lambert, Belgium
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, Leuven, Belgium
| | - Magalie Feyeux
- Nantes Université, CHU Nantes, CNRS, Inserm, BioCore, US16, SFR Bonamy, Nantes, France
| | - Joëlle Veziers
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
| | - Pierre Guihard
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
| | - Boris Halgand
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
| | - Sophie Sourice
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
| | - Jérôme Guicheux
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
| | - Xavier Prieur
- Nantes Université, CNRS, Inserm, l’Institut du Thorax, Nantes, France
| | - Laurent Beck
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
| | - Sarah Beck-Cormier
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, SFR Bonamy, Nantes, France
- *Correspondence: Sarah Beck-Cormier,
| |
Collapse
|
9
|
Ko FC, Kobelski MM, Zhang W, Grenga GM, Martins JS, Demay MB. Phosphate restriction impairs mTORC1 signaling leading to increased bone marrow adipose tissue and decreased bone in growing mice. J Bone Miner Res 2021; 36:1510-1520. [PMID: 33900666 DOI: 10.1002/jbmr.4312] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 04/14/2021] [Accepted: 04/17/2021] [Indexed: 01/08/2023]
Abstract
Bone marrow stromal cells (BMSCs) are multipotent cells that differentiate into cells of the osteogenic and adipogenic lineage. A striking inverse relationship between bone marrow adipose tissue (BMAT) and bone volume is seen in several conditions, suggesting that differentiation of BMSCs into bone marrow adipocytes diverts cells from the osteogenic lineage, thereby compromising the structural and mechanical properties of bone. Phosphate restriction of growing mice acutely decreases bone formation, blocks osteoblast differentiation and increases BMAT. Studies performed to evaluate the cellular and molecular basis for the effects of acute phosphate restriction demonstrate that it acutely increases 5' adenosine monophosphate-activated protein kinase (AMPK) phosphorylation and inhibits mammalian target of rapamycin complex 1 (mTORC1) signaling in osteoblasts. This is accompanied by decreased expression of Wnt10b in BMSCs. Phosphate restriction also promotes expression of the key adipogenic transcription factors, peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT-enhancer binding protein α (CEBPα), in CXCL12 abundant reticular (CAR) cells, which represent undifferentiated BMSCs and are the main source of BMAT and osteoblasts in the adult murine skeleton. Consistent with this, lineage tracing studies reveal that the BMAT observed in phosphate-restricted mice is of CAR cell origin. To determine whether circumventing the decrease in mTORC1 signaling in maturing osteoblasts attenuates the osteoblast and BMAT phenotype, phosphate-restricted mice with OSX-CreERT2 -mediated haploinsufficiency of the mTORC1 inhibitor, TSC2, were generated. TSC2 haploinsufficiency in preosteoblasts/osteoblasts normalized bone volume and osteoblast number in phosphate-restricted mice and attenuated the increase in BMAT observed. Thus, acute phosphate restriction leads to decreased bone and increases BMAT by impairing mTORC1 signaling in osterix-expressing cells. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Frank C Ko
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | | | - Wanlin Zhang
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Gina M Grenga
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Janaina S Martins
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Marie B Demay
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
10
|
Wang H, Wei P, Zhang Y, Li Y, Yin L. LncRNA TCONS_00023297 Regulates the Balance of Osteogenic and Adipogenic Differentiation in Bone Marrow Mesenchymal Stem Cells and the Coupling Process of Osteogenesis and Angiogenesis. Front Cell Dev Biol 2021; 9:697858. [PMID: 34262909 PMCID: PMC8274487 DOI: 10.3389/fcell.2021.697858] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 05/27/2021] [Indexed: 12/01/2022] Open
Abstract
Long noncoding RNA (lncRNA) is a noncoding RNA with a length of more than 200 bases. It plays an important role in the occurrence and development of diseases. Research on lncRNAs has received increasing attention. Bone is an important organ of the human body. As the population ages, the incidence of osteoporosis gradually increases. The mechanism of action of lncRNAs in the development of osteoporosis is unclear. The imbalance between osteogenic and adipogenic differentiation in bone marrow mesenchymal stem cells (hBMSCs) and the coupling process of osteogenesis and angiogenesis plays an important role in the development of osteoporosis. Therefore, this study focused on the mechanism by which lncRNAs regulate the osteogenic differentiation of bone marrow mesenchymal stem cells and the mechanism of action of lncRNAs in bone metabolism. The expression of lncRNAs in the osteogenic differentiation of hBMSCs was detected by lncRNA microarray. Real-time quantitative PCR was used to detect the expression changes of lncRNA and osteogenic genes during hBMSC osteogenic and adipogenic differentiation. The ceRNA mechanisms were detected by RIP and luciferase reporter gene assays. The effect of lncRNAs on the osteogenesis–angiogenesis coupling process was detected by Transwell assays. TCONS_00023297 increased expression during osteogenic differentiation; TCONS_00023297 overexpression promoted osteogenic differentiation of hBMSCs; BMP2 regulated TCONS_00023297 expression in a concentration- and time-dependent manner; TCONS_00023297 regulated miR-608 via a ceRNA mechanism; TCONS_00023297 inhibited hBMSC adipogenic differentiation; and TCONS_00023297 promoted VEGF secretion by hBMSCs. TCONS_00023297 regulates osteogenic differentiation, adipogenic differentiation, and osteogenic–angiogenic coupling of hBMSCs via the TCONS_00023297/miR-608/RUNX2/SHH signaling axis.
Collapse
Affiliation(s)
- Haitao Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Peng Wei
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuebai Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences of Zhengzhou University, Zhengzhou, China
| | - Li Yin
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Yuan Y, Jagga S, Martins JS, Rana R, Pajevic PD, Liu ES. Impaired 1,25 dihydroxyvitamin D3 action and hypophosphatemia underlie the altered lacuno-canalicular remodeling observed in the Hyp mouse model of XLH. PLoS One 2021; 16:e0252348. [PMID: 34043707 PMCID: PMC8158930 DOI: 10.1371/journal.pone.0252348] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/12/2021] [Indexed: 11/28/2022] Open
Abstract
Osteocytes remodel the perilacunar matrix and canaliculi. X-linked hypophosphatemia (XLH) is characterized by elevated serum levels of fibroblast growth factor 23 (FGF23), leading to decreased 1,25 dihydroxyvitamin D3 (1,25D) production and hypophosphatemia. Bones from mice with XLH (Hyp) have enlarged osteocyte lacunae, enhanced osteocyte expression of genes of bone remodeling, and impaired canalicular structure. The altered lacuno-canalicular (LCN) phenotype is improved with 1,25D or anti-FGF23 antibody treatment, pointing to roles for 1,25D and/or phosphate in regulating this process. To address whether impaired 1,25D action results in LCN alterations, the LCN phenotype was characterized in mice lacking the vitamin D receptor (VDR) in osteocytes (VDRf/f;DMP1Cre+). Mice lacking the sodium phosphate transporter NPT2a (NPT2aKO) have hypophosphatemia and high serum 1,25D levels, therefore the LCN phenotype was characterized in these mice to determine if increased 1,25D compensates for hypophosphatemia in regulating LCN remodeling. Unlike Hyp mice, neither VDRf/f;DMP1Cre+ nor NPT2aKO mice have dramatic alterations in cortical microarchitecture, allowing for dissecting 1,25D and phosphate specific effects on LCN remodeling in tibial cortices. Histomorphometric analyses demonstrate that, like Hyp mice, tibiae and calvariae in VDRf/f;DMP1Cre+ and NPT2aKO mice have enlarged osteocyte lacunae (tibiae: 0.15±0.02μm2(VDRf/f;DMP1Cre-) vs 0.19±0.02μm2(VDRf/f;DMP1Cre+), 0.12±0.02μm2(WT) vs 0.18±0.0μm2(NPT2aKO), calvariae: 0.09±0.02μm2(VDRf/f;DMP1Cre-) vs 0.11±0.02μm2(VDRf/f;DMP1Cre+), 0.08±0.02μm2(WT) vs 0.13±0.02μm2(NPT2aKO), p<0.05 all comparisons) and increased immunoreactivity of bone resorption marker Cathepsin K (Ctsk). The osteocyte enriched RNA isolated from tibiae in VDRf/f;DMP1Cre+ and NPT2aKO mice have enhanced expression of matrix resorption genes that are classically expressed by osteoclasts (Ctsk, Acp5, Atp6v0d2, Nhedc2). Treatment of Ocy454 osteocytes with 1,25D or phosphate inhibits the expression of these genes. Like Hyp mice, VDRf/f;DMP1Cre+ and NPT2aKO mice have impaired canalicular organization in tibia and calvaria. These studies demonstrate that hypophosphatemia and osteocyte-specific 1,25D actions regulate LCN remodeling. Impaired 1,25D action and low phosphate levels contribute to the abnormal LCN phenotype observed in XLH.
Collapse
Affiliation(s)
- Ye Yuan
- Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Endocrinology, Diabetes, Hypertension, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Supriya Jagga
- Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Endocrinology, Diabetes, Hypertension, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Janaina S. Martins
- Harvard Medical School, Boston, Massachusetts, United States of America
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Rakshya Rana
- Division of Endocrinology, Diabetes, Hypertension, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Paola Divieti Pajevic
- Department of Translational Dental Medicine, Boston University School of Dental Medicine, Boston, Massachusetts, United States of America
| | - Eva S. Liu
- Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Endocrinology, Diabetes, Hypertension, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
12
|
You M, Zhang L, Zhang X, Fu Y, Dong X. MicroRNA-197-3p Inhibits the Osteogenic Differentiation in Osteoporosis by Down-Regulating KLF 10. Clin Interv Aging 2021; 16:107-117. [PMID: 33469278 PMCID: PMC7810594 DOI: 10.2147/cia.s269171] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/03/2020] [Indexed: 12/19/2022] Open
Abstract
Background Studies have shown that microRNA (miRNA) regulates gene expression of osteoporosis (OS). It is known that miR-197-3p is abnormally expressed in osteoporosis. This study is to investigate the mechanism of miR-197-3p in regulating osteoblast differentiation. Methods Rats were ovariectomized to establish an animal model of postmenopausal osteoporosis. The expression of miR-197-3p and KLF10 was detected in ovariectomized rat models. Primary osteoblasts and MC3T-E1 cells were divided into the control group, miR-197-3p inhibitor group, NC inhibitor group and miR-197-3p inhibitor + si-KLF10 group. The expression of miR-197-3p and Kruppel-like factor 10 (KLF10) was detected by qRT-PCR and Western blot. The relationship between miR-197-3p and KLF10 was analyzed by bioinformatics and luciferase reporter assay. Cell viability was evaluated by MTT assay. The ALP activity measurement and mineralization analysis were performed. Results The expression of miR-197-3p was significantly raised in ovariectomized osteoporosis rats. During the differentiation of osteoblasts, the expression of miR-197-3p was significantly decreased, while the expression of KLF10 was significantly raised in primary osteoblasts and MC3E3T1 cells. The expression of RUNX2, ALP, OCN and OSX in miR-197-3p inhibitor group and MC3T3-E1 group was significantly raised, and the cell survival rate and mineralized nodule were raised as well. KLF10 may be the downstream target gene of miR-197-3p. After co-transfection of miR-197-3p inhibitor and si-klf10, ALP, Runx2, OCN and OSX mRNA, cell survival rate and mineralized nodule were significantly decreased in primary osteoblasts and MC3T3-E1 cells. Conclusion MiR-197-3p Inhibition promoted osteoblast differentiation and reduced OS by up-regulating KLF10.
Collapse
Affiliation(s)
- Murong You
- Department of Orthopedics, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi Province 330006, People's Republic of China
| | - Liang Zhang
- Department of Orthopedics, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi Province 330006, People's Republic of China
| | - Xiaoxiang Zhang
- Department of Orthopedics, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi Province 330006, People's Republic of China
| | - Yang Fu
- Department of Orthopedics, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi Province 330006, People's Republic of China
| | - Xieping Dong
- Department of Orthopedics, Jiangxi Provincial People's Hospital, Nanchang, Jiangxi Province 330006, People's Republic of China
| |
Collapse
|
13
|
Little-Letsinger SE, Pagnotti GM, McGrath C, Styner M. Exercise and Diet: Uncovering Prospective Mediators of Skeletal Fragility in Bone and Marrow Adipose Tissue. Curr Osteoporos Rep 2020; 18:774-789. [PMID: 33068251 PMCID: PMC7736569 DOI: 10.1007/s11914-020-00634-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/29/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW To highlight recent basic, translational, and clinical works demonstrating exercise and diet regulation of marrow adipose tissue (MAT) and bone and how this informs current understanding of the relationship between marrow adiposity and musculoskeletal health. RECENT FINDINGS Marrow adipocytes accumulate in the bone in the setting of not only hypercaloric intake (calorie excess; e.g., diet-induced obesity) but also with hypocaloric intake (calorie restriction; e.g., anorexia), despite the fact that these states affect bone differently. With hypercaloric intake, bone quantity is largely unaffected, whereas with hypocaloric intake, bone quantity and quality are greatly diminished. Voluntary running exercise in rodents was found to lower MAT and promote bone in eucaloric and hypercaloric states, while degrading bone in hypocaloric states, suggesting differential modulation of MAT and bone, dependent upon whole-body energy status. Energy status alters bone metabolism and bioenergetics via substrate availability or excess, which plays a key role in the response of bone and MAT to mechanical stimuli. Marrow adipose tissue (MAT) is a fat depot with a potential role in-as well as responsivity to-whole-body energy metabolism. Understanding the localized function of this depot in bone cell bioenergetics and substrate storage, principally in the exercised state, will aid to uncover putative therapeutic targets for skeletal fragility.
Collapse
Affiliation(s)
- Sarah E Little-Letsinger
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina, Chapel Hill, NC, USA.
| | - Gabriel M Pagnotti
- Department of Medicine, Division of Endocrinology, Indiana University, Indianapolis, IN, USA
| | - Cody McGrath
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina, Chapel Hill, NC, USA
| | - Maya Styner
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
14
|
Platko K, Lebeau PF, Gyulay G, Lhoták Š, MacDonald ME, Pacher G, Hyun Byun J, Boivin FJ, Igdoura SA, Cutz JC, Bridgewater D, Ingram AJ, Krepinsky JC, Austin RC. TDAG51 (T-Cell Death-Associated Gene 51) Is a Key Modulator of Vascular Calcification and Osteogenic Transdifferentiation of Arterial Smooth Muscle Cells. Arterioscler Thromb Vasc Biol 2020; 40:1664-1679. [PMID: 32434409 DOI: 10.1161/atvbaha.119.313779] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Cardiovascular disease is the primary cause of mortality in patients with chronic kidney disease. Vascular calcification (VC) in the medial layer of the vessel wall is a unique and prominent feature in patients with advanced chronic kidney disease and is now recognized as an important predictor and independent risk factor for cardiovascular and all-cause mortality in these patients. VC in chronic kidney disease is triggered by the transformation of vascular smooth muscle cells (VSMCs) into osteoblasts as a consequence of elevated circulating inorganic phosphate (Pi) levels, due to poor kidney function. The objective of our study was to investigate the role of TDAG51 (T-cell death-associated gene 51) in the development of medial VC. METHODS AND RESULTS Using primary mouse and human VSMCs, we found that TDAG51 is induced in VSMCs by Pi and is expressed in the medial layer of calcified human vessels. Furthermore, the transcriptional activity of RUNX2 (Runt-related transcription factor 2), a well-established driver of Pi-mediated VC, is reduced in TDAG51-/- VSMCs. To explain these observations, we identified that TDAG51-/- VSMCs express reduced levels of the type III sodium-dependent Pi transporter, Pit-1, a solute transporter, a solute transporter, a solute transporter responsible for cellular Pi uptake. Significantly, in response to hyperphosphatemia induced by vitamin D3, medial VC was attenuated in TDAG51-/- mice. CONCLUSIONS Our studies highlight TDAG51 as an important mediator of Pi-induced VC in VSMCs through the downregulation of Pit-1. As such, TDAG51 may represent a therapeutic target for the prevention of VC and cardiovascular disease in patients with chronic kidney disease.
Collapse
Affiliation(s)
- Khrystyna Platko
- From the Division of Nephrology, Department of Medicine (K.P., P.F.L., G.G., Š.L., M.E.M., G.P., J.H.B., A.J.I., J.C.K., R.C.A.), McMaster University, and The Research Institute of St. Joseph's Hamilton, ON, Canada
| | - Paul F Lebeau
- From the Division of Nephrology, Department of Medicine (K.P., P.F.L., G.G., Š.L., M.E.M., G.P., J.H.B., A.J.I., J.C.K., R.C.A.), McMaster University, and The Research Institute of St. Joseph's Hamilton, ON, Canada
| | - Gabriel Gyulay
- From the Division of Nephrology, Department of Medicine (K.P., P.F.L., G.G., Š.L., M.E.M., G.P., J.H.B., A.J.I., J.C.K., R.C.A.), McMaster University, and The Research Institute of St. Joseph's Hamilton, ON, Canada
| | - Šárka Lhoták
- From the Division of Nephrology, Department of Medicine (K.P., P.F.L., G.G., Š.L., M.E.M., G.P., J.H.B., A.J.I., J.C.K., R.C.A.), McMaster University, and The Research Institute of St. Joseph's Hamilton, ON, Canada
| | - Melissa E MacDonald
- From the Division of Nephrology, Department of Medicine (K.P., P.F.L., G.G., Š.L., M.E.M., G.P., J.H.B., A.J.I., J.C.K., R.C.A.), McMaster University, and The Research Institute of St. Joseph's Hamilton, ON, Canada
| | - Giusepina Pacher
- From the Division of Nephrology, Department of Medicine (K.P., P.F.L., G.G., Š.L., M.E.M., G.P., J.H.B., A.J.I., J.C.K., R.C.A.), McMaster University, and The Research Institute of St. Joseph's Hamilton, ON, Canada
| | - Jae Hyun Byun
- From the Division of Nephrology, Department of Medicine (K.P., P.F.L., G.G., Š.L., M.E.M., G.P., J.H.B., A.J.I., J.C.K., R.C.A.), McMaster University, and The Research Institute of St. Joseph's Hamilton, ON, Canada
| | - Felix J Boivin
- Department of Pathology and Molecular Medicine (F.J.B., S.A.I., D.B.), McMaster University Medical Centre, Hamilton, ON, Canada
| | - Suleiman A Igdoura
- Department of Pathology and Molecular Medicine (F.J.B., S.A.I., D.B.), McMaster University Medical Centre, Hamilton, ON, Canada.,Department of Biology (S.A.I.), McMaster University Medical Centre, Hamilton, ON, Canada
| | - Jean-Claude Cutz
- Department of Pathology and Molecular Medicine (J.-C.C.), McMaster University, and The Research Institute of St. Joseph's Hamilton, ON, Canada
| | - Darren Bridgewater
- Department of Pathology and Molecular Medicine (F.J.B., S.A.I., D.B.), McMaster University Medical Centre, Hamilton, ON, Canada
| | - Alistair J Ingram
- From the Division of Nephrology, Department of Medicine (K.P., P.F.L., G.G., Š.L., M.E.M., G.P., J.H.B., A.J.I., J.C.K., R.C.A.), McMaster University, and The Research Institute of St. Joseph's Hamilton, ON, Canada
| | - Joan C Krepinsky
- From the Division of Nephrology, Department of Medicine (K.P., P.F.L., G.G., Š.L., M.E.M., G.P., J.H.B., A.J.I., J.C.K., R.C.A.), McMaster University, and The Research Institute of St. Joseph's Hamilton, ON, Canada
| | - Richard C Austin
- From the Division of Nephrology, Department of Medicine (K.P., P.F.L., G.G., Š.L., M.E.M., G.P., J.H.B., A.J.I., J.C.K., R.C.A.), McMaster University, and The Research Institute of St. Joseph's Hamilton, ON, Canada
| |
Collapse
|
15
|
Tratwal J, Labella R, Bravenboer N, Kerckhofs G, Douni E, Scheller EL, Badr S, Karampinos DC, Beck-Cormier S, Palmisano B, Poloni A, Moreno-Aliaga MJ, Fretz J, Rodeheffer MS, Boroumand P, Rosen CJ, Horowitz MC, van der Eerden BCJ, Veldhuis-Vlug AG, Naveiras O. Reporting Guidelines, Review of Methodological Standards, and Challenges Toward Harmonization in Bone Marrow Adiposity Research. Report of the Methodologies Working Group of the International Bone Marrow Adiposity Society. Front Endocrinol (Lausanne) 2020; 11:65. [PMID: 32180758 PMCID: PMC7059536 DOI: 10.3389/fendo.2020.00065] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 01/31/2020] [Indexed: 12/14/2022] Open
Abstract
The interest in bone marrow adiposity (BMA) has increased over the last decade due to its association with, and potential role, in a range of diseases (osteoporosis, diabetes, anorexia, cancer) as well as treatments (corticosteroid, radiation, chemotherapy, thiazolidinediones). However, to advance the field of BMA research, standardization of methods is desirable to increase comparability of study outcomes and foster collaboration. Therefore, at the 2017 annual BMA meeting, the International Bone Marrow Adiposity Society (BMAS) founded a working group to evaluate methodologies in BMA research. All BMAS members could volunteer to participate. The working group members, who are all active preclinical or clinical BMA researchers, searched the literature for articles investigating BMA and discussed the results during personal and telephone conferences. According to the consensus opinion, both based on the review of the literature and on expert opinion, we describe existing methodologies and discuss the challenges and future directions for (1) histomorphometry of bone marrow adipocytes, (2) ex vivo BMA imaging, (3) in vivo BMA imaging, (4) cell isolation, culture, differentiation and in vitro modulation of primary bone marrow adipocytes and bone marrow stromal cell precursors, (5) lineage tracing and in vivo BMA modulation, and (6) BMA biobanking. We identify as accepted standards in BMA research: manual histomorphometry and osmium tetroxide 3D contrast-enhanced μCT for ex vivo quantification, specific MRI sequences (WFI and H-MRS) for in vivo studies, and RT-qPCR with a minimal four gene panel or lipid-based assays for in vitro quantification of bone marrow adipogenesis. Emerging techniques are described which may soon come to complement or substitute these gold standards. Known confounding factors and minimal reporting standards are presented, and their use is encouraged to facilitate comparison across studies. In conclusion, specific BMA methodologies have been developed. However, important challenges remain. In particular, we advocate for the harmonization of methodologies, the precise reporting of known confounding factors, and the identification of methods to modulate BMA independently from other tissues. Wider use of existing animal models with impaired BMA production (e.g., Pfrt-/-, KitW/W-v) and development of specific BMA deletion models would be highly desirable for this purpose.
Collapse
Affiliation(s)
- Josefine Tratwal
- Laboratory of Regenerative Hematopoiesis, Institute of Bioengineering and Swiss Institute for Experimental Cancer Research, Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Rossella Labella
- Tissue and Tumour Microenvironments Lab, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Nathalie Bravenboer
- Department of Clinical Chemistry, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam Movement Sciences, Amsterdam, Netherlands
- Section of Endocrinology, Department of Internal Medicine, Center for Bone Quality, Leiden University Medical Center, Leiden, Netherlands
| | - Greet Kerckhofs
- Biomechanics Lab, Institute of Mechanics, Materials and Civil Engineering, UCLouvain, Louvain-la-Neuve, Belgium
- Department Materials Engineering, KU Leuven, Leuven, Belgium
| | - Eleni Douni
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, Athens, Greece
- Institute for Bioinnovation, Biomedical Sciences Research Center Alexander Fleming, Athens, Greece
| | - Erica L. Scheller
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, MO, United States
| | - Sammy Badr
- Univ. Lille, EA 4490 - PMOI - Physiopathologie des Maladies Osseuses Inflammatoires, Lille, France
- CHU Lille, Service de Radiologie et Imagerie Musculosquelettique, Lille, France
| | - Dimitrios C. Karampinos
- Department of Diagnostic and Interventional Radiology, Technical University of Munich, Munich, Germany
| | - Sarah Beck-Cormier
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
| | - Biagio Palmisano
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, United States
| | - Antonella Poloni
- Hematology, Department of Clinic and Molecular Science, Università Politecnica Marche-AOU Ospedali Riuniti, Ancona, Italy
| | - Maria J. Moreno-Aliaga
- Centre for Nutrition Research and Department of Nutrition, Food Science and Physiology, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- IdiSNA, Navarra's Health Research Institute, Pamplona, Spain
- CIBERobn Physiopathology of Obesity and Nutrition, Centre of Biomedical Research Network, ISCIII, Madrid, Spain
| | - Jackie Fretz
- Department of Orthopaedics and Rehabilitation, Cellular and Developmental Biology, Yale University School of Medicine, New Haven, CT, United States
| | - Matthew S. Rodeheffer
- Department of Comparative Medicine and Molecular, Cellular and Developmental Biology, Yale University School of Medicine, New Haven, CT, United States
| | - Parastoo Boroumand
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Clifford J. Rosen
- Maine Medical Center Research Institute, Center for Clinical and Translational Research, Scarborough, ME, United States
| | - Mark C. Horowitz
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, United States
| | - Bram C. J. van der Eerden
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Annegreet G. Veldhuis-Vlug
- Section of Endocrinology, Department of Internal Medicine, Center for Bone Quality, Leiden University Medical Center, Leiden, Netherlands
- Maine Medical Center Research Institute, Center for Clinical and Translational Research, Scarborough, ME, United States
- Jan van Goyen Medical Center/OLVG Hospital, Department of Internal Medicine, Amsterdam, Netherlands
- *Correspondence: Annegreet G. Veldhuis-Vlug
| | - Olaia Naveiras
- Laboratory of Regenerative Hematopoiesis, Institute of Bioengineering and Swiss Institute for Experimental Cancer Research, Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Hematology Service, Departments of Oncology and Laboratory Medicine, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- Olaia Naveiras ;
| |
Collapse
|
16
|
Khan R, Raza SHA, Junjvlieke Z, Wang H, Cheng G, Smith SB, Jiang Z, Li A, Zan L. RNA-seq reveal role of bovine TORC2 in the regulation of adipogenesis. Arch Biochem Biophys 2019; 680:108236. [PMID: 31893525 DOI: 10.1016/j.abb.2019.108236] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 12/05/2019] [Accepted: 12/20/2019] [Indexed: 12/18/2022]
Abstract
Low intramuscular adipose tissue (marbling) continues to be challenge for improving beef quality in Chinese cattle. Highly marbled meat is very desirable; hence, methods to increase IMF content have become a key aspect of improving meat quality. Therefore, research on the mechanism of adipogenesis provides invaluable information for the improvement of meat quality. This study investigated the effect of TORC2 and its underlying mechanism on lipid metabolism in bovine adipocytes. The TORC2 gene was downregulated in bovine adipocytes by siRNA, and RNA sequencing was performed. Downregulation of TORC2 negatively affected bovine adipocyte differentiation. In addition, a total of 577 DEGs were found, containing 146 up-regulated and 376 down-regulated genes. KEGG pathway analysis revealed that the DEGs were linked with neuroactive ligand-receptor interaction pathway, calcium signaling pathway, cAMP pathway, chemokine signaling pathway and Wnt signaling pathway. Gene Ontology (GO) term analysis of the DEGs showed that down-regulation of TORC2 gene significantly suppressed the genes regulating important GO terms of adipogenesis-related processes in bovine adipocytes, especially regulation of biological activity, regulation of primary metabolic process, regulation of multicellular organismal process, cell adhesion, lipid metabolic process, secretion, chemical homeostasis, regulation of transport, cell-cell signaling, cAMP metabolic process, cellular calcium ion homeostasis, fat cell differentiation, and cell maturation. In conclusion, our results suggest that TORC2 at least in part regulates lipid metabolism in bovine adipocytes. The results of this study provide a basis for studying the function and molecular mechanism of the TORC2 gene in regulating adipogenesis.
Collapse
Affiliation(s)
- Rajwali Khan
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| | - Sayed Haidar Abbas Raza
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| | - Zainaguli Junjvlieke
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Hongbao Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Gong Cheng
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Stephen B Smith
- Department of Animal Science, Texas A&M University, College Station, TX, 77843, USA
| | - Zhongliang Jiang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Anning Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China; National Beef Cattle Improvement Center, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
17
|
Abstract
The skeleton harbors an array of lineage cells that have an essential role in whole body homeostasis. Adipocytes start the colonization of marrow space early in postnatal life, expanding progressively and influencing other components of the bone marrow through paracrine signaling. In this unique, closed, and hypoxic environment close to the endosteal surface and adjacent to the microvascular space the marrow adipocyte can store or provide energy, secrete adipokines, and target neighboring bone cells. Adipocyte progenitors can also migrate from the bone marrow to populate white adipose tissue, a process that accelerates during weight gain. The marrow adipocyte also has an endocrine role in whole body homeostasis through its varied secretome that targets distant adipose depots, skeletal muscle, and the nervous system. Further insights into the biology of this unique and versatile cell will undoubtedly lead to novel therapeutic approaches to metabolic and age-related disorders such as osteoporosis and diabetes mellitus.
Collapse
Affiliation(s)
- Francisco J A de Paula
- Department of Internal Medicine, Ribeirao Preto Medical School, University of São Paulo, São Paulo 14049-900, Brazil;
| | - Clifford J Rosen
- Center for Clinical and Translational Research, Maine Medical Center Research Institute, Scarborough, Maine 04074, USA;
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW This article reviews the past 2 years of research on Notch signaling as it relates to bone physiology, with the goal of reconciling seemingly discrepant findings and identifying fruitful areas of potential future research. RECENT FINDINGS Conditional animal models and high-throughput omics have contributed to a greater understanding of the context-dependent role of Notch signaling in bone. However, significant gaps remain in our understanding of how spatiotemporal context and epigenetic state dictate downstream Notch phenotypes. Biphasic activation of Notch signaling orchestrates progression of mesenchymal progenitor cells through the osteoblast lineage, but there is a limited understanding of ligand- and receptor-specific functions. Paracrine Notch signaling through non-osteoblastic cell types contributes additional layers of complexity, and we anticipate impactful future work related to the integration of these cell types and signaling mechanisms.
Collapse
Affiliation(s)
- Daniel W Youngstrom
- Department of Orthopaedic Surgery, University of Michigan Medical School, 109 Zina Pitcher Pl, Ann Arbor, MI, 48872, USA.
| | - Kurt D Hankenson
- Department of Orthopaedic Surgery, University of Michigan Medical School, 109 Zina Pitcher Pl, Ann Arbor, MI, 48872, USA
| |
Collapse
|
19
|
Beck L. Expression and function of Slc34 sodium-phosphate co-transporters in skeleton and teeth. Pflugers Arch 2018; 471:175-184. [PMID: 30511265 DOI: 10.1007/s00424-018-2240-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/22/2018] [Accepted: 11/23/2018] [Indexed: 12/20/2022]
Abstract
Under normal physiological condition, the biomineralization process is limited to skeletal tissues and teeth and occurs throughout the individual's life. Biomineralization is an actively regulated process involving the progressive mineralization of the extracellular matrix secreted by osteoblasts in bone or odontoblasts and ameloblasts in tooth. Although the detailed molecular mechanisms underlying the formation of calcium-phosphate apatite crystals are still debated, it is suggested that calcium and phosphate may need to be transported across the membrane of the mineralizing cell, suggesting a pivotal role of phosphate transporters in bone and tooth mineralization. In this context, this short review describes the current knowledge on the role of Slc34 Na+-phosphate transporters in skeletal and tooth mineralization.
Collapse
Affiliation(s)
- Laurent Beck
- INSERM, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Faculté de Chirurgie Dentaire, Université de Nantes, ONIRIS, 1 place Alexis Ricordeau, 44042, Nantes, France. .,Université de Nantes, UFR Odontologie, 44042, Nantes, France.
| |
Collapse
|
20
|
Sroga GE, Vashishth D. Phosphorylation of Extracellular Bone Matrix Proteins and Its Contribution to Bone Fragility. J Bone Miner Res 2018; 33:2214-2229. [PMID: 30001467 DOI: 10.1002/jbmr.3552] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 07/05/2018] [Accepted: 07/08/2018] [Indexed: 01/22/2023]
Abstract
Phosphorylation of bone matrix proteins is of fundamental importance to all vertebrates including humans. However, it is currently unknown whether increase or decline of total protein phosphorylation levels, particularly in hypophosphatemia-related osteoporosis, osteomalacia, and rickets, contribute to bone fracture. To address this gap, we combined biochemical measurements with mechanical evaluation of bone to discern fracture characteristics associated with age-related development of skeletal fragility in relation to total phosphorylation levels of bone matrix proteins and one of the key representatives of bone matrix phosphoproteins, osteopontin (OPN). Here for the first time, we report that as people age the total phosphorylation level declines by approximately 20% for bone matrix proteins and approximately 30% for OPN in the ninth decade of human life. Moreover, our results suggest that the decline of total protein phosphorylation of extracellular matrix (ECM) contributes to bone fragility, but less pronouncedly than glycation. We theorize that the separation of two sources of OPN negative charges, acidic backbone amino acids and phosphorylation, would be nature's means of assuring that OPN functions in both energy dissipation and biomineralization. We propose that total phosphorylation decline could be an important contributor to the development of osteoporosis, increased fracture risk and skeletal fragility. Targeting the enzymes kinase FamC20 and bone alkaline phosphatase involved in the regulation of matrix proteins' phosphorylation could be a means for the development of suitable therapeutic treatments. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Grażyna E Sroga
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Deepak Vashishth
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| |
Collapse
|
21
|
Noguchi T, Hussein AI, Horowitz N, Carroll D, Gower AC, Demissie S, Gerstenfeld LC. Hypophosphatemia Regulates Molecular Mechanisms of Circadian Rhythm. Sci Rep 2018; 8:13756. [PMID: 30213970 PMCID: PMC6137060 DOI: 10.1038/s41598-018-31830-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 08/13/2018] [Indexed: 12/31/2022] Open
Abstract
Transcriptomic analysis showed that the central circadian pathway genes had significantly altered expression in fracture calluses from mice fed a low phosphate diet. This led us to hypothesize that phosphate deficiency altered the circadian cycle in peripheral tissues. Analysis of the expression of the central clock genes over a 24-36 hour period in multiple peripheral tissues including fracture callus, proximal tibia growth plate and cardiac tissues after 12 days on a low phosphate diet showed higher levels of gene expression in the hypophosphatemia groups (p < 0.001) and a 3 to 6 hour elongation of the circadian cycle. A comparative analysis of the callus tissue transcriptome genes that were differentially regulated by hypophosphatemia with published data for the genes in bone that are diurnally regulated identified 1879 genes with overlapping differential regulation, which were shown by ontology assessment to be associated with oxidative metabolism and apoptosis. Network analysis of the central circadian pathway genes linked their expression to the up regulated expression of the histone methyltransferase gene EZH2, a gene that when mutated in both humans and mice controls overall skeletal growth. These data suggest that phosphate is an essential metabolite that controls circadian function in both skeletal and non skeletal peripheral tissues and associates its levels with the overall oxidative metabolism and skeletal growth of animals.
Collapse
Affiliation(s)
- Takashi Noguchi
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, USA
| | - Amira I Hussein
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, USA
| | - Nina Horowitz
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, USA
| | - Deven Carroll
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, USA
| | - Adam C Gower
- Clinical and Translational Science Institute, Boston University School of Medicine, Boston, USA
| | - Serkalem Demissie
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Louis C Gerstenfeld
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, USA.
| |
Collapse
|
22
|
Abstract
Marrow adipocytes, collectively termed marrow adipose tissue (MAT), reside in the bone marrow in close contact to bone cells and haematopoietic cells. Marrow adipocytes arise from the mesenchymal stem cell and share their origin with the osteoblast. Shifts in the lineage allocation of the mesenchymal stromal cell could potentially explain the association between increased MAT and increased fracture risk in diseases such as postmenopausal osteoporosis, anorexia nervosa and diabetes. Functionally, marrow adipocytes secrete adipokines, such as adiponectin, and cytokines, such as RANK ligand and stem cell factor. These mediators can influence both bone remodelling and haematopoiesis by promoting bone resorption and haematopoietic recovery following chemotherapy. In addition, marrow adipocytes can secrete free fatty acids, acting as a energy supply for bone and haematopoietic cells. However, this induced lipolysis is also used by neoplastic cells to promote survival and proliferation. Therefore, MAT could represent a new therapeutic target for multiple diseases from osteoporosis to leukaemia, although the exact characteristics and role of the marrow adipocyte in health and diseases remain to be determined.
Collapse
Affiliation(s)
- A G Veldhuis-Vlug
- Department of Endocrinology and Metabolism, Academic Medical Center, Amsterdam, The Netherlands
- Center for Clinical and Translational Research, Maine Medical Center Research Institute, Scarborough, ME, USA
| | - C J Rosen
- Center for Clinical and Translational Research, Maine Medical Center Research Institute, Scarborough, ME, USA
| |
Collapse
|
23
|
Ang KH, Patel AD, Berkwitt AK. An Unusual Presentation of Hypophosphatemic Rickets. AACE Clin Case Rep 2018. [DOI: 10.4158/ep171853.cr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
24
|
Styner M, Pagnotti GM, McGrath C, Wu X, Sen B, Uzer G, Xie Z, Zong X, Styner MA, Rubin CT, Rubin J. Exercise Decreases Marrow Adipose Tissue Through ß-Oxidation in Obese Running Mice. J Bone Miner Res 2017; 32:1692-1702. [PMID: 28436105 PMCID: PMC5550355 DOI: 10.1002/jbmr.3159] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 03/17/2017] [Accepted: 04/20/2017] [Indexed: 12/23/2022]
Abstract
The relationship between marrow adipose tissue (MAT) and bone health is poorly understood. We used running exercise to ask whether obesity-associated MAT can be attenuated via exercise and whether this correlates with gains in bone quantity and quality. C57BL/6 mice were divided into diet-induced obesity (DIO, n = 14) versus low-fat diet (LFD, n = 14). After 3 months, 16-week-old mice were allocated to an exercise intervention (LFD-E, DIO-E) or a control group (LFD, DIO) for 6 weeks (4 groups, n = 7/group). Marrow adipocyte area was 44% higher with obesity (p < 0.0001) and after exercise 33% lower in LFD (p < 0.0001) and 39% lower in DIO (p < 0.0001). In LFD, exercise did not affect adipocyte number; however, in DIO, the adipocyte number was 56% lower (p < 0.0001). MAT was 44% higher in DIO measured by osmium-μCT, whereas exercise associated with reduced MAT (-23% in LFD, -48% in DIO, p < 0.05). MAT was additionally quantified by 9.4TMRI, and correlated with osmium-µCT (r = 0.645; p < 0.01). Consistent with higher lipid beta oxidation, perilipin 3 (PLIN3) rose with exercise in tibial mRNA (+92% in LFD, +60% in DIO, p < 0.05). Tibial µCT-derived trabecular bone volume (BV/TV) was not influenced by DIO but responded to exercise with an increase of 19% (p < 0.001). DIO was associated with higher cortical periosteal and endosteal volumes of 15% (p = 0.012) and 35% (p < 0.01), respectively, but Ct.Ar/Tt.Ar was lower by 2.4% (p < 0.05). There was a trend for higher stiffness (N/m) in DIO, and exercise augmented this further. In conclusion, obesity associated with increases in marrow lipid-measured by osmium-μCT and MRI-and partially due to an increase in adipocyte size, suggesting increased lipid uptake into preexisting adipocytes. Exercise associated with smaller adipocytes and less bone lipid, likely invoking increased ß-oxidation and basal lipolysis as evidenced by higher levels of PLIN3. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Maya Styner
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina, Chapel Hill, NC, USA
| | - Gabriel M Pagnotti
- Department of Biomedical Engineering, State University of New York, Stony Brook, Stony Brook, NY, USA
| | - Cody McGrath
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina, Chapel Hill, NC, USA
| | - Xin Wu
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina, Chapel Hill, NC, USA
| | - Buer Sen
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina, Chapel Hill, NC, USA
| | - Gunes Uzer
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina, Chapel Hill, NC, USA
- Department of Mechanical and Biomedical Engineering, Boise State University, Boise, ID, USA
| | - Zhihui Xie
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina, Chapel Hill, NC, USA
| | - Xiaopeng Zong
- Department of Radiology and Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC, USA
| | - Martin A Styner
- Department of Computer Science, University of North Carolina, Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA
| | - Clinton T Rubin
- Department of Biomedical Engineering, State University of New York, Stony Brook, Stony Brook, NY, USA
| | - Janet Rubin
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
25
|
Papaioannou G, Petit ET, Liu ES, Baccarini M, Pritchard C, Demay MB. Raf Kinases Are Essential for Phosphate Induction of ERK1/2 Phosphorylation in Hypertrophic Chondrocytes and Normal Endochondral Bone Development. J Biol Chem 2017; 292:3164-3171. [PMID: 28073913 PMCID: PMC5336153 DOI: 10.1074/jbc.m116.763342] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 01/04/2017] [Indexed: 01/18/2023] Open
Abstract
Hypophosphatemia causes rickets by impairing hypertrophic chondrocyte apoptosis. Phosphate induction of MEK1/2-ERK1/2 phosphorylation in hypertrophic chondrocytes is required for phosphate-mediated apoptosis and growth plate maturation. MEK1/2 can be activated by numerous molecules including Raf isoforms. A- and B-Raf ablation in chondrocytes does not alter skeletal development, whereas ablation of C-Raf decreases hypertrophic chondrocyte apoptosis and impairs vascularization of the growth plate. However, ablation of C-Raf does not impair phosphate-induced ERK1/2 phosphorylation in vitro, but leads to rickets by decreasing VEGF protein stability. To determine whether Raf isoforms are required for phosphate-induced hypertrophic chondrocyte apoptosis, mice lacking all three Raf isoforms in chondrocytes were generated. Raf deletion caused neonatal death and a significant expansion of the hypertrophic chondrocyte layer of the growth plate, accompanied by decreased cleaved caspase-9. This was associated with decreased phospho-ERK1/2 immunoreactivity in the hypertrophic chondrocyte layer and impaired vascular invasion. These data further demonstrated that Raf kinases are required for phosphate-induced ERK1/2 phosphorylation in cultured hypertrophic chondrocytes and perform essential, but partially redundant roles in growth plate maturation.
Collapse
Affiliation(s)
- Garyfallia Papaioannou
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts 02114; Harvard Medical School, Boston, Massachusetts 02115
| | - Elizabeth T Petit
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts 02114
| | - Eva S Liu
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts 02114; Harvard Medical School, Boston, Massachusetts 02115; Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Boston, Massachusetts 02115
| | - Manuela Baccarini
- Department of Microbiology, Immunobiology and Genetics, Center of Molecular Biology, Max F. Perutz Laboratories, University of Vienna, Doktor-Bohr-Gasse 9, Vienna 1030, Austria
| | - Catrin Pritchard
- Department of Molecular and Cell Biology, University of Leicester, University Road, Leicester LE1 7RH, United Kingdom
| | - Marie B Demay
- Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts 02114; Harvard Medical School, Boston, Massachusetts 02115.
| |
Collapse
|
26
|
Zhou ZF, Sun TW, Chen F, Zuo DQ, Wang HS, Hua YQ, Cai ZD, Tan J. Calcium phosphate-phosphorylated adenosine hybrid microspheres for anti-osteosarcoma drug delivery and osteogenic differentiation. Biomaterials 2016; 121:1-14. [PMID: 28063979 DOI: 10.1016/j.biomaterials.2016.12.031] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 12/19/2016] [Accepted: 12/27/2016] [Indexed: 01/02/2023]
Abstract
Biocompatibility, biodegradability and bioactivity are significantly important in practical applications of various biomaterials for bone tissue engineering. Herein, we develop a functional inorganic-organic hybrid system of calcium phosphate-phosphorylated adenosine (CPPA). Both calcium phosphate and phosphorylated adenosine molecules in CPPA are fundamental components in mammalians and play important roles in biological metabolism. In this work, we report our three leading research qualities: (1) CPPA hybrid microspheres with hollow and porous structure are synthesized by a facile one-step microwave-assisted solvothermal method; (2) CPPA hybrid microspheres show high doxorubicin loading capacity and pH-responsive drug release properties, and demonstrate positive therapeutic effects on six osteosarcoma cell lines in vitro and a mouse model of 143B osteosarcoma subcutaneous tumor in vivo; (3) CPPA hybrid microspheres are favorable to promote osteogenic differentiation of human bone mesenchymal stem cells (hBMSCs) by activating the AMPK pathway, with satisfactory evidences from cellular alkaline phosphatase staining, alizarin red staining, real time PCR and western analysis. The as-prepared CPPA hybrid microspheres are promising in anti-osteosarcoma and bone regeneration, which simultaneously display excellent properties on drug delivery and osteogenic differentiation of hBMSCs.
Collapse
Affiliation(s)
- Zi-Fei Zhou
- Department of Orthopedic Surgery, Shanghai East Hospital, Tongji University, Shanghai 200120, PR China
| | - Tuan-Wei Sun
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, PR China
| | - Feng Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, PR China.
| | - Dong-Qing Zuo
- Department of Orthopedic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, PR China
| | - Hong-Sheng Wang
- Department of Orthopedic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, PR China
| | - Ying-Qi Hua
- Department of Orthopedic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, PR China
| | - Zheng-Dong Cai
- Department of Orthopedic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai 200080, PR China.
| | - Jun Tan
- Department of Orthopedic Surgery, Shanghai East Hospital, Tongji University, Shanghai 200120, PR China.
| |
Collapse
|