50 results on '"Janet L. Crane"'
Search Results
2. Editorial: Management of Bone Disorders in Children
- Author
-
Janet L. Crane and Madhusmita Misra
- Subjects
bone disorders ,osteoporosis management ,X-linked hypophosphatemia ,oral contraception ,giant cell tumor ,vitamin d deficient rickets ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Published
- 2021
- Full Text
- View/download PDF
3. Case Report: Safety and Efficacy of Denosumab in Four Children With Noonan Syndrome With Multiple Giant Cell Lesions of the Jaw
- Author
-
Kristen Ferriero, Biraj Shah, Yun Yan, Surya Khatri, John Caccamese, Joseph A. Napoli, Michael B. Bober, and Janet L. Crane
- Subjects
Noonan syndrome ,multiple giant cell lesions ,denosumab ,jaw ,child ,Pediatrics ,RJ1-570 - Abstract
Noonan syndrome is a genetic disorder caused by mutations in the RAS/MAPK pathway. Multiple giant cell lesions are a rare sequelae of disruptions in this pathway, termed Noonan-like multiple giant cell lesions (NL/MGCLs). Medical management of these tumors rather than surgical intervention is preferential as the lesions are benign but locally destructive and recurring. This case series describes four male pediatric patients with Noonan syndrome and multiple giant cell lesions of the jaw treated with denosumab, a monoclonal antibody to receptor activator of nuclear factor kappa B ligand (RANKL), which has been approved for the treatment of malignant giant cell tumors in adults but not evaluated for safety or efficacy in children. All four pediatric patients responded clinically and radiographically to the treatment. Adverse events occurred in a predictable pattern and included hypocalcemia and joint pain during the initiation of treatment and symptomatic hypercalcemia after the cessation of treatment. Growth was not significantly impaired in these skeletally immature patients. This case series demonstrates how a weight-adjusted denosumab dose can effectively treat NL/MGCLs and provides laboratory data for consideration of the timing of monitoring for known side effects.
- Published
- 2020
- Full Text
- View/download PDF
4. Programmed cell senescence in skeleton during late puberty
- Author
-
Changjun Li, Yu Chai, Lei Wang, Bo Gao, Hao Chen, Peisong Gao, Feng-Quan Zhou, Xianghang Luo, Janet L. Crane, Bin Yu, Xu Cao, and Mei Wan
- Subjects
Science - Abstract
Mesenchymal stem cells are essential for bone development, but it is unclear if their activity is maintained after late puberty, when bone growth decelerates. The authors show that during late puberty in mice, these cells undergo senescence under the epigenetic control of Ezh2.
- Published
- 2017
- Full Text
- View/download PDF
5. RhoA determines lineage fate of mesenchymal stem cells by modulating CTGF–VEGF complex in extracellular matrix
- Author
-
Changjun Li, Gehua Zhen, Yu Chai, Liang Xie, Janet L. Crane, Emily Farber, Charles R. Farber, Xianghang Luo, Peisong Gao, Xu Cao, and Mei Wan
- Subjects
Science - Abstract
It is unclear what regulates the fate of mesenchymal stem cells (MSCs) in arterial repair following injury. Here, the authors show that MSC differentiation following injury is triggered by RhoA which in turn stimulates the release of connective tissue growth factor and vascular endothelial growth factor.
- Published
- 2016
- Full Text
- View/download PDF
6. Author Correction: Sensory innervation in porous endplates by Netrin-1 from osteoclasts mediates PGE2-induced spinal hypersensitivity in mice
- Author
-
Shuangfei Ni, Zemin Ling, Xiao Wang, Yong Cao, Tianding Wu, Ruoxian Deng, Janet L. Crane, Richard Skolasky, Shadpour Demehri, Gehua Zhen, Amit Jain, Panfeng Wu, Dayu Pan, Bo Hu, Xiao Lyu, Yusheng Li, Hao Chen, Huabin Qi, Yun Guan, Xinzhong Dong, Mei Wan, Xuenong Zou, Hongbin Lu, Jianzhong Hu, and Xu Cao
- Subjects
Science - Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2020
- Full Text
- View/download PDF
7. Kaposiform lymphangiomatosis: Diagnosis, pathogenesis, and treatment
- Author
-
C. Griffin McDaniel, Denise M. Adams, Kimberley E. Steele, Adrienne M. Hammill, A. Carl Merrow, Janet L. Crane, Christopher L. Smith, Harry P. W. Kozakewich, and Timothy D. Le Cras
- Subjects
Oncology ,Pediatrics, Perinatology and Child Health ,Hematology - Published
- 2023
- Full Text
- View/download PDF
8. Glucocorticoids Disrupt Skeletal Angiogenesis Through Transrepression of NF‐κB–Mediated Preosteoclast Pdgfb Transcription in Young Mice
- Author
-
Shijie Chen, Gehua Zhen, Song Wu, Yusheng Li, Xu Cao, Shan Lv, Janet L. Crane, Jianxi Zhu, and Yi Peng
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Angiogenesis ,Endocrinology, Diabetes and Metabolism ,Osteoporosis ,030209 endocrinology & metabolism ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Glucocorticoid receptor ,Osteogenesis ,Internal medicine ,medicine ,Animals ,Orthopedics and Sports Medicine ,Glucocorticoids ,Cells, Cultured ,Transrepression ,Bone growth ,Tube formation ,PDGFB ,Chemistry ,NF-kappa B ,Proto-Oncogene Proteins c-sis ,medicine.disease ,030104 developmental biology ,Endocrinology ,Prednisolone ,medicine.drug - Abstract
In the growing skeleton, angiogenesis is intimately coupled with osteogenesis. Chronic, high doses of glucocorticoids (GCs) are associated with decreased bone vasculature and induce osteoporosis and growth failure. The mechanism of GC-suppression of angiogenesis and relationship to osteoporosis and growth retardation remains largely unknown. Type H vessels, which are regulated by preosteoclast (POC) platelet-derived growth factor-BB (PDGF-BB), are specifically coupled with bone formation and development. We determined the effect of GCs on POC synthesis of PDGF-BB in relation to type H vessel formation, bone mass, and bone growth in the distal femur of 2-week-old young mice receiving prednisolone or vehicle for 2, 4, or 6 weeks. After 2 weeks of prednisolone, the number of POCs were unchanged while POC synthesis of PDGF-BB was reduced. Longer treatment with prednisolone reduced POCs numbers and PDGF-BB. These changes were associated with a reduction in type H vessels, bone formation rate, bone mass, and bone length at each time point. In vitro, excessive concentrations of prednisolone (10-6 M) resulted in decreased PDGF-BB concentration and POC numbers. Conditioned medium from POC cultures treated with control concentration of prednisolone (10-7 M) or recombinant PDGF-BB stimulated endothelial tube formation, whereas conditioned medium from control concentration of prednisolone-treated POC cultures neutralized by PDGF-BB antibody or excessive prednisolone inhibited endothelial tube formation. Administration of excessive prednisolone attenuated the P65 subunit of nuclear factor kappa B (NF-κB) binding to the Pdgfb promoter, resulting in lower Pdgfb transcription. Co-treatment with excessive prednisolone and the glucocorticoid receptor (GR) antagonist (RU486), GR siRNA, or TNFα rescued NF-κB binding to the Pdgfb promoter and endothelial tube formation. These results indicate that PDGF-BB synthesis in POCs is suppressed by GCs through transrepression of GR/NF-κB, thus inhibiting type H vessel formation and associated osteoporosis and growth failure. © 2020 American Society for Bone and Mineral Research.
- Published
- 2020
- Full Text
- View/download PDF
9. Type H blood vessels in bone modeling and remodeling
- Author
-
Yusheng Li, Janet L. Crane, Yi Peng, and Song Wu
- Subjects
0301 basic medicine ,CD31 ,Angiogenesis ,medicine.medical_treatment ,Sialoglycoproteins ,Medicine (miscellaneous) ,Neovascularization, Physiologic ,CD31hiEmcnhi endothelial cells ,Bone healing ,Review ,Bone and Bones ,Bone remodeling ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Osteogenesis ,medicine ,Animals ,Humans ,H-type vessels ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,Bone growth ,Regeneration (biology) ,Growth factor ,Type H vessels ,Cell biology ,Vascular endothelial growth factor ,Platelet Endothelial Cell Adhesion Molecule-1 ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Bone formation ,Blood Vessels ,Bone Remodeling - Abstract
In the mammalian skeletal system, osteogenesis and angiogenesis are intimately linked during bone growth and regeneration in bone modeling and during bone homeostasis in bone remodeling. Recent studies have expanded our knowledge about the molecular and cellular mechanisms responsible for coupling angiogenesis and bone formation. Type H vessels, termed such because of high expression of Endomucin (Emcn) and CD31, have recently been identified and have the ability to induce bone formation. Factors including platelet-derived growth factor type BB (PDGF-BB), slit guidance ligand 3 (SLIT3), hypoxia-inducible factor 1-alpha (HIF-1α), Notch, and vascular endothelial growth factor (VEGF) are involved in the coupling of angiogenesis and osteogenesis. This review summarizes the current understanding of signaling pathways that regulate type H vessels and how type H vessels modulate osteogenesis. Further studies dissecting the regulation and function of type H vessels will provide new insights into the role of bone vasculature in the metabolism of the skeleton. We also discuss considerations for therapeutic approaches targeting type H vessels to promote fracture healing, prevent pathological bone loss, osteonecrosis, osteoarthritis, and bone metastases.
- Published
- 2020
10. Insulin Glargine Dose and Weight Changes in Underweight, Normal Weight, and Overweight Children Newly Diagnosed with Type 1 Diabetes Mellitus
- Author
-
Bethany Sharpless Chalk, Carlton K. K. Lee, Gayane Yenokyan, Janet L. Crane, and Erika May Pineda
- Subjects
Blood Glucose ,Male ,0301 basic medicine ,Pediatrics ,medicine.medical_specialty ,Percentile ,Adolescent ,030106 microbiology ,Insulin Glargine ,030204 cardiovascular system & hematology ,Overweight ,Medical Records ,Article ,Body Mass Index ,03 medical and health sciences ,0302 clinical medicine ,Thinness ,medicine ,Humans ,Hypoglycemic Agents ,Pharmacology (medical) ,Dosing ,Child ,Retrospective Studies ,Academic Medical Centers ,Type 1 diabetes ,Dose-Response Relationship, Drug ,Insulin glargine ,business.industry ,Body Weight ,nutritional and metabolic diseases ,medicine.disease ,Diabetes Mellitus, Type 1 ,Normal weight ,Child, Preschool ,Female ,medicine.symptom ,Underweight ,business ,Body mass index ,medicine.drug - Abstract
STUDY OBJECTIVE: Newly diagnosed pediatric patients with type 1 diabetes mellitus (T1D) can be underweight, overweight, or normal weight at presentation. Study objectives were to determine if, across weight categories, admission body weight (ABW)-based initial insulin glargine dosing resulted in similar fasting blood glucose responses on day of discharge, how initial ABW-based doses differed from doses at outpatient follow-up, and whether an ideal body weight (IBW) would provide a better estimate of body weight after discharge. DESIGN: Retrospective chart review. SETTING: Urban tertiary academic medical center. PATIENTS: Eighty-one pediatric patients newly diagnosed with T1D who started therapy with subcutaneous insulin glargine between October 2014 and October 2016; patients were categorized by weight using body mass index (BMI) percentiles (underweight, normal weight, overweight/obese). MEASUREMENTS AND MAIN RESULTS: Data on patient parameters from hospitalization and outpatient physician follow-up were collected. The McLaren, Moore, and BMI IBW methods were used to calculate IBW for each patient; these IBWs were compared with weights at outpatient follow-up. Initial insulin glargine doses were similar among all weight groups: median (range) 0.299 (0.227–0.4), 0.297 (0.204–0.421), and 0.291 (0.194–0.394) units/kg/dose, respectively, for the underweight, normal weight, overweight/obese groups. No significant differences in discharge fasting glucose level or insulin glargine dose change from admission to first outpatient follow-up visit were noted. Underweight patients gained significantly more weight within 60 days after discharge compared with normal and overweight/obese patients, (median 16.3% vs 7.7% and 5.7%, respectively, p=0.002), aligning closest with the McLaren IBW. ABW was the best estimate of weight at outpatient follow-up in the overweight/obese patient group. CONCLUSION: For children who presented underweight, the McLaren IBW method was the best predictor of outpatient dose and body weight, whereas ABW was the best estimate in overweight and obese patients. Further investigation of the role of IBW or adjusted body weight–based dosing methods in underweight pediatric patients with T1D may assist in optimal dosing.
- Published
- 2019
- Full Text
- View/download PDF
11. Editorial: Management of Bone Disorders in Children
- Author
-
Madhusmita Misra and Janet L. Crane
- Subjects
medicine.medical_specialty ,X-linked hypophosphatemia ,business.industry ,oral contraception ,Endocrinology, Diabetes and Metabolism ,Osteoporosis ,Rickets ,bone disorders ,medicine.disease ,Epidermal nevus syndrome ,RC648-665 ,Dermatology ,Diseases of the endocrine glands. Clinical endocrinology ,Denosumab ,medicine ,business ,vitamin d deficient rickets ,Oral contraception ,osteoporosis management ,giant cell tumor ,medicine.drug - Published
- 2021
- Full Text
- View/download PDF
12. Case Report: Safety and Efficacy of Denosumab in Four Children With Noonan Syndrome With Multiple Giant Cell Lesions of the Jaw
- Author
-
Joseph A. Napoli, Kristen Ferriero, Biraj Shah, John F. Caccamese, Janet L. Crane, Yun Yan, Michael B. Bober, and Surya Khatri
- Subjects
Oncology ,medicine.medical_specialty ,Case Report ,030204 cardiovascular system & hematology ,Pediatrics ,03 medical and health sciences ,0302 clinical medicine ,jaw ,030225 pediatrics ,Internal medicine ,medicine ,Noonan syndrome ,Giant Cell Tumors ,Adverse effect ,Not evaluated ,child ,business.industry ,multiple giant cell lesions ,Genetic disorder ,lcsh:RJ1-570 ,denosumab ,lcsh:Pediatrics ,medicine.disease ,Denosumab ,Giant cell ,Joint pain ,Pediatrics, Perinatology and Child Health ,medicine.symptom ,business ,medicine.drug - Abstract
Noonan syndrome is a genetic disorder caused by mutations in the RAS/MAPK pathway. Multiple giant cell lesions are a rare sequelae of disruptions in this pathway, termed Noonan-like multiple giant cell lesions (NL/MGCLs). Medical management of these tumors rather than surgical intervention is preferential as the lesions are benign but locally destructive and recurring. This case series describes four male pediatric patients with Noonan syndrome and multiple giant cell lesions of the jaw treated with denosumab, a monoclonal antibody to receptor activator of nuclear factor kappa B ligand (RANKL), which has been approved for the treatment of malignant giant cell tumors in adults but not evaluated for safety or efficacy in children. All four pediatric patients responded clinically and radiographically to the treatment. Adverse events occurred in a predictable pattern and included hypocalcemia and joint pain during the initiation of treatment and symptomatic hypercalcemia after the cessation of treatment. Growth was not significantly impaired in these skeletally immature patients. This case series demonstrates how a weight-adjusted denosumab dose can effectively treat NL/MGCLs and provides laboratory data for consideration of the timing of monitoring for known side effects.
- Published
- 2020
- Full Text
- View/download PDF
13. Sensory nerves regulate mesenchymal stromal cell lineage commitment by tuning sympathetic tones
- Author
-
Ruoxian Deng, Wen Yuan, Xiao Wang, Gehua Zhen, Mi Yang, Mei Wan, Peng Xue, Huajiang Chen, Dayu Pan, Zemin Ling, Shen Liu, Bo Hu, Bo Gao, Weiping Su, Hao Chen, Janet L. Crane, Xiaonan Liu, Shuangfei Ni, Panfeng Wu, Zengwu Shao, Ying Zhang, Xu Cao, Xiao Lv, Yusheng Li, and Lei Wang
- Subjects
0301 basic medicine ,Stromal cell ,Sensory Receptor Cells ,Bone healing ,Dinoprostone ,03 medical and health sciences ,Gene Knockout Techniques ,Mice ,0302 clinical medicine ,Osteogenesis ,medicine ,Animals ,Prostaglandin E2 ,Mice, Knockout ,Adipogenesis ,Osteoblasts ,Chemistry ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,General Medicine ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Cyclooxygenase 2 ,030220 oncology & carcinogenesis ,lipids (amino acids, peptides, and proteins) ,Bone marrow ,Receptors, Prostaglandin E, EP4 Subtype ,medicine.drug ,Sensory nerve ,Adult stem cell ,Research Article - Abstract
The sensory nerve was recently identified as being involved in regulation of bone mass accrual. We previously discovered that prostaglandin E(2) (PGE2) secreted by osteoblasts could activate sensory nerve EP4 receptor to promote bone formation by inhibiting sympathetic activity. However, the fundamental units of bone formation are active osteoblasts, which originate from mesenchymal stromal/stem cells (MSCs). Here, we found that after sensory denervation, knockout of the EP4 receptor in sensory nerves, or knockout of COX-2 in osteoblasts, could significantly promote adipogenesis and inhibit osteogenesis in adult mice. Furthermore, injection of SW033291 (a small molecule that locally increases the PGE2 level) or propranolol (a beta blocker) significantly promoted osteogenesis and inhibited adipogenesis. This effect of SW033291, but not propranolol, was abolished in conditional EP4-KO mice under normal conditions or in the bone repair process. We conclude that the PGE2/EP4 sensory nerve axis could regulate MSC differentiation in bone marrow of adult mice.
- Published
- 2020
14. Kaposiform lymphangiomatosis treated with multimodal therapy improves coagulopathy and reduces blood angiopoietin-2 levels
- Author
-
Maxim Itkin, Clifford Takemoto, Mara Coyan, Elisa Boscolo, Jackie Manfredo, Janet L. Crane, Timothy D. Le Cras, and Denise M. Adams
- Subjects
Male ,medicine.medical_specialty ,Vincristine ,Kasabach-Merritt Syndrome ,Gastroenterology ,Article ,Angiopoietin-2 ,03 medical and health sciences ,0302 clinical medicine ,Prednisone ,Internal medicine ,Consumptive Coagulopathy ,medicine ,Coagulopathy ,Humans ,Child ,Lymphangiomatosis ,Sarcoma, Kaposi ,business.industry ,Multimodal therapy ,Thrombosis ,Hematology ,medicine.disease ,Prognosis ,Combined Modality Therapy ,Regimen ,Oncology ,030220 oncology & carcinogenesis ,Pediatrics, Perinatology and Child Health ,Hemangioendothelioma ,Biomarker (medicine) ,business ,030215 immunology ,medicine.drug - Abstract
Kaposiform lymphangiomatosis (KLA) is a rare, life-threatening congenital lymphatic malformation. Diagnosis is often delayed due to complex indistinct symptoms. Blood angiopoietin-2 (ANG2) levels are elevated in KLA and may be useful as a biomarker to monitor disease status. We report a 7-year-old male child with easy bruising, inguinal swelling, and consumptive coagulopathy, diagnosed with KLA. A multimodal treatment regimen of prednisone, sirolimus, vincristine, and adjunctive zoledronate was used. Plasma ANG2 levels were highly elevated at diagnosis but decreased during treatment. The patient showed significant clinical improvement over a 38-month period and normalization of ANG2 levels correlated with resolution of the coagulopathy.
- Published
- 2020
15. Preservation of type H vessels and osteoblasts by enhanced preosteoclast platelet-derived growth factor type BB attenuates glucocorticoid-induced osteoporosis in growing mice
- Author
-
Zixing Ye, Zhuying Xia, Shan Lv, Xu Cao, Guoxian Ding, Ping Yang, Yi Peng, Yan Wang, and Janet L. Crane
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Mice, 129 Strain ,Histology ,Physiology ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Cathepsin K ,Becaplermin ,Osteoclasts ,Mice, Transgenic ,Article ,Bone resorption ,Bone remodeling ,Mice ,Random Allocation ,03 medical and health sciences ,Osteoclast ,Internal medicine ,medicine ,Animals ,Glucocorticoids ,Osteoblasts ,Chemistry ,Growth factor ,Age Factors ,Osteopetrosis ,Osteoblast ,medicine.disease ,Resorption ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Blood Vessels ,Osteoporosis ,Bone Remodeling - Abstract
Survival of chronic diseases in childhood is often achieved utilizing glucocorticoids, but comes with significant side effects, including glucocorticoid-induced osteoporosis (GIO). Knowledge of the mechanism of GIO is limited to the adult skeleton. We explored the effect of genetic loss and inhibition of cathepsin K (Ctsk) as a potential treatment target in a young GIO mouse model as genetic loss of cathepsin K results in a mild form of osteopetrosis secondary to impaired osteoclast bone resorption with maintenance of bone formation. We first characterized the temporal osteoclast and osteoblast progenitor populations in Ctsk(−/−) and wild type (WT) mice in the primary and secondary spongiosa, as sites representative of trabecular bone modeling and remodeling, respectively. In the primary spongiosa, Ctsk(−/−) mice had decreased numbers of osteoclasts at young ages (2 and 4 weeks) and increased osteoblast lineage cells at later age (8 weeks) relative to WT littermates. In the secondary spongiosa, Ctsk(−/−) mice had greater numbers of osteoclasts and preosteoblasts relative to WT littermates. We next developed a young GIO mouse model with prednisolone 10 mg/m(2)/day injected intraperitoneally daily from 2 through 6 weeks of age. Overall, WT-prednisolone mice had lower bone volume per tissue volume, whereas Ctsk(−/−)-prednisolone mice maintained a similar bone volume relative to Ctsk(−/−)-vehicle controls. WT-prednisolone mice exhibited a decreased number of osteoclasts, tartrate-resistant acid phosphatase and platelet-derived growth factor type BB (PDGF-BB) co-positive cells, type H endothelial cells, and osteoblasts relative to WT-vehicle mice in both the primary and secondary spongiosa. Interestingly, Ctsk(−/−)-prednisolone mice demonstrated a paradoxical response with increased numbers of all parameters in primary spongiosa and no change in secondary spongiosa. Finally, treatment with a cathepsin K inhibitor prevented WT-prednioslone decline in osteoclasts, osteoblasts, type H vessels, and bone volume. These data demonstrate that cells in the primary and secondary spongiosa respond differently to glucocorticoids and genetic manipulation. Inhibition of osteoclast resorption that preserves osteoclast coupling factors, such as through inhibition of cathepsin K, may be a potential preventive treatment strategy against GIO in the growing skeleton.
- Published
- 2018
- Full Text
- View/download PDF
16. Multiple endocrine neoplasia type 1 presenting with concurrent insulinoma and prolactinoma in early-adolescence
- Author
-
Janet L. Crane, Yasmin Akhtar, and Angela Verardo
- Subjects
Pediatrics ,medicine.medical_specialty ,endocrine system diseases ,030209 endocrinology & metabolism ,Case Report ,Neuroendocrine tumors ,Hypoglycemia ,lcsh:Diseases of the endocrine glands. Clinical endocrinology ,03 medical and health sciences ,0302 clinical medicine ,medicine ,MEN1 ,Prolactinoma ,030212 general & internal medicine ,Multiple endocrine neoplasia ,Insulinoma ,lcsh:RC648-665 ,business.industry ,lcsh:RJ1-570 ,Autosomal dominant trait ,lcsh:Pediatrics ,medicine.disease ,business ,Primary hyperparathyroidism - Abstract
Background Multiple Endocrine Neoplasia Type 1 (MEN1) is a rare autosomal dominant disease that generally presents with primary hyperparathyroidism. However, initial presentation may vary and continued reevaluation of etiology of symptoms is required for appropriate diagnosis. Case Presentation Twelve year old female presented with altered mental status that self-resolved and hypoglycemia. Laboratory evaluation revealed pituitary dysfunction with central hypothyroidism and adrenal insufficiency in the setting of hyperprolactinemia. Macroadenoma was confirmed on imaging. Despite medical treatment of pituitary hormone disorders, she continued to have significant hypoglycemia and further workup revealed hyperinsulinism. Insulinoma was identified and confirmed by endoscopic ultrasound. Hypoglycemia resolved after laproscopic enucleation of the insulinoma. Conclusion Children presenting with one endocrine tumor should be investigated for other potential endocrine tumors. Multiple imaging modalities may be required to confidently identify neuroendocrine tumors for appropriate surgical intervention.
- Published
- 2018
- Full Text
- View/download PDF
17. Oxidized phospholipids are ligands for LRP6
- Author
-
Weiqi Lei, Haiyun Liu, Bing Yu, Changjun Li, Xu Cao, Yu Chai, Weiping Su, Lei Wang, Bin Yu, Xiaonan Liu, Janet L. Crane, and Mei Wan
- Subjects
0301 basic medicine ,Histology ,Physiology ,Endocrinology, Diabetes and Metabolism ,030209 endocrinology & metabolism ,Endocytosis ,Article ,lcsh:Physiology ,03 medical and health sciences ,0302 clinical medicine ,Lipid oxidation ,medicine ,Receptor ,lcsh:QH301-705.5 ,lcsh:QP1-981 ,Chemistry ,Mesenchymal stem cell ,Wnt signaling pathway ,LRP6 ,LRP5 ,Osteoblast ,3. Good health ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Biology (General) - Abstract
Low-density lipoprotein receptor–related protein 6 (LRP6) is a co-receptor for Wnt signaling and can be recruited by multiple growth factors/hormones to their receptors facilitating intracellular signaling activation. The ligands that bind directly to LRP6 have not been identified. Here, we report that bioactive oxidized phospholipids (oxPLs) are native ligands of LRP6, but not the closely related LRP5. oxPLs are products of lipid oxidation involving in pathological conditions such as hyperlipidemia, atherosclerosis, and inflammation. We found that cell surface LRP6 in bone marrow mesenchymal stromal cells (MSCs) decreased rapidly in response to increased oxPLs in marrow microenvironment. LRP6 directly bound and mediated the uptake of oxPLs by MSCs. oxPL-LRP6 binding induced LRP6 endocytosis through a clathrin-mediated pathway, decreasing responses of MSCs to osteogenic factors and diminishing osteoblast differentiation ability. Thus, LRP6 functions as a receptor and molecular target of oxPLs for their adverse effect on MSCs, revealing a potential mechanism underlying atherosclerosis-associated bone loss., Bone loss: revealing a molecular cause of ‘numb’ stem cells A constituent of oxidized ‘bad cholesterol’ blocks essential signaling processes leading to pathogenic bone loss. LRP6 is a crucial receptor involved in multiple physiological processes, including bone loss; however, direct pathogenic modulators of the receptor have yet to be elucidated. Now, John Hopkins University School of Medicine’s Mei Wan, with a US and Chinese research team, has discovered that oxPL, a byproduct of the oxidization of cholesterol carrier LDL, binds directly to LRP6 and causes its removal from the surface of bone marrow stem cells. As a result, these stem cells are unable to sense the external signaling molecules that drive bone growth. oxPLs are products of diseases such as hyperlipidemia and atherosclerosis, and this paper helps to reveal their pathogenesis and offers potential targets for therapeutic interventions.
- Published
- 2018
- Full Text
- View/download PDF
18. Bone Matrix IGF-1 in Bone Remodeling
- Author
-
Janet L. Crane
- Subjects
Paracrine signalling ,medicine.anatomical_structure ,Chemistry ,Growth factor ,medicine.medical_treatment ,Osteocyte ,medicine ,Osteoblast ,Bone marrow ,Autocrine signalling ,Bone resorption ,Cell biology ,Bone remodeling - Abstract
In bone remodeling, bone resorption and formation are coupled to ensure that the same amount of bone that is resorbed is replaced. Insulin-like growth factor type 1 (IGF-1) is a critical factor involved in coupled bone remodeling. IGF-1 is a unique peptide that can mediate endocrine, paracrine, and autocrine effects. IGF-1 is most often bound to proteins which aid in tissue-specific targeting, including deposition into the bone matrix. Bone matrix IGF-1 can act in a delayed paracrine manner to stimulate osteoblast and osteocyte differentiation, aiding in bone formation. IGF-1 interacts with multiple factors in the bone marrow microenvironment to regulate the bone remodeling process.
- Published
- 2020
- Full Text
- View/download PDF
19. Bisphosphonate Therapy for Treating Osteonecrosis in Pediatric Leukemia Patients: A Systematic Review
- Author
-
Kathy Ruble, Janet L. Crane, Shanaz M. Daneshdoost, Jad M. El Abiad, Carol D. Morris, Lynne C. Jones, and Adam S. Levin
- Subjects
medicine.medical_specialty ,MEDLINE ,Antineoplastic Agents ,CINAHL ,Pediatrics ,Article ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Vitamin D and neurology ,Medicine ,Humans ,Vitamin D ,Adverse effect ,Child ,Pediatric leukemia ,Leukemia ,Bone Density Conservation Agents ,Diphosphonates ,business.industry ,Osteonecrosis ,Hematology ,medicine.disease ,Systematic review ,Oncology ,030220 oncology & carcinogenesis ,Pediatrics, Perinatology and Child Health ,Calcium ,Bisphosphonate therapy ,business ,030215 immunology - Abstract
BACKGROUND: Despite improved outcomes in children with leukemia, complications such as osteonecrosis are common. We conducted a systematic review to investigate the role of bisphosphonates in reducing pain, improving mobility, and stabilizing lesions in pediatric leukemia survivors. METHODS: Using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines, we searched the PubMed, Embase, Cochrane, Web of Science, Scopus, CINAHL, and ClinicalTrials.gov databases. Five of 221 articles retrieved met our inclusion criteria. RESULTS: Bisphosphonates, especially when combined with dietary calcium and vitamin D supplements and physical therapy (“supplements/PT”) were associated with improved pain and mobility in 54% and 50% of patients, respectively. A significantly greater proportion of patients treated with bisphosphonates (83%) reported mild/moderate pain or no pain compared with those with supplements/PT alone (36%) (P < 0.001). Sixty-six percent of patients treated with bisphosphonates achieved improved/full mobility compared with 27% of those treated with supplements/PT alone (P = 0.02). However, 46% of patients showed progressive joint destruction despite bisphosphonate therapy. No adverse events were reported, except for acute phase reactions to intravenous therapies. CONCLUSIONS: Bisphosphonates, when combined with supplements/PT, were associated with less pain and improved mobility, but not prevention of joint destruction in pediatric leukemia patients with osteonecrosis.
- Published
- 2019
20. Sensory innervation in porous endplates by Netrin-1 from osteoclasts mediates PGE2-induced spinal hypersensitivity in mice
- Author
-
Bo Hu, Ruoxian Deng, Yong Cao, Shuangfei Ni, Xinzhong Dong, Dayu Pan, Gehua Zhen, Amit Jain, Shadpour Demehri, Huabin Qi, Janet L. Crane, Xiao Lyu, Yusheng Li, Tianding Wu, Hao Chen, Panfeng Wu, Xu Cao, Xiao Wang, Hongbin Lu, Xuenong Zou, Mei Wan, Jianzhong Hu, Yun Guan, Richard L. Skolasky, and Zemin Ling
- Subjects
0301 basic medicine ,Male ,Lumbar spine instability ,Aging ,General Physics and Astronomy ,Osteoclasts ,Chronic pain ,0302 clinical medicine ,Netrin ,Medicine ,lcsh:Science ,Mice, Knockout ,Multidisciplinary ,Lumbar Vertebrae ,biology ,Behavior, Animal ,Netrin-1 ,Spinal pain ,medicine.anatomical_structure ,RANKL ,Hyperalgesia ,Signal transduction ,Porosity ,Signal Transduction ,musculoskeletal diseases ,Sensory Receptor Cells ,Prostaglandin E2 receptor ,Science ,Pain ,Sensory system ,Motor Endplate ,General Biochemistry, Genetics and Molecular Biology ,Article ,Dinoprostone ,03 medical and health sciences ,Osteoclast ,Hypersensitivity ,Animals ,Humans ,Author Correction ,Bone ,business.industry ,technology, industry, and agriculture ,General Chemistry ,Spine ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,biology.protein ,lcsh:Q ,business ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Spinal pain is a major clinical problem, however, its origins and underlying mechanisms remain unclear. Here we report that in mice, osteoclasts induce sensory innervation in the porous endplates which contributes to spinal hypersensitivity in mice. Sensory innervation of the porous areas of sclerotic endplates in mice was confirmed. Lumbar spine instability (LSI), or aging, induces spinal hypersensitivity in mice. In these conditions, we show that there are elevated levels of PGE2 which activate sensory nerves, leading to sodium influx through Nav 1.8 channels. We show that knockout of PGE2 receptor 4 in sensory nerves significantly reduces spinal hypersensitivity. Inhibition of osteoclast formation by knockout Rankl in the osteocytes significantly inhibits LSI-induced porosity of endplates, sensory innervation, and spinal hypersensitivity. Knockout of Netrin-1 in osteoclasts abrogates sensory innervation into porous endplates and spinal hypersensitivity. These findings suggest that osteoclast-initiated porosity of endplates and sensory innervation are potential therapeutic targets for spinal pain., Spinal pain is a major clinical problem. Here the authors show that osteoclasts create porous area of endplates of the vertebrae and sensory innervation of porous endplates by Netrin-1 release from osteoclasts mediates PGE2-induced spinal hypersensitivity in mice.
- Published
- 2019
21. Subchondral bone osteoclasts induce sensory innervation and osteoarthritis pain
- Author
-
Yun Guan, Yusheng Li, Zhiyong Chen, Gehua Zhen, Mei Wan, Qin Zheng, Holger K. Eltzschig, Mi Yang, Chuanlong Wu, Xinzhong Dong, Jianxi Zhu, Manman Gao, Julia S. Kuliwaba, Richard L. Skolasky, Yihe Hu, Hongwei Ouyang, Shouan Zhu, Lei Wang, David M. Findlay, Ya Yang, Feng Quan Zhou, Shuangfei Ni, Janet L. Crane, Tianding Wu, Bo Gao, Xu Cao, Yong Cao, and Senbo An
- Subjects
0301 basic medicine ,musculoskeletal diseases ,Male ,medicine.medical_specialty ,Deleted in Colorectal Cancer ,Sensory Receptor Cells ,Osteoclasts ,Pain ,Osteoarthritis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Dorsal root ganglion ,Osteoclast ,Internal medicine ,Ganglia, Spinal ,medicine ,Animals ,biology ,business.industry ,General Medicine ,Netrin-1 ,medicine.disease ,DCC Receptor ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,RANKL ,030220 oncology & carcinogenesis ,Joint pain ,biology.protein ,Neuron ,Bone Remodeling ,medicine.symptom ,business ,Sensory nerve ,Research Article - Abstract
Joint pain is the defining symptom of osteoarthritis (OA) but its origin and mechanisms remain unclear. Here, we investigated an unprecedented role of osteoclast-initiated subchondral bone remodeling in sensory innervation for OA pain. We show that osteoclasts secrete netrin-1 to induce sensory nerve axonal growth in subchondral bone. Reduction of osteoclast formation by knockout of receptor activator of nuclear factor kappa-B ligand (Rankl) in osteocytes inhibited the growth of sensory nerves into subchondral bone, dorsal root ganglion neuron hyperexcitability, and behavioral measures of pain hypersensitivity in OA mice. Moreover, we demonstrated a possible role for netrin-1 secreted by osteoclasts during aberrant subchondral bone remodeling in inducing sensory innervation and OA pain through its receptor DCC (deleted in colorectal cancer). Importantly, knockout of Netrin1 in tartrate-resistant acid phosphatase-positive (TRAP-positive) osteoclasts or knockdown of Dcc reduces OA pain behavior. In particular, inhibition of osteoclast activity by alendronate modifies aberrant subchondral bone remodeling and reduces innervation and pain behavior at the early stage of OA. These results suggest that intervention of the axonal guidance molecules (e.g., netrin-1) derived from aberrant subchondral bone remodeling may have therapeutic potential for OA pain.
- Published
- 2018
22. Systemic neutralization of TGF-β attenuates osteoarthritis
- Author
-
Xu Cao, Xiao Wang, Gehua Zhen, Liang Xie, Janet L. Crane, Tao Qiu, Qianming Chen, Francis Tintani, Fengfeng Li, and Mei Wan
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Stromal cell ,business.industry ,General Neuroscience ,Mesenchymal stem cell ,Osteoblast ,Osteoarthritis ,medicine.disease ,General Biochemistry, Genetics and Molecular Biology ,Pathogenesis ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,History and Philosophy of Science ,Systemic administration ,medicine ,Stem cell ,business ,Transforming growth factor - Abstract
Osteoarthritis (OA) is a major source of pain and disability worldwide with no effective medical therapy due to poor understanding of its pathogenesis. Transforming growth factor β (TGF-β) has been reported to play a role in subchondral bone pathology and articular cartilage degeneration during the progression of OA. In this study, we demonstrated that systemic use of a TGF-β-neutralizing antibody (1D11) attenuates OA progression by targeting subchondral bone pathological features in rodent OA models. Systemic administration of 1D11 preserves the subchondral bone microarchitecture, preventing articular cartilage degeneration by inhibition of excessive TGF-β activity, in both subchondral bone and the circulation. Moreover, the aberrant increases in the numbers of blood vessels, nestin(+) mesenchymal stromal/stem cells, and osterix(+) osteoblast progenitors were normalized by 1D11 systemic injection. Thus, systemic neutralization of excessive TGF-β ligands effectively prevented OA progression in animal models, with promising clinical implications for OA treatment.
- Published
- 2016
- Full Text
- View/download PDF
23. Halofuginone attenuates osteoarthritis by inhibition of TGF-β activity and H-type vessel formation in subchondral bone
- Author
-
Xu Cao, Xin Jin, Mei Wan, Min Xie, Changjun Li, Hui Xie, Long Wang, Tao Qiu, Zhuang Cui, Liang Xie, Gehua Zhen, Sheng Ding, Qin Bian, Bin Yu, and Janet L. Crane
- Subjects
Cartilage, Articular ,Male ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Immunology ,Osteoclasts ,Arthritis ,Osteoarthritis ,Bone and Bones ,General Biochemistry, Genetics and Molecular Biology ,Bone resorption ,Bone remodeling ,Mice ,Random Allocation ,03 medical and health sciences ,Piperidines ,Rheumatology ,Transforming Growth Factor beta ,Animals ,Immunology and Allergy ,Medicine ,Anterior Cruciate Ligament ,Bone Resorption ,Basic and Translational Research ,Aggrecan ,Quinazolinones ,Halofuginone ,business.industry ,ADAMTS ,medicine.disease ,Rats ,Mice, Inbred C57BL ,Treatment ,Disease Models, Animal ,030104 developmental biology ,Rats, Inbred Lew ,Disease Progression ,Bone Remodeling ,business ,medicine.drug ,Calcification - Abstract
ObjectivesExamine whether osteoarthritis (OA) progression can be delayed by halofuginone in anterior cruciate ligament transection (ACLT) rodent models.Methods3-month-old male C57BL/6J (wild type; WT) mice and Lewis rats were randomised to sham-operated, ACLT-operated, treated with vehicle, or ACLT-operated, treated with halofuginone. Articular cartilage degeneration was graded using the Osteoarthritis Research Society International (OARSI)-modified Mankin criteria. Immunostaining, flow cytometry, RT-PCR and western blot analyses were conducted to detect relative protein and RNA expression. Bone micro CT (μCT) and CT-based microangiography were quantitated to detect alterations of microarchitecture and vasculature in tibial subchondral bone.ResultsHalofuginone attenuated articular cartilage degeneration and subchondral bone deterioration, resulting in substantially lower OARSI scores. Specifically, we found that proteoglycan loss and calcification of articular cartilage were significantly decreased in halofuginone-treated ACLT rodents compared with vehicle-treated ACLT controls. Halofuginone reduced collagen X (Col X), matrix metalloproteinase-13 and A disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS 5) and increased lubricin, collagen II and aggrecan. In parallel, halofuginone-attenuated uncoupled subchondral bone remodelling as defined by reduced subchondral bone tissue volume, lower trabecular pattern factor (Tb.pf) and increased thickness of subchondral bone plate compared with vehicle-treated ACLT controls. We found that halofuginone exerted protective effects in part by suppressing Th17-induced osteoclastic bone resorption, inhibiting Smad2/3-dependent TGF-β signalling to restore coupled bone remodelling and attenuating excessive angiogenesis in subchondral bone.ConclusionsHalofuginone attenuates OA progression by inhibition of subchondral bone TGF-β activity and aberrant angiogenesis as a potential preventive therapy for OA.
- Published
- 2015
- Full Text
- View/download PDF
24. Aberrant Activation of TGF-β in Subchondral Bone at the Onset of Rheumatoid Arthritis Joint Destruction
- Author
-
Li-Wei Zheng, Wenlong Liu, Xin Xu, Xu Cao, Janet L. Crane, Xuedong Zhou, Gehua Zhen, Liang Xie, and Qin Bian
- Subjects
musculoskeletal diseases ,Autoimmune disease ,biology ,business.industry ,Endocrinology, Diabetes and Metabolism ,Mesenchymal stem cell ,Transforming growth factor beta ,medicine.disease ,Bone resorption ,Immune system ,medicine.anatomical_structure ,Rheumatoid arthritis ,Immunology ,Cancer research ,biology.protein ,Medicine ,Orthopedics and Sports Medicine ,Tibia ,Bone marrow ,business - Abstract
Rheumatoid arthritis (RA) is an autoimmune disease that often leads to joint destruction. A myriad of drugs targeting the immune abnormalities and downstream inflammatory cascades have been developed, but the joint destruction is not effectively halted. Here we report that aberrant activation of TGF-β in the subchondral bone marrow by immune response increases osteoprogenitors and uncoupled bone resorption and formation in RA mouse/rat models. Importantly, either systemic or local blockade of TGF-β activity in the subchondral bone attenuated articular cartilage degeneration in RA. Moreover, conditional deletion of TGF-β receptor II (Tgfbr2) in nestin-positive cells also effectively halted progression of RA joint destruction. Our data demonstrate that aberrant activation of TGF-β in the subchondral bone is involved at the onset of RA joint cartilage degeneration. Thus, modulation of subchondral bone TGF-β activity could be a potential therapy for RA joint destruction.
- Published
- 2015
- Full Text
- View/download PDF
25. Transforming growth factor-β in stem cells and tissue homeostasis
- Author
-
Xu Cao, Xuedong Zhou, Janet L. Crane, Quan Yuan, Liwei Zheng, Gehua Zhen, and Xin Xu
- Subjects
0301 basic medicine ,Histology ,lcsh:QP1-981 ,Physiology ,Endocrinology, Diabetes and Metabolism ,Context (language use) ,Review Article ,Biology ,medicine.disease ,lcsh:Physiology ,3. Good health ,Cell biology ,Extracellular matrix ,03 medical and health sciences ,030104 developmental biology ,Immune system ,lcsh:Biology (General) ,Fibrosis ,medicine ,Progenitor cell ,Stem cell ,lcsh:QH301-705.5 ,Tissue homeostasis ,Transforming growth factor - Abstract
TGF-β 1–3 are unique multi-functional growth factors that are only expressed in mammals, and mainly secreted and stored as a latent complex in the extracellular matrix (ECM). The biological functions of TGF-β in adults can only be delivered after ligand activation, mostly in response to environmental perturbations. Although involved in multiple biological and pathological processes of the human body, the exact roles of TGF-β in maintaining stem cells and tissue homeostasis have not been well-documented until recent advances, which delineate their functions in a given context. Our recent findings, along with data reported by others, have clearly shown that temporal and spatial activation of TGF-β is involved in the recruitment of stem/progenitor cell participation in tissue regeneration/remodeling process, whereas sustained abnormalities in TGF-β ligand activation, regardless of genetic or environmental origin, will inevitably disrupt the normal physiology and lead to pathobiology of major diseases. Modulation of TGF-β signaling with different approaches has proven effective pre-clinically in the treatment of multiple pathologies such as sclerosis/fibrosis, tumor metastasis, osteoarthritis, and immune disorders. Thus, further elucidation of the mechanisms by which TGF-β is activated in different tissues/organs and how targeted cells respond in a context-dependent way can likely be translated with clinical benefits in the management of a broad range of diseases with the involvement of TGF-β., Growth factor: Activation in health and disease Targeting a critical growth factor involved in bone and other tissue remodeling could help treat osteoarthritis and other skeletal disorders. A team led by Zhou Xuedong from Sichuan University in Chengdu, China, and Xu Cao from the Johns Hopkins University School of Medicine in Baltimore, Maryland, USA, review the ways in which temporal and spatial activation of transforming growth factor-β (TGF-β), a multi-functional signaling molecule, are needed for proper tissue development and regulation of stem cells throughout the body. Looking at the skeletal system in particular, the researchers discuss how TGF-β controls the balance between bone resorption and bone formation. Faulty TGF-β signaling can lead to numerous bone-associated disorders, including rare genetic diseases and metastatic cancers. The authors also summarize clinical efforts to modulate TGF-β with drugs for the treatment of osteoarthritis and other conditions.
- Published
- 2018
- Full Text
- View/download PDF
26. Aberrant TGF-β activation in bone tendon insertion induces enthesopathy-like disease
- Author
-
Fengfeng Li, Liang Xie, Xu Cao, Ruoxian Deng, Gehua Zhen, Yiguo Wang, Manman Gao, Janet L. Crane, Mei Wan, Cunyi Fan, Xiao Wang, Ping Yang, and Xiaohua Jia
- Subjects
0301 basic medicine ,Male ,Pathology ,medicine.medical_specialty ,Stromal cell ,Bone disease ,Osteoclasts ,Mice, Transgenic ,Enthesopathy ,Bone and Bones ,Bone remodeling ,Tendons ,03 medical and health sciences ,Mice ,Transforming Growth Factor beta ,medicine ,Animals ,Mice, Knockout ,Chemistry ,Receptor, Transforming Growth Factor-beta Type II ,Fibrocartilage ,Mesenchymal Stem Cells ,General Medicine ,X-Ray Microtomography ,medicine.disease ,Enthesis ,Antibodies, Neutralizing ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Cartilage ,Phenotype ,Bone marrow ,Bone Remodeling ,Collagen ,Calcification ,Research Article ,Signal Transduction - Abstract
Enthesopathy is a disorder of bone, tendon, or ligament insertion. It represents one-fourth of all tendon-ligament diseases and is one of the most difficult tendon-ligament disorders to treat. Despite its high prevalence, the exact pathogenesis of this condition remains unknown. Here, we show that TGF-β was activated in both a semi-Achilles tendon transection (SMTS) mouse model and in a dorsiflexion immobilization (DI) mouse model of enthesopathy. High concentrations of active TGF-β recruited mesenchymal stromal stem cells (MSCs) and led to excessive vessel formation, bone deterioration, and fibrocartilage calcification. Transgenic expression of active TGF-β1 in bone also induced enthesopathy with a phenotype similar to that observed in SMTS and DI mice. Systemic inhibition of TGF-β activity by injection of 1D11, a TGF-β-neutralizing antibody, but not a vehicle antibody, attenuated the excessive vessel formation and restored uncoupled bone remodeling in SMTS mice. 1D11-treated SMTS fibrocartilage had increased proteoglycan and decreased collagen X and matrix metalloproteinase 13 expression relative to control antibody treatment. Notably, inducible knockout of the TGF-β type II receptor in mouse MSCs preserved the bone microarchitecture and fibrocartilage composition after SMTS relative to the WT littermate controls. Thus, elevated levels of active TGF-β in the enthesis bone marrow induce the initial pathological changes of enthesopathy, indicating that TGF-β inhibition could be a potential therapeutic strategy.
- Published
- 2018
27. PTH Receptor Signaling in Osteoblasts Regulates Endochondral Vascularization in Maintenance of Postnatal Growth Plate
- Author
-
Tao Qiu, Peter J. Newman, Xu Cao, William W. Lu, Lingling Xian, Matthew J. Hilton, Janet L. Crane, and Chunyi Wen
- Subjects
medicine.medical_specialty ,Parathyroid hormone receptor ,Chemistry ,Angiogenesis ,Endocrinology, Diabetes and Metabolism ,Cartilage ,Parathyroid hormone ,Chondrocyte ,Bone remodeling ,Endocrinology ,medicine.anatomical_structure ,Internal medicine ,Bone cell ,medicine ,Orthopedics and Sports Medicine ,Endochondral ossification - Abstract
Longitudinal growth of postnatal bone requires precise control of growth plate cartilage chondrocytes and subsequent osteogenesis and bone formation. Little is known about the role of angiogenesis and bone remodeling in maintenance of cartilaginous growth plate. Parathyroid hormone (PTH) stimulates bone remodeling by activating PTH receptor (PTH1R). Mice with conditional deletion of PTH1R in osteoblasts showed disrupted trabecular bone formation. The mice also exhibited postnatal growth retardation with profound defects in growth plate cartilage, ascribable predominantly to a decrease in number of hypertrophic chondrocytes, resulting in premature fusion of the growth plate and shortened long bones. Further characterization of hypertrophic zone and primary spongiosa revealed that endochondral angiogenesis and vascular invasion of the cartilage were impaired, which was associated with aberrant chondrocyte maturation and cartilage development. These studies reveal that PTH1R signaling in osteoblasts regulates cartilaginous growth plate for postnatal growth of bone.
- Published
- 2015
- Full Text
- View/download PDF
28. Programmed cell senescence in skeleton during late puberty
- Author
-
Yu Chai, Xu Cao, Feng Quan Zhou, Changjun Li, Bin Yu, Hao Chen, Mei Wan, Lei Wang, Xiang-Hang Luo, Janet L. Crane, Bo Gao, and Peisong Gao
- Subjects
0301 basic medicine ,Senescence ,Male ,Adolescent ,Science ,General Physics and Astronomy ,Mice, Transgenic ,macromolecular substances ,Cell fate determination ,Biology ,Methylation ,General Biochemistry, Genetics and Molecular Biology ,Article ,Epigenesis, Genetic ,Histones ,Nestin ,03 medical and health sciences ,Histone H3 ,Mice ,Osteogenesis ,Animals ,Humans ,Enhancer of Zeste Homolog 2 Protein ,Sexual Maturation ,Progenitor cell ,lcsh:Science ,Cellular Senescence ,Bone growth ,Mice, Knockout ,Multidisciplinary ,Bone Development ,EZH2 ,Puberty ,Mesenchymal Stem Cells ,General Chemistry ,Cell biology ,030104 developmental biology ,Histone ,Histone methyltransferase ,biology.protein ,Osteoporosis ,lcsh:Q ,Female - Abstract
Mesenchymal stem/progenitor cells (MSPCs) undergo rapid self-renewal and differentiation, contributing to fast skeletal growth during childhood and puberty. It remains unclear whether these cells change their properties during late puberty to young adulthood, when bone growth and accrual decelerate. Here we show that MSPCs in primary spongiosa of long bone in mice at late puberty undergo normal programmed senescence, characterized by loss of nestin expression. MSPC senescence is epigenetically controlled by the polycomb histone methyltransferase enhancer of zeste homolog 2 (Ezh2) and its trimethylation of histone H3 on Lysine 27 (H3K27me3) mark. Ezh2 maintains the repression of key cell senescence inducer genes through H3K27me3, and deletion of Ezh2 in early pubertal mice results in premature cellular senescence, depleted MSPCs pool, and impaired osteogenesis as well as osteoporosis in later life. Our data reveals a programmed cell fate change in postnatal skeleton and unravels a regulatory mechanism underlying this phenomenon., Mesenchymal stem cells are essential for bone development, but it is unclear if their activity is maintained after late puberty, when bone growth decelerates. The authors show that during late puberty in mice, these cells undergo senescence under the epigenetic control of Ezh2.
- Published
- 2017
29. Cervical cancer cell-derived angiopoietins promote tumor progression
- Author
-
Zehua Wang, Ruiqing Dong, Jing Guo, Na Chen, Ping Yang, Dongyun Yang, Bangxing Huang, Jing Cai, Janet L. Crane, and Xiaoqing Yi
- Subjects
0301 basic medicine ,Adult ,Angiogenesis ,Carcinogenesis ,Vesicular Transport Proteins ,Uterine Cervical Neoplasms ,Vimentin ,medicine.disease_cause ,03 medical and health sciences ,Mice ,Adjuvant therapy ,medicine ,Angiopoietin-1 ,Animals ,Humans ,Neoplasm Invasiveness ,RC254-282 ,Aged ,Cell Proliferation ,Cervical cancer ,biology ,Neovascularization, Pathologic ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Angiopoietins ,General Medicine ,Middle Aged ,medicine.disease ,Prognosis ,Angiopoietin receptor ,Receptor, TIE-2 ,Xenograft Model Antitumor Assays ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Tumor progression ,Cancer research ,biology.protein ,Female ,Neoplasm Recurrence, Local ,business ,HeLa Cells - Abstract
Metastatic or recurrent cervical cancer has limited treatment options and a high rate of mortality. Although anti-vascular endothelial growth factor drugs have shown great promise as a therapeutic target for treatment of advanced cervical cancer, drug resistance and class-specific side effects negate long-term benefits. The identification of alternative anti-angiogenic factors will be critical for future drug development for advanced or recurrent cervical cancer. In this study, we found that angiopoietins and Tie receptors were highly expressed in cervical cancer cells. Tie-2 expression in tumor cells predicted poorer prognosis. Wound closure assay and Transwell assay showed that upregulated or downregulated Ang-1 and Ang-2 expression promoted or reduced cervical cancer cell lines migration and invasion, respectively. In subcutaneous xenograft models of cervical cancer, downregulation of Ang-1 and Ang-2 attenuated tumor growth. The expression of vimentin and endomucin and microvessel density were all significantly decreased in the siAng-1 group and siAng-2 group relative to the infection control group. Our data support that dual inhibition of Ang-1 and Ang-2 may be an alternative target for anti-angiogenic adjuvant therapy in advanced or recurrent cervical squamous cell cancer.
- Published
- 2017
30. Inhibition of overactive TGF-β attenuates progression of heterotopic ossification in mice
- Author
-
Cunyi Fan, Xiao Wang, Janet L. Crane, Liang Xie, Manli Tu, Mei Wan, Ruoxian Deng, Fengfeng Li, Yiguo Wang, Yuji Mishina, Hao Chen, Manman Gao, Gehua Zhen, Xu Cao, Shen Liu, Ping Yang, and Bo Gao
- Subjects
0301 basic medicine ,Male ,Becaplermin ,General Physics and Astronomy ,Osteoclasts ,Bone remodeling ,Tendons ,Fracture Fixation, Internal ,Fractures, Bone ,Mice ,Osteogenesis ,Transforming Growth Factor beta ,Fracture fixation ,Brain Injuries, Traumatic ,Elbow Joint ,lcsh:Science ,Mice, Knockout ,Multidisciplinary ,Chemistry ,Middle Aged ,3. Good health ,medicine.anatomical_structure ,Female ,Bone Remodeling ,medicine.symptom ,Adult ,Stromal cell ,Science ,Mice, Transgenic ,Achilles Tendon ,General Biochemistry, Genetics and Molecular Biology ,Article ,Transforming Growth Factor beta1 ,03 medical and health sciences ,Young Adult ,Tendon Injuries ,medicine ,Animals ,Humans ,Progenitor cell ,Muscle, Skeletal ,Spinal Cord Injuries ,Ossification ,Cartilage ,Ossification, Heterotopic ,Mesenchymal stem cell ,Receptor, Transforming Growth Factor-beta Type II ,Mesenchymal Stem Cells ,General Chemistry ,medicine.disease ,Antibodies, Neutralizing ,Disease Models, Animal ,030104 developmental biology ,Myositis Ossificans ,Case-Control Studies ,Cancer research ,Heterotopic ossification ,lcsh:Q ,Elbow Injuries - Abstract
Acquired heterotopic ossification (HO) is a painful and debilitating disease characterized by extraskeletal bone formation after injury. The exact pathogenesis of HO remains unknown. Here we show that TGF-β initiates and promotes HO in mice. We find that calcified cartilage and newly formed bone resorb osteoclasts after onset of HO, which leads to high levels of active TGF-β that recruit mesenchymal stromal/progenitor cells (MSPCs) in the HO microenvironment. Transgenic expression of active TGF-β in tendon induces spontaneous HO, whereas systemic injection of a TGF-β neutralizing antibody attenuates ectopic bone formation in traumatic and BMP-induced mouse HO models, and in a fibrodysplasia ossificans progressive mouse model. Moreover, inducible knockout of the TGF-β type II receptor in MSPCs inhibits HO progression in HO mouse models. Our study points toward elevated levels of active TGF-β as inducers and promoters of ectopic bone formation, and suggest that TGF-β might be a therapeutic target in HO., Heterotopic ossification (HO) is a painful disease of unknown etiology characterized by extraskeletal bone formation after injury. Here the authors show that TGF-β is increased in HO lesions, where it promotes the early stages of HO pathology, and demonstrate that TGF-β inhibition ameliorates HO in mice.
- Published
- 2017
31. Mechanosignaling activation of TGFβ maintains intervertebral disc homeostasis
- Author
-
Yongjun Wang, Mei Wan, Janet L. Crane, Amit Jain, Paul D. Sponseller, Qin Bian, Lee H. Riley, Khaled M. Kebaish, Zhengdong Zhang, Xu Cao, X. Edward Guo, Lei Ma, and Cheryle A. Séguin
- Subjects
musculoskeletal diseases ,0301 basic medicine ,Histology ,biology ,Physiology ,Chemistry ,Endocrinology, Diabetes and Metabolism ,Cell ,Integrin ,Intervertebral disc ,Transforming growth factor beta ,musculoskeletal system ,medicine.disease ,Article ,Degenerative disc disease ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Medicine and Health Sciences ,biology.protein ,medicine ,Signal transduction ,Receptor ,Homeostasis - Abstract
Intervertebral disc (IVD) degeneration is the leading cause of disability with no disease-modifying treatment. IVD degeneration is associated with instable mechanical loading in the spine, but little is known about how mechanical stress regulates nucleus notochordal (NC) cells to maintain IVD homeostasis. Here we report that mechanical stress can result in excessive integrin αvβ6-mediated activation of transforming growth factor beta (TGFβ), decreased NC cell vacuoles, and increased matrix proteoglycan production, and results in degenerative disc disease (DDD). Knockout of TGFβ type II receptor (TβRII) or integrin αv in the NC cells inhibited functional activity of postnatal NC cells and also resulted in DDD under mechanical loading. Administration of RGD peptide, TGFβ, and αvβ6-neutralizing antibodies attenuated IVD degeneration. Thus, integrin-mediated activation of TGFβ plays a critical role in mechanical signaling transduction to regulate IVD cell function and homeostasis. Manipulation of this signaling pathway may be a potential therapeutic target to modify DDD.
- Published
- 2017
- Full Text
- View/download PDF
32. IGF-I induced phosphorylation of PTH receptor enhances osteoblast to osteocyte transition
- Author
-
Tao Qiu, Xu Cao, Lingling Xian, Janet L. Crane, Hui Xie, and Liang Xie
- Subjects
0301 basic medicine ,Histology ,Physiology ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Parathyroid hormone ,macromolecular substances ,lcsh:Physiology ,Article ,03 medical and health sciences ,medicine ,lcsh:QH301-705.5 ,Insulin-like growth factor 1 receptor ,lcsh:QP1-981 ,Chemistry ,Parathyroid hormone receptor ,Growth factor ,Osteoblast ,3. Good health ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Biology (General) ,Osteocyte ,Phosphorylation ,Mesenchymal stem cell differentiation - Abstract
Parathyroid hormone (PTH) regulates bone remodeling by activating PTH type 1 receptor (PTH1R) in osteoblasts/osteocytes. Insulin-like growth factor type 1 (IGF-1) stimulates mesenchymal stem cell differentiation to osteoblasts. However, little is known about the signaling mechanisms that regulates the osteoblast-to-osteocyte transition. Here we report that PTH and IGF-I synergistically enhance osteoblast-to-osteocyte differentiation. We identified that a specific tyrosine residue, Y494, on the cytoplasmic domain of PTH1R can be phosphorylated by insulin-like growth factor type I receptor (IGF1R) in vitro. Phosphorylated PTH1R localized to the barbed ends of actin filaments and increased actin polymerization during morphological change of osteoblasts into osteocytes. Disruption of the phosphorylation site reduced actin polymerization and dendrite length. Mouse models with conditional ablation of PTH1R in osteoblasts demonstrated a reduction in the number of osteoctyes and dendrites per osteocyte, with complete overlap of PTH1R with phosphorylated-PTH1R positioning in osteocyte dendrites in wild-type mice. Thus, our findings reveal a novel signaling mechanism that enhances osteoblast-to-osteocyte transition by direct phosphorylation of PTH1R by IGF1R., Bone formation: Hormone and growth factor work together A key hormone and growth factor work together to help turn bone-forming cells into mature bone. Janet Crane and colleagues from Johns Hopkins University School of Medicine in Baltimore, Maryland, USA, tested the effects of parathyroid hormone (PTH) and insulin like-growth factor type 1 (IGF-1) signaling on the differentiation of bone-forming osteoblasts by modulating the activity of their receptors in genetically engineered mice. They found a specific part of the PTH type 1 receptor has a phosphate group added to it by the IGF-1 receptor. This chemical tagging leads to changes in the cytoskeleton of osteoblasts that enhance the formation of mature bone cells known as osteocytes. Mice without this PTH receptor had reduced numbers of osteocytes in their bone. The findings reveal a novel signaling mechanism behind this cellular transition during bone building.
- Published
- 2017
33. PDGF-BB secreted by preosteoclasts induces angiogenesis during coupling with osteogenesis
- Author
-
Mei Wan, Changjun Li, Long Wang, Liang Xie, Zhuang Cui, Janet L. Crane, Weizhong Chang, Qin Bian, Bin Yu, Maureen Pickarski, Hui Xie, Xiang-Hang Luo, Gehua Zhen, Yin Hu, Zhuying Xia, Jolene J. Windle, Er-Yuan Liao, Tao Qiu, Le Thi Duong, Xu Cao, and Lingling Xian
- Subjects
CD31 ,Angiogenesis ,Cathepsin K ,Becaplermin ,Osteoclasts ,Cell Count ,Bone remodeling ,Neovascularization ,0302 clinical medicine ,Cell Movement ,Osteogenesis ,Femur ,Phosphorylation ,Tartrate-resistant acid phosphatase ,0303 health sciences ,Proto-Oncogene Proteins c-sis ,General Medicine ,3. Good health ,Isoenzymes ,Platelet Endothelial Cell Adhesion Molecule-1 ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,cardiovascular system ,Female ,medicine.symptom ,medicine.medical_specialty ,Ovariectomy ,Acid Phosphatase ,Neovascularization, Physiologic ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Internal medicine ,medicine ,Animals ,Protease Inhibitors ,030304 developmental biology ,Tartrate-Resistant Acid Phosphatase ,Endothelial Cells ,Mesenchymal Stem Cells ,X-Ray Microtomography ,Mice, Inbred C57BL ,Endocrinology ,Culture Media, Conditioned ,Focal Adhesion Protein-Tyrosine Kinases ,Cortical bone ,Bone marrow ,Proto-Oncogene Proteins c-akt - Abstract
Osteogenesis during bone modeling and remodeling is coupled with angiogenesis. A recent study showed that a specific vessel subtype, strongly positive for CD31 and endomucin (CD31(hi)Emcn(hi)), couples angiogenesis and osteogenesis. Here, we found that platelet-derived growth factor-BB (PDGF-BB) secreted by preosteoclasts induces CD31(hi)Emcn(hi) vessel formation during bone modeling and remodeling. Mice with depletion of PDGF-BB in the tartrate-resistant acid phosphatase-positive cell lineage show significantly lower trabecular and cortical bone mass, serum and bone marrow PDGF-BB concentrations, and fewer CD31(hi)Emcn(hi) vessels compared to wild-type mice. In the ovariectomy (OVX)-induced osteoporotic mouse model, serum and bone marrow levels of PDGF-BB and numbers of CD31(hi)Emcn(hi) vessels are significantly lower compared to sham-operated controls. Treatment with exogenous PDGF-BB or inhibition of cathepsin K to increase the number of preosteoclasts, and thus the endogenous levels of PDGF-BB, increases CD31(hi)Emcn(hi) vessel number and stimulates bone formation in OVX mice. Thus, pharmacotherapies that increase PDGF-BB secretion from preosteoclasts offer a new therapeutic target for treating osteoporosis by promoting angiogenesis and thus bone formation.
- Published
- 2014
- Full Text
- View/download PDF
34. Bone marrow mesenchymal stem cells and TGF-β signaling in bone remodeling
- Author
-
Xu Cao and Janet L. Crane
- Subjects
Bone remodeling period ,Time Factors ,Osteoclasts ,Bone Marrow Cells ,Bone Neoplasms ,Bone healing ,Bone resorption ,Bone remodeling ,03 medical and health sciences ,0302 clinical medicine ,Osteogenesis ,Transforming Growth Factor beta ,Osteoclast ,Osteoarthritis ,Bone cell ,medicine ,Animals ,Homeostasis ,Humans ,Cell Lineage ,Bone Resorption ,030304 developmental biology ,0303 health sciences ,Chemistry ,Cell Differentiation ,Mesenchymal Stem Cells ,General Medicine ,Anatomy ,Cell biology ,Bone morphogenetic protein 7 ,medicine.anatomical_structure ,Gene Expression Regulation ,Parathyroid Hormone ,030220 oncology & carcinogenesis ,Science in Medicine ,Bone Remodeling ,Bone marrow ,Signal Transduction - Abstract
During bone resorption, abundant factors previously buried in the bone matrix are released into the bone marrow microenvironment, which results in recruitment and differentiation of bone marrow mesenchymal stem cells (MSCs) for subsequent bone formation, temporally and spatially coupling bone remodeling. Parathyroid hormone (PTH) orchestrates the signaling of many pathways that direct MSC fate. The spatiotemporal release and activation of matrix TGF-β during osteoclast bone resorption recruits MSCs to bone-resorptive sites. Dysregulation of TGF-β alters MSC fate, uncoupling bone remodeling and causing skeletal disorders. Modulation of TGF-β or PTH signaling may reestablish coupled bone remodeling and be a potential therapy.
- Published
- 2014
- Full Text
- View/download PDF
35. Function of matrix IGF-1 in coupling bone resorption and formation
- Author
-
Xu Cao and Janet L. Crane
- Subjects
Bone remodeling period ,medicine.medical_specialty ,Osteoporosis ,Osteoclasts ,Biology ,Bone tissue ,Models, Biological ,Article ,Bone resorption ,Bone remodeling ,Osteogenesis ,Internal medicine ,Drug Discovery ,medicine ,Humans ,Bone Resorption ,Insulin-Like Growth Factor I ,Genetics (clinical) ,Bone mineral ,Osteoblasts ,medicine.disease ,Cell biology ,Bone morphogenetic protein 7 ,medicine.anatomical_structure ,Endocrinology ,Molecular Medicine ,Bone Remodeling ,Bone marrow ,Signal Transduction - Abstract
Balancing bone resorption and formation is the quintessential component for the prevention of osteoporosis. Signals that determine the recruitment, replication, differentiation, function, and apoptosis of osteoblasts and osteoclasts direct bone remodeling and determine whether bone tissue is gained, lost, or balanced. Therefore, understanding the signaling pathways involved in the coupling process will help develop further targets for osteoporosis therapy, by blocking bone resorption or enhancing bone formation in a space- and time-dependent manner. Insulin-like growth factor type 1 (IGF-1) has long been known to play a role in bone strength. It is one of the most abundant substances in the bone matrix, circulates systemically and is secreted locally, and has a direct relationship with bone mineral density. Recent data has helped further our understanding of the direct role of IGF-1 signaling in coupling bone remodeling which will be discussed in this review. The bone marrow microenvironment plays a critical role in the fate of mesenchymal stem cells and hematopoietic stem cells and thus how IGF-1 interacts with other factors in the microenvironment are equally important. While previous clinical trials with IGF-1 administration have been unsuccessful at enhancing bone formation, advances in basic science studies have provided insight into further mechanisms that should be considered for future trials. Additional basic science studies dissecting the regulation and the function of matrix IGF-1 in modeling and remodeling will continue to provide further insight for future directions for anabolic therapies for osteoporosis.
- Published
- 2013
- Full Text
- View/download PDF
36. Disruption of LRP6 in osteoblasts blunts the bone anabolic activity of PTH
- Author
-
Mei Wan, Xu Cao, Weishan Wang, Qiujuan Xing, Hui Xie, Shi Chenhui, Janet L. Crane, Bing Yu, and Changjun Li
- Subjects
musculoskeletal diseases ,medicine.medical_specialty ,Anabolism ,Chemistry ,Endocrinology, Diabetes and Metabolism ,Cellular differentiation ,Parathyroid hormone ,LRP6 ,Osteoblast ,Bone resorption ,Bone remodeling ,medicine.anatomical_structure ,Endocrinology ,Internal medicine ,medicine ,Orthopedics and Sports Medicine ,Cortical bone - Abstract
Mutations in low-density lipoprotein receptor-related protein 6 (LRP6) are associated with human skeletal disorders. LRP6 is required for parathyroid hormone (PTH)-stimulated signaling pathways in osteoblasts. We investigated whether LRP6 in osteoblasts directly regulates bone remodeling and mediates the bone anabolic effects of PTH by specifically deleting LRP6 in mature osteoblasts in mice (LRP6 KO). Three-month-old LRP6 KO mice had a significant reduction in bone mass in the femora secondary spongiosa relative to their wild-type littermates, whereas marginal changes were found in femoral tissue of 1-month-old LRP6 KO mice. The remodeling area of the 3-month-old LRP6 KO mice showed a decreased bone formation rate as detected by Goldner's Trichrome staining and calcein double labeling. Bone histomorphometric and immumohistochemical analysis revealed a reduction in osteoblasts but little change in the numbers of osteoclasts and osteoprogenitors/osteoblast precursors in LRP6 KO mice compared with wild-type littermates. In addition, the percentage of the apoptotic osteoblasts on the bone surface was higher in LRP6 KO mice compared with wild-type littermates. Intermittent injection of PTH had no effect on bone mass or osteoblastic bone formation in either trabecular and cortical bone in LRP6 KO mice, whereas all were enhanced in wild-type littermates. Additionally, the anti-apoptotic effect of PTH on osteoblasts in LRP6 KO mice was less significant compared with wild-type mice. Therefore, our findings demonstrate that LRP6 in osteoblasts is essential for osteoblastic differentiation during bone remodeling and the anabolic effects of PTH.
- Published
- 2013
- Full Text
- View/download PDF
37. Inhibition of TGF-β signaling in mesenchymal stem cells of subchondral bone attenuates osteoarthritis
- Author
-
John A. Carrino, Weizhong Chang, William W. Lu, Mei Wan, Chunyi Wen, Frederic B. Askin, Simon C. Mears, Dmitri Artemov, Lee H. Riley, Frank J. Frassica, Xiaofeng Jia, Andrew J. Cosgarea, Qianming Chen, Jie Yao, Yu Li, Xuedong Zhou, Janet L. Crane, Paul D. Sponseller, Zhihe Zhao, Gehua Zhen, and Xu Cao
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Angiogenesis ,Osteoarthritis ,Protein Serine-Threonine Kinases ,Bone and Bones ,General Biochemistry, Genetics and Molecular Biology ,Pathogenesis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Transforming Growth Factor beta ,Animals ,Medicine ,Cells, Cultured ,030304 developmental biology ,0303 health sciences ,biology ,business.industry ,Osteoid ,Cartilage ,Mesenchymal stem cell ,Receptor, Transforming Growth Factor-beta Type II ,Mesenchymal Stem Cells ,General Medicine ,Transforming growth factor beta ,medicine.disease ,Rats ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Rats, Inbred Lew ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,business ,Receptors, Transforming Growth Factor beta ,Signal Transduction ,Transforming growth factor - Abstract
Osteoarthritis is a highly prevalent and debilitating joint disorder. There is no effective medical therapy for the condition because of limited understanding of its pathogenesis. We show that transforming growth factor β1 (TGF-β1) is activated in subchondral bone in response to altered mechanical loading in an anterior cruciate ligament transection (ACLT) mouse model of osteoarthritis. TGF-β1 concentrations are also high in subchondral bone from humans with osteoarthritis. High concentrations of TGF-β1 induced formation of nestin-positive mesenchymal stem cell (MSC) clusters, leading to formation of marrow osteoid islets accompanied by high levels of angiogenesis. We found that transgenic expression of active TGF-β1 in osteoblastic cells induced osteoarthritis, whereas inhibition of TGF-β activity in subchondral bone attenuated the degeneration of articular cartilage. In particular, knockout of the TGF-β type II receptor (TβRII) in nestin-positive MSCs led to less development of osteoarthritis relative to wild-type mice after ACLT. Thus, high concentrations of active TGF-β1 in subchondral bone seem to initiate the pathological changes of osteoarthritis, and inhibition of this process could be a potential therapeutic approach to treating this disease.
- Published
- 2013
- Full Text
- View/download PDF
38. Parathyroid hormone induces differentiation of mesenchymal stromal/stem cells by enhancing bone morphogenetic protein signaling
- Author
-
Xiaoli Zhao, Chaozhe Yang, Mei Wan, Lingling Xian, Bing Yu, Xu Cao, William W. Lu, and Janet L. Crane
- Subjects
medicine.medical_specialty ,animal structures ,Stromal cell ,Endocrinology, Diabetes and Metabolism ,Cellular differentiation ,Green Fluorescent Proteins ,Parathyroid hormone ,Biology ,Bone Morphogenetic Protein Receptors, Type II ,Bone morphogenetic protein ,Models, Biological ,Article ,Smad1 Protein ,Mice ,Internal medicine ,medicine ,Animals ,Humans ,Orthopedics and Sports Medicine ,Bone morphogenetic protein receptor ,Phosphorylation ,Receptor, Parathyroid Hormone, Type 1 ,CD11b Antigen ,Osteoblasts ,Mesenchymal stem cell ,Cell Differentiation ,Mesenchymal Stem Cells ,Endocytosis ,BMPR2 ,Cell biology ,Mice, Inbred C57BL ,HEK293 Cells ,Endocrinology ,Parathyroid Hormone ,Low Density Lipoprotein Receptor-Related Protein-6 ,Bone Morphogenetic Proteins ,embryonic structures ,Leukocyte Common Antigens ,Signal transduction ,Carrier Proteins ,Signal Transduction - Abstract
Parathyroid hormone (PTH) stimulates bone remodeling and induces differentiation of bone marrow mesenchymal stromal/stem cells (MSCs) by orchestrating activities of local factors such as bone morphogenetic proteins (BMPs). The activity and specificity of different BMP ligands are controlled by various extracellular antagonists that prevent binding of BMPs to their receptors. Low-density lipoprotein receptor-related protein 6 (LRP6) has been shown to interact with both the PTH and BMP extracellular signaling pathways by forming a complex with parathyroid hormone 1 receptor (PTH1R) and sharing common antagonists with BMPs. We hypothesized that PTH-enhanced differentiation of MSCs into the osteoblast lineage through enhancement of BMP signaling occurs by modifying the extracellular antagonist network via LRP6. In vitro studies using multiple cell lines, including Sca-1(+) CD45(-) CD11b(-) MSCs, showed that a single injection of PTH enhanced phosphorylation of Smad1 and could also antagonize the inhibitory effect of noggin. PTH treatment induced endocytosis of a PTH1R/LRP6 complex and resulted in enhancement of phosphorylation of Smad1 that was abrogated by deletion of PTH1R, β-arrestin, or chlorpromazine. Deletion of LRP6 alone led to enhancement of pSmad1 levels that could not be further increased with PTH treatment. Finally, knockdown of LRP6 increased the exposure of endogenous cell-surface BMP receptor type II (BMPRII) significantly in C2C12 cells, and PTH treatment significantly enhanced cell-surface binding of (125) I-BMP2 in a dose- and time-dependent manner, implying that LRP6 organizes an extracellular network of BMP antagonists that prevent access of BMPs to BMP receptors. In vivo studies in C57BL/6J mice and of transplanted green fluorescent protein (GFP)-labeled Sca-1(+) CD45(-) CD11b(-) MSCs into the bone marrow cavity of Rag2(-/-) immunodeficient mice showed that PTH enhanced phosphorylation of Smad1 and increased commitment of MSCs to osteoblast lineage, respectively. These data demonstrate that PTH enhancement of MSC differentiation to the osteoblast lineage occurs through a PTH- and LRP6-dependent pathway by endocytosis of the PTH1R/LRp6 complex, allowing enhancement of BMP signaling.
- Published
- 2012
- Full Text
- View/download PDF
39. Excessive Activation of TGFβ by Spinal Instability Causes Vertebral Endplate Sclerosis
- Author
-
Lei Ma, Khaled M. Kebaish, Yongjun Wang, Amit Jain, Xin Xu, Paul D. Sponseller, Willian Weijia Lu, Janet L. Crane, X. Edward Guo, Mei Wan, Zhendong Zhang, Xu Cao, Qin Bian, and Lee H. Riley
- Subjects
Male ,musculoskeletal diseases ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Osteocalcin ,Receptor, Transforming Growth Factor-beta Type I ,Mice, Transgenic ,Dioxoles ,Intervertebral Disc Degeneration ,Protein Serine-Threonine Kinases ,Mechanotransduction, Cellular ,Article ,Muscle hypertrophy ,Nestin ,Weight-Bearing ,Pathogenesis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Transforming Growth Factor beta ,medicine ,Animals ,Humans ,Mechanotransduction ,Intervertebral Disc ,Receptor ,Sclerosis ,Multidisciplinary ,biology ,Ossification ,business.industry ,Intervertebral disc ,Transforming growth factor beta ,musculoskeletal system ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Sp7 Transcription Factor ,Benzamides ,biology.protein ,Stress, Mechanical ,medicine.symptom ,business ,Receptors, Transforming Growth Factor beta ,030217 neurology & neurosurgery - Abstract
Narrowed intervertebral disc (IVD) space is a characteristic of IVD degeneration. EP sclerosis is associated with IVD, however the pathogenesis of EP hypertrophy is poorly understood. Here, we employed two spine instability mouse models to investigate temporal and spatial EP changes associated with IVD volume, considering them as a functional unit. We found that aberrant mechanical loading leads to accelerated ossification and hypertrophy of EP, decreased IVD volume and increased activation of TGFβ. Overexpression of active TGFβ in CED mice showed a similar phenotype of spine instability model. Administration of TGFβ Receptor I inhibitor attenuates pathologic changes of EP and prevents IVD narrowing. The aberrant activation of TGFβ resulting in EPs hypertrophy-induced IVD space narrowing provides a pharmacologic target that could have therapeutic potential to delay DDD.
- Published
- 2016
40. Role of TGF-β Signaling in Coupling Bone Remodeling
- Author
-
Lingling Xian, Xu Cao, and Janet L. Crane
- Subjects
0301 basic medicine ,Chemistry ,Mesenchymal stem cell migration ,Osteoblast ,Bioinformatics ,Bone morphogenetic protein 2 ,Bone resorption ,Resorption ,Bone remodeling ,Cell biology ,Bone morphogenetic protein 7 ,03 medical and health sciences ,030104 developmental biology ,medicine.anatomical_structure ,Osteoclast ,medicine - Abstract
TGF-β signaling plays a key role in the temporal and spatial regulation of bone remodeling. During osteoclast bone resorption, TGF-β is released from the bone matrix and activated. Active TGF-β recruits mesenchymal stem cells to the bone resorption pit through the SMAD signaling pathway. Mesenchymal stem cells undergo osteoblast differentiation and deposit new bone filling in the resorption pit and maintaining the structural integrity of the skeleton. Thus, TGF-β signaling plays a key role in the coupling process and disruptions to the TGF-β signaling pathway lead to loss of skeletal integrity. This chapter describes methods on how to quantitate bone matrix TGF-β and assess its role in mesenchymal stem cell migration both in vitro and in vivo.
- Published
- 2016
- Full Text
- View/download PDF
41. Role of TGF-β Signaling in Coupling Bone Remodeling
- Author
-
Janet L, Crane, Lingling, Xian, and Xu, Cao
- Subjects
Mice ,Bone Marrow ,Cell Movement ,Transforming Growth Factor beta ,Animals ,Bone Remodeling ,In Vitro Techniques ,Immunohistochemistry ,Extracellular Matrix ,Rats ,Signal Transduction - Abstract
TGF-β signaling plays a key role in the temporal and spatial regulation of bone remodeling. During osteoclast bone resorption, TGF-β is released from the bone matrix and activated. Active TGF-β recruits mesenchymal stem cells to the bone resorption pit through the SMAD signaling pathway. Mesenchymal stem cells undergo osteoblast differentiation and deposit new bone filling in the resorption pit and maintaining the structural integrity of the skeleton. Thus, TGF-β signaling plays a key role in the coupling process and disruptions to the TGF-β signaling pathway lead to loss of skeletal integrity. This chapter describes methods on how to quantitate bone matrix TGF-β and assess its role in mesenchymal stem cell migration both in vitro and in vivo.
- Published
- 2015
42. A Mouse Model of Albright Hereditary Osteodystrophy Generated by Targeted Disruption of Exon 1 of the Gnas Gene
- Author
-
Gary S. Wand, William F. Schwindinger, Matthew D. Ringel, Michael A. Levine, Motoyasu Saji, Larry S. Zweifel, Emily L. Germain-Lee, Rediet Zewdu, David L. Huso, and Janet L. Crane
- Subjects
musculoskeletal diseases ,endocrine system ,medicine.medical_specialty ,Litter Size ,Thyrotropin ,Fibrous Dysplasia, Polyostotic ,Bone and Bones ,Genomic Imprinting ,Mice ,Exon ,Endocrinology ,Internal medicine ,Chromogranins ,GTP-Binding Protein alpha Subunits, Gs ,medicine ,GNAS complex locus ,Animals ,Humans ,Imprinting (psychology) ,Allele ,Paternal Inheritance ,Receptor ,Mice, Knockout ,biology ,Body Weight ,Heterozygote advantage ,Exons ,Survival Analysis ,Molecular biology ,Body Height ,Disease Models, Animal ,Fertility ,Phenotype ,Parathyroid Hormone ,biology.protein ,Genomic imprinting ,hormones, hormone substitutes, and hormone antagonists ,Adenylyl Cyclases - Abstract
Albright hereditary osteodystrophy is caused by heterozygous inactivating mutations in GNAS, a gene that encodes not only the alpha-chain of Gs (Galphas), but also NESP55 and XLalphas through use of alternative first exons. Patients with GNAS mutations on maternally inherited alleles are resistant to multiple hormones such as PTH, TSH, LH/FSH, GHRH, and glucagon, whose receptors are coupled to Gs. This variant of Albright hereditary osteodystrophy is termed pseudohypoparathyroidism type 1a and is due to presumed tissue-specific paternal imprinting of Galphas. Previous studies have shown that mice heterozygous for a targeted disruption of exon 2 of Gnas, the murine homolog of GNAS, showed unique phenotypes dependent on the parent of origin of the mutated allele. However, hormone resistance occurred only when the disrupted gene was maternally inherited. Because disruption of exon 2 is predicted to inactivate Galphas as well as NESP55 and XLalphas, we created transgenic mice with disruption of exon 1 to investigate the effects of isolated loss of Galphas. Heterozygous mice that inherited the disruption maternally (-m/+) exhibited PTH and TSH resistance, whereas those with paternal inheritance (+/-p) had normal hormone responsiveness. Heterozygous mice were shorter and, when the disrupted allele was inherited maternally, weighed more than wild-type littermates. Galphas protein and mRNA expression was consistent with paternal imprinting in the renal cortex and thyroid, but there was no imprinting in renal medulla, heart, or adipose. These findings confirm the tissue-specific paternal imprinting of GNAS and demonstrate that Galphas deficiency alone is sufficient to account for the hormone resistance of pseudohypoparathyroidism type 1a.
- Published
- 2005
- Full Text
- View/download PDF
43. Growth Hormone Deficiency in Pseudohypoparathyroidism Type 1a: Another Manifestation of Multihormone Resistance
- Author
-
Janet L. Crane, Suzanne M. Jan de Beur, Michael A. Levine, Emily L. Germain-Lee, and Joshua D. Groman
- Subjects
Adult ,Male ,endocrine system ,medicine.medical_specialty ,Adolescent ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Biology ,Biochemistry ,Short stature ,Growth hormone deficiency ,Absorptiometry, Photon ,Endocrinology ,Internal medicine ,GTP-Binding Protein alpha Subunits, Gs ,medicine ,Humans ,Obesity ,Insulin-Like Growth Factor I ,Child ,Pseudohypoparathyroidism ,Bone Development ,Anthropometry ,Human Growth Hormone ,Biochemistry (medical) ,Brachydactyly ,Genetic disorder ,medicine.disease ,Body Height ,Growth hormone secretion ,Blotting, Southern ,Phenotype ,Child, Preschool ,Female ,STX16 ,Pseudopseudohypoparathyroidism ,medicine.symptom - Abstract
Albright hereditary osteodystrophy (AHO) is a genetic disorder caused by heterozygous inactivating mutations in GNAS1, the gene encoding the -chain of Gs, and is associated with short stature, obesity, brachydactyly, and sc ossifications. AHO patients with GNAS1 mutations on maternally inherited alleles also manifest resistance to multiple hormones (e.g. PTH, TSH, LH, FSH), a variant termed pseudohypoparathyroidism (PHP) type 1a, due to paternal imprinting of Gs transcripts in specific tissues. Recent evidence has shown that Gs transcripts are also imprinted in the pituitary somatotrophs that secrete GH. Because this imprinting could influence GHRH-dependent stimulation of somatotrophs, we hypothesized that maternally inherited GNAS1 mutations would impair GH secretion. We studied GH status in 13 subjects with PHP type 1a. GH responses to arginine/L-dopa and arginine/ GHRH were deficient in nine subjects, all of whom were obese and had low serum concentrations of IGF-I. By contrast, none of the four GH-sufficient subjects were obese, and all had normal IGF-I levels. Our data indicate that GH deficiency is common (69%) in PHP type 1a and may contribute to the obesity and short stature typical of AHO. We propose that GH status be evaluated in all patients with PHP type 1a. (J Clin Endocrinol Metab 88: 4059 – 4069, 2003)
- Published
- 2003
- Full Text
- View/download PDF
44. The ratio of serum Angiopoietin-1 to Angiopoietin-2 in patients with cervical cancer is a valuable diagnostic and prognostic biomarker
- Author
-
Lisha Xie, Shouhua Yang, Ping Yang, Xiaoqing Yi, Na Chen, Zehua Wang, Hangyu Wang, Guo Jing, Janet L. Crane, Jing Cai, Ruiqing Dong, and Dongyun Yang
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,Survival ,Angiogenesis ,lcsh:Medicine ,Women’s Health ,Cervical intraepithelial neoplasia ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Medicine ,Gynecology and Obstetrics ,Cervix ,Cervical cancer ,Receiver operating characteristic ,business.industry ,General Neuroscience ,lcsh:R ,Diagnostic biomarker ,Serum angiopoietin ,General Medicine ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Angiopoietin-1 ,030220 oncology & carcinogenesis ,Adenocarcinoma ,Immunohistochemistry ,General Agricultural and Biological Sciences ,business - Abstract
Objectives Angiopoietins have been found to play essential roles in tumor angiogenesis. The present study was aimed at investigating the diagnostic and prognostic values of serum angiopoietin 1 and 2 (sAng-1 and sAng-2) in cervical cancer. Methods The sAng-1 and sAng-2 concentrations were analyzed in 77 women with cervical cancer, 44 women with cervical intraepithelial neoplasia (CIN) and 43 women without cervical lesions by enzyme-linked immunosorbent assay. The diagnostic values of sAng-1, sAng-2 and sAng-1/sAng-2 were evaluated by receiver operating characteristic (ROC) curves. The Ang-1 and Ang-2 expression in cervical cancer tissues as well as microvessel density (MVD), were assessed by immunohistochemistry. Results The concentration of sAng-2 gradually increased and the sAng-1/Ang-2 ratio was gradually decreased from normal control to CIN, then to squamous cell cancer, and the sAng-1/sAng-2 ratio was also significantly decreased in adenocarcinoma. The area under ROC curves of sAng-2 and sAng-1/sAng-2 ratio for discriminating cervical cancer from normal were 0.744 and 0.705, respectively. Decreased sAng-1/sAng-2 was significantly associated with advanced tumor stage, poor differentiation, lymph-vascular space invasion and high MVD. sAng-2 was positively correlated with the Ang-2 expression in cervix epithelia. A high sAng-1/sAng-2 ratio was associated with a longer progression-free survival and a longer overall survival in cervical cancer patients. Conclusions These findings suggest that sAng-2 and the sAng-1/sAng-2 ratio may be valuable diagnostic and prognostic biomarkers for cervical cancer.
- Published
- 2017
- Full Text
- View/download PDF
45. MicroRNA 224 Regulates Ion Transporter Expression in Ameloblasts To Coordinate Enamel Mineralization
- Author
-
Lei Cheng, Bo Gao, Xin Xu, Feifei Sun, Xuedong Zhou, Jianxun Sun, Mian Wan, Yi Fan, Yachuan Zhou, Liwei Zheng, Janet L. Crane, and Xin Zhou
- Subjects
Cystic Fibrosis Transmembrane Conductance Regulator ,Cell Line ,Mice ,Downregulation and upregulation ,stomatognathic system ,Amelogenesis ,Ameloblasts ,Animals ,Humans ,Dental Enamel ,Molecular Biology ,3' Untranslated Regions ,Ion transporter ,biology ,Enamel paint ,Three prime untranslated region ,Sodium-Bicarbonate Symporters ,Cell Biology ,Articles ,Cell biology ,Up-Regulation ,Enamel mineralization ,stomatognathic diseases ,MicroRNAs ,Biochemistry ,Gene Expression Regulation ,visual_art ,biology.protein ,visual_art.visual_art_medium ,SLC4A4 ,Ameloblast ,Tooth - Abstract
Enamel mineralization is accompanied by the release of protons into the extracellular matrix, which is buffered to regulate the pH value in the local microenvironment. The present study aimed to investigate the role of microRNA 224 (miR-224) as a regulator of SLC4A4 and CFTR, encoding the key buffering ion transporters, in modulating enamel mineralization. miR-224 was significantly downregulated as ameloblasts differentiated, in parallel with upregulation of SLC4A4 and CFTR. Overexpression of miR-224 downregulated SLC4A4 and CFTR expression in cultured human epithelial cells. A microRNA luciferase assay confirmed the specific binding of miR-224 to the 3' untranslated regions (UTRs) of SLC4A4 and CFTR mRNAs, thereby inhibiting protein translation. miR-224 agomir injection in mouse neonatal incisors resulted in normal enamel length and thickness, but with disturbed organization of the prism structure and deficient crystal growth. Moreover, the enamel Ca/P ratio and microhardness were markedly reduced after miR-224 agomir administration. These results demonstrate that miR-224 plays a pivotal role in fine tuning enamel mineralization by modulating SLC4A4 and CFTR to maintain pH homeostasis and support enamel mineralization.
- Published
- 2014
46. IGF-1 Signaling is Essential for Differentiation of Mesenchymal Stem Cells for Peak Bone Mass
- Author
-
Luo Zhao, Joseph S. Frye, Xu Cao, Janet L. Crane, Lingling Xian, and Tao Qiu
- Subjects
Peak bone mass ,medicine.medical_specialty ,Histology ,Physiology ,Endocrinology, Diabetes and Metabolism ,Growth factor ,medicine.medical_treatment ,Mesenchymal stem cell ,Wild type ,Cell migration ,Biology ,Bone remodeling ,Endocrinology ,Internal medicine ,Knockout mouse ,Immunology ,medicine ,Original Article ,Secondary osteoporosis - Abstract
Survival of children with chronic medical illnesses is leading to an increase in secondary osteoporosis due to impaired peak bone mass (PBM). Insulin-like growth factor type 1 (IGF-1) levels correlate with the pattern of bone mass accrual and many chronic illnesses are associated with low IGF-1 levels. Reduced serum levels of IGF-1 minimally affect the integrity of the skeleton, whereas recent studies suggest that skeletal IGF-I regulates PBM. To determine the role of IGF-1 in postnatal bone mass accrual regardless of source, we established an inducible type 1 Igf receptor Cre/lox knockout mouse model, in which the type 1 Igf receptor was deleted inducibely in the mesenchymal stem cells (MSCs) from 3–7 weeks of age. The size of the mouse was not affected as knockout and wild type mice had similar body weights and nasoanal and femoral lengths. However, bone volume and trabecular bone thickness were decreased in the secondary spongiosa of female knockout mice relative to wild type controls, indicating that IGF-1 is critical for bone mass. IGF-1 signaling in MSCs in vitro has been implicated to be involved in both migration to the bone surface and differentiation into bone forming osteoblasts. To clarify the exact role of IGF-1 in bone, we found by immunohistochemical analysis that a similar number of Osterix–positive osteoprogenitors were on the bone perimeter, indicating migration of MSCs was not affected. Most importantly, 56% fewer osteocalcin-positive mature osteoblasts were present on the bone perimeter in the secondary spongiosa in knockout mice versus wild type littermates. These in vivo data demonstrate that the primary role of skeletal IGF-1 is for the terminal differentiation of osteoprogenitors, but refute the role of IGF-1 in MSC migration in vivo. Additionally, these findings confirm that impaired IGF-1 signaling in bone MSCs is sufficient to impair bone mass acquisition.
- Published
- 2013
47. Disruption of LRP6 in osteoblasts blunts the bone anabolic activity of PTH
- Author
-
Changjun, Li, Qiujuan, Xing, Bing, Yu, Hui, Xie, Weishan, Wang, Chenhui, Shi, Janet L, Crane, Xu, Cao, and Mei, Wan
- Subjects
musculoskeletal diseases ,Mice, Knockout ,Osteoblasts ,Integrases ,Apoptosis ,Cell Differentiation ,Organ Size ,Cyclic AMP-Dependent Protein Kinases ,Bone and Bones ,GTP-Binding Protein alpha Subunits ,Article ,Mice ,Anabolic Agents ,Phenotype ,Gene Expression Regulation ,Osteogenesis ,Parathyroid Hormone ,Low Density Lipoprotein Receptor-Related Protein-6 ,Animals ,Humans ,Bone Remodeling ,Bone Resorption ,Gene Deletion ,beta Catenin ,Signal Transduction - Abstract
Mutations in low-density lipoprotein receptor-related protein 6 (LRP6) are associated with human skeletal disorders. LRP6 is required for parathyroid hormone (PTH)-stimulated signaling pathways in osteoblasts. We investigated whether LRP6 in osteoblasts directly regulates bone remodeling and mediates the bone anabolic effects of PTH by specifically deleting LRP6 in mature osteoblasts in mice (LRP6 KO). Three-month-old LRP6 KO mice had a significant reduction in bone mass in the femora secondary spongiosa relative to their wild-type littermates, whereas marginal changes were found in femoral tissue of 1-month-old LRP6 KO mice. The remodeling area of the 3-month-old LRP6 KO mice showed a decreased bone formation rate as detected by Goldner's Trichrome staining and calcein double labeling. Bone histomorphometric and immumohistochemical analysis revealed a reduction in osteoblasts but little change in the numbers of osteoclasts and osteoprogenitors/osteoblast precursors in LRP6 KO mice compared with wild-type littermates. In addition, the percentage of the apoptotic osteoblasts on the bone surface was higher in LRP6 KO mice compared with wild-type littermates. Intermittent injection of PTH had no effect on bone mass or osteoblastic bone formation in either trabecular and cortical bone in LRP6 KO mice, whereas all were enhanced in wild-type littermates. Additionally, the anti-apoptotic effect of PTH on osteoblasts in LRP6 KO mice was less significant compared with wild-type mice. Therefore, our findings demonstrate that LRP6 in osteoblasts is essential for osteoblastic differentiation during bone remodeling and the anabolic effects of PTH.
- Published
- 2012
48. Imprinting Status of GαS, NESP55, and XLαs in Cell Cultures Derived from Human Embryonic Germ Cells: GNAS Imprinting in Human Embryonic Germ Cells
- Author
-
Michael J. Shamblott, Janet L. Crane, Michael A. Levine, Emily L. Germain-Lee, Stephanie Hsu, and Joyce Axelman
- Subjects
Male ,Embryonic Germ Cells ,Time Factors ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Epigenesis, Genetic ,Genomic Imprinting ,GNAS complex locus ,Chromogranins ,GTP-Binding Protein alpha Subunits, Gs ,Humans ,General Pharmacology, Toxicology and Pharmaceutics ,Allele ,Imprinting (psychology) ,Gene ,Research Articles ,Alleles ,Embryonic Stem Cells ,Regulation of gene expression ,Models, Genetic ,General Neuroscience ,Chromosome Mapping ,General Medicine ,DNA Methylation ,Molecular biology ,Gene Expression Regulation ,DNA methylation ,Mutation ,biology.protein ,Female ,Genomic imprinting - Abstract
GNAS is a complex gene that through use of alternative first exons encodes signaling proteins Galpha(s) and XLalphas plus neurosecretory protein NESP55. Tissue-specific expression of these proteins is regulated through reciprocal genomic imprinting in fully differentiated and developed tissue. Mutations in GNAS account for several human disorders, including McCune-Albright syndrome and Albright hereditary osteodystrophy, and further knowledge of GNAS imprinting may provide insights into variable phenotypes of these disorders. We therefore analyzed expression of Galpha(s), NESP55, and XLalphas prior to tissue differentiation in cell cultures derived from human primordia germ cells. We found that the expression of Galpha(s) was biallelic (maternal allele: 52.6%+/- 2.5%; paternal allele: 47.2%+/- 2.5%; p= 0.07), whereas NESP55 was expressed preferentially from the maternal allele (maternal allele: 81.9%+/- 10%; paternal allele: 18.1%+/- 10%; p= 0.002) and XLalphas was preferentially expressed from the paternal allele (maternal allele: 2.7%+/- 0.3%; paternal allele: 97.3%+/- 0.3%; p= 0.007). These results demonstrate that imprinting of NESP55 occurs very early in development, although complete imprinting appears to take place later than 5-11 weeks postfertilization, and that imprinting of XLalphas occurs very early postfertilization. By contrast, imprinting of Galpha(s) most likely occurs after 11 weeks postfertilization and after tissue differentiation.
- Published
- 2009
49. Paternal imprinting of Galpha(s) in the human thyroid as the basis of TSH resistance in pseudohypoparathyroidism type 1a
- Author
-
Zhichao Deng, Michael A. Levine, Emily L. Germain-Lee, Chang Lin Ding, Motoyasu Saji, Janet L. Crane, and Matthew D. Ringel
- Subjects
Male ,Thyroid Hormone Resistance Syndrome ,endocrine system ,medicine.medical_specialty ,endocrine system diseases ,Biophysics ,Thyroid Gland ,Nerve Tissue Proteins ,Biology ,Biochemistry ,Genomic Imprinting ,Internal medicine ,medicine ,GNAS complex locus ,Chromogranins ,GTP-Binding Protein alpha Subunits, Gs ,Humans ,RNA, Messenger ,Imprinting (psychology) ,Molecular Biology ,Pseudohypoparathyroidism ,DNA Primers ,Base Sequence ,Reverse Transcriptase Polymerase Chain Reaction ,Thyroid ,Cell Biology ,medicine.disease ,Heterotrimeric GTP-Binding Proteins ,Endocrinology ,medicine.anatomical_structure ,Mutation ,biology.protein ,STX16 ,Pseudopseudohypoparathyroidism ,Genomic imprinting ,Hormone - Abstract
Albright hereditary osteodystrophy (AHO) is characterized by multiple somatic defects secondary to mutations in the GNAS1 gene. AHO patients with mutations on maternally inherited alleles are resistant to multiple hormones (e.g., PTH, TSH), a variant termed pseudohypoparathyroidism (PHP) type 1a, due to presumed tissue-specific paternal imprinting of the a chain of Gs as demonstrated in murine renal proximal tubule and fat cells. Studies in human tissues thus far revealed imprinting only in pituitary. Because mild hypothyroidism due to TSH resistance occurs in most PHP type 1a patients, we investigated whether Gas is imprinted in thyroid. Examination of eight normal thyroids demonstrated significantly greater expression from the maternal GNAS1 allele, with paternal Gas transcripts accounting for only 25.9–40.4%. Expression of NESP55, XLas, and 1A was uniallelic. We conclude that Gas is incompletely imprinted in the thyroid, which provides an explanation for mild TSH resistance in PHP type 1a. 2002 Elsevier Science (USA). All rights reserved.
- Published
- 2002
50. Sensory nerves regulate mesenchymal stromal cell lineage commitment by tuning sympathetic tones.
- Author
-
Bo Hu, Xiao Lv, Hao Chen, Peng Xue, Bo Gao, Xiao Wang, Gehua Zhen, Janet L. Crane, Dayu Pan, Shen Liu, Shuangfei Ni, Panfeng Wu, Weiping Su, Xiaonan Liu, Mi Yang, Ruoxian Deng, Yusheng Li, Lei Wang, Ying Zhang, and Mei Wan
- Abstract
The sensory nerve was recently identified as being involved in regulation of bone mass accrual. We previously discovered that prostaglandin E2 (PGE2) secreted by osteoblasts could activate sensory nerve EP4 receptor to promote bone formation by inhibiting sympathetic activity. However, the fundamental units of bone formation are active osteoblasts, which originate from mesenchymal stromal/stem cells (MSCs). Here, we found that after sensory denervation, knockout of the EP4 receptor in sensory nerves, or knockout of COX-2 in osteoblasts, could significantly promote adipogenesis and inhibit osteogenesis in adult mice. Furthermore, injection of SW033291 (a small molecule that locally increases the PGE2 level) or propranolol (a beta blocker) significantly promoted osteogenesis and inhibited adipogenesis. This effect of SW033291, but not propranolol, was abolished in conditional EP4-KO mice under normal conditions or in the bone repair process. We conclude that the PGE2/EP4 sensory nerve axis could regulate MSC differentiation in bone marrow of adult mice. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.