1
|
Ye Q, Taleb SJ, Zhao J, Zhao Y. Emerging role of BMPs/BMPR2 signaling pathway in treatment for pulmonary fibrosis. Biomed Pharmacother 2024; 178:117178. [PMID: 39142248 PMCID: PMC11364484 DOI: 10.1016/j.biopha.2024.117178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/16/2024] Open
Abstract
Pulmonary fibrosis is a fatal and chronic lung disease that is characterized by accumulation of thickened scar in the lungs and impairment of gas exchange. The cases with unknown etiology are referred as idiopathic pulmonary fibrosis (IPF). There are currently no effective therapeutics to cure the disease; thus, the investigation of the pathogenesis of IPF is of great importance. Recent studies on bone morphogenic proteins (BMPs) and their receptors have indicated that reduction of BMP signaling in lungs may play a significant role in the development of lung fibrosis. BMPs are members of TGF-β superfamily, and they have been shown to play an anti-fibrotic role in combating TGF-β-mediated pathways. The impact of BMP receptors, in particular BMPR2, on pulmonary fibrosis is growing attraction to researchers. Previous studies on BMPR2 have often focused on pulmonary arterial hypertension (PAH). Given the strong clinical association between PAH and lung fibrosis, understanding BMPs/BMPR2-mediated signaling pathway is important for development of therapeutic strategies to treat IPF. In this review, we comprehensively review recent studies regarding the biological functions of BMPs and their receptors in lungs, especially focusing on their roles in the pathogenesis of pulmonary fibrosis and fibrosis resolution.
Collapse
Affiliation(s)
- Qinmao Ye
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, United States
| | - Sarah J Taleb
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, United States
| | - Jing Zhao
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, United States; Department of internal Medicine, the Ohio State University, Columbus, OH, United States
| | - Yutong Zhao
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, United States; Department of internal Medicine, the Ohio State University, Columbus, OH, United States.
| |
Collapse
|
2
|
Che Z, Sun Q, Zhao Z, Wu Y, Xing H, Song K, Chen A, Wang B, Cai M. Growth factor-functionalized titanium implants for enhanced bone regeneration: A review. Int J Biol Macromol 2024; 274:133153. [PMID: 38897500 DOI: 10.1016/j.ijbiomac.2024.133153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/02/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024]
Abstract
Titanium and titanium alloys are widely favored materials for orthopedic implants due to their exceptional mechanical properties and biological inertness. The additional benefit of sustained local release of bioactive substances further promotes bone tissue formation, thereby augmenting the osseointegration capacity of titanium implants and attracting increasing attention in bone tissue engineering. Among these bioactive substances, growth factors have shown remarkable osteogenic and angiogenic induction capabilities. Consequently, researchers have developed various physical, chemical, and biological loading techniques to incorporate growth factors into titanium implants, ensuring controlled release kinetics. In contrast to conventional treatment modalities, the localized release of growth factors from functionalized titanium implants not only enhances osseointegration but also reduces the risk of complications. This review provides a comprehensive examination of the types and mechanisms of growth factors, along with a detailed exploration of the methodologies used to load growth factors onto the surface of titanium implants. Moreover, it highlights recent advancements in the application of growth factors to the surface of titanium implants (Scheme 1). Finally, the review discusses current limitations and future prospects for growth factor-functionalized titanium implants. In summary, this paper presents cutting-edge design strategies aimed at enhancing the bone regenerative capacity of growth factor-functionalized titanium implants-a significant advancement in the field of enhanced bone regeneration.
Collapse
Affiliation(s)
- Zhenjia Che
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China.
| | - Qi Sun
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Zhenyu Zhao
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Yanglin Wu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Hu Xing
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Kaihang Song
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Aopan Chen
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Bo Wang
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China.
| | - Ming Cai
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China.
| |
Collapse
|
3
|
Wei D, Su Y, Leung PCK, Li Y, Chen ZJ. Roles of bone morphogenetic proteins in endometrial remodeling during the human menstrual cycle and pregnancy. Hum Reprod Update 2024; 30:215-237. [PMID: 38037193 DOI: 10.1093/humupd/dmad031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/17/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND During the human menstrual cycle and pregnancy, the endometrium undergoes a series of dynamic remodeling processes to adapt to physiological changes. Insufficient endometrial remodeling, characterized by inadequate endometrial proliferation, decidualization and spiral artery remodeling, is associated with infertility, endometriosis, dysfunctional uterine bleeding, and pregnancy-related complications such as preeclampsia and miscarriage. Bone morphogenetic proteins (BMPs), a subset of the transforming growth factor-β (TGF-β) superfamily, are multifunctional cytokines that regulate diverse cellular activities, such as differentiation, proliferation, apoptosis, and extracellular matrix synthesis, are now understood as integral to multiple reproductive processes in women. Investigations using human biological samples have shown that BMPs are essential for regulating human endometrial remodeling processes, including endometrial proliferation and decidualization. OBJECTIVE AND RATIONALE This review summarizes our current knowledge on the known pathophysiological roles of BMPs and their underlying molecular mechanisms in regulating human endometrial proliferation and decidualization, with the goal of promoting the development of innovative strategies for diagnosing, treating and preventing infertility and adverse pregnancy complications associated with dysregulated human endometrial remodeling. SEARCH METHODS A literature search for original articles published up to June 2023 was conducted in the PubMed, MEDLINE, and Google Scholar databases, identifying studies on the roles of BMPs in endometrial remodeling during the human menstrual cycle and pregnancy. Articles identified were restricted to English language full-text papers. OUTCOMES BMP ligands and receptors and their transduction molecules are expressed in the endometrium and at the maternal-fetal interface. Along with emerging technologies such as tissue microarrays, 3D organoid cultures and advanced single-cell transcriptomics, and given the clinical availability of recombinant human proteins and ongoing pharmaceutical development, it is now clear that BMPs exert multiple roles in regulating human endometrial remodeling and that these biomolecules (and their receptors) can be targeted for diagnostic and therapeutic purposes. Moreover, dysregulation of these ligands, their receptors, or signaling determinants can impact endometrial remodeling, contributing to infertility or pregnancy-related complications (e.g. preeclampsia and miscarriage). WIDER IMPLICATIONS Although further clinical trials are needed, recent advancements in the development of recombinant BMP ligands, synthetic BMP inhibitors, receptor antagonists, BMP ligand sequestration tools, and gene therapies have underscored the BMPs as candidate diagnostic biomarkers and positioned the BMP signaling pathway as a promising therapeutic target for addressing infertility and pregnancy complications related to dysregulated human endometrial remodeling.
Collapse
Affiliation(s)
- Daimin Wei
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, China
| | - Yaxin Su
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
| | - Peter C K Leung
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Yan Li
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong, China
| |
Collapse
|
4
|
Che Z, Song Y, Zhu L, Liu T, Li X, Huang L. Emerging roles of growth factors in osteonecrosis of the femoral head. Front Genet 2022; 13:1037190. [PMID: 36452155 PMCID: PMC9702520 DOI: 10.3389/fgene.2022.1037190] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/24/2022] [Indexed: 12/20/2023] Open
Abstract
Osteonecrosis of the femoral head (ONFH) is a potentially disabling orthopedic condition that requires total hip arthroplasty in most late-stage cases. However, mechanisms underlying the development of ONFH remain unknown, and the therapeutic strategies remain limited. Growth factors play a crucial role in different physiological processes, including cell proliferation, invasion, metabolism, apoptosis, and stem cell differentiation. Recent studies have reported that polymorphisms of growth factor-related genes are involved in the pathogenesis of ONFH. Tissue and genetic engineering are attractive strategies for treating early-stage ONFH. In this review, we summarized dysregulated growth factor-related genes and their role in the occurrence and development of ONFH. In addition, we discussed their potential clinical applications in tissue and genetic engineering for the treatment of ONFH.
Collapse
Affiliation(s)
- Zhenjia Che
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Yang Song
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Liwei Zhu
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Tengyue Liu
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Xudong Li
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Lanfeng Huang
- Department of Orthopaedics, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
5
|
Wu CK, Wei MT, Wu HC, Wu CL, Wu CJ, Liaw H, Su WP. BMP2 promotes lung adenocarcinoma metastasis through BMP receptor 2-mediated SMAD1/5 activation. Sci Rep 2022; 12:16310. [PMID: 36175474 PMCID: PMC9522928 DOI: 10.1038/s41598-022-20788-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 09/19/2022] [Indexed: 12/04/2022] Open
Abstract
Bone morphogenetic protein 2 (BMP2) is highly overexpressed in human non-small cell lung cancer (NSCLC) and correlates with tumor stage and metastatic burden. Although several lines of evidence suggest that BMP2 promotes cell migration and invasiveness in vitro, the in vivo role of BMP2 in the metastasis of lung adenocarcinoma cells remains less well understood. Here, we revealed that BMP2 is highly overexpressed in lung adenocarcinoma patients with lymph node metastasis compared with patients without lymph node metastasis. Using an in vivo orthotopic mouse model, we clearly demonstrated that BMP2 promotes lung adenocarcinoma metastasis. The depletion of BMP2 or its receptor BMPR2 significantly reduced cell migration and invasiveness. We further identified that BMP2/BMPR2-mediated cell migration involves the activation of the SMAD1/5/8 signaling pathway, independent of the KRAS signaling pathway. Significantly, the depletion of SMAD1/5/8 or the inhibition of SMAD1/5/8 by LDN193189 inhibitor significantly reduced cell migration. These findings show that BMP2 promotes NSCLC metastasis, indicating that targeting the BMP2 signaling pathway may represent a potential therapeutic strategy for treating patients with metastatic NSCLC.
Collapse
Affiliation(s)
- Cheng-Kuei Wu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, No. 35, Xiao-Tong Road, Tainan, 704, Taiwan
| | - Man-Ting Wei
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, No. 35, Xiao-Tong Road, Tainan, 704, Taiwan
| | - Hung-Chang Wu
- Division of Hematology and Oncology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan.,Department of Pharmacy, Chia-Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Cheng-Lin Wu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, No. 35, Xiao-Tong Road, Tainan, 704, Taiwan.,Department of Pathology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Cheng-Ju Wu
- Departments of Oncology and Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hungjiun Liaw
- Department of Life Sciences, National Cheng Kung University, No. 1 University Road, Tainan City, 701, Taiwan.
| | - Wen-Pin Su
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, No. 35, Xiao-Tong Road, Tainan, 704, Taiwan. .,Departments of Oncology and Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
6
|
Carlson WD, Keck PC, Bosukonda D, Carlson FR. A Process for the Design and Development of Novel Bone Morphogenetic Protein-7 (BMP-7) Mimetics With an Example: THR-184. Front Pharmacol 2022; 13:864509. [PMID: 35873578 PMCID: PMC9306349 DOI: 10.3389/fphar.2022.864509] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 06/13/2022] [Indexed: 11/29/2022] Open
Abstract
Growth Factors have been evaluated as therapeutic targets for the treatment of a broad spectrum of diseases. Because they are proteins with pleiotropic effects, the quest to harness their beneficial effects has presented challenges. Most Growth Factors operate at the extracellular-receptor level and have natural feedback mechanisms that modulate their effects. As proteins, they are difficult and expensive to manufacture. Frequently proteins must be administered parenterally, may invoke an immune response, and may be neutralized by naturally occurring inhibitors. To circumvent these limitations, we have undertaken an effort to develop mimetics for the Bone Morphogenetic Protein (BMP) signaling pathway effects that incorporate the beneficial effects, eliminate the deleterious effects, and thereby create effective drug-like compounds.To this end, we have designed and tested a family of small peptide BMP mimetics. The design used the three-dimensional structure of BMP-7 to identify likely active surface regions. Lead sequences were then optimized based on in vitro assays that examine the selective binding to BMP receptors, demonstrate the phosphorylation of Smad-1,5,8, detect anti-apoptosis and anti-inflammation, and block the epithelial to mesenchymal transition (EMT) in renal tubular epithelial cells. These sequences were further optimized using in vivo assays of the attenuation of acute kidney injury in a rat-model of unilateral clamp ischemic reperfusion. This process uses a Structure Variance Analysis algorithm (SVA) to identify structure/activity relationships. One member of this family, THR-184, is an agonist of BMP signaling and a potent antagonist of TGFβ signaling. This small peptide mimetic inhibits inflammation, apoptosis, fibrosis and reverses epithelial to mesenchymal transition (EMT) by regulating multiple signaling pathways involved in the cellular injury of multiple organs. Its effects have been shown to control Acute Kidney Injury (AKI). THR-184 has progressed through phase I and II clinical trials for the prevention of Cardio-Vascular Surgery (CVS) associated AKI. This work provides a roadmap for the development of other growth factor mimetics and demonstrates how we might harness their therapeutic potential.
Collapse
Affiliation(s)
- William D. Carlson
- Division of Cardiology, Mass General Hospital/Harvard, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
- Therapeutics By Design, Boston, MA, United States
- Thrasos Therapeutics, Hopkinton, MA, United States
- *Correspondence: William D. Carlson,
| | - Peter C. Keck
- Therapeutics By Design, Boston, MA, United States
- Thrasos Therapeutics, Hopkinton, MA, United States
| | - Dattatreyamurty Bosukonda
- Division of Cardiology, Mass General Hospital/Harvard, Boston, MA, United States
- Therapeutics By Design, Boston, MA, United States
- Thrasos Therapeutics, Hopkinton, MA, United States
| | - Frederic Roy Carlson
- Therapeutics By Design, Boston, MA, United States
- Thrasos Therapeutics, Hopkinton, MA, United States
| |
Collapse
|
7
|
Zhu L, Liu Y, Wang A, Zhu Z, Li Y, Zhu C, Che Z, Liu T, Liu H, Huang L. Application of BMP in Bone Tissue Engineering. Front Bioeng Biotechnol 2022; 10:810880. [PMID: 35433652 PMCID: PMC9008764 DOI: 10.3389/fbioe.2022.810880] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 03/01/2022] [Indexed: 01/15/2023] Open
Abstract
At present, bone nonunion and delayed union are still difficult problems in orthopaedics. Since the discovery of bone morphogenetic protein (BMP), it has been widely used in various studies due to its powerful role in promoting osteogenesis and chondrogenesis. Current results show that BMPs can promote healing of bone defects and reduce the occurrence of complications. However, the mechanism of BMP in vivo still needs to be explored, and application of BMP alone to a bone defect site cannot achieve good therapeutic effects. It is particularly important to modify implants to carry BMP to achieve slow and sustained release effects by taking advantage of the nature of the implant. This review aims to explain the mechanism of BMP action in vivo, its biological function, and how BMP can be applied to orthopaedic implants to effectively stimulate bone healing in the long term. Notably, implantation of a system that allows sustained release of BMP can provide an effective method to treat bone nonunion and delayed bone healing in the clinic.
Collapse
Affiliation(s)
- Liwei Zhu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
- Orthopaedic Research Institute of Jilin Province, Changchun, China
| | - Yuzhe Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Ao Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Zhengqing Zhu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Youbin Li
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Chenyi Zhu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Zhenjia Che
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Tengyue Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - He Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
- Orthopaedic Research Institute of Jilin Province, Changchun, China
- *Correspondence: He Liu, ; Lanfeng Huang,
| | - Lanfeng Huang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: He Liu, ; Lanfeng Huang,
| |
Collapse
|
8
|
Sugii H, Albougha MS, Adachi O, Tomita H, Tomokiyo A, Hamano S, Hasegawa D, Yoshida S, Itoyama T, Maeda H. Activin A Promotes Osteoblastic Differentiation of Human Preosteoblasts through the ALK1-Smad1/5/9 Pathway. Int J Mol Sci 2021; 22:13491. [PMID: 34948289 PMCID: PMC8704413 DOI: 10.3390/ijms222413491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/10/2021] [Accepted: 12/14/2021] [Indexed: 11/17/2022] Open
Abstract
Activin A, a member of transforming growth factor-β superfamily, is involved in the regulation of cellular differentiation and promotes tissue healing. Previously, we reported that expression of activin A was upregulated around the damaged periodontal tissue including periodontal ligament (PDL) tissue and alveolar bone, and activin A promoted PDL-related gene expression of human PDL cells (HPDLCs). However, little is known about the biological function of activin A in alveolar bone. Thus, this study analyzed activin A-induced biological functions in preosteoblasts (Saos2 cells). Activin A promoted osteoblastic differentiation of Saos2 cells. Activin receptor-like kinase (ALK) 1, an activin type I receptor, was more strongly expressed in Saos2 cells than in HPDLCs, and knockdown of ALK1 inhibited activin A-induced osteoblastic differentiation of Saos2 cells. Expression of ALK1 was upregulated in alveolar bone around damaged periodontal tissue when compared with a nondamaged site. Furthermore, activin A promoted phosphorylation of Smad1/5/9 during osteoblastic differentiation of Saos2 cells and knockdown of ALK1 inhibited activin A-induced phosphorylation of Smad1/5/9 in Saos2 cells. Collectively, these findings suggest that activin A promotes osteoblastic differentiation of preosteoblasts through the ALK1-Smad1/5/9 pathway and could be used as a therapeutic product for the healing of alveolar bone as well as PDL tissue.
Collapse
Affiliation(s)
- Hideki Sugii
- Department of Endodontology and Operative Dentistry, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan; (M.S.A.); (O.A.); (H.T.); (S.H.); (H.M.)
| | - Mhd Safwan Albougha
- Department of Endodontology and Operative Dentistry, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan; (M.S.A.); (O.A.); (H.T.); (S.H.); (H.M.)
| | - Orie Adachi
- Department of Endodontology and Operative Dentistry, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan; (M.S.A.); (O.A.); (H.T.); (S.H.); (H.M.)
| | - Hiroka Tomita
- Department of Endodontology and Operative Dentistry, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan; (M.S.A.); (O.A.); (H.T.); (S.H.); (H.M.)
| | - Atsushi Tomokiyo
- Department of Endodontology, Kyushu University Hospital, Fukuoka 812-8582, Japan; (A.T.); (D.H.); (S.Y.); (T.I.)
| | - Sayuri Hamano
- Department of Endodontology and Operative Dentistry, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan; (M.S.A.); (O.A.); (H.T.); (S.H.); (H.M.)
- OBT Center, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan
| | - Daigaku Hasegawa
- Department of Endodontology, Kyushu University Hospital, Fukuoka 812-8582, Japan; (A.T.); (D.H.); (S.Y.); (T.I.)
| | - Shinichiro Yoshida
- Department of Endodontology, Kyushu University Hospital, Fukuoka 812-8582, Japan; (A.T.); (D.H.); (S.Y.); (T.I.)
| | - Tomohiro Itoyama
- Department of Endodontology, Kyushu University Hospital, Fukuoka 812-8582, Japan; (A.T.); (D.H.); (S.Y.); (T.I.)
| | - Hidefumi Maeda
- Department of Endodontology and Operative Dentistry, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka 812-8582, Japan; (M.S.A.); (O.A.); (H.T.); (S.H.); (H.M.)
- Department of Endodontology, Kyushu University Hospital, Fukuoka 812-8582, Japan; (A.T.); (D.H.); (S.Y.); (T.I.)
| |
Collapse
|
9
|
Lee J, Jang EH, Kim JH, Park S, Kang Y, Park S, Lee K, Kim JH, Youn YN, Ryu W. Highly flexible and porous silk fibroin microneedle wraps for perivascular drug delivery. J Control Release 2021; 340:125-135. [PMID: 34688718 DOI: 10.1016/j.jconrel.2021.10.024] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/17/2021] [Accepted: 10/19/2021] [Indexed: 12/29/2022]
Abstract
Various perivascular drug delivery techniques have been demonstrated for localized post-treatment of intimal hyperplasia: a vascular inflammatory response caused by endothelial damages. Although most perivascular devices have focused on controlling the delivery duration of anti-proliferation drug, the confined and unidirectional delivery of the drug to the target tissue has become increasingly important. In addition, careful attention should also be paid to the luminal stability and the adequate exchange of vascular protein or cell between the blood vessel and extravascular tissue to avoid any side effect from the long-term application of any perivascular device. Here, a highly flexible and porous silk fibroin microneedle wrap (Silk MN wrap) is proposed to directly inject antiproliferative drug to the anastomosis sites while ensuring sufficient vascular exchanges. Drug-embedded silk MNs were transfer-molded on a highly flexible and porous silk wrap. The enhanced cell compatibility, molecular permeability, and flexibility of silk MN wrap guaranteed the structural integrity of blood vessels. Silk wrap successfully supported the silk MNs and induced multiple MN penetration to the target tissue. Over 28 days, silk MN wrap significantly inhibited intimal hyperplasia with a 62.1% reduction in neointimal formation.
Collapse
Affiliation(s)
- JiYong Lee
- School of Mechanical Engineering, YONSEI University, Seoul 03722, South Korea
| | - Eui Hwa Jang
- Division of Cardiovascular Surgery, Severance Cardiovascular Hospital, YONSEI University College of Medicine, Seoul 03722, South Korea
| | - Jae Ho Kim
- School of Mechanical Engineering, YONSEI University, Seoul 03722, South Korea
| | - SeungHyun Park
- School of Mechanical Engineering, YONSEI University, Seoul 03722, South Korea
| | - Yosup Kang
- School of Mechanical Engineering, YONSEI University, Seoul 03722, South Korea
| | - Sanghyun Park
- School of Mechanical Engineering, YONSEI University, Seoul 03722, South Korea
| | - KangJu Lee
- Department of Healthcare and Biomedical Engineering, Chonnam National University, Yeosu 59626, South Korea; Terasaki Institute for Biomedical Innovation, Los Angeles, CA 90005, USA
| | - Jung-Hwan Kim
- Division of Cardiovascular Surgery, Severance Cardiovascular Hospital, YONSEI University College of Medicine, Seoul 03722, South Korea
| | - Young-Nam Youn
- Division of Cardiovascular Surgery, Severance Cardiovascular Hospital, YONSEI University College of Medicine, Seoul 03722, South Korea.
| | - WonHyoung Ryu
- School of Mechanical Engineering, YONSEI University, Seoul 03722, South Korea.
| |
Collapse
|
10
|
Migliorini E, Guevara-Garcia A, Albiges-Rizo C, Picart C. Learning from BMPs and their biophysical extracellular matrix microenvironment for biomaterial design. Bone 2020; 141:115540. [PMID: 32730925 PMCID: PMC7614069 DOI: 10.1016/j.bone.2020.115540] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/17/2020] [Accepted: 07/18/2020] [Indexed: 01/19/2023]
Abstract
It is nowadays well-accepted that the extracellular matrix (ECM) is not a simple reservoir for growth factors but is an organization center of their biological activity. In this review, we focus on the ability of the ECM to regulate the biological activity of BMPs. In particular, we survey the role of the ECM components, notably the glycosaminoglycans and fibrillary ECM proteins, which can be promoters or repressors of the biological activities mediated by the BMPs. We examine how a process called mechano-transduction induced by the ECM can affect BMP signaling, including BMP internalization by the cells. We also focus on the spatio-temporal regulation of the BMPs, including their release from the ECM, which enables to modulate their spatial localization as well as their local concentration. We highlight how biomaterials can recapitulate some aspects of the BMPs/ECM interactions and help to answer fundamental questions to reveal previously unknown molecular mechanisms. Finally, the design of new biomaterials inspired by the ECM to better present BMPs is discussed, and their use for a more efficient bone regeneration in vivo is also highlighted.
Collapse
Affiliation(s)
- Elisa Migliorini
- CNRS, Grenoble Institute of Technology, LMGP, UMR 5628, 3 Parvis Louis Néel, 38016 Grenoble, France; CEA, Institute of Interdisciplinary Research of Grenoble (IRIG), Biomimetism and Regenerative Medicine Lab, ERL 5000, Université Grenoble-Alpes (UGA)/CEA/CNRS, Grenoble France.
| | - Amaris Guevara-Garcia
- CNRS, Grenoble Institute of Technology, LMGP, UMR 5628, 3 Parvis Louis Néel, 38016 Grenoble, France; CEA, Institute of Interdisciplinary Research of Grenoble (IRIG), Biomimetism and Regenerative Medicine Lab, ERL 5000, Université Grenoble-Alpes (UGA)/CEA/CNRS, Grenoble France; Université Grenoble Alpes, Institut for Advances Biosciences, Institute Albert Bonniot, INSERM U1209, CNRS 5309, La Tronche, France
| | - Corinne Albiges-Rizo
- Université Grenoble Alpes, Institut for Advances Biosciences, Institute Albert Bonniot, INSERM U1209, CNRS 5309, La Tronche, France
| | - Catherine Picart
- CNRS, Grenoble Institute of Technology, LMGP, UMR 5628, 3 Parvis Louis Néel, 38016 Grenoble, France; CEA, Institute of Interdisciplinary Research of Grenoble (IRIG), Biomimetism and Regenerative Medicine Lab, ERL 5000, Université Grenoble-Alpes (UGA)/CEA/CNRS, Grenoble France.
| |
Collapse
|
11
|
Posa F, Baha-Schwab EH, Wei Q, Di Benedetto A, Neubauer S, Reichart F, Kessler H, Spatz JP, Albiges-Rizo C, Mori G, Cavalcanti-Adam EA. Surface Co-presentation of BMP-2 and integrin selective ligands at the nanoscale favors α 5β 1 integrin-mediated adhesion. Biomaterials 2020; 267:120484. [PMID: 33142116 DOI: 10.1016/j.biomaterials.2020.120484] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 10/14/2020] [Accepted: 10/21/2020] [Indexed: 12/17/2022]
Abstract
Here we present the use of surface nanopatterning of covalently immobilized BMP-2 and integrin selective ligands to determine the specificity of their interactions in regulating cell adhesion and focal adhesion assembly. Gold nanoparticle arrays carrying single BMP-2 dimers are prepared by block-copolymer micellar nanolithography and azide-functionalized integrin ligands (cyclic-RGD peptides or α5β1 integrin peptidomimetics) are immobilized on the surrounding polyethylene glycol alkyne by click chemistry. Compared to BMP-2 added to the media, surface immobilized BMP-2 (iBMP-2) favors the spatial segregation of adhesion clusters and enhances focal adhesion (FA) size in cells adhering to α5β1 integrin selective ligands. Moreover, iBMP-2 copresented with α5β1 integrin ligands induces the recruitment of αvβ3 integrins in FAs. When copresented with RGD, iBMP-2 induces the assembly of a higher number of FAs, which are not affected by α5β1 integrin blocking. Our dual-functionalized platforms offer the possibility to study the crosstalk between integrins and BMP receptors, and more in general they could be used to address the spatial regulation of growth factors and adhesion receptors crosstalk on biomimetic surfaces.
Collapse
Affiliation(s)
- Francesca Posa
- Max Planck Institute for Medical Research, Department of Cellular Biophysics, Jahnstraße 29, Heidelberg, 69120, Germany
| | - Elisabeth H Baha-Schwab
- Max Planck Institute for Medical Research, Department of Cellular Biophysics, Jahnstraße 29, Heidelberg, 69120, Germany
| | - Qiang Wei
- Max Planck Institute for Medical Research, Department of Cellular Biophysics, Jahnstraße 29, Heidelberg, 69120, Germany
| | - Adriana Di Benedetto
- University of Foggia, Department of Clinical and Experimental Medicine, viale Pinto 1, Foggia, 71122, Italy
| | - Stefanie Neubauer
- Institute for Advanced Study and Center of Integrated Protein Science (CIPSM), Department of Chemistry, Technische Universität München, Lichtenbergstraße 4, Garching, 85748, Germany
| | - Florian Reichart
- Institute for Advanced Study and Center of Integrated Protein Science (CIPSM), Department of Chemistry, Technische Universität München, Lichtenbergstraße 4, Garching, 85748, Germany
| | - Horst Kessler
- Institute for Advanced Study and Center of Integrated Protein Science (CIPSM), Department of Chemistry, Technische Universität München, Lichtenbergstraße 4, Garching, 85748, Germany
| | - Joachim P Spatz
- Max Planck Institute for Medical Research, Department of Cellular Biophysics, Jahnstraße 29, Heidelberg, 69120, Germany
| | - Corinne Albiges-Rizo
- Institut Albert Bonniot, Université Joseph Fourier, INSERM U823, CNRS ERL 5284, Grenoble Alpessite Santé, Grenoble Cedex, 09, F38042, France
| | - Giorgio Mori
- University of Foggia, Department of Clinical and Experimental Medicine, viale Pinto 1, Foggia, 71122, Italy
| | - Elisabetta Ada Cavalcanti-Adam
- Max Planck Institute for Medical Research, Department of Cellular Biophysics, Jahnstraße 29, Heidelberg, 69120, Germany.
| |
Collapse
|
12
|
Mikai A, Ono M, Tosa I, Nguyen HTT, Hara ES, Nosho S, Kimura-Ono A, Nawachi K, Takarada T, Kuboki T, Oohashi T. BMP-2/β-TCP Local Delivery for Bone Regeneration in MRONJ-Like Mouse Model. Int J Mol Sci 2020; 21:ijms21197028. [PMID: 32987737 PMCID: PMC7583034 DOI: 10.3390/ijms21197028] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/14/2020] [Accepted: 09/22/2020] [Indexed: 12/25/2022] Open
Abstract
Medication-related osteonecrosis of the jaw (MRONJ) is a severe pathological condition associated mainly with the long-term administration of bone resorption inhibitors, which are known to induce suppression of osteoclast activity and bone remodeling. Bone Morphogenetic Protein (BMP)-2 is known to be a strong inducer of bone remodeling, by directly regulating osteoblast differentiation and osteoclast activity. This study aimed to evaluate the effects of BMP-2 adsorbed onto beta-tricalcium phosphate (β-TCP), which is an osteoinductive bioceramic material and allows space retention, on the prevention and treatment of MRONJ in mice. Tooth extraction was performed after 3 weeks of zoledronate (ZA) and cyclophosphamide (CY) administration. For prevention studies, BMP-2/β-TCP was transplanted immediately after tooth extraction, and the mice were administered ZA and CY for an additional 4 weeks. The results showed that while the tooth extraction socket was mainly filled with a sparse tissue in the control group, bone formation was observed at the apex of the tooth extraction socket and was filled with a dense connective tissue rich in cellular components in the BMP-2/β-TCP transplanted group. For treatment studies, BMP-2/β-TCP was transplanted 2 weeks after tooth extraction, and bone formation was followed up for the subsequent 4 weeks under ZA and CY suspension. The results showed that although the tooth extraction socket was mainly filled with soft tissue in the control group, transplantation of BMP-2/β-TCP could significantly accelerate bone formation, as shown by immunohistochemical analysis for osteopontin, and reduce the bone necrosis in tooth extraction sockets. These data suggest that the combination of BMP-2/β-TCP could become a suitable therapy for the management of MRONJ.
Collapse
Affiliation(s)
- Akihiro Mikai
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (A.M.); (H.T.T.N.); (S.N.); (T.O.)
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (I.T.); (A.K.-O.); (K.N.); (T.K.)
| | - Mitsuaki Ono
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (A.M.); (H.T.T.N.); (S.N.); (T.O.)
- Correspondence: ; Tel.: +81-86-235-7129; Fax: +81-86-222-7768
| | - Ikue Tosa
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (I.T.); (A.K.-O.); (K.N.); (T.K.)
| | - Ha Thi Thu Nguyen
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (A.M.); (H.T.T.N.); (S.N.); (T.O.)
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (I.T.); (A.K.-O.); (K.N.); (T.K.)
| | - Emilio Satoshi Hara
- Department of Biomaterials, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan;
| | - Shuji Nosho
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (A.M.); (H.T.T.N.); (S.N.); (T.O.)
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (I.T.); (A.K.-O.); (K.N.); (T.K.)
| | - Aya Kimura-Ono
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (I.T.); (A.K.-O.); (K.N.); (T.K.)
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama 700-8558, Japan
| | - Kumiko Nawachi
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (I.T.); (A.K.-O.); (K.N.); (T.K.)
| | - Takeshi Takarada
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan;
| | - Takuo Kuboki
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (I.T.); (A.K.-O.); (K.N.); (T.K.)
| | - Toshitaka Oohashi
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (A.M.); (H.T.T.N.); (S.N.); (T.O.)
| |
Collapse
|
13
|
Halloran D, Durbano HW, Nohe A. Bone Morphogenetic Protein-2 in Development and Bone Homeostasis. J Dev Biol 2020; 8:E19. [PMID: 32933207 PMCID: PMC7557435 DOI: 10.3390/jdb8030019] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/01/2020] [Accepted: 09/11/2020] [Indexed: 12/11/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are multi-functional growth factors belonging to the Transforming Growth Factor-Beta (TGF-β) superfamily. These proteins are essential to many developmental processes, including cardiogenesis, neurogenesis, and osteogenesis. Specifically, within the BMP family, Bone Morphogenetic Protein-2 (BMP-2) was the first BMP to be characterized and has been well-studied. BMP-2 has important roles during embryonic development, as well as bone remodeling and homeostasis in adulthood. Some of its specific functions include digit formation and activating osteogenic genes, such as Runt-Related Transcription Factor 2 (RUNX2). Because of its diverse functions and osteogenic potential, the Food and Drug Administration (FDA) approved usage of recombinant human BMP-2 (rhBMP-2) during spinal fusion surgery, tibial shaft repair, and maxillary sinus reconstructive surgery. However, shortly after initial injections of rhBMP-2, several adverse complications were reported, and alternative therapeutics have been developed to limit these side-effects. As the clinical application of BMP-2 is largely implicated in bone, we focus primarily on its role in bone. However, we also describe briefly the role of BMP-2 in development. We then focus on the structure of BMP-2, its activation and regulation signaling pathways, BMP-2 clinical applications, and limitations of using BMP-2 as a therapeutic. Further, this review explores other potential treatments that may be useful in treating bone disorders.
Collapse
Affiliation(s)
| | | | - Anja Nohe
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA; (D.H.); (H.W.D.)
| |
Collapse
|
14
|
Association of distinct type 1 bone morphogenetic protein receptors with different molecular pathways and survival outcomes in neuroblastoma. Neuronal Signal 2020; 4:NS20200006. [PMID: 32714600 PMCID: PMC7366490 DOI: 10.1042/ns20200006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/14/2020] [Accepted: 04/15/2020] [Indexed: 12/21/2022] Open
Abstract
Neuroblastoma (NB) is a paediatric cancer that arises in the sympathetic nervous system. Patients with stage 4 tumours have poor outcomes and 20% of high-risk cases have MYCN amplification. The bone morphogenetic proteins (BMPs) play roles in sympathetic neuritogenesis, by signalling through bone morphogenetic protein receptor (BMPR)2 and either BMPR1A or BMPR1B. Alterations in BMPR2 expression have been reported in NB; it is unknown if the expression of BMPR1A or BMPR1B is altered. We report lower BMPR2 and BMPR1B, and higher BMPR1A, expression in stage 4 and in MYCN-amplified NB. Kaplan–Meier plots showed that high BMPR2 or BMPR1B expression was linked to better survival, while high BMPR1A was linked to worse survival. Gene ontology enrichment and pathway analyses revealed that BMPR2 and BMPR1B co-expressed genes were enriched in those associated with NB differentiation. BMPR1A co-expressed genes were enriched in those associated with cell proliferation. Moreover, the correlation between BMPR2 and BMPR1A was strengthened, while the correlation between BMPR2 and BMPR1B was lost, in MYCN-amplified NB. This suggested that differentiation should decrease BMPR1A and increase BMPR1B expression. In agreement, nerve growth factor treatment of cultured sympathetic neurons decreased Bmpr1a expression and increased Bmpr1b expression. Overexpression of dominant negative BMPR1B, treatment with a BMPR1B inhibitor and treatment with GDF5, which signals via BMPR1B, showed that BMPR1B signalling is required for optimal neuritogenesis in NB cells, suggesting that loss of BMPR1B may alter neuritogenesis. The present study shows that expression of distinct BMPRs is associated with different survival outcomes in NB.
Collapse
|
15
|
Son S, Yoon SH, Kim MH, Yun X. Activin A and BMP chimera (AB204) induced bone fusion in osteoporotic spine using an ovariectomized rat model. Spine J 2020; 20:809-820. [PMID: 31899374 DOI: 10.1016/j.spinee.2019.12.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 12/22/2019] [Accepted: 12/23/2019] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Recombinant human bone morphogenic protein 2 (rhBMP2) has been used to induce bone fusion in patients with spinal fusion surgery. However, the effectiveness of rhBMP2 in the bone fusion process is limited in osteoporosis patients, and a high dose of rhBMP2 for enough bone fusion sometimes provokes side effects. Therefore, substitutes for rhBMP2 with a higher therapeutic potency are needed, and already several studies have published the effectiveness of Activin A/BMP2 chimera (AB204) in new bone formation process in vitro and in vivo. PURPOSE In the present study, we provide evidence that bone fusion activity of AB204 is superior to that of rhBMP2 in osteoporotic rat models. STUDY DESIGN/SETTINGS An in vivo animal study was carried out. METHODS A total of 40 Sprague-Dawley rats underwent bilateral ovariectomy. At 6 weeks after ovariectomy, a lumbar spinal bone fusion model of bilateral intertransverse process was performed. All rats were randomly divided into four groups as follows: rats receiving 5 µg of rhBMP2 (Group I), rats receiving 10 µg of rhBMP2 (Group II), rats receiving 5 µg of AB204 (Group III), and rats receiving 10 µg of AB204 (Group IV). Simple radiographs were performed at 6 and 12 weeks after bone fusion, and direct palpation, micro-CT, and immunohistochemistry (hematoxylin-eosin stain and Masson's trichrome stain) were performed at 12 weeks after bone fusion. The qualitative degree of bone fusion was assessed as manual fusion score from direct palpation, and radio-histologic fusion score from simple radiographs, micro-CT, and immunohistochemistry. Also, the quantitative degree of bone fusion was assessed using fusion bone volume by micro-CT and serum osteocalcin level as bone turnover markers. RESULTS The change of body weight was not different among the groups during follow-up. The qualitative degree of bone fusion assessed by direct palpation, simple radiographs, micro-CT, and histologic evaluation was significantly different among the four groups. Also, the quantitative degree of bone fusion including fusion bone volume and serum osteocalcin was significantly different among the groups. Especially, in manual fusion score, radio-histologic fusion score, and fusion bone volume, the AB204 group revealed superior results to the rhBMP2 group when using the same dose. Furthermore, even the low-dose AB204 group (Group III) showed superior results to the high-dose rhBMP2 group (Group II) in radio-histologic fusion score and fusion bone volume. CONCLUSION The effect of bone fusion in osteoporotic rats was significantly higher in the AB204 group than in the rhBMP2 group. CLINICAL SIGNIFICANCE If further organized animal studies and clinical trials are provided, AB204 may be a good substitute for rhBMP2 in osteoporotic spinal fusion surgery, as a superior osteogenesis inducer.
Collapse
Affiliation(s)
- Seong Son
- Department of Neurosurgery, Gil Medical Center, Gachon University College of Medicine, Incheon, South Korea
| | - Seung Hwan Yoon
- Department of Neurosurgery, Inha University Hospital, Inha University College of Medicine, Incheon, South Korea.
| | - Moon Hang Kim
- Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
| | - Xiang Yun
- Department of Orthopedic Surgery, School of Medicine, Ajou University, Suwon, South Korea
| |
Collapse
|
16
|
Hu J, Wu J, Li Y, Wang Z, Tang J, Li Z, Hu L, Huang Q, Ye L, Xu L. Sclerostin domain-containing protein 1 is dispensable for the differentiation of follicular helper and follicular regulatory T cells during acute viral infection. Am J Transl Res 2019; 11:3722-3736. [PMID: 31312383 PMCID: PMC6614606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 05/07/2019] [Indexed: 06/10/2023]
Abstract
T follicular helper (TFH) cells are crucial for effective humoral immunity by providing the required signals to cognate B cells and promoting germinal center (GC) formation. Many intrinsic and extrinsic factors have been reported to be involved in the multistage, multifactorial differentiation process of TFH cells. By comparing gene expression between TFH cells and TH1 cells based on published GEO data, we found selective and high expression of sclerostin domain-containing protein 1 (SOSTDC1) in TFH cells but not in TH1 cells; however, it is unclear whether SOSTDC1 is important for the differentiation and/or function of TFH cells. Using a mouse model of acute lymphocytic choriomeningitis virus (LCMV) infection, we confirmed the selective expression of SOSTDC1 in TFH cells compared to that in TH1 cells, but the ablation of SOSTDC1 did not affect TFH cell differentiation or effector function. Thus, our results indicate that the SOSTDC1 protein is merely a specific marker of TFH cells but does not play a functional role in the differentiation of TFH cells during acute viral infection.
Collapse
Affiliation(s)
- Jianjun Hu
- Institute of Immunology, Third Military Medical UniversityChongqing 400038, China
| | - Jialin Wu
- Institute of Immunology, Third Military Medical UniversityChongqing 400038, China
| | - Yiding Li
- Institute of Immunology, Third Military Medical UniversityChongqing 400038, China
| | - Zhiming Wang
- Institute of Immunology, Third Military Medical UniversityChongqing 400038, China
| | - Jianfang Tang
- Institute of Immunology, Third Military Medical UniversityChongqing 400038, China
| | - Zhirong Li
- Institute of Immunology, Third Military Medical UniversityChongqing 400038, China
| | - Li Hu
- Institute of Immunology, Third Military Medical UniversityChongqing 400038, China
| | - Qizhao Huang
- Department of Oncology, General Hospital of Western Theater CommandChengdu, China
| | - Lilin Ye
- Institute of Immunology, Third Military Medical UniversityChongqing 400038, China
| | - Lifan Xu
- Institute of Immunology, Third Military Medical UniversityChongqing 400038, China
| |
Collapse
|
17
|
Gu H, Xue Z, Wang M, Yang M, Wang K, Xu D. Effect of Hydroxyapatite Surface on BMP-2 Biological Properties by Docking and Molecular Simulation Approaches. J Phys Chem B 2019; 123:3372-3382. [DOI: 10.1021/acs.jpcb.9b01982] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
| | | | - Menghao Wang
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan 610031, P. R. China
| | | | | | | |
Collapse
|
18
|
Ning J, Zhao Y, Ye Y, Yu J. Opposing roles and potential antagonistic mechanism between TGF-β and BMP pathways: Implications for cancer progression. EBioMedicine 2019; 41:702-710. [PMID: 30808576 PMCID: PMC6442991 DOI: 10.1016/j.ebiom.2019.02.033] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 02/05/2019] [Accepted: 02/15/2019] [Indexed: 02/08/2023] Open
Abstract
The transforming growth factor β (TGF-β) superfamily participates in tumour proliferation, apoptosis, differentiation, migration, invasion, immune evasion and extracellular matrix remodelling. Genetic deficiency in distinct components of TGF-β and BMP-induced signalling pathways or their excessive activation has been reported to regulate the development and progression of some cancers. As more in-depth studies about this superfamily have been conducted, more evidence suggests that the TGF-β and BMP pathways play an opposing role. The cross-talk of these 2 pathways has been widely studied in kidney disease and bone formation, and the opposing effects have also been observed in some cancers. However, the antagonistic mechanisms are still insufficiently investigated in cancer. In this review, we aim to display more evidences and possible mechanisms accounting for the antagonism between these 2 pathways, which might provide some clues for further study in cancer. Describe the basics of TGF-β and BMP signalling Summarize the potential mechanisms accounting for the antagonism between TGF-β and BMP pathways Provide some evidence about the antagonistic effects between pathways observed in some cancers
Collapse
Affiliation(s)
- Junya Ning
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China; Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - Yi Zhao
- Key Laboratory of Intelligent Information Processing, Advanced Computer Research Center, State Key Laboratory of Computer Architecture, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, PR China
| | - Yingnan Ye
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China
| | - Jinpu Yu
- Cancer Molecular Diagnostics Core, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China; Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, Tianjin, PR China.
| |
Collapse
|
19
|
Abstract
Bone morphogenetic proteins (BMPs) constitute the largest subdivision of the transforming growth factor-β family of ligands. BMPs exhibit widespread utility and pleiotropic, context-dependent effects, and the strength and duration of BMP pathway signaling is tightly regulated at numerous levels via mechanisms operating both inside and outside the cell. Defects in the BMP pathway or its regulation underlie multiple human diseases of different organ systems. Yet much remains to be discovered about the BMP pathway in its original context, i.e., the skeleton. In this review, we provide a comprehensive overview of the intricacies of the BMP pathway and its inhibitors in bone development, homeostasis, and disease. We frame the content of the review around major unanswered questions for which incomplete evidence is available. First, we consider the gene regulatory network downstream of BMP signaling in osteoblastogenesis. Next, we examine why some BMP ligands are more osteogenic than others and what factors limit BMP signaling during osteoblastogenesis. Then we consider whether specific BMP pathway components are required for normal skeletal development, and if the pathway exerts endogenous effects in the aging skeleton. Finally, we propose two major areas of need of future study by the field: greater resolution of the gene regulatory network downstream of BMP signaling in the skeleton, and an expanded repertoire of reagents to reliably and specifically inhibit individual BMP pathway components.
Collapse
Affiliation(s)
- Jonathan W Lowery
- Division of Biomedical Science, Marian University College of Osteopathic Medicine , Indianapolis, Indiana ; and Department of Developmental Biology, Harvard School of Dental Medicine , Boston, Massachusetts
| | - Vicki Rosen
- Division of Biomedical Science, Marian University College of Osteopathic Medicine , Indianapolis, Indiana ; and Department of Developmental Biology, Harvard School of Dental Medicine , Boston, Massachusetts
| |
Collapse
|
20
|
Ryu D, Yoon BH, Oh CH, Kim MH, Kim JY, Yoon SH, Choe S. Activin A/BMP2 Chimera (AB204) Exhibits Better Spinal Bone Fusion Properties than rhBMP2. J Korean Neurosurg Soc 2018; 61:669-679. [PMID: 30396241 PMCID: PMC6280059 DOI: 10.3340/jkns.2017.0295] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 02/20/2018] [Indexed: 11/27/2022] Open
Abstract
Objective To compare the spinal bone fusion properties of activin A/BMP2 chimera (AB204) with recombinant human bone morphogenetic protein (rhBMP2) using a rat posterolateral spinal fusion model.
Methods The study was designed to compare the effects and property at different dosages of AB204 and rhBMP2 on spinal bone fusion. Sixty-one male Sprague-Dawley rats underwent posterolateral lumbar spinal fusion using one of nine treatments during the study, that is, sham; osteon only; 3.0 μg, 6.0 μg, or 10.0 μg of rhBMP2 with osteon; and 1.0 μg, 3.0 μg, 6.0 μg, or 10.0 μg of AB204 with osteon. The effects and property on spinal bone fusion was calculated at 4 and 8 weeks after treatment using the scores of physical palpation, simple radiograph, micro-computed tomography, and immunohistochemistry.
Results Bone fusion scores were significantly higher for 10.0 μg AB204 and 10.0 μg rhBMP2 than for osteon only or 1.0 μg AB204. AB204 exhibited more prolonged osteoblastic activity than rhBMP2. Bone fusion properties of AB204 were similar with the properties of rhBMP2 at doses of 6.0 and 10.0 μg, but, the properties of AB204 at doses of 3.0 μg exhibited better than the properties of rhBMP2 at doses of 3.0 μg.
Conclusion AB204 chimeras could to be more potent for treating spinal bone fusion than rhBMP2 substitutes with increased osteoblastic activity for over a longer period.
Collapse
Affiliation(s)
- Dalsung Ryu
- Department of Neurosurgery, Inha University College of Medicine, Incheon, Korea
| | - Byung-Hak Yoon
- Protein Engineering Laboratory, joint Center for Biosciences at Songdo Global University, Incheon, Korea
| | - Chang-Hyun Oh
- Department of Neurosurgery, Inha University College of Medicine, Incheon, Korea.,Department of Neurosurgery, Cham Teun Teun Research Institute, Seoul, Korea
| | - Moon-Hang Kim
- Department of Physiology, Inha University College of Medicine, Incheon, Korea
| | - Ji-Yong Kim
- Department of Neurosurgery, Inha University College of Medicine, Incheon, Korea
| | - Seung Hwan Yoon
- Department of Neurosurgery, Inha University College of Medicine, Incheon, Korea
| | - Senyon Choe
- Protein Engineering Laboratory, joint Center for Biosciences at Songdo Global University, Incheon, Korea.,Structural Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
| |
Collapse
|
21
|
Kim MJ, Park SY, Chang HR, Jung EY, Munkhjargal A, Lim JS, Lee MS, Kim Y. Clinical significance linked to functional defects in bone morphogenetic protein type 2 receptor, BMPR2. BMB Rep 2018; 50:308-317. [PMID: 28391780 PMCID: PMC5498141 DOI: 10.5483/bmbrep.2017.50.6.059] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Indexed: 12/18/2022] Open
Abstract
Bone morphogenetic protein type 2 receptor (BMPR2) is one of the transforming growth factor-β (TGF-β) superfamily receptors, performing diverse roles during embryonic development, vasculogenesis, and osteogenesis. Human BMPR2 consists of 1,038 amino acids, and contains functionally conserved extracellular, transmembrane, kinase, and C-terminal cytoplasmic domains. Bone morphogenetic proteins (BMPs) engage the tetrameric complex, composed of BMPR2 and its corresponding type 1 receptors, which initiates SMAD proteins-mediated signal transduction leading to the expression of target genes implicated in the development or differentiation of the embryo, organs and bones. In particular, genetic alterations of BMPR2 gene are associated with several clinical disorders, including representative pulmonary arterial hypertension, cancers, and metabolic diseases, thus demonstrating the physiological importance of BMPR2. In this mini review, we summarize recent findings regarding the molecular basis of BMPR2 functions in BMP signaling, and the versatile roles of BMPR2. In addition, various aspects of experimentally validated pathogenic mutations of BMPR2 and the linked human diseases will also be discussed, which are important in clinical settings for diagnostics and treatment.
Collapse
Affiliation(s)
- Myung-Jin Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul 04310, Korea
| | - Seon Young Park
- Department of Biological Sciences, Sookmyung Women's University, Seoul 04310, Korea
| | - Hae Ryung Chang
- Department of Biological Sciences, Sookmyung Women's University, Seoul 04310, Korea
| | - Eun Young Jung
- Department of Biological Sciences, Sookmyung Women's University, Seoul 04310, Korea
| | - Anudari Munkhjargal
- Department of Biological Sciences, Sookmyung Women's University, Seoul 04310, Korea
| | - Jong-Seok Lim
- Department of Biological Sciences, Sookmyung Women's University, Seoul 04310, Korea
| | - Myeong-Sok Lee
- Department of Biological Sciences, Sookmyung Women's University, Seoul 04310, Korea
| | - Yonghwan Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul 04310, Korea
| |
Collapse
|
22
|
Lee YH, Lee BW, Jung YC, Yoon BI, Woo HM, Kang BJ. Application of alginate microbeads as a carrier of bone morphogenetic protein-2 for bone regeneration. J Biomed Mater Res B Appl Biomater 2018; 107:286-294. [PMID: 29569344 DOI: 10.1002/jbm.b.34119] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 02/21/2018] [Accepted: 03/08/2018] [Indexed: 01/13/2023]
Abstract
Bone morphogenetic protein-2 (BMP-2) is commonly used to enhance bone regeneration. The potential of BMP-2 for bone regeneration varies according to the concentration and release kinetics on the implanted site. Therefore, it is important to determine appropriate carriers of BMP-2. However, no optimal delivery vehicles have been identified. In the present study, we used alginate microbeads as a delivery vehicle for BMP-2. Alginate microbeads can be implanted onto the disease site through surgery or injection. The objective of this study was to evaluate that the osteoinductive properties of BMP-2 are effective in alginate microbeads as a carrier. In this study, the release kinetics of BMP-2 in alginate microbeads was evaluated using an enzyme-linked immunosorbent assay. BMP-2 released from alginate microbeads induced high alkaline phosphatase activity in canine adipose tissue-derived mesenchymal stem cells. Injection of alginate microbeads with BMP-2 into mouse subcutaneous tissue, as well as surgical implantation into the 5-mm circular calvarial defects in rats, was conducted and the results showed extensive new bone formation. In conclusion, alginate microbeads can be utilized as an effective BMP-2 delivery vehicle for use in orthopedic surgery and as an injectable vehicle for a minimally invasive therapy. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 107B: 286-294, 2019.
Collapse
Affiliation(s)
- Yun Hwan Lee
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Byung-Woo Lee
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Republic of Korea
| | | | - Byung-Il Yoon
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Heung-Myong Woo
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Byung-Jae Kang
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Republic of Korea
| |
Collapse
|
23
|
Ahsan S, Ge Y, Tainsky MA. Combinatorial therapeutic targeting of BMP2 and MEK-ERK pathways in NF1-associated malignant peripheral nerve sheath tumors. Oncotarget 2018; 7:57171-57185. [PMID: 27494873 PMCID: PMC5302981 DOI: 10.18632/oncotarget.11036] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 07/19/2016] [Indexed: 12/22/2022] Open
Abstract
The clinical management of malignant peripheral nerve sheath tumors (MPNSTs) is challenging not only due to its aggressive and invasive nature, but also limited therapeutic options. Using gene expression profiling, our lab identified BMP2-SMAD1/5/8 pathway as a potential therapeutic target for treating MPNSTs. In this study, we explored the therapeutic impact of targeting BMP2-SMAD1/5/8 pathway in conjunction with RAS-MEK-ERK signaling, which is constitutively activated in MPNSTs. Our results indicated that single agent treatment with LDN-193189, a BMP2 Type I receptor inhibitor, did not affect the growth and survival of MPNST cells at biochemically relevant inhibitory concentrations. However, addition of a MEK1/2 inhibitor, selumetinib, to LDN-193189-treated cells resulted in significant inhibition of cell growth and induction of cell death. LDN-193189 at biochemically effective concentrations significantly inhibited motility and invasiveness of MPNST cells, and these effects were enhanced by the addition of selumetinib. Overall, our results advocate for a combinatorial therapeutic approach for MPNSTs that not only targets the growth and survival via inhibition of MEK1/2, but also its malignant spread by suppressing the activation of BMP2-SMAD1/5/8 pathway. Importantly, these studies were conducted in low-passage patient-derived MPNST cells, allowing for an investigation of the effects of the proposed drug treatments in a biologically-relevant context.
Collapse
Affiliation(s)
- Sidra Ahsan
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI 48201, USA.,Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA.,Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yubin Ge
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA.,Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Michael A Tainsky
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI 48201, USA.,Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA.,Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA.,Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
24
|
Xu X, Zheng L, Yuan Q, Zhen G, Crane JL, Zhou X, Cao X. Transforming growth factor-β in stem cells and tissue homeostasis. Bone Res 2018; 6:2. [PMID: 29423331 PMCID: PMC5802812 DOI: 10.1038/s41413-017-0005-4] [Citation(s) in RCA: 239] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 11/12/2017] [Accepted: 11/15/2017] [Indexed: 02/05/2023] Open
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.
Collapse
Affiliation(s)
- Xin Xu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liwei Zheng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Gehua Zhen
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Janet L. Crane
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD USA
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD USA
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xu Cao
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD USA
| |
Collapse
|
25
|
Genthe JR, Min J, Farmer DM, Shelat AA, Grenet JA, Lin W, Finkelstein D, Vrijens K, Chen T, Guy RK, Clements WK, Roussel MF. Ventromorphins: A New Class of Small Molecule Activators of the Canonical BMP Signaling Pathway. ACS Chem Biol 2017; 12:2436-2447. [PMID: 28787124 DOI: 10.1021/acschembio.7b00527] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Here, we describe three new small-molecule activators of BMP signaling found by high throughput screening of a library of ∼600 000 small molecules. Using a cell-based luciferase assay in the BMP4-responsive human cervical carcinoma clonal cell line, C33A-2D2, we identified three compounds with similar chemotypes that each ventralize zebrafish embryos and stimulate increased expression of the BMP target genes, bmp2b and szl. Because these compounds ventralize zebrafish embryos, we have termed them "ventromorphins." As expected for a BMP pathway activator, they induce the differentiation of C2C12 myoblasts to osteoblasts. Affymetrix RNA analysis confirmed the differentiation results and showed that ventromorphins treatment elicits a genetic response similar to BMP4 treatment. Unlike isoliquiritigenin (SJ000286237), a flavone that maximally activates the pathway after 24 h of treatment, all three ventromorphins induced SMAD1/5/8 phosphorylation within 30 min of treatment and achieved peak activity within 1 h, indicating that their responses are consistent with directly activating BMP signaling.
Collapse
Affiliation(s)
- Jamie R. Genthe
- Departments
of Hematology, ‡Chemical Biology and Therapeutics, §Tumor Cell Biology,
and ∥Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, United States
| | - Jaeki Min
- Departments
of Hematology, ‡Chemical Biology and Therapeutics, §Tumor Cell Biology,
and ∥Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, United States
| | - Dana M. Farmer
- Departments
of Hematology, ‡Chemical Biology and Therapeutics, §Tumor Cell Biology,
and ∥Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, United States
| | - Anang A. Shelat
- Departments
of Hematology, ‡Chemical Biology and Therapeutics, §Tumor Cell Biology,
and ∥Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, United States
| | - Jose A. Grenet
- Departments
of Hematology, ‡Chemical Biology and Therapeutics, §Tumor Cell Biology,
and ∥Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, United States
| | - Wenwei Lin
- Departments
of Hematology, ‡Chemical Biology and Therapeutics, §Tumor Cell Biology,
and ∥Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, United States
| | - David Finkelstein
- Departments
of Hematology, ‡Chemical Biology and Therapeutics, §Tumor Cell Biology,
and ∥Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, United States
| | - Karen Vrijens
- Departments
of Hematology, ‡Chemical Biology and Therapeutics, §Tumor Cell Biology,
and ∥Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, United States
| | - Taosheng Chen
- Departments
of Hematology, ‡Chemical Biology and Therapeutics, §Tumor Cell Biology,
and ∥Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, United States
| | - R. Kiplin Guy
- Departments
of Hematology, ‡Chemical Biology and Therapeutics, §Tumor Cell Biology,
and ∥Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, United States
| | - Wilson K. Clements
- Departments
of Hematology, ‡Chemical Biology and Therapeutics, §Tumor Cell Biology,
and ∥Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, United States
| | - Martine F. Roussel
- Departments
of Hematology, ‡Chemical Biology and Therapeutics, §Tumor Cell Biology,
and ∥Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, United States
| |
Collapse
|
26
|
Lee JC, Volpicelli EJ. Bioinspired Collagen Scaffolds in Cranial Bone Regeneration: From Bedside to Bench. Adv Healthc Mater 2017; 6:10.1002/adhm.201700232. [PMID: 28585295 PMCID: PMC5831258 DOI: 10.1002/adhm.201700232] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/11/2017] [Indexed: 12/24/2022]
Abstract
Calvarial defects are common reconstructive dilemmas secondary to a variety of etiologies including traumatic brain injury, cerebrovascular disease, oncologic resection, and congenital anomalies. Reconstruction of the calvarium is generally undertaken for the purposes of cerebral protection, contour restoration for psychosocial well-being, and normalization of neurological dysfunction frequently found in patients with massive cranial defects. Current methods for reconstruction using autologous grafts, allogeneic grafts, or alloplastic materials have significant drawbacks that are unique to each material. The combination of wide medical relevance and the need for a better clinical solution render defects of the cranial skeleton an ideal target for development of regenerative strategies focused on calvarial bone. With the improved understanding of the instructive properties of tissue-specific extracellular matrices and the advent of precise nanoscale modulation in materials science, strategies in regenerative medicine have shifted in paradigm. Previously considered to be simple carriers of stem cells and growth factors, increasing evidence exists for differential materials directing lineage specific differentiation of progenitor cells and tissue regeneration. In this work, we review the clinical challenges for calvarial reconstruction, the anatomy and physiology of bone, and extracellular matrix-inspired, collagen-based materials that have been tested for in vivo cranial defect healing.
Collapse
Affiliation(s)
- Justine C Lee
- Greater Los Angeles Veterans Affairs Research Service, Los Angeles, California
- University of California Los Angeles Division of Plastic and Reconstructive Surgery, Los Angeles, California
| | - Elizabeth J Volpicelli
- Greater Los Angeles Veterans Affairs Research Service, Los Angeles, California
- University of California Los Angeles Division of Plastic and Reconstructive Surgery, Los Angeles, California
| |
Collapse
|
27
|
A synthetic BMP-2 mimicking peptide induces glioblastoma stem cell differentiation. Biochim Biophys Acta Gen Subj 2017; 1861:2282-2292. [PMID: 28687190 DOI: 10.1016/j.bbagen.2017.07.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 06/09/2017] [Accepted: 07/03/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND Glioblastoma (GBM) is the most aggressive type of primary brain tumor, characterized by the intrinsic resistance to chemotherapy due to the presence of a highly aggressive Cancer Stem Cell (CSC) sub-population. In this context, Bone Morphogenetic Proteins (BMPs) have been demonstrated to induce CSC differentiation and to sensitize GBM cells to treatments. METHODS The BMP-2 mimicking peptide, named GBMP1a, was synthesized on solid-phase by Fmoc chemistry. Structural characterization and prediction of receptor binding were obtained by Circular Dicroism (CD) and NRM analyses. Activation of BMP signalling was evaluated by a luciferase reporter assay and western blot. Pro-differentiating effects of GBMP1a were verified by immunostaining and neurosphere assay in primary glioblastoma cultures. RESULTS CD and NMR showed that GBMP1a correctly folds into expected tridimensional structures and predicted its binding to BMPR-IA to the same epitope as in the native complex. Reporter analysis disclosed that GBMP1a is able to activate BMP signalling in GBM cells. Moreover, BMP-signalling activation was specifically dependent on smad1/5/8 phosphorylation. Finally, we confirmed that GBMP1a treatment is sufficient to enhance osteogenic differentiation of Mesenchymal Stem Cells and to induce astroglial differentiation of glioma stem cells (GSCs) in vitro. CONCLUSIONS GBMP1a was demonstrated to be a good inducer of GSC differentiation, thus being considered a potential anti-cancer tool to be further developed for GBM treatment. GENERAL SIGNIFICANCE These data highlight the role of BMP-mimicking peptides as potential anti-cancer agents against GBM and stimulate the further development of GBMP1a-based structures in order to enhance its stability and activity.
Collapse
|
28
|
Pan H, Zhang H, Abraham P, Komatsu Y, Lyons K, Kaartinen V, Mishina Y. BmpR1A is a major type 1 BMP receptor for BMP-Smad signaling during skull development. Dev Biol 2017. [PMID: 28641928 DOI: 10.1016/j.ydbio.2017.06.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Craniosynostosis is caused by premature fusion of one or more sutures in an infant skull, resulting in abnormal facial features. The molecular and cellular mechanisms by which genetic mutations cause craniosynostosis are incompletely characterized, and many of the causative genes for diverse types of syndromic craniosynostosis have not yet been identified. We previously demonstrated that augmentation of BMP signaling mediated by a constitutively active BMP type IA receptor (ca-BmpR1A) in neural crest cells (ca1A hereafter) causes craniosynostosis and superimposition of heterozygous null mutation of Bmpr1a rescues premature suture fusion (ca1A;1aH hereafter). In this study, we superimposed heterozygous null mutations of the other two BMP type I receptors, Bmpr1b and Acvr1 (ca1A;1bH and ca1A;AcH respectively hereafter) to further dissect involvement of BMP-Smad signaling. Unlike caA1;1aH, ca1A;1bH and ca1A;AcH did not restore the craniosynostosis phenotypes. In our in vivo study, Smad-dependent BMP signaling was decreased to normal levels in mut;1aH mice. However, BMP receptor-regulated Smads (R-Smads; pSmad1/5/9 hereafter) levels were comparable between ca1A, ca1A;1bH and ca1A;AcH mice, and elevated compared to control mice. Bmpr1a, Bmpr1b and Acvr1 null cells were used to examine potential mechanisms underlying the differences in ability of heterozygosity for Bmpr1a vs. Bmpr1b or Acvr1 to rescue the mut phenotype. pSmad1/5/9 level was undetectable in Bmpr1a homozygous null cells while pSmad1/5/9 levels did not decrease in Bmpr1b or Acvr1 homozygous null cells. Taken together, our study indicates that different levels of expression and subsequent activation of Smad signaling differentially contribute each BMP type I receptor to BMP-Smad signaling and craniofacial development. These results also suggest differential involvement of each type 1 receptor in pathogenesis of syndromic craniosynostoses.
Collapse
Affiliation(s)
- Haichun Pan
- Department of Biologic & Materials Sciences, School of Dentistry, University of Michigan, 1011 N. University Ave., Ann Arbor, MI 48109, USA
| | - Honghao Zhang
- Department of Biologic & Materials Sciences, School of Dentistry, University of Michigan, 1011 N. University Ave., Ann Arbor, MI 48109, USA
| | - Ponnu Abraham
- Department of Biologic & Materials Sciences, School of Dentistry, University of Michigan, 1011 N. University Ave., Ann Arbor, MI 48109, USA
| | - Yoshihiro Komatsu
- Department of Biologic & Materials Sciences, School of Dentistry, University of Michigan, 1011 N. University Ave., Ann Arbor, MI 48109, USA; Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, USA
| | - Karen Lyons
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Vesa Kaartinen
- Department of Biologic & Materials Sciences, School of Dentistry, University of Michigan, 1011 N. University Ave., Ann Arbor, MI 48109, USA
| | - Yuji Mishina
- Department of Biologic & Materials Sciences, School of Dentistry, University of Michigan, 1011 N. University Ave., Ann Arbor, MI 48109, USA.
| |
Collapse
|
29
|
ARX polyalanine expansion mutations lead to migration impediment in the rostral cortex coupled with a developmental deficit of calbindin-positive cortical GABAergic interneurons. Neuroscience 2017. [PMID: 28627419 DOI: 10.1016/j.neuroscience.2017.06.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The Aristaless-related homeobox gene (ARX) is indispensable for interneuron development. Patients with ARX polyalanine expansion mutations of the first two tracts (namely PA1 and PA2) suffer from intellectual disability of varying severity, with seizures a frequent comorbidity. The impact of PA1 and PA2 mutations on the brain development is unknown, hindering the search for therapeutic interventions. Here, we characterized the disturbances to cortical interneuron development in mice modeling the two most common ARX polyalanine expansion mutations in human. We found a consistent ∼40-50% reduction of calbindin-positive interneurons, but not Stt+ or Cr+ interneurons, within the cortex of newborn hemizygous mice (p=0.024) for both mutant strains compared to wildtype (p=0.011). We demonstrate that this was a consequence of calbindin precursor cells being arrested or delayed at the ventral subpallium en route of tangential migration. Ex-vivo assay validated this migration deficit in PA1 cells (p=0.0002) suggesting that the defect is contributed by intrinsic loss of Arx function within migrating cells. Both humans and mice with PA1 mutations present with severe clinical features, including intellectual disability and infantile spasms. Our data further demonstrated the pathogenic mechanism was robustly shared between PA1 and PA2 mutations, as previously reported including Arx protein reduction and overlapping transcriptome profiles within the developing mouse brains. Data from our study demonstrated that cortical calbindin interneuron development and migration is negatively affected by ARX polyalanine expansion mutations. Understanding the cellular pathogenesis contributing to disease manifestation is necessary to screen efficacy of potential therapeutic interventions.
Collapse
|
30
|
Targeting bone morphogenetic protein signalling in midbrain dopaminergic neurons as a therapeutic approach in Parkinson's disease. Neuronal Signal 2017; 1:NS20170027. [PMID: 32714578 PMCID: PMC7373244 DOI: 10.1042/ns20170027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 02/20/2017] [Accepted: 02/23/2017] [Indexed: 11/17/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, characterized by the degeneration of midbrain dopaminergic (mDA) neurons and their axons, and aggregation of α-synuclein, which leads to motor and late-stage cognitive impairments. As the motor symptoms of PD are caused by the degeneration of a specific population of mDA neurons, PD lends itself to neurotrophic factor therapy. The goal of this therapy is to apply a neurotrophic factor that can slow down, halt or even reverse the progressive degeneration of mDA neurons. While the best known neurotrophic factors are members of the glial cell line-derived neurotrophic factor (GDNF) family, their lack of clinical efficacy to date means that it is important to continue to study other neurotrophic factors. Bone morphogenetic proteins (BMPs) are naturally secreted proteins that play critical roles during nervous system development and in the adult brain. In this review, we provide an overview of the BMP ligands, BMP receptors (BMPRs) and their intracellular signalling effectors, the Smad proteins. We review the available evidence that BMP-Smad signalling pathways play an endogenous role in mDA neuronal survival in vivo, before outlining how exogenous application of BMPs exerts potent effects on mDA neuron survival and axon growth in vitro and in vivo. We discuss the molecular mechanisms that mediate these effects, before highlighting the potential of targeting the downstream effectors of BMP-Smad signalling as a novel neuroprotective approach to slow or stop the degeneration of mDA neurons in PD.
Collapse
|
31
|
CILLER IM, PALANISAMY SKA, CILLER UA, MCFARLANE JR. Postnatal Expression of Bone Morphogenetic Proteins and Their Receptors in the Mouse Testis. Physiol Res 2016; 65:673-682. [DOI: 10.33549/physiolres.933193] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
TGF-β superfamily members including bone morphogenetic proteins (BMPs) and their receptors (BMPR-1A, -1B and -2) have been shown to be important for reproductive function in both males and females, while information on the role of BMPs in males is limited. Functional studies on select BMPs and BMP receptors have demonstrated vital roles for these proteins in somatic and germ cell proliferation, steroidogenesis and overall fertility. In order to gain insight into the importance of these genes during postnatal reproductive development in males, our study was undertaken to specify the distribution of BMP and BMPR mRNA in male reproductive and steroidogenic tissues and quantify these genes in the testis using the mouse as our model. We screened testis at two, four, six and eight weeks of age for the expression of ten BMPs and three BMP receptors using RT-qPCR. All three BMP receptor mRNAs – Bmpr1a, Bmpr1b and Bmpr2, and ten BMP mRNAs – Bmp2, Bmp3, Bmp3b, Bmp4, Bmp5, Bmp6, Bmp7, Bmp8a, Bmp8b and Bmp15 were expressed in mouse testis at all stages screened. Testicular expression of genes varied within age groups and at specific developmental stages. Our study establishes an extensive BMP system in mouse reproductive and steroidogenic tissues.
Collapse
Affiliation(s)
- I. M. CILLER
- Centre for Bioactive Discovery in Health and Ageing, School of Science and Technology, University of New England, Armidale, Australia
| | | | | | | |
Collapse
|
32
|
An investigation of BMP-7 mediated alterations to BMP signalling components in human tenocyte-like cells. Sci Rep 2016; 6:29703. [PMID: 27406972 PMCID: PMC4942578 DOI: 10.1038/srep29703] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 06/21/2016] [Indexed: 12/12/2022] Open
Abstract
The incidence of tendon re-tears post-surgery is an ever present complication. It is suggested that the application of biological factors, such as bone morphogenetic protein 7 (BMP-7), can reduce complication rates by promoting tenogenic characteristics in in vitro studies. However, there remains a dearth of information in regards to the mechanisms of BMP-7 signalling in tenocytes. Using primary human tenocyte-like cells (hTLCs) from the supraspinatus tendon the BMP-7 signalling pathway was investigated: induction of the BMP associated Smad pathway and non-Smad pathways (AKT, p38, ERK1/2 and JNK); alterations in gene expression of BMP-7 associated receptors, Smad pathway components, Smad target gene (ID1) and tenogenic marker scleraxis. BMP-7 increases the expression of specific BMP associated receptors, BMPR-Ib and BMPR-II, and Smad8. Additionally, BMP-7 activates significantly Smad1/5/8 and slightly p38 pathways as indicated by an increase in phosphorylation and proven by inhibition experiments, where p-ERK1/2 and p-JNK pathways remain mainly unresponsive. Furthermore, BMP-7 increases the expression of the Smad target gene ID1, and the tendon specific transcription factor scleraxis. The study shows that tenocyte-like cells undergo primarily Smad8 and p38 signalling after BMP-7 stimulation. The up-regulation of tendon related marker genes and matrix proteins such as Smad8/9, scleraxis and collagen I might lead to positive effects of BMP-7 treatment for rotator cuff repair, without significant induction of osteogenic and chondrogenic markers.
Collapse
|
33
|
Manson SR, Austin PF, Guo Q, Moore KH. BMP-7 Signaling and its Critical Roles in Kidney Development, the Responses to Renal Injury, and Chronic Kidney Disease. VITAMINS AND HORMONES 2016; 99:91-144. [PMID: 26279374 DOI: 10.1016/bs.vh.2015.05.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chronic kidney disease (CKD) is a significant health problem that most commonly results from congenital abnormalities in children and chronic renal injury in adults. The therapeutic potential of BMP-7 was first recognized nearly two decades ago with studies demonstrating its requirement for kidney development and ability to inhibit the pathogenesis of renal injury in models of CKD. Since this time, our understanding of CKD has advanced considerably and treatment strategies have evolved with the identification of many additional signaling pathways, cell types, and pathologic processes that contribute to disease progression. The purpose of this review is to revisit the seminal studies that initially established the importance of BMP-7, highlight recent advances in BMP-7 research, and then integrate this knowledge with current research paradigms. We will provide an overview of the evolutionarily conserved roles of BMP proteins and the features that allow BMP signaling pathways to function as critical signaling nodes for controlling biological processes, including those related to CKD. We will discuss the multifaceted functions of BMP-7 during kidney development and the potential for alterations in BMP-7 signaling to result in congenital abnormalities and pediatric kidney disease. We will summarize the renal protective effects of recombinant BMP-7 in experimental models of CKD and then propose a model to describe the potential physiological role of endogenous BMP-7 in the innate repair mechanisms of the kidneys that respond to renal injury. Finally, we will highlight emerging clinical approaches for applying our knowledge of BMP-7 toward improving the treatment of patients with CKD.
Collapse
Affiliation(s)
- Scott R Manson
- Department of Surgery, Division of Urology, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, Missouri, USA.
| | - Paul F Austin
- Department of Surgery, Division of Urology, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, Missouri, USA
| | - Qiusha Guo
- Department of Surgery, Division of Urology, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, Missouri, USA
| | - Katelynn H Moore
- Department of Surgery, Division of Urology, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, Missouri, USA
| |
Collapse
|
34
|
The biological function of type I receptors of bone morphogenetic protein in bone. Bone Res 2016; 4:16005. [PMID: 27088043 PMCID: PMC4820739 DOI: 10.1038/boneres.2016.5] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 02/04/2016] [Accepted: 02/20/2016] [Indexed: 12/13/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) have multiple roles in skeletal development, homeostasis and regeneration. BMPs signal via type I and type II serine/threonine kinase receptors (BMPRI and BMPRII). In recent decades, genetic studies in humans and mice have demonstrated that perturbations in BMP signaling via BMPRI resulted in various diseases in bone, cartilage, and muscles. In this review, we focus on all three types of BMPRI, which consist of activin-like kinase 2 (ALK2, also called type IA activin receptor), activin-like kinase 3 (ALK3, also called BMPRIA), and activin-like kinase 6 (ALK6, also called BMPRIB). The research areas covered include the current progress regarding the roles of these receptors during myogenesis, chondrogenesis, and osteogenesis. Understanding the physiological and pathological functions of these receptors at the cellular and molecular levels will advance drug development and tissue regeneration for treating musculoskeletal diseases and bone defects in the future.
Collapse
|
35
|
Schille C, Heller J, Schambony A. Differential requirement of bone morphogenetic protein receptors Ia (ALK3) and Ib (ALK6) in early embryonic patterning and neural crest development. BMC DEVELOPMENTAL BIOLOGY 2016; 16:1. [PMID: 26780949 PMCID: PMC4717534 DOI: 10.1186/s12861-016-0101-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 01/05/2016] [Indexed: 01/01/2023]
Abstract
Background Bone morphogenetic proteins regulate multiple processes in embryonic development, including early dorso-ventral patterning and neural crest development. BMPs activate heteromeric receptor complexes consisting of type I and type II receptor-serine/threonine kinases. BMP receptors Ia and Ib, also known as ALK3 and ALK6 respectively, are the most common type I receptors that likely mediate most BMP signaling events. Since early expression patterns and functions in Xenopus laevis development have not been described, we have addressed these questions in the present study. Results Here we have analyzed the temporal and spatial expression patterns of ALK3 and ALK6; we have also carried out loss-of-function studies to define the function of these receptors in early Xenopus development. We detected both redundant and non-redundant roles of ALK3 and ALK6 in dorso-ventral patterning. From late gastrula stages onwards, their expression patterns diverged, which correlated with a specific, non-redundant requirement of ALK6 in post-gastrula neural crest cells. ALK6 was essential for induction of neural crest cell fate and further development of the neural crest and its derivatives. Conclusions ALK3 and ALK6 both contribute to the gene regulatory network that regulates dorso-ventral patterning; they play partially overlapping and partially non-redundant roles in this process. ALK3 and ALK6 are independently required for the spatially restricted activation of BMP signaling and msx2 upregulation at the neural plate border, whereas in post-gastrula development ALK6 exerts a highly specific, conserved function in neural crest development. Electronic supplementary material The online version of this article (doi:10.1186/s12861-016-0101-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Carolin Schille
- Biology Department, Developmental Biology, Friedrich-Alexander University Erlangen-Nuremberg, 91058, Erlangen, Germany.
| | - Jens Heller
- Biology Department, Developmental Biology, Friedrich-Alexander University Erlangen-Nuremberg, 91058, Erlangen, Germany.
| | - Alexandra Schambony
- Biology Department, Developmental Biology, Friedrich-Alexander University Erlangen-Nuremberg, 91058, Erlangen, Germany.
| |
Collapse
|
36
|
Abstract
Muscle and bone are two intimately connected tissues. A coordinated interplay between these tissues at mechanical levels is required for their development, function and ageing. Evidence is emerging that several genes and molecular pathways exert a pleiotropic effect on both muscle and bone. Bone morphogenetic proteins (BMPs) are secreted signal factors belonging to the transforming growth factor β (TGFβ) superfamily. BMPs have an essential role during bone and cartilage formation and maintenance. Recently, we and others have demonstrated that the BMP pathway also has a role in controlling adult skeletal muscle mass. Thus, BMPs become crucial regulators of both bone and muscle formation and homeostasis. In this review we will discuss the signalling downstream BMP and its role in muscle-bone interaction. This article is part of a Special Issue entitled "Muscle Bone Interactions".
Collapse
Affiliation(s)
- Roberta Sartori
- Dulbecco Telethon Institute, Venetian Institute of Molecular Medicine, 35129 Padova, Italy; Department of Biomedical Sciences, University of Padova, 35121 Padova, Italy.
| | - Marco Sandri
- Dulbecco Telethon Institute, Venetian Institute of Molecular Medicine, 35129 Padova, Italy; Department of Biomedical Sciences, University of Padova, 35121 Padova, Italy; Telethon Institute of Genetics and Medicine (TIGEM), 80131 Napoli, Italy.
| |
Collapse
|
37
|
Matsubara T, Araki M, Abe H, Ueda O, Jishage KI, Mima A, Goto C, Tominaga T, Kinosaki M, Kishi S, Nagai K, Iehara N, Fukushima N, Kita T, Arai H, Doi T. Bone Morphogenetic Protein 4 and Smad1 Mediate Extracellular Matrix Production in the Development of Diabetic Nephropathy. Diabetes 2015; 64:2978-90. [PMID: 25995358 DOI: 10.2337/db14-0893] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Accepted: 04/12/2015] [Indexed: 01/20/2023]
Abstract
Diabetic nephropathy is the leading cause of end-stage renal disease. It is pathologically characterized by the accumulation of extracellular matrix in the mesangium, of which the main component is α1/α2 type IV collagen (Col4a1/a2). Recently, we identified Smad1 as a direct regulator of Col4a1/a2 under diabetic conditions in vitro. Here, we demonstrate that Smad1 plays a key role in diabetic nephropathy through bone morphogenetic protein 4 (BMP4) in vivo. Smad1-overexpressing mice (Smad1-Tg) were established, and diabetes was induced by streptozotocin. Nondiabetic Smad1-Tg did not exhibit histological changes in the kidney; however, the induction of diabetes resulted in an ∼1.5-fold greater mesangial expansion, consistent with an increase in glomerular phosphorylated Smad1. To address regulatory factors of Smad1, we determined that BMP4 and its receptor are increased in diabetic glomeruli and that diabetic Smad1-Tg and wild-type mice treated with a BMP4-neutralizing antibody exhibit decreased Smad1 phosphorylation and ∼40% less mesangial expansion than those treated with control IgG. Furthermore, heterozygous Smad1 knockout mice exhibit attenuated mesangial expansion in the diabetic condition. The data indicate that BMP4/Smad1 signaling is a critical cascade for the progression of mesangial expansion and that blocking this signal could be a novel therapeutic strategy for diabetic nephropathy.
Collapse
Affiliation(s)
| | - Makoto Araki
- Department of Nephrology, Kyoto University, Kyoto, Japan
| | - Hideharu Abe
- Department of Nephrology, Tokushima University, Tokushima, Japan
| | - Otoya Ueda
- Chugai Pharmaceutical Co., Ltd., Shizuoka, Japan
| | | | - Akira Mima
- Department of Nephrology, Tokushima University, Tokushima, Japan
| | - Chisato Goto
- Chugai Research Institute for Medical Science, Inc., Shizuoka, Japan
| | - Tatsuya Tominaga
- Department of Nephrology, Tokushima University, Tokushima, Japan
| | | | - Seiji Kishi
- Department of Nephrology, Tokushima University, Tokushima, Japan
| | - Kojiro Nagai
- Department of Nephrology, Tokushima University, Tokushima, Japan
| | | | | | - Toru Kita
- Kobe City Medical Center General Hospital, Kyoto, Japan
| | - Hidenori Arai
- National Center for Geriatrics and Gerontology, Aichi, Japan
| | - Toshio Doi
- Department of Nephrology, Tokushima University, Tokushima, Japan
| |
Collapse
|
38
|
Abstract
Primordial follicles (PF) are formed when somatic cells differentiate into flattened pregranulosa cells, invaginate into the oocyte nests and encircle individual oocytes. We hypothesize that BMP2 regulates PF formation by promoting the transition of germ cells into oocytes and somatic cells into pregranulosa cells. E15 hamster ovaries were cultured for 8 days corresponding to postnatal day 8 (P8) in vivo, with or without BMP2, and the formation of PF was examined. BMP2 was expressed in the oocytes as well as ovarian somatic cells during development. BMP2 exposure for the first two days or the last two days or the entire 8 days of culture led to increase in PF formation suggesting that BMP2 affected both germ cell transition and somatic cell differentiation. Whereas an ALK2/3 inhibitor completely blocked BMP2-induced PF formation, an ALK2-specific inhibitor was partially effective, suggesting that BMP2 affected PF formation via both ALK2 and ALK3. BMP2 also reduced apoptosis in vitro. Further, more meiotic oocytes were present in BMP2 exposed ovaries. In summary, the results provide the first evidence that BMP2 regulates primordial follicle formation by promoting germ cell to oocyte transition and somatic cell to pre-granulosa cells formation and it acts via both ALK2 and ALK3.
Collapse
Affiliation(s)
| | - Shyamal K Roy
- 1] Department of Cellular and Integrative Physiology, and Obstetrics and Gynecology [2] University of Nebraska Medical Center, Omaha, NE
| |
Collapse
|
39
|
Skripitz R, Kurth A, Roth A. [Supportive drugs for improved implant healing]. DER ORTHOPADE 2015; 44:703-709. [PMID: 26160658 DOI: 10.1007/s00132-015-3136-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Improvement of implant healing in orthopedic and trauma surgery serves to improve the life expectancy of the implant. Good primary stability by clamping is a prerequisite for secondary stability and for the actual integration and healing of the implant. RESULTS Possible causes of implant loosening are abrasive particles, which arrive at non-integrated implants at the unsealed prosthesis-bone interface and provoke a macrophage-mediated foreign body reaction, resulting in periprosthetic osteolysis. Numerous animal studies have already described the use of bisphosphonates to inhibit osteolysis induced by abrasion and secondary instability. In patients with total knee arthroplasty, a decrease in prosthetic migration under the influence of bisphosphonates could be shown. The stimulation of bone formation around the implants and the resulting implant healing was demonstrated both in animal experiments for bone morphogenetic proteins (BMP) and in case reports for intermittent parathyroid hormone administration. CONCLUSION By using supportive drugs, it is possible to achieve an improvement in the osseointegration of implants; thus, more rapid secondary stability and load-bearing are expected.
Collapse
Affiliation(s)
- R Skripitz
- Orthopädische Klinik und Poliklinik, Universitätsmedizin Rostock, Doberaner Str. 142, 18057, Rostock, Deutschland.
| | - A Kurth
- Themistocles Gluck Hospital, Ratingen, Deutschland
| | - A Roth
- Bereich Endoprothetik/Orthopädie, Klinik für Orthopädie, Unfallchirurgie und Plastische Chirurgie, Universitätsklinik Leipzig AöR, Leipzig, Deutschland
| |
Collapse
|
40
|
Wei Q, Pohl TLM, Seckinger A, Spatz JP, Cavalcanti-Adam EA. Regulation of integrin and growth factor signaling in biomaterials for osteodifferentiation. Beilstein J Org Chem 2015; 11:773-83. [PMID: 26124879 PMCID: PMC4464188 DOI: 10.3762/bjoc.11.87] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 05/07/2015] [Indexed: 12/21/2022] Open
Abstract
Stem cells respond to the microenvironment (niche) they are located in. Under natural conditions, the extracellular matrix (ECM) is the essential component the in stem cell niche, in which both integrin ligands and growth factors are important regulators to directly or indirectly modulate the cell behavior. In this review, we summarize the current knowledge about the potential of integrin ligands and growth factors to induce osteogenic differentiation of stem cells, and discuss the signaling pathways that are initiated by both individual and cooperative parameters. The joint effect of integrin ligands and growth factors is highlighted as the multivalent interactions for bone therapy.
Collapse
Affiliation(s)
- Qiang Wei
- Department of Biophysical Chemistry, Institute for Physical Chemistry, University of Heidelberg, INF 253, 69120 Heidelberg, Germany ; Department of New Materials and Biosystems, Max-Planck Institute for Intelligent Systems, Stuttgart, Germany
| | - Theresa L M Pohl
- Department of Biophysical Chemistry, Institute for Physical Chemistry, University of Heidelberg, INF 253, 69120 Heidelberg, Germany ; Department of New Materials and Biosystems, Max-Planck Institute for Intelligent Systems, Stuttgart, Germany
| | - Anja Seckinger
- Department of Internal Medicine V, Oncology, Hematology, and Rheumatology, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Joachim P Spatz
- Department of Biophysical Chemistry, Institute for Physical Chemistry, University of Heidelberg, INF 253, 69120 Heidelberg, Germany ; Department of New Materials and Biosystems, Max-Planck Institute for Intelligent Systems, Stuttgart, Germany
| | - Elisabetta A Cavalcanti-Adam
- Department of Biophysical Chemistry, Institute for Physical Chemistry, University of Heidelberg, INF 253, 69120 Heidelberg, Germany ; Department of New Materials and Biosystems, Max-Planck Institute for Intelligent Systems, Stuttgart, Germany
| |
Collapse
|
41
|
Schwab EH, Pohl TLM, Haraszti T, Schwaerzer GK, Hiepen C, Spatz JP, Knaus P, Cavalcanti-Adam EA. Nanoscale control of surface immobilized BMP-2: toward a quantitative assessment of BMP-mediated signaling events. NANO LETTERS 2015; 15:1526-1534. [PMID: 25668064 DOI: 10.1021/acs.nanolett.5b00315] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
In this work we determine the impact of surface density of immobilized BMP-2 on intracellular signal transduction. We use block copolymer micellar nanolithography to fabricate substrates with precisely spaced and tunable gold nanoparticle arrays carrying single BMP-2 molecules. We found that the immobilized growth factor triggers prolonged and elevated Smad signaling pathway activation compared to the same amount of soluble protein. This approach is suitable for achieving controlled and sustained local delivery of BMP-2 and other growth factors.
Collapse
Affiliation(s)
- Elisabeth H Schwab
- Department of Biophysical Chemistry, Institute of Physical Chemistry, University of Heidelberg , INF 253, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Ma WH, Liu YJ, Wang W, Zhang YZ. Neuropeptide Y, substance P, and human bone morphogenetic protein 2 stimulate human osteoblast osteogenic activity by enhancing gap junction intercellular communication. ACTA ACUST UNITED AC 2015; 48:299-307. [PMID: 25714881 PMCID: PMC4418359 DOI: 10.1590/1414-431x20144226] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 11/13/2014] [Indexed: 01/05/2023]
Abstract
Bone homeostasis seems to be controlled by delicate and subtle “cross talk” between
the nervous system and “osteo-neuromediators” that control bone remodeling. The
purpose of this study was to evaluate the effect of interactions between
neuropeptides and human bone morphogenetic protein 2 (hBMP2) on human osteoblasts. We
also investigated the effects of neuropeptides and hBMP2 on gap junction
intercellular communication (GJIC). Osteoblasts were treated with neuropeptide Y
(NPY), substance P (SP), or hBMP2 at three concentrations. At various intervals after
treatment, cell viability was measured by the MTT assay. In addition, cellular
alkaline phosphatase (ALP) activity and osteocalcin were determined by colorimetric
assay and radioimmunoassay, respectively. The effects of NPY, SP and hBMP on GJIC
were determined by laser scanning confocal microscopy. The viability of cells treated
with neuropeptides and hBMP2 increased significantly in a time-dependent manner, but
was inversely associated with the concentration of the treatments. ALP activity and
osteocalcin were both reduced in osteoblasts exposed to the combination of
neuropeptides and hBMP2. The GJIC of osteoblasts was significantly increased by the
neuropeptides and hBMP2. These results suggest that osteoblast activity is increased
by neuropeptides and hBMP2 through increased GJIC. Identification of the
GJIC-mediated signal transduction capable of modulating the cellular activities of
bone cells represents a novel approach to studying the biology of skeletal
innervation.
Collapse
Affiliation(s)
- W H Ma
- The Third Hospital of Hebei Medical University, The Provincial Key Laboratory for Orthopedic Biomechanics of Hebei, Shijiazhuang, Hebei Province, China
| | - Y J Liu
- The Third Hospital of Hebei Medical University, The Provincial Key Laboratory for Orthopedic Biomechanics of Hebei, Shijiazhuang, Hebei Province, China
| | - W Wang
- The Third Hospital of Hebei Medical University, The Provincial Key Laboratory for Orthopedic Biomechanics of Hebei, Shijiazhuang, Hebei Province, China
| | - Y Z Zhang
- The Third Hospital of Hebei Medical University, The Provincial Key Laboratory for Orthopedic Biomechanics of Hebei, Shijiazhuang, Hebei Province, China
| |
Collapse
|
43
|
Maxhimer JB, Bradley JP, Lee JC. Signaling pathways in osteogenesis and osteoclastogenesis: Lessons from cranial sutures and applications to regenerative medicine. Genes Dis 2015; 2:57-68. [PMID: 25961069 PMCID: PMC4425620 DOI: 10.1016/j.gendis.2014.12.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
One of the simplest models for examining the interplay between bone formation and resorption is the junction between the cranial bones. Although only roughly a quarter of patients diagnosed with craniosynostosis have been linked to known genetic disturbances, the molecular mechanisms elucidated from these studies have provided basic knowledge of bone homeostasis. This work has translated to methods and advances in bone tissue engineering. In this review, we examine the current knowledge of cranial suture biology derived from human craniosynostosis syndromes and discuss its application to regenerative medicine.
Collapse
Affiliation(s)
- Justin B. Maxhimer
- Division of Plastic and Reconstructive Surgery, UCLA David Geffen School of Medicine, CA, USA
| | - James P. Bradley
- Division of Plastic and Reconstructive Surgery, Temple University/St. Christopher's Hospital for Children, PA, USA
| | - Justine C. Lee
- Division of Plastic and Reconstructive Surgery, UCLA David Geffen School of Medicine, CA, USA
- Division of Plastic and Reconstructive Surgery, Greater Los Angeles VA Healthcare System, USA
- Corresponding author. UCLA Division of Plastic and Reconstructive Surgery, 200 UCLA Medical Plaza, Suite 465, Los Angeles, CA 90095, USA. Tel.: +1 310 794 7616; fax: +1 310 206 6833.
| |
Collapse
|
44
|
Yoon BH, Esquivies L, Ahn C, Gray PC, Ye SK, Kwiatkowski W, Choe S. An activin A/BMP2 chimera, AB204, displays bone-healing properties superior to those of BMP2. J Bone Miner Res 2014; 29:1950-9. [PMID: 24692083 PMCID: PMC4276739 DOI: 10.1002/jbmr.2238] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 03/02/2014] [Accepted: 03/13/2014] [Indexed: 12/27/2022]
Abstract
Recombinant bone morphogenetic protein 2 (rhBMP2) has been used clinically to treat bone fractures in human patients. However, the high doses of rhBMP2 required for a therapeutic response can cause undesirable side effects. Here, we demonstrate that a novel Activin A/BMP2 (AB2) chimera, AB204, promotes osteogenesis and bone healing much more potently and effectively than rhBMP2. Remarkably, 1 month of AB204 treatment completely heals tibial and calvarial defects of critical size in mice at a concentration 10-fold lower than a dose of rhBMP2 that only partially heals the defect. We determine the structure of AB204 to 2.3 Å that reveals a distinct BMP2-like fold in which the Activin A sequence segments confer insensitivity to the BMP2 antagonist Noggin and an affinity for the Activin/BMP type II receptor ActRII that is 100-fold greater than that of BMP2. The structure also led to our identification of a single Activin A-derived amino acid residue, which, when mutated to the corresponding BMP2 residue, resulted in a significant increase in the affinity of AB204 for its type I receptor BMPRIa and a further enhancement in AB204's osteogenic potency. Together, these findings demonstrate that rationally designed AB2 chimeras can provide BMP2 substitutes with enhanced potency for treating non-union bone fractures.
Collapse
Affiliation(s)
- Byung-Hak Yoon
- Protein Engineering Laboratory, Joint Center for Biosciences at Songdo Global University Campus, Incheon, Republic of Korea; Department of Pharmacology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
45
|
Bhatt RS, Atkins MB. Molecular pathways: can activin-like kinase pathway inhibition enhance the limited efficacy of VEGF inhibitors? Clin Cancer Res 2014; 20:2838-45. [PMID: 24714770 DOI: 10.1158/1078-0432.ccr-13-2788] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The vascular endothelial growth factor (VEGF) pathway is critical for tumor angiogenesis. However, VEGF pathway inhibition has been limited by intrinsic and acquired resistance. Simultaneously targeting multiple steps involved in tumor angiogenesis is a potential means of overcoming this resistance. Activin like kinase 1 (ALK1) and endoglin (ENG) have effects on angiogenesis that are distinct from those of VEGF. Whereas VEGF is important for vessel initiation, ALK1 and endoglin are involved in vessel network formation. Thus, ALK1 and endoglin pathway inhibitors are attractive partners for VEGF-based combination antiangiogenic therapy. Genetic evidence supports a role for this receptor family and its ligands, bone morphogenetic proteins (BMP) 9 and 10, in vascular development. Patients with genetic alterations in ALK1 or endoglin develop hereditary hemorrhagic telangiectasia, a disorder characterized by abnormal vessel development. There are several inhibitors of the ALK1 pathway advancing in clinical development for treatment of various tumor types, including renal cell and ovarian carcinomas. Targeting of alternate angiogenic pathways, particularly in combination with VEGF pathway blockade, holds the promise of optimally inhibiting angiogenically driven tumor progression. Clin Cancer Res; 20(11); 2838-45. ©2014 AACR.
Collapse
Affiliation(s)
- Rupal S Bhatt
- Authors' Affiliations: Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts and Departments of Oncology and Medicine, Georgetown-Lombardi Comprehensive Cancer Center, Washington, District of Columbia
| | - Michael B Atkins
- Authors' Affiliations: Division of Hematology-Oncology and Cancer Biology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts and Departments of Oncology and Medicine, Georgetown-Lombardi Comprehensive Cancer Center, Washington, District of Columbia
| |
Collapse
|
46
|
Hegarty SV, Collins LM, Gavin AM, Roche SL, Wyatt SL, Sullivan AM, O'Keeffe GW. Canonical BMP-Smad signalling promotes neurite growth in rat midbrain dopaminergic neurons. Neuromolecular Med 2014; 16:473-89. [PMID: 24682653 DOI: 10.1007/s12017-014-8299-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 03/07/2014] [Indexed: 01/01/2023]
Abstract
Ventral midbrain (VM) dopaminergic (DA) neurons project to the dorsal striatum via the nigrostriatal pathway to regulate voluntary movements, and their loss leads to the motor dysfunction seen in Parkinson's disease (PD). Despite recent progress in the understanding of VM DA neurogenesis, the factors regulating nigrostriatal pathway development remain largely unknown. The bone morphogenetic protein (BMP) family regulates neurite growth in the developing nervous system and may contribute to nigrostriatal pathway development. Two related members of this family, BMP2 and growth differentiation factor (GDF)5, have neurotrophic effects, including promotion of neurite growth, on cultured VM DA neurons. However, the molecular mechanisms regulating their effects on DA neurons are unknown. By characterising the temporal expression profiles of endogenous BMP receptors (BMPRs) in the developing and adult rat VM and striatum, this study identified BMP2 and GDF5 as potential regulators of nigrostriatal pathway development. Furthermore, through the use of noggin, dorsomorphin and BMPR/Smad plasmids, this study demonstrated that GDF5- and BMP2-induced neurite outgrowth from cultured VM DA neurons is dependent on BMP type I receptor activation of the Smad 1/5/8 signalling pathway.
Collapse
Affiliation(s)
- Shane V Hegarty
- Department of Anatomy and Neuroscience, Biosciences Institute, University College Cork, Cork, Ireland
| | | | | | | | | | | | | |
Collapse
|
47
|
Constam DB. Regulation of TGFβ and related signals by precursor processing. Semin Cell Dev Biol 2014; 32:85-97. [PMID: 24508081 DOI: 10.1016/j.semcdb.2014.01.008] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 01/29/2014] [Indexed: 10/25/2022]
Abstract
Secreted cytokines of the TGFβ family are found in all multicellular organisms and implicated in regulating fundamental cell behaviors such as proliferation, differentiation, migration and survival. Signal transduction involves complexes of specific type I and II receptor kinases that induce the nuclear translocation of Smad transcription factors to regulate target genes. Ligands of the BMP and Nodal subgroups act at a distance to specify distinct cell fates in a concentration-dependent manner. These signaling gradients are shaped by multiple factors, including proteases of the proprotein convertase (PC) family that hydrolyze one or several peptide bonds between an N-terminal prodomain and the C-terminal domain that forms the mature ligand. This review summarizes information on the proteolytic processing of TGFβ and related precursors, and its spatiotemporal regulation by PCs during development and various diseases, including cancer. Available evidence suggests that the unmasking of receptor binding epitopes of TGFβ is only one (and in some cases a non-essential) function of precursor processing. Future studies should consider the impact of proteolytic maturation on protein localization, trafficking and turnover in cells and in the extracellular space.
Collapse
Affiliation(s)
- Daniel B Constam
- Ecole Polytechnique Fédérale de Lausanne (EPFL), Bâtiment SV ISREC, Station 19, CH-1015 Lausanne, Switzerland.
| |
Collapse
|
48
|
Okuma-Yoshioka C, Seto H, Kadono Y, Hikita A, Oshima Y, Kurosawa H, Nakamura K, Tanaka S. Tumor necrosis factor-α inhibits chondrogenic differentiation of synovial fibroblasts through p38 mitogen activating protein kinase pathways. Mod Rheumatol 2014; 18:366-378. [DOI: 10.3109/s10165-008-0069-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Chiaki Okuma-Yoshioka
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Department of Orthopaedics, School of Medicine, Juntendo University, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hiroaki Seto
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Department of Orthopaedics, School of Medicine, Juntendo University, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yuho Kadono
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Atsuhiko Hikita
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yasushi Oshima
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hisashi Kurosawa
- Department of Orthopaedics, School of Medicine, Juntendo University, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kozo Nakamura
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Sakae Tanaka
- Department of Orthopaedic Surgery, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
49
|
Holtzhausen A, Golzio C, How T, Lee YH, Schiemann WP, Katsanis N, Blobe GC. Novel bone morphogenetic protein signaling through Smad2 and Smad3 to regulate cancer progression and development. FASEB J 2013; 28:1248-67. [PMID: 24308972 DOI: 10.1096/fj.13-239178] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The bone morphogenetic protein (BMP) signaling pathways have important roles in embryonic development and cellular homeostasis, with aberrant BMP signaling resulting in a broad spectrum of human disease. We report that BMPs unexpectedly signal through the canonical transforming growth factor β (TGF-β)-responsive Smad2 and Smad3. BMP-induced Smad2/3 signaling occurs preferentially in embryonic cells and transformed cells. BMPs signal to Smad2/3 by stimulating complex formation between the BMP-binding TGF-β superfamily receptors, activin receptor-like kinase (ALK)3/6, and the Smad2/3 phosphorylating receptors ALK5/7. BMP signaling through Smad2 mediates, in part, dorsoventral axis patterning in zebrafish embryos, whereas BMP signaling through Smad3 facilitates cancer cell invasion. Consistent with increased BMP-mediated Smad2/3 signaling during cancer progression, Smad1/5 and Smad 2/3 signaling converge in human cancer specimens. Thus, the signaling mechanisms used by BMPs and TGF-β superfamily receptors are broader than previously appreciated.
Collapse
Affiliation(s)
- Alisha Holtzhausen
- 1Duke University Medical Center, 450 Research Drive, LSRC B354, Box 91004, Durham, NC 27708, USA.
| | | | | | | | | | | | | |
Collapse
|
50
|
Vrijens K, Lin W, Cui J, Farmer D, Low J, Pronier E, Zeng FY, Shelat AA, Guy K, Taylor MR, Chen T, Roussel MF. Identification of small molecule activators of BMP signaling. PLoS One 2013; 8:e59045. [PMID: 23527084 PMCID: PMC3602516 DOI: 10.1371/journal.pone.0059045] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2012] [Accepted: 02/11/2013] [Indexed: 12/13/2022] Open
Abstract
Bone Morphogenetic Proteins (BMPs) are morphogens that play a major role in regulating development and homeostasis. Although BMPs are used for the treatment of bone and kidney disorders, their clinical use is limited due to the supra-physiological doses required for therapeutic efficacy causing severe side effects. Because recombinant BMPs are expensive to produce, small molecule activators of BMP signaling would be a cost-effective alternative with the added benefit of being potentially more easily deliverable. Here, we report our efforts to identify small molecule activators of BMP signaling. We have developed a cell-based assay to monitor BMP signaling by stably transfecting a BMP-responsive human cervical carcinoma cell line (C33A) with a reporter construct in which the expression of luciferase is driven by a multimerized BMP-responsive element from the Id1 promoter. A BMP-responsive clone C33A-2D2 was used to screen a bioactive library containing ∼5,600 small molecules. We identified four small molecules of the family of flavonoids all of which induced luciferase activity in a dose-dependent manner and ventralized zebrafish embryos. Two of the identified compounds induced Smad1, 5 phosphorylation (P-Smad), Id1 and Id2 expression in a dose-dependent manner demonstrating that our assays identified small molecule activators of BMP signaling.
Collapse
Affiliation(s)
- Karen Vrijens
- Departments of Tumor Cell Biology, Memphis, Tennessee, United States of America
| | - Wenwei Lin
- Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Jimmy Cui
- Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Dana Farmer
- Departments of Tumor Cell Biology, Memphis, Tennessee, United States of America
| | - Jonathan Low
- Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Elodie Pronier
- Departments of Tumor Cell Biology, Memphis, Tennessee, United States of America
- Institut National de la Santé et de la Recherche Medicale, U1009, Institut Gustave Roussy, Villejuif, France
| | - Fu-Yue Zeng
- Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Anang A. Shelat
- Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Kiplin Guy
- Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Michael R. Taylor
- Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Taosheng Chen
- Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Martine F. Roussel
- Departments of Tumor Cell Biology, Memphis, Tennessee, United States of America
- * E-mail:
| |
Collapse
|