1
|
Xinyi Y, Vladimirovich RI, Beeraka NM, Satyavathi A, Kamble D, Nikolenko VN, Lakshmi AN, Basappa B, Reddy Y P, Fan R, Liu J. Emerging insights into epigenetics and hematopoietic stem cell trafficking in age-related hematological malignancies. Stem Cell Res Ther 2024; 15:401. [PMID: 39506818 PMCID: PMC11539620 DOI: 10.1186/s13287-024-04008-4] [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: 07/31/2024] [Accepted: 10/22/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Hematopoiesis within the bone marrow (BM) is a complex and tightly regulated process predominantly influenced by immune factors. Aging, diabetes, and obesity are significant contributors to BM niche damage, which can alter hematopoiesis and lead to the development of clonal hematopoiesis of intermediate potential (CHIP). Genetic/epigenetic alterations during aging could influence BM niche reorganization for hematopoiesis or clonal hematopoiesis. CHIP is driven by mutations in genes such as Tet2, Dnmt3a, Asxl1, and Jak2, which are associated with age-related hematological malignancies. OBJECTIVE This literature review aims to provide an updated exploration of the functional aspects of BM niche cells within the hematopoietic microenvironment in the context of age-related hematological malignancies. The review specifically focuses on how immunological stressors modulate different signaling pathways that impact hematopoiesis. METHODS An extensive review of recent studies was conducted, examining the roles of various BM niche cells in hematopoietic stem cell (HSC) trafficking and the development of age-related hematological malignancies. Emphasis was placed on understanding the influence of immunological stressors on these processes. RESULTS Recent findings reveal a significant microheterogeneity and temporal stochasticity of niche cells across the BM during hematopoiesis. These studies demonstrate that niche cells, including mesenchymal stem cells, osteoblasts, and endothelial cells, exhibit dynamic interactions with HSCs, significantly influenced by the BM microenvironment as the age increases. Immunosurveillance plays a crucial role in maintaining hematopoietic homeostasis, with alterations in immune signaling pathways contributing to the onset of hematological malignancies. Novel insights into the interaction between niche cells and HSCs under stress/aging conditions highlight the importance of niche plasticity and adaptability. CONCLUSION The involvement of age-induced genetic/epigenetic alterations in BM niche cells and immunological stressors in hematopoiesis is crucial for understanding the development of age-related hematological malignancies. This comprehensive review provides new insights into the complex interplay between niche cells and HSCs, emphasizing the potential for novel therapeutic approaches that target niche cell functionality and resilience to improve hematopoietic outcomes in the context of aging and metabolic disorders. NOVELTY STATEMENT This review introduces novel concepts regarding the plasticity and adaptability of BM niche cells in response to immunological stressors and epigenetics. It proposes that targeted therapeutic strategies aimed at enhancing niche cell resilience could mitigate the adverse effects of aging, diabetes, and obesity on hematopoiesis and clonal hematopoiesis. Additionally, the review suggests that understanding the precise temporal and spatial dynamics of niche-HSC interactions and epigenetics influence may lead to innovative treatments for age-related hematological malignancies.
Collapse
Affiliation(s)
- Yang Xinyi
- Department of Oncology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str, Moscow, 119991, Russia
| | - Reshetov Igor Vladimirovich
- Department of Oncology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str, Moscow, 119991, Russia
| | - Narasimha M Beeraka
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str, Moscow, 119991, Russia.
- Raghavendra Institute of Pharmaceutical Education and Research (RIPER), Anantapuramu, Chiyyedu, Andhra Pradesh, 515721, India.
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut Street, R4-168, Indianapolis, IN, 46202, USA.
- Department of Studies in Molecular Biology, Faculty of Science and Technology, University of Mysore, Mysore, Karnataka, 570006, India.
| | - Allaka Satyavathi
- Department of Chemistry, Faculty of science, Dr B R Ambedkar Open University, Wanaparthy, Telangana, 509103, India
| | - Dinisha Kamble
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut Street, R4-168, Indianapolis, IN, 46202, USA
| | - Vladimir N Nikolenko
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str, Moscow, 119991, Russia
| | - Allaka Naga Lakshmi
- Department of Computer Science, St Philomena's College (Autonomous), Bangalore - Mysore Rd, Bannimantap, Mysuru, Karnataka, 570015, India
| | - Basappa Basappa
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Mysore, Karnataka, 570006, India
| | - Padmanabha Reddy Y
- Raghavendra Institute of Pharmaceutical Education and Research (RIPER), Anantapuramu, Chiyyedu, Andhra Pradesh, 515721, India
| | - Ruitai Fan
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, China.
| | - Junqi Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, No. 1, Jianshe East Road, Zhengzhou, 450000, China
| |
Collapse
|
2
|
Yuan Y, Zou M, Wu S, Liu C, Hao L. Recent advances in nanomaterials for the treatment of femoral head necrosis. Hum Cell 2024; 37:1290-1305. [PMID: 38995503 DOI: 10.1007/s13577-024-01102-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 07/05/2024] [Indexed: 07/13/2024]
Abstract
Osteonecrosis of the femoral head (ONFH) is a condition that causes considerable pain and discomfort for patients, and its pathogenic mechanisms are not yet fully understood. While there have been many studies that suggest multiple factors may contribute to its development, current treatments involve both surgical and nonsurgical options. However, there is still much room for improvement in these treatment methods, particularly when it comes to preventing postoperative complications and optimizing surgical procedures. Nanomaterials, as a type of small molecule material, have shown great promise in treating bone tissue diseases, including ONFH. In fact, several nanocomposite materials have demonstrated specific effects in preventing ONFH, promoting bone tissue repair and growth, and optimizing surgical treatment. This article provides a comprehensive overview of current treatments for ONFH, including their advantages and limitations, and reviews the latest advances in nanomaterials for treating this condition. Additionally, this article explores the therapeutic mechanisms involved in using nanomaterials to treat ONFH and to identify new methods and ideas for improving outcomes for patients.
Collapse
Affiliation(s)
- Yalin Yuan
- Department of Orthopedics, Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- The Second Clinical Medical College, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Mi Zou
- Department of Orthopedics, Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- The Second Clinical Medical College, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Shuqin Wu
- Department of Orthopedics, Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- The Second Clinical Medical College, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Congcong Liu
- Department of Orthopedics, Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- The Second Clinical Medical College, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Liang Hao
- Department of Orthopedics, Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
3
|
Strasser AS, Gonzalez-Reiche AS, Zhou X, Valdebenito-Maturana B, Ye X, Zhang B, Wu M, van Bakel H, Jabs EW. Limb reduction in an Esco2 cohesinopathy mouse model is mediated by p53-dependent apoptosis and vascular disruption. Nat Commun 2024; 15:7154. [PMID: 39168984 PMCID: PMC11339411 DOI: 10.1038/s41467-024-51328-3] [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: 09/01/2023] [Accepted: 08/01/2024] [Indexed: 08/23/2024] Open
Abstract
Roberts syndrome (RBS) is an autosomal recessive disorder with profound growth deficiency and limb reduction caused by ESCO2 loss-of-function variants. Here, we elucidate the pathogenesis of limb reduction in an Esco2fl/fl;Prrx1-CreTg/0 mouse model using bulk- and single-cell-RNA-seq and gene co-expression network analyses during embryogenesis. Our results reveal morphological and vascular defects culminating in hemorrhage of mutant limbs at E12.5. Underlying this abnormal developmental progression is a pre-apoptotic, mesenchymal cell population specific to mutant limb buds enriched for p53-related signaling beginning at E9.5. We then characterize these p53-related processes of cell cycle arrest, DNA damage, cell death, and the inflammatory leukotriene signaling pathway in vivo. In utero treatment with pifithrin-α, a p53 inhibitor, rescued the hemorrhage in mutant limbs. Lastly, significant enrichments were identified among genes associated with RBS, thalidomide embryopathy, and other genetic limb reduction disorders, suggesting a common vascular etiology among these conditions.
Collapse
Affiliation(s)
- Arielle S Strasser
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Ana Silvia Gonzalez-Reiche
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Xianxiao Zhou
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Braulio Valdebenito-Maturana
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Xiaoqian Ye
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Meng Wu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
- Department of Clinical Genomics, Mayo Clinic, 200 First Street, Rochester, MN, USA.
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street, Rochester, MN, USA.
| | - Harm van Bakel
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
| | - Ethylin Wang Jabs
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
- Department of Clinical Genomics, Mayo Clinic, 200 First Street, Rochester, MN, USA.
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 First Street, Rochester, MN, USA.
- Department of Cell, Development and Regenerative Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA.
| |
Collapse
|
4
|
Wang D, Guo Y, Heng BC, Zhang X, Wei Y, He Y, Xu M, Xia B, Deng X. Cell membrane vesicles derived from hBMSCs and hUVECs enhance bone regeneration. Bone Res 2024; 12:23. [PMID: 38594236 PMCID: PMC11003965 DOI: 10.1038/s41413-024-00325-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 02/04/2024] [Accepted: 03/08/2024] [Indexed: 04/11/2024] Open
Abstract
Bone tissue renewal can be enhanced through co-transplantation of bone mesenchymal stem cells (BMSCs) and vascular endothelial cells (ECs). However, there are apparent limitations in stem cell-based therapy which hinder its clinic translation. Hence, we investigated the potential of alternative stem cell substitutes for facilitating bone regeneration. In this study, we successfully prepared cell membrane vesicles (CMVs) from BMSCs and ECs. The results showed that BMSC-derived cell membrane vesicles (BMSC-CMVs) possessed membrane receptors involved in juxtacrine signaling and growth factors derived from their parental cells. EC-derived cell membrane vesicles (EC-CMVs) also contained BMP2 and VEGF derived from their parental cells. BMSC-CMVs enhanced tube formation and migration ability of hUVECs, while EC-CMVs promoted the osteogenic differentiation of hBMSCs in vitro. Using a rat skull defect model, we found that co-transplantation of BMSC-CMVs and EC-CMVs could stimulate angiogenesis and bone formation in vivo. Therefore, our research might provide an innovative and feasible approach for cell-free therapy in bone tissue regeneration.
Collapse
Affiliation(s)
- Dandan Wang
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Yaru Guo
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, P. R. China
| | - Boon Chin Heng
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, P. R. China
| | - Xuehui Zhang
- Department of Dental Materials & Dental Medical Devices Testing Center, Peking University School and Hospital of Stomatology, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, P. R. China
| | - Yan Wei
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, P. R. China
| | - Ying He
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, P. R. China
| | - Mingming Xu
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, P. R. China
| | - Bin Xia
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China.
| | - Xuliang Deng
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, P. R. China.
| |
Collapse
|
5
|
Eazer J, Barsoum M, Smith C, Hotta K, Behnke B, Holmes C, Caldwell J, Ghosh P, Reid-Foley E, Park H, Delp M, Muller-Delp J. Adaptations of bone and bone vasculature to muscular stretch training. JBMR Plus 2024; 8:ziad019. [PMID: 38741608 PMCID: PMC11090128 DOI: 10.1093/jbmrpl/ziad019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/16/2023] [Accepted: 12/07/2023] [Indexed: 05/16/2024] Open
Abstract
The magnitude of bone formation and remodeling is linked to both the magnitude of strain placed on the bone and the perfusion of bone. It was previously reported that an increase in bone perfusion and bone density occurs in the femur of old rats with moderate aerobic exercise training. This study determined the acute and chronic effects of static muscle stretching on bone blood flow and remodeling. Old male Fischer 344 rats were randomized to either a naive or stretch-trained group. Static stretching of ankle flexor muscles was achieved by placement of a dorsiflexion splint on the left ankle for 30 min/d, 5d/wk for 4wk. The opposite hindlimb served as a contralateral control (nonstretched) limb. Bone blood flow was assessed during and after acute stretching in naive rats, and at rest and during exercise in stretch-trained rats. Vascular reactivity of the nutrient artery of the proximal tibia was also assessed in stretch-trained rats. MicroCT analysis was used to assess bone volume and micro-architecture of the trabecular bone of both tibias near that growth plate. In naive rats, static stretching increased blood flow to the proximal tibial metaphasis. Blood flow to the proximal tibial metaphysis during treadmill exercise was higher in the stretched limb after 4 wk of daily stretching. Daily stretching also increased tibial bone weight and increased total volume in both the proximal and distal tibial metaphyses. In the trabecular bone immediately below the proximal tibial growth plate, total volume and bone volume increased, but bone volume/total volume was unchanged and trabecular connectivity decreased. In contrast, intravascular volume increased in this region of the bone. These data suggest that blood flow to the tibia increases during bouts of static stretching of the hindlimb muscles, and that 4 wk of daily muscle stretching leads to bone remodeling and an increase in intravascular volume of the tibial bone.
Collapse
Affiliation(s)
- Julia Eazer
- Department of Biomedical Sciences, Florida State University, Tallahassee, Fl, 32304, United States
| | - Mina Barsoum
- Department of Chemical Engineering, Florida State University, Tallahassee, Fl, 32304, United States
| | - Cole Smith
- Department of Biomedical Sciences, Florida State University, Tallahassee, Fl, 32304, United States
| | - Kazuki Hotta
- Department of Biomedical Sciences, Florida State University, Tallahassee, Fl, 32304, United States
- Department of Physical Therapy, Niigata University of Health and Welfare, Niigata, Japan
| | - Brad Behnke
- Department of Kinesiology & Johnson Cancer Research Center, Kansas State University, Manhattan, KS, 66506, United States
| | - Christina Holmes
- Department of Biomedical Engineering, Florida State University, Tallahassee, FL, 32310 United States
| | - Jacob Caldwell
- Department of Biomedical Sciences, Florida State University, Tallahassee, Fl, 32304, United States
| | - Payal Ghosh
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Fl, 32304, United States
| | - Emily Reid-Foley
- Department of Biomedical Sciences, Florida State University, Tallahassee, Fl, 32304, United States
| | - Hyerim Park
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Fl, 32304, United States
| | - Michael Delp
- Department of Nutrition and Integrative Physiology, Florida State University, Tallahassee, Fl, 32304, United States
| | - Judy Muller-Delp
- Department of Biomedical Sciences, Florida State University, Tallahassee, Fl, 32304, United States
| |
Collapse
|
6
|
Pang Y, Ma Y, Zheng K, Zhu S, Sui H, Ren H, Liu K, Li W, Huang Y, Du D, Gao J, Zhang C. Costal Cartilage Graft Repair Osteochondral Defect in a Mouse Model. Cartilage 2023:19476035231209404. [PMID: 37881954 DOI: 10.1177/19476035231209404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2023] Open
Abstract
OBJECTIVE Osteochondral defects develop into osteoarthritis without intervention. Costal cartilage can be utilized as an alternative source for repairing osteochondral defect. Our previous clinical study has shown the successful osteochondral repair by costal cartilage graft with integration into host bone bed. In this study, we investigate how cartilaginous graft adapt to osteochondral environment and the mechanism of bone-cartilage interface formation. DESIGN Costal cartilage grafting was performed in C57BL/6J mice and full-thickness osteochondral defect was made as control. 3D optical profiles and micro-CT were applied to evaluate the reconstruction of articular cartilage surface and subchondral bone as well as gait analysis to evaluate articular function. Histological staining was performed at 2, 4, and 8 weeks after surgery. Moreover, costal cartilage from transgenic mice with fluorescent markers were transplanted into wild-type mice to observe the in vivo changes of costal chondrocytes. RESULTS At 8 weeks after surgery, 3D optical profiles and micro-CT showed that in the graft group, the articular surface and subchondral bone were well preserved. Gait analysis and International Cartilage Repair Society (ICRS) score evaluation showed a good recovery of joint function and histological repair in the graft group. Safranin O staining showed the gradual integration of graft and host tissue. Costal cartilage from transgenic mice with fluorescent markers showed that donor-derived costal chondrocytes turned into osteocytes in the subchondral area of host femur. CONCLUSION Costal cartilage grafting shows both functional and histological repair of osteochondral defect in mice. Graft-derived costal chondrocytes differentiate into osteocytes and contribute to endochondral ossification.
Collapse
Affiliation(s)
- Yidan Pang
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiyang Ma
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kaiwen Zheng
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Siyuan Zhu
- Department of General Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongyu Sui
- School of Information Science and Technology, ShanghaiTech University, Shanghai, China
| | - Hao Ren
- School of Information Science and Technology, ShanghaiTech University, Shanghai, China
| | - Kang Liu
- Beixcell (Beijing) Biotechnology Ltd, Beijing, China
| | - Wei Li
- Beixcell (Beijing) Biotechnology Ltd, Beijing, China
| | - Yigang Huang
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dajiang Du
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junjie Gao
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Jinjiang Municipal Hospital (Shanghai Sixth People's Hospital Fujian), Jinjiang City, Quanzhou, China
| | - Changqing Zhang
- Department of Orthopaedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Du Y, Chen M, Jiang J, Wang L, Wu G, Feng J. Hst1/Gel-MA Scaffold Significantly Promotes the Quality of Osteochondral Regeneration in the Temporomandibular Joint. J Funct Biomater 2023; 14:513. [PMID: 37888178 PMCID: PMC10607535 DOI: 10.3390/jfb14100513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/28/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
OBJECTIVE Our aim was to evaluate the capacity of the human salivary histatin-1-functionalized methacrylic gelatin scaffold to control osteochondral tissue regeneration and repair in vivo in rabbits with major temporomandibular joint dimensional abnormalities. MATERIALS AND METHODS In order to compare human salivary histatin-1-functionalized methacrylic gelatin scaffolds to the Blank and Gel-MA hydrogel groups, scaffolds were implanted into osteochondral lesions of a critical size (3 × 3 mm) in the anterior region of the condyle of the temporomandibular joint in New Zealand white rabbits. At 4 weeks after implantation, the repair was evaluated using macroscopic examination, histology, and micro-CT analysis. RESULTS In the comparison of the composite scaffold group with the Blank and Gel-MA groups, analysis of the healed tissue revealed an improved macroscopic appearance in the composite scaffold group. Regeneration was induced by host cell migration in the Hst1/Gel-MA scaffold group. CONCLUSIONS The current study offers a viable method for in vivo cartilage repair that does not require cell transplantation. Future clinical applications of this strategy's optimization have many potential advantages.
Collapse
Affiliation(s)
- Yiyang Du
- School/Hospital of Stomatology, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.D.); (M.C.); (J.J.)
| | - Menghan Chen
- School/Hospital of Stomatology, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.D.); (M.C.); (J.J.)
| | - Jing Jiang
- School/Hospital of Stomatology, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.D.); (M.C.); (J.J.)
| | - Lei Wang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China;
- Department of Oral Implantology and Prosthetic Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam (UvA) and Vrije Universiteit Amsterdam (VU), 1081 LA Amsterdam, The Netherlands
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Center for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam (VU), Amsterdam Movement Science, 1081 LA Amsterdam, The Netherlands
| | - Gang Wu
- Department of Oral Implantology and Prosthetic Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam (UvA) and Vrije Universiteit Amsterdam (VU), 1081 LA Amsterdam, The Netherlands
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Center for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam (VU), Amsterdam Movement Science, 1081 LA Amsterdam, The Netherlands
| | - Jianying Feng
- School/Hospital of Stomatology, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.D.); (M.C.); (J.J.)
| |
Collapse
|
8
|
Watanabe H, Maishi N, Hoshi-Numahata M, Nishiura M, Nakanishi-Kimura A, Hida K, Iimura T. Skeletal-Vascular Interactions in Bone Development, Homeostasis, and Pathological Destruction. Int J Mol Sci 2023; 24:10912. [PMID: 37446097 DOI: 10.3390/ijms241310912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 06/28/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Bone is a highly vascularized organ that not only plays multiple roles in supporting the body and organs but also endows the microstructure, enabling distinct cell lineages to reciprocally interact. Recent studies have uncovered relevant roles of the bone vasculature in bone patterning, morphogenesis, homeostasis, and pathological bone destruction, including osteoporosis and tumor metastasis. This review provides an overview of current topics in the interactive molecular events between endothelial cells and bone cells during bone ontogeny and discusses the future direction of this research area to find novel ways to treat bone diseases.
Collapse
Affiliation(s)
- Haruhisa Watanabe
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Nako Maishi
- Department of Vascular Biology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Marie Hoshi-Numahata
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Mai Nishiura
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Atsuko Nakanishi-Kimura
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Kyoko Hida
- Department of Vascular Biology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| | - Tadahiro Iimura
- Department of Pharmacology, Faculty and Graduate School of Dental Medicine, Hokkaido University, N13 W7, Sapporo 060-8586, Hokkaido, Japan
| |
Collapse
|
9
|
Tsutsumi R, Eiraku M. How might we build limbs in vitro informed by the modular aspects and tissue-dependency in limb development? Front Cell Dev Biol 2023; 11:1135784. [PMID: 37283945 PMCID: PMC10241304 DOI: 10.3389/fcell.2023.1135784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 05/10/2023] [Indexed: 06/08/2023] Open
Abstract
Building limb morphogenesis in vitro would substantially open up avenues for research and applications of appendage development. Recently, advances in stem cell engineering to differentiate desired cell types and produce multicellular structures in vitro have enabled the derivation of limb-like tissues from pluripotent stem cells. However, in vitro recapitulation of limb morphogenesis is yet to be achieved. To formulate a method of building limbs in vitro, it is critically important to understand developmental mechanisms, especially the modularity and the dependency of limb development on the external tissues, as those would help us to postulate what can be self-organized and what needs to be externally manipulated when reconstructing limb development in vitro. Although limbs are formed on the designated limb field on the flank of embryo in the normal developmental context, limbs can also be regenerated on the amputated stump in some animals and experimentally induced at ectopic locations, which highlights the modular aspects of limb morphogenesis. The forelimb-hindlimb identity and the dorsal-ventral, proximal-distal, and anterior-posterior axes are initially instructed by the body axis of the embryo, and maintained in the limb domain once established. In contrast, the aspects of dependency on the external tissues are especially underscored by the contribution of incoming tissues, such as muscles, blood vessels, and peripheral nerves, to developing limbs. Together, those developmental mechanisms explain how limb-like tissues could be derived from pluripotent stem cells. Prospectively, the higher complexity of limb morphologies is expected to be recapitulated by introducing the morphogen gradient and the incoming tissues in the culture environment. Those technological developments would dramatically enhance experimental accessibility and manipulability for elucidating the mechanisms of limb morphogenesis and interspecies differences. Furthermore, if human limb development can be modeled, drug development would be benefited by in vitro assessment of prenatal toxicity on congenital limb deficiencies. Ultimately, we might even create a future in which the lost appendage would be recovered by transplanting artificially grown human limbs.
Collapse
Affiliation(s)
- Rio Tsutsumi
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
- Laboratory of Developmental Systems, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Mototsugu Eiraku
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
- Laboratory of Developmental Systems, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
10
|
Descoteaux AE, Zuch DT, Bradham CA. Polychrome labeling reveals skeletal triradiate and elongation dynamics and abnormalities in patterning cue-perturbed embryos. Dev Biol 2023; 498:1-13. [PMID: 36948411 DOI: 10.1016/j.ydbio.2023.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/24/2023]
Abstract
The larval skeleton of the sea urchin Lytechinus variegatus is an ideal model system for studying skeletal patterning; however, our understanding of the etiology of skeletal patterning in sea urchin larvae is limited due to the lack of approaches to live-image skeleton formation. Calcium-binding fluorochromes have been used to study the temporal dynamics of bone growth and healing. To date, only calcein green has been used in sea urchin larvae to fluorescently label the larval skeleton. Here, we optimize labeling protocols for two additional calcium-binding fluorochromes: xylenol orange and calcein blue- and demonstrate that these fluorochromes can be used individually or in nested pulse-chase experiments to understand the temporal dynamics of skeletogenesis and patterning. Using a pulse-chase approach, we show that the initiation of skeletogenesis begins around 15 h post fertilization. We also assess the timing of triradiate formation in embryos treated with a range of patterning perturbagens and demonstrate that triradiate formation is delayed and asynchronous in embryos ventralized via treatment with either nickel or chlorate. Finally, we measure the extent of fluorochrome incorporation in triple-labeled embryos to determine the elongation rate of numerous skeletal elements throughout early skeletal patterning and compare this to the rate of skeletal growth in embryos treated with axitinib to inhibit VEGFR. We find that skeletal elements elongate much more slowly in axitinib-treated embryos, and that axitinib treatment is sufficient to induce abnormal orientation of the triradiates.
Collapse
Affiliation(s)
- Abigail E Descoteaux
- Department of Biology, Boston University, Boston, MA, 02215, United States; Program in Molecular Biology, Cell Biology, and Biochemistry, Boston University, Boston, MA, 02215, United States; Biological Design Center, Boston University, Boston, MA, 02215, United States
| | - Daniel T Zuch
- Department of Biology, Boston University, Boston, MA, 02215, United States; Program in Molecular Biology, Cell Biology, and Biochemistry, Boston University, Boston, MA, 02215, United States
| | - Cynthia A Bradham
- Department of Biology, Boston University, Boston, MA, 02215, United States; Program in Molecular Biology, Cell Biology, and Biochemistry, Boston University, Boston, MA, 02215, United States; Biological Design Center, Boston University, Boston, MA, 02215, United States; Program in Bioinformatics, Boston University, Boston, MA, 02215, United States.
| |
Collapse
|
11
|
Fuiten AM, Yoshimoto Y, Shukunami C, Stadler HS. Digits in a dish: An in vitro system to assess the molecular genetics of hand/foot development at single-cell resolution. Front Cell Dev Biol 2023; 11:1135025. [PMID: 36994104 PMCID: PMC10040768 DOI: 10.3389/fcell.2023.1135025] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 02/21/2023] [Indexed: 03/16/2023] Open
Abstract
In vitro models allow for the study of developmental processes outside of the embryo. To gain access to the cells mediating digit and joint development, we identified a unique property of undifferentiated mesenchyme isolated from the distal early autopod to autonomously re-assemble forming multiple autopod structures including: digits, interdigital tissues, joints, muscles and tendons. Single-cell transcriptomic analysis of these developing structures revealed distinct cell clusters that express canonical markers of distal limb development including: Col2a1, Col10a1, and Sp7 (phalanx formation), Thbs2 and Col1a1 (perichondrium), Gdf5, Wnt5a, and Jun (joint interzone), Aldh1a2 and Msx1 (interdigital tissues), Myod1 (muscle progenitors), Prg4 (articular perichondrium/articular cartilage), and Scx and Tnmd (tenocytes/tendons). Analysis of the gene expression patterns for these signature genes indicates that developmental timing and tissue-specific localization were also recapitulated in a manner similar to the initiation and maturation of the developing murine autopod. Finally, the in vitro digit system also recapitulates congenital malformations associated with genetic mutations as in vitro cultures of Hoxa13 mutant mesenchyme produced defects present in Hoxa13 mutant autopods including digit fusions, reduced phalangeal segment numbers, and poor mesenchymal condensation. These findings demonstrate the robustness of the in vitro digit system to recapitulate digit and joint development. As an in vitro model of murine digit and joint development, this innovative system will provide access to the developing limb tissues facilitating studies to discern how digit and articular joint formation is initiated and how undifferentiated mesenchyme is patterned to establish individual digit morphologies. The in vitro digit system also provides a platform to rapidly evaluate treatments aimed at stimulating the repair or regeneration of mammalian digits impacted by congenital malformation, injury, or disease.
Collapse
Affiliation(s)
- Allison M. Fuiten
- Research Center, Shriners Children’s, Portland, OR, United States
- Department of Orthopaedics and Rehabilitation, Oregon Health and Science University, Portland, OR, United States
| | - Yuki Yoshimoto
- Department of Molecular Biology and Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Chisa Shukunami
- Department of Molecular Biology and Biochemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - H. Scott Stadler
- Research Center, Shriners Children’s, Portland, OR, United States
- Department of Orthopaedics and Rehabilitation, Oregon Health and Science University, Portland, OR, United States
- *Correspondence: H. Scott Stadler,
| |
Collapse
|
12
|
Bonsang B, Maksimovic L, Maille P, Martin N, Laurendeau I, Pasmant E, Bièche I, Deschamps J, Wolkenstein P, Ortonne N. VEGF and VEGFR family members are expressed by neoplastic cells of NF1-associated tumors and may play an oncogenic role in malignant peripheral nerve sheath tumor growth through an autocrine loop. Ann Diagn Pathol 2022; 60:151997. [DOI: 10.1016/j.anndiagpath.2022.151997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 06/17/2022] [Indexed: 11/01/2022]
|
13
|
Vitamin B 2 Prevents Glucocorticoid-Caused Damage of Blood Vessels in Osteonecrosis of the Femoral Head. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4006184. [PMID: 35845964 PMCID: PMC9279053 DOI: 10.1155/2022/4006184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 04/25/2022] [Accepted: 05/10/2022] [Indexed: 12/19/2022]
Abstract
Osteonecrosis of the femoral head (ONFH) is a disorder that can cause collapse of the femoral head. The damage and dysfunction of femoral head microvascular endothelial cells are related to the pathogenesis of glucocorticoid-induced ONFH. Reports suggest that vitamin B2 can promote osteoblast differentiation and prevent low bone mineral density and prevent reperfusion oxidative injury. To explore the effect and possible molecular mechanism of vitamin B2 on the ONFH and Human Umbilical Vein Endothelial Cells (HUVECs), we performed a rat model of ONFH by dexamethasone. The rats were randomly divided into four groups: control group, vitamin B2 group, dexamethasone group, and dexamethasone combined with vitamin B2 treatment group. HUVECs were used to further prove the role and mechanism of vitamin B2 in vitro. In patients, according to immunohistochemical and qRT-PCR of the femoral head, the angiogenic capacity of the ONFH femoral head is compromised. In vivo, it showed that vitamin B2 could inhibit glucocorticoid-induced ONFH-like changes in rats by suppressing cell apoptosis, promoting the regeneration of blood vessels, and increasing bone mass. According to in vitro results, vitamin B2 could induce the migration of HUVECs, enhance the expression of angiogenesis-related factors, and inhibit glucocorticoid-induced apoptosis. The underlying mechanism may be that vitamin B2 activates the PI3K signaling pathway. Vitamin B2 alleviated dexamethasone-induced ONFH, and vitamin B2 could promote the proliferation and migration of HUVECs and inhibit their apoptosis by activating the PI3K/Akt signaling pathway. Vitamin B2 may be a potentially effective treatment for ONFH.
Collapse
|
14
|
Liao C, Lu M, Hong Y, Mao C, Chen J, Ren C, Lin M, Chen W. Osteogenic and angiogenic profiles of the palatal process of the maxilla and the palatal process of the palatine bone. J Anat 2022; 240:385-397. [PMID: 34569061 PMCID: PMC8742962 DOI: 10.1111/joa.13545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/28/2021] [Accepted: 08/31/2021] [Indexed: 11/30/2022] Open
Abstract
Hard palate consists anteriorly of the palatal process of the maxilla (ppmx) and posteriorly of the palatal process of the palatine (ppp). Currently, palatal osteogenesis is receiving increasing attention. This is the first study to provide an overview of the osteogenesis process of the mouse hard palate. We found that the period in which avascular mesenchymal condensation becomes a vascularized bone structure corresponds to embryonic day (E) 14.5 to E16.5 in the hard palate. The ppmx and ppp differ remarkably in morphology and molecular respects during osteogenesis. Osteoclasts in the ppmx and ppp are heterogeneous. There was a multinucleated giant osteoclast on the bone surface at the lateral-nasal side of the ppmx, while osteoclasts in the ppp were more abundant and adjacent to blood vessels but were smaller and had fewer nuclei. In addition, bone remodeling in the hard palate was asymmetric and exclusively occurred on the nasal side of the hard palate at E18.5. During angiogenesis, CD31-positive endothelial cells were initially localized in the surrounding of palatal mesenchymal condensation and then invaded the condensation in a sprouting fashion. At the transcriptome level, we found 78 differentially expressed genes related to osteogenesis and angiogenesis between the ppmx and ppp. Fifty-five related genes were up/downregulated from E14.5 to E16.5. Here, we described the morphogenesis and the heterogeneity in the osteogenic and angiogenic genes profiles of the ppmx and ppp, which are significant for subsequent studies of normal and abnormal subjects.
Collapse
Affiliation(s)
- Caiyu Liao
- Department of Oral and Maxillofacial SurgeryFujian Medical University Union HospitalFuzhouFujianChina
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and UniversitySchool and Hospital of StomatologyFujian Medical UniversityFuzhouFujianChina
| | - Meng Lu
- Department of Oral and Maxillofacial SurgeryFujian Medical University Union HospitalFuzhouFujianChina
| | - Yuhang Hong
- Department of Oral and Maxillofacial SurgeryFujian Medical University Union HospitalFuzhouFujianChina
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and UniversitySchool and Hospital of StomatologyFujian Medical UniversityFuzhouFujianChina
| | - Chuanqing Mao
- Department of Oral and Maxillofacial SurgeryFujian Medical University Union HospitalFuzhouFujianChina
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and UniversitySchool and Hospital of StomatologyFujian Medical UniversityFuzhouFujianChina
| | - Jiangping Chen
- Department of Oral and Maxillofacial SurgeryFujian Medical University Union HospitalFuzhouFujianChina
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and UniversitySchool and Hospital of StomatologyFujian Medical UniversityFuzhouFujianChina
| | - Chengyan Ren
- Department of Oral and Maxillofacial SurgeryFujian Medical University Union HospitalFuzhouFujianChina
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and UniversitySchool and Hospital of StomatologyFujian Medical UniversityFuzhouFujianChina
| | - Minkui Lin
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and UniversitySchool and Hospital of StomatologyFujian Medical UniversityFuzhouFujianChina
| | - Weihui Chen
- Department of Oral and Maxillofacial SurgeryFujian Medical University Union HospitalFuzhouFujianChina
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and UniversitySchool and Hospital of StomatologyFujian Medical UniversityFuzhouFujianChina
| |
Collapse
|
15
|
Bone morphogenetic protein 9 enhances osteogenic and angiogenic responses of human amniotic mesenchymal stem cells cocultured with umbilical vein endothelial cells through the PI3K/AKT/m-TOR signaling pathway. Aging (Albany NY) 2021; 13:24829-24849. [PMID: 34837694 PMCID: PMC8660623 DOI: 10.18632/aging.203718] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 10/25/2021] [Indexed: 01/06/2023]
Abstract
Background: Neovascularization plays an essential part in bone fracture and defect healing, constructing tissue engineered bone that targets bone regeneration. Bone morphogenetic protein 9 (BMP9) is a regular indicator that potentiates osteogenic and angiogenic differentiation of MSCs. Objectives: To investigate the effects of BMP9 on osteogenesis and angiogenesis of human amniotic mesenchymal stem cells (hAMSCs) cocultured with human umbilical vein endothelial cells (HUVECs) and determine the possible underlying molecular mechanism. Results: The isolated hAMSCs expressed surface markers of MSCs. hAMSCs cocultured with HUVECs enhance osteogenic differentiation and upregulate the expression of angiogenic factors. BMP9 not only potentiates angiogenic signaling of hAMSCs cocultured with HUVECs also increases ectopic bone formation and subcutaneous vessel invasion. Mechanically, the coupling effect between osteogenesis and angiogenesis induced by BMP9 was activated by the BMP/Smad and PI3K/AKT/m-TOR signaling pathways. Conclusions: BMP9-enhanced osteoblastic and angiogenic differentiation in cocultivation with hAMSCs and HUVECs in vitro and in vivo also provide a chance to harness the BMP9-regulated coordinated effect between osteogenic and angiogenic pathways through BMP/Smad and PI3K/AKT/m-TOR signalings. Materials and Methods: The ALP and Alizarin Red S staining assay to determine the effects of osteoblastic differentiation. RT-qPCR and western blot was measured the expression of angiogenesis-related factors. Ectopic bone formation was established and retrieved bony masses were subjected to histochemical staining. The angiogenesis ability and vessel invasion were subsequently determined by immunofluorescence staining. Molecular mechanisms such as the BMP/Smad and PI3K/AKT/m-TOR signaling pathways were detected by ELISA and western blot analysis.
Collapse
|
16
|
Secchi C, Belli M, Harrison TNH, Swift J, Ko C, Duleba AJ, Stupack D, Chang RJ, Shimasaki S. Effect of the spatial-temporal specific theca cell Cyp17 overexpression on the reproductive phenotype of the novel TC17 mouse. J Transl Med 2021; 19:428. [PMID: 34654452 PMCID: PMC8520195 DOI: 10.1186/s12967-021-03103-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/01/2021] [Indexed: 12/15/2022] Open
Abstract
Background In the ovarian follicle, the Theca Cells (TCs) have two main functions: preserving morphological integrity and, importantly, secreting steroid androgen hormones. TCs express the essential enzyme 17α-hydroxylase/17,20-desmolase (CYP17), which permits the conversion of pregnenolone and progesterone into androgens. Dysregulation of CYP17 enzyme activity due to an intrinsic ovarian defect is hypothesized to be a cause of hyperandrogenism in women. Androgen excess is observed in women with polycystic ovary syndrome (PCOS) resulting from excess endogenous androgen production, and in transgender males undergoing exogenous testosterone therapy after female sex assignment at birth. However, the molecular and morphological effects of Cyp17 overexpression and androgen excess on folliculogenesis is unknown. Methods In this work, seeking a comprehensive profiling of the local outcomes of the androgen excess in the ovary, we generated a transgenic mouse model (TC17) with doxycycline (Dox)-induced Cyp17 overexpression in a local and temporal manner. TC17 mice were obtained by a combination of the Tet-dependent expression system and the Cre/LoxP gene control system. Results Ovaries of Dox-treated TC17 mice overexpressed Cyp17 specifically in TCs, inducing high testosterone levels. Surprisingly, TC17 ovarian morphology resembled the human ovarian features of testosterone-treated transgender men (partially impaired folliculogenesis, hypertrophic or luteinized stromal cells, atretic follicles, and collapsed clusters). We additionally assessed TC17 fertility denoting a perturbation of the normal reproductive functions (e.g., low pregnancy rate and numbers of pups per litter). Finally, RNAseq analysis permitted us to identify dysregulated genes (Lhcgr, Fshr, Runx1) and pathways (Extra Cellular Matrix and Steroid Synthesis). Conclusions Our novel mouse model is a versatile tool to provide innovative insights into study the effects of Cyp17 overexpression and hyperandrogenism in the ovary. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03103-x.
Collapse
Affiliation(s)
- Christian Secchi
- Department of Obstetrics, Gynecology and Reproductive Sciences, School of Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Martina Belli
- Department of Obstetrics, Gynecology and Reproductive Sciences, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Tracy N H Harrison
- Department of Obstetrics, Gynecology and Reproductive Sciences, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Joseph Swift
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - CheMyong Ko
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Antoni J Duleba
- Department of Obstetrics, Gynecology and Reproductive Sciences, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Dwayne Stupack
- Department of Obstetrics, Gynecology and Reproductive Sciences, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - R Jeffrey Chang
- Department of Obstetrics, Gynecology and Reproductive Sciences, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Shunichi Shimasaki
- Department of Obstetrics, Gynecology and Reproductive Sciences, School of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
17
|
Han X, Sun M, Chen B, Saiding Q, Zhang J, Song H, Deng L, Wang P, Gong W, Cui W. Lotus seedpod-inspired internal vascularized 3D printed scaffold for bone tissue repair. Bioact Mater 2021; 6:1639-1652. [PMID: 33313444 PMCID: PMC7701916 DOI: 10.1016/j.bioactmat.2020.11.019] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/27/2020] [Accepted: 11/12/2020] [Indexed: 12/16/2022] Open
Abstract
In the field of bone defect repair, 3D printed scaffolds have the characteristics of personalized customization and accurate internal structure. However, how to construct a well-structured vascular network quickly and effectively inside the scaffold is essential for bone repair after transplantation. Herein, inspired by the unique biological structure of "lotus seedpod", hydrogel microspheres encapsulating deferoxamine (DFO) liposomes were prepared through microfluidic technology as "lotus seeds", and skillfully combined with a three-dimensional (3D) printed bioceramic scaffold with biomimetic "lotus" biological structure which can internally grow blood vessels. In this composite scaffold system, DFO was effectively released by 36% in the first 6 h, which was conducive to promote the growth of blood vessels inside the scaffold quickly. In the following 7 days, the release rate of DFO reached 69%, which was fundamental in the formation of blood vessels inside the scaffold as well as osteogenic differentiation of bone mesenchymal stem cells (BMSCs). It was confirmed that the composite scaffold could significantly promote the human umbilical vein endothelial cells (HUVECs) to form the vascular morphology within 6 h in vitro. In vivo, the composite scaffold increased the expression of vascularization and osteogenic related proteins Hif1-α, CD31, OPN, and OCN in the rat femoral defect model, significantly cutting down the time of bone repair. To sum up, this "lotus seedpod" inspired porous bioceramic 3D printed scaffold with internal vascularization functionality has broad application prospects in the future.
Collapse
Affiliation(s)
- Xiaoyu Han
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, Shandong, 250013, PR China
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Mingjie Sun
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, Shandong, 250013, PR China
| | - Bo Chen
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Qimanguli Saiding
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Junyue Zhang
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, Shandong, 250013, PR China
| | - Hongliang Song
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, Shandong, 250013, PR China
| | - Lianfu Deng
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| | - Peng Wang
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, Shandong, 250013, PR China
| | - Weiming Gong
- Department of Orthopedics, Jinan Central Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, 105 Jiefang Road, Lixia District, Jinan, Shandong, 250013, PR China
| | - Wenguo Cui
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, PR China
| |
Collapse
|
18
|
Schott NG, Friend NE, Stegemann JP. Coupling Osteogenesis and Vasculogenesis in Engineered Orthopedic Tissues. TISSUE ENGINEERING. PART B, REVIEWS 2021; 27:199-214. [PMID: 32854589 PMCID: PMC8349721 DOI: 10.1089/ten.teb.2020.0132] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/25/2020] [Indexed: 12/20/2022]
Abstract
Inadequate vascularization of engineered tissue constructs is a main challenge in developing a clinically impactful therapy for large, complex, and recalcitrant bone defects. It is well established that bone and blood vessels form concomitantly during development, as well as during repair after injury. Endothelial cells (ECs) and mesenchymal stromal cells (MSCs) are known to be key players in orthopedic tissue regeneration and vascularization, and these cell types have been used widely in tissue engineering strategies to create vascularized bone. Coculture studies have demonstrated that there is crosstalk between ECs and MSCs that can lead to synergistic effects on tissue regeneration. At the same time, the complexity in fabricating, culturing, and characterizing engineered tissue constructs containing multiple cell types presents a challenge in creating multifunctional tissues. In particular, the timing, spatial distribution, and cell phenotypes that are most conducive to promoting concurrent bone and vessel formation are not well understood. This review describes the processes of bone and vascular development, and how these have been harnessed in tissue engineering strategies to create vascularized bone. There is an emphasis on interactions between ECs and MSCs, and the culture systems that can be used to understand and control these interactions within a single engineered construct. Developmental engineering strategies to mimic endochondral ossification are discussed as a means of generating vascularized orthopedic tissues. The field of tissue engineering has made impressive progress in creating tissue replacements. However, the development of larger, more complex, and multifunctional engineered orthopedic tissues will require a better understanding of how osteogenesis and vasculogenesis are coupled in tissue regeneration. Impact statement Vascularization of large engineered tissue volumes remains a challenge in developing new and more biologically functional bone grafts. A better understanding of how blood vessels develop during bone formation and regeneration is needed. This knowledge can then be applied to develop new strategies for promoting both osteogenesis and vasculogenesis during the creation of engineered orthopedic tissues. This article summarizes the processes of bone and blood vessel development, with a focus on how endothelial cells and mesenchymal stromal cells interact to form vascularized bone both during development and growth, as well as tissue healing. It is meant as a resource for tissue engineers who are interested in creating vascularized tissue, and in particular to those developing cell-based therapies for large, complex, and recalcitrant bone defects.
Collapse
Affiliation(s)
- Nicholas G. Schott
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Nicole E. Friend
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Jan P. Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
19
|
Bhatti FUR, Dadwal UC, Valuch CR, Tewari NP, Awosanya OD, de Andrade Staut C, Sun S, Mendenhall SK, Perugini AJ, Nagaraj RU, Battina HL, Nazzal MK, Blosser RJ, Maupin KA, Childress PJ, Li J, Kacena MA. The effects of high fat diet, bone healing, and BMP-2 treatment on endothelial cell growth and function. Bone 2021; 146:115883. [PMID: 33581374 PMCID: PMC8009863 DOI: 10.1016/j.bone.2021.115883] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 02/08/2023]
Abstract
Angiogenesis is a vital process during the regeneration of bone tissue. The aim of this study was to investigate angiogenesis at the fracture site as well as at distal locations from obesity-induced type 2 diabetic mice that were treated with bone morphogenetic protein-2 (BMP-2, local administration at the time of surgery) to heal a femoral critical sized defect (CSD) or saline as a control. Mice were fed a high fat diet (HFD) to induce a type 2 diabetic-like phenotype while low fat diet (LFD) animals served as controls. Endothelial cells (ECs) were isolated from the lungs (LECs) and bone marrow (BMECs) 3 weeks post-surgery, and the fractured femurs were also examined. Our studies demonstrate that local administration of BMP-2 at the fracture site in a CSD model results in complete bone healing within 3 weeks for all HFD mice and 66.7% of LFD mice, whereas those treated with saline remain unhealed. At the fracture site, vessel parameters and adipocyte numbers were significantly increased in BMP-2 treated femurs, irrespective of diet. At distal sites, LEC and BMEC proliferation was not altered by diet or BMP-2 treatment. HFD increased the tube formation ability of both LECs and BMECs. Interestingly, BMP-2 treatment at the time of surgery reduced tube formation in LECs and humeri BMECs. However, migration of BMECs from HFD mice treated with BMP-2 was increased compared to BMECs from HFD mice treated with saline. BMP-2 treatment significantly increased the expression of CD31, FLT-1, and ANGPT2 in LECs and BMECs in LFD mice, but reduced the expression of these same genes in HFD mice. To date, this is the first study that depicts the systemic influence of fracture surgery and local BMP-2 treatment on the proliferation and angiogenic potential of ECs derived from the bone marrow and lungs.
Collapse
Affiliation(s)
- Fazal Ur Rehman Bhatti
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA; Richard L. Roudebush VA Medical Center, IN, USA
| | - Ushashi C Dadwal
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA; Richard L. Roudebush VA Medical Center, IN, USA
| | - Conner R Valuch
- Department of Biology, Indiana University Purdue University Indianapolis, IN, USA
| | - Nikhil P Tewari
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Olatundun D Awosanya
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | | | - Seungyup Sun
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Stephen K Mendenhall
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Anthony J Perugini
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Rohit U Nagaraj
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Hanisha L Battina
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Murad K Nazzal
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Rachel J Blosser
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA; Richard L. Roudebush VA Medical Center, IN, USA
| | - Kevin A Maupin
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA
| | - Paul J Childress
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA; Richard L. Roudebush VA Medical Center, IN, USA
| | - Jiliang Li
- Department of Biology, Indiana University Purdue University Indianapolis, IN, USA
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, IN, USA; Richard L. Roudebush VA Medical Center, IN, USA.
| |
Collapse
|
20
|
Heinemann-Yerushalmi L, Bentovim L, Felsenthal N, Vinestock RC, Michaeli N, Krief S, Silberman A, Cohen M, Ben-Dor S, Brenner O, Haffner-Krausz R, Itkin M, Malitsky S, Erez A, Zelzer E. BCKDK regulates the TCA cycle through PDC in the absence of PDK family during embryonic development. Dev Cell 2021; 56:1182-1194.e6. [PMID: 33773101 DOI: 10.1016/j.devcel.2021.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 12/10/2020] [Accepted: 03/01/2021] [Indexed: 12/15/2022]
Abstract
Pyruvate dehydrogenase kinases (PDK1-4) inhibit the TCA cycle by phosphorylating pyruvate dehydrogenase complex (PDC). Here, we show that PDK family is dispensable for murine embryonic development and that BCKDK serves as a compensatory mechanism by inactivating PDC. First, we knocked out all four Pdk genes one by one. Surprisingly, Pdk total KO embryos developed and were born in expected ratios but died by postnatal day 4 because of hypoglycemia or ketoacidosis. Moreover, PDC was phosphorylated in these embryos, suggesting that another kinase compensates for PDK family. Bioinformatic analysis implicated branched-chain ketoacid dehydrogenase kinase (Bckdk), a key regulator of branched-chain amino acids (BCAAs) catabolism. Indeed, knockout of Bckdk and Pdk family led to the loss of PDC phosphorylation, an increase in PDC activity and pyruvate entry into the TCA cycle, and embryonic lethality. These findings reveal a regulatory crosstalk hardwiring BCAA and glucose catabolic pathways, which feed the TCA cycle.
Collapse
Affiliation(s)
| | - Lital Bentovim
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Neta Felsenthal
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ron Carmel Vinestock
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Nofar Michaeli
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Sharon Krief
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Alon Silberman
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Marina Cohen
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Shifra Ben-Dor
- Bioinformatics and Biological Computing Unit, Biological Services, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Ori Brenner
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Rebecca Haffner-Krausz
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Maxim Itkin
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Sergey Malitsky
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ayelet Erez
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Elazar Zelzer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
21
|
Galea GL, Zein MR, Allen S, Francis-West P. Making and shaping endochondral and intramembranous bones. Dev Dyn 2020; 250:414-449. [PMID: 33314394 PMCID: PMC7986209 DOI: 10.1002/dvdy.278] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/13/2020] [Accepted: 11/20/2020] [Indexed: 12/13/2022] Open
Abstract
Skeletal elements have a diverse range of shapes and sizes specialized to their various roles including protecting internal organs, locomotion, feeding, hearing, and vocalization. The precise positioning, size, and shape of skeletal elements is therefore critical for their function. During embryonic development, bone forms by endochondral or intramembranous ossification and can arise from the paraxial and lateral plate mesoderm or neural crest. This review describes inductive mechanisms to position and pattern bones within the developing embryo, compares and contrasts the intrinsic vs extrinsic mechanisms of endochondral and intramembranous skeletal development, and details known cellular processes that precisely determine skeletal shape and size. Key cellular mechanisms are employed at distinct stages of ossification, many of which occur in response to mechanical cues (eg, joint formation) or preempting future load‐bearing requirements. Rapid shape changes occur during cellular condensation and template establishment. Specialized cellular behaviors, such as chondrocyte hypertrophy in endochondral bone and secondary cartilage on intramembranous bones, also dramatically change template shape. Once ossification is complete, bone shape undergoes functional adaptation through (re)modeling. We also highlight how alterations in these cellular processes contribute to evolutionary change and how differences in the embryonic origin of bones can influence postnatal bone repair. Compares and contrasts Endochondral and intramembranous bone development Reviews embryonic origins of different bones Describes the cellular and molecular mechanisms of positioning skeletal elements. Describes mechanisms of skeletal growth with a focus on the generation of skeletal shape
Collapse
Affiliation(s)
- Gabriel L Galea
- Developmental Biology and Cancer, UCL GOS Institute of Child Health, London, UK.,Comparative Bioveterinary Sciences, Royal Veterinary College, London, UK
| | - Mohamed R Zein
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| | - Steven Allen
- Comparative Bioveterinary Sciences, Royal Veterinary College, London, UK
| | - Philippa Francis-West
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| |
Collapse
|
22
|
Horton JA, Hootnick DR. The vascular origins of antero-medial tibial bowing in congenital fibular deficiency. Anat Rec (Hoboken) 2020; 304:1889-1900. [PMID: 33314725 DOI: 10.1002/ar.24580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/06/2020] [Accepted: 10/27/2020] [Indexed: 11/10/2022]
Abstract
Anteromedial bowing and shortening of the tibia are intrinsic features of limbs with congenital fibular deficiency (CFD). Tibial bowing occurs more frequently when the fibula is radiographically absent rather than deficient. The bowing has been attributed to rapid longitudinal growth of the tibial anlage coupled with anteromedial tibial bending moments of the posterior crural and lateral peroneal musculature unopposed in the absence of a fibular strut. Eccentric mechanical loading results in asymmetric mineral deposition and thickening of the diaphyseal cortex. Skeletogenesis depends upon an intimate interplay between the normally prefigured tibial cartilage anlage and beginning muscular contractile actions during initial vascularization of the anlage, while the embryonic limb vasculature is undergoing a series of transitions. A diaphyseal periosteal collar normally forms at the site of nutrient artery invasion and stabilizes the growing anlage. In CFD however, arteriography consistently reveals anomalous tibial nutrient arterial branches, which originate from a primitive axial artery rather than from the usual posterior tibial artery. These anomalous nutrient arteries enter the tibial shaft at the posterior aspect of the proximal metaphysis, establishing an eccentric bone collar. The developing vasculature of the embryonic limb is responsive to the then most metabolically active tissues. Disruption of the reciprocal relationship between the transitioning vasculature and the developing long bones is pivotal in producing the diverse skeletal malformations of the congenital short limb (CSL). Embryonic vascular dysgenesis contributes not only to the well-recognized congenital tibial and fibular shortenings but also predisposes to congenital anteromedial bowing of the tibia.
Collapse
Affiliation(s)
- Jason A Horton
- Departments of Orthopedic Surgery, SUNY Upstate Medical University, Syracuse, New York, USA.,Departments of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - David R Hootnick
- Departments of Orthopedic Surgery, SUNY Upstate Medical University, Syracuse, New York, USA.,Departments of Pediatrics, SUNY Upstate Medical University, Syracuse, New York, USA.,Departments of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
23
|
Chen J, Hendriks M, Chatzis A, Ramasamy SK, Kusumbe AP. Bone Vasculature and Bone Marrow Vascular Niches in Health and Disease. J Bone Miner Res 2020; 35:2103-2120. [PMID: 32845550 DOI: 10.1002/jbmr.4171] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/21/2020] [Accepted: 08/05/2020] [Indexed: 12/20/2022]
Abstract
Bone vasculature and bone marrow vascular niches supply oxygen, nutrients, and secrete angiocrine factors required for the survival, maintenance, and self-renewal of stem and progenitor cells. In the skeletal system, vasculature creates nurturing niches for bone and blood-forming stem cells. Blood vessels regulate hematopoiesis and drive bone formation during development, repair, and regeneration. Dysfunctional vascular niches induce skeletal aging, bone diseases, and hematological disorders. Recent cellular and molecular characterization of the bone marrow microenvironment has provided unprecedented insights into the complexity, heterogeneity, and functions of the bone vasculature and vascular niches. The bone vasculature is composed of distinct vessel subtypes that differentially regulate osteogenesis, hematopoiesis, and disease conditions in bones. Further, bone marrow vascular niches supporting stem cells are often complex microenvironments involving multiple different cell populations and vessel subtypes. This review provides an overview of the emerging vascular cell heterogeneity in bone and the new roles of the bone vasculature and associated vascular niches in health and disease. © 2020 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Junyu Chen
- Tissue and Tumor Microenvironments Group, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Michelle Hendriks
- Institute of Clinical Sciences, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Alexandros Chatzis
- Tissue and Tumor Microenvironments Group, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Saravana K Ramasamy
- Institute of Clinical Sciences, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Anjali P Kusumbe
- Tissue and Tumor Microenvironments Group, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| |
Collapse
|
24
|
Ko FC, Sumner DR. How faithfully does intramembranous bone regeneration recapitulate embryonic skeletal development? Dev Dyn 2020; 250:377-392. [PMID: 32813296 DOI: 10.1002/dvdy.240] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 07/19/2020] [Accepted: 08/13/2020] [Indexed: 02/06/2023] Open
Abstract
Postnatal intramembranous bone regeneration plays an important role during a wide variety of musculoskeletal regeneration processes such as fracture healing, joint replacement and dental implant surgery, distraction osteogenesis, stress fracture healing, and repair of skeletal defects caused by trauma or resection of tumors. The molecular basis of intramembranous bone regeneration has been interrogated using rodent models of most of these conditions. These studies reveal that signaling pathways such as Wnt, TGFβ/BMP, FGF, VEGF, and Notch are invoked, reminiscent of embryonic development of membranous bone. Discoveries of several skeletal stem cell/progenitor populations using mouse genetic models also reveal the potential sources of postnatal intramembranous bone regeneration. The purpose of this review is to compare the underlying molecular signals and progenitor cells that characterize embryonic development of membranous bone and postnatal intramembranous bone regeneration.
Collapse
Affiliation(s)
- Frank C Ko
- Department of Cell & Molecular Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - D Rick Sumner
- Department of Cell & Molecular Medicine, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
25
|
Cordeiro IR, Tanaka M. Environmental Oxygen is a Key Modulator of Development and Evolution: From Molecules to Ecology. Bioessays 2020; 42:e2000025. [DOI: 10.1002/bies.202000025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 06/09/2020] [Indexed: 12/19/2022]
Affiliation(s)
- Ingrid Rosenburg Cordeiro
- Department of Life Science and Technology Tokyo Institute of Technology B‐17, 4259 Nagatsuta‐cho, Midori‐ku Yokohama 226‐8501 Japan
| | - Mikiko Tanaka
- Department of Life Science and Technology Tokyo Institute of Technology B‐17, 4259 Nagatsuta‐cho, Midori‐ku Yokohama 226‐8501 Japan
| |
Collapse
|
26
|
Fowler DA, Larsson HCE. The tissues and regulatory pattern of limb chondrogenesis. Dev Biol 2020; 463:124-134. [PMID: 32417169 DOI: 10.1016/j.ydbio.2020.04.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 12/24/2022]
Abstract
Initial limb chondrogenesis offers the first differentiated tissues that resemble the mature skeletal anatomy. It is a developmental progression of three tissues. The limb begins with undifferentiated mesenchyme-1, some of which differentiates into condensations-2, and this tissue then transforms into cartilage-3. Each tissue is identified by physical characteristics of cell density, shape, and extracellular matrix composition. Tissue specific regimes of gene regulation underlie the diagnostic physical and chemical properties of these three tissues. These three tissue based regimes co-exist amid a background of other gene regulatory regimes within the same tissues and time-frame of limb development. The bio-molecular indicators of gene regulation reveal six identifiable patterns. Three of these patterns describe the unique bio-molecular indicators of each of the three tissues. A fourth pattern shares bio-molecular indicators between condensation and cartilage. Finally, a fifth pattern is composed of bio-molecular indicators that are found in undifferentiated mesenchyme prior to any condensation differentiation, then these bio-molecular indicators are upregulated in condensations and downregulated in undifferentiated mesenchyme. The undifferentiated mesenchyme that remains in between the condensations and cartilage, the interdigit, contains a unique set of bio-molecular indicators that exhibit dynamic behaviour during chondrogenesis and therefore argue for its own inclusion as a tissue in its own right and for more study into this process of differentiation.
Collapse
Affiliation(s)
- Donald A Fowler
- Redpath Museum, McGill University, 859 Sherbrooke St W, Montréal, QC, H3A 0C4, Canada; Department of Biology, McGill University, Stewart Biology Building, 1205 Docteur Penfield, Montréal, QC, H3A 1B1, Canada.
| | - Hans C E Larsson
- Redpath Museum, McGill University, 859 Sherbrooke St W, Montréal, QC, H3A 0C4, Canada.
| |
Collapse
|
27
|
Zhao Y, Xie L. Unique bone marrow blood vessels couple angiogenesis and osteogenesis in bone homeostasis and diseases. Ann N Y Acad Sci 2020; 1474:5-14. [PMID: 32242943 DOI: 10.1111/nyas.14348] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/10/2020] [Accepted: 03/18/2020] [Indexed: 02/05/2023]
Abstract
Blood vessels serve as a versatile transport system and play crucial roles in organ development, regeneration, and stem cell behavior. In the skeletal system, certain capillaries support perivascular stem cells or osteoprogenitor cells and thereby regulate bone formation. Recent studies reported that a specialized capillary subtype, termed type H vessels, is shown to couple angiogenesis and osteogenesis in rodents and humans. They can be distinguished by specific cell surface markers, as the endothelial cells in the metaphysis and endosteum highly express the junctional protein CD31 and the sialoglycoprotein endomucin. Here, we provide an overview of the role of type H vessels in bone homeostasis and summarize their linkage with various cytokines to control bone cell behavior and bone formation. We also discuss the potential clinical application for bone disorders by targeting these specific vessels according to their physiological and pathobiological settings.
Collapse
Affiliation(s)
- Yifan Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Liang Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
28
|
Sivaraj KK, Dharmalingam B, Mohanakrishnan V, Jeong HW, Kato K, Schröder S, Adams S, Koh GY, Adams RH. YAP1 and TAZ negatively control bone angiogenesis by limiting hypoxia-inducible factor signaling in endothelial cells. eLife 2020; 9:50770. [PMID: 31958058 PMCID: PMC6970532 DOI: 10.7554/elife.50770] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 12/21/2019] [Indexed: 12/22/2022] Open
Abstract
Blood vessels are integrated into different organ environments with distinct properties and physiology (Augustin and Koh, 2017). A striking example of organ-specific specialization is the bone vasculature where certain molecular signals yield the opposite effect as in other tissues (Glomski et al., 2011; Kusumbe et al., 2014; Ramasamy et al., 2014). Here, we show that the transcriptional coregulators Yap1 and Taz, components of the Hippo pathway, suppress vascular growth in the hypoxic microenvironment of bone, in contrast to their pro-angiogenic role in other organs. Likewise, the kinase Lats2, which limits Yap1/Taz activity, is essential for bone angiogenesis but dispensable in organs with lower levels of hypoxia. With mouse genetics, RNA sequencing, biochemistry, and cell culture experiments, we show that Yap1/Taz constrain hypoxia-inducible factor 1α (HIF1α) target gene expression in vivo and in vitro. We propose that crosstalk between Yap1/Taz and HIF1α controls angiogenesis depending on the level of tissue hypoxia, resulting in organ-specific biological responses.
Collapse
Affiliation(s)
- Kishor K Sivaraj
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany.,Faculty of Medicine, University of Münster, Münster, Germany
| | - Backialakshmi Dharmalingam
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany.,Faculty of Medicine, University of Münster, Münster, Germany
| | - Vishal Mohanakrishnan
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany.,Faculty of Medicine, University of Münster, Münster, Germany
| | - Hyun-Woo Jeong
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany.,Faculty of Medicine, University of Münster, Münster, Germany
| | - Katsuhiro Kato
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany.,Faculty of Medicine, University of Münster, Münster, Germany
| | - Silke Schröder
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany.,Faculty of Medicine, University of Münster, Münster, Germany
| | - Susanne Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany.,Faculty of Medicine, University of Münster, Münster, Germany
| | - Gou Young Koh
- Center for Vascular Research, Institute of Basic Science (IBS), Daejeon, Republic of Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.,Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany.,Faculty of Medicine, University of Münster, Münster, Germany
| |
Collapse
|
29
|
Willis CR, Ames RM, Deane CS, Phillips BE, Boereboom CL, Abdulla H, Bukhari SS, Lund JN, Williams JP, Wilkinson DJ, Smith K, Kadi F, Szewczyk NJ, Atherton PJ, Etheridge T. Network analysis of human muscle adaptation to aging and contraction. Aging (Albany NY) 2020; 12:740-755. [PMID: 31910159 PMCID: PMC6977671 DOI: 10.18632/aging.102653] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 12/24/2019] [Indexed: 12/21/2022]
Abstract
Resistance exercise (RE) remains a primary approach for minimising aging muscle decline. Understanding muscle adaptation to individual contractile components of RE (eccentric, concentric) might optimise RE-based intervention strategies. Herein, we employed a network-driven pipeline to identify putative molecular drivers of muscle aging and contraction mode responses. RNA-sequencing data was generated from young (21±1 y) and older (70±1 y) human skeletal muscle before and following acute unilateral concentric and contralateral eccentric contractions. Application of weighted gene co-expression network analysis identified 33 distinct gene clusters ('modules') with an expression profile regulated by aging, contraction and/or linked to muscle strength. These included two contraction 'responsive' modules (related to 'cell adhesion' and 'transcription factor' processes) that also correlated with the magnitude of post-exercise muscle strength decline. Module searches for 'hub' genes and enriched transcription factor binding sites established a refined set of candidate module-regulatory molecules (536 hub genes and 60 transcription factors) as possible contributors to muscle aging and/or contraction responses. Thus, network-driven analysis can identify new molecular candidates of functional relevance to muscle aging and contraction mode adaptations.
Collapse
Affiliation(s)
- Craig R.G. Willis
- Department of Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter EX1 2LU, UK
| | - Ryan M. Ames
- Biosciences, University of Exeter, Exeter EX4 4QD, UK
| | - Colleen S. Deane
- Department of Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter EX1 2LU, UK
| | - Bethan E. Phillips
- MRC-ARUK Centre for Musculoskeletal aging Research and National Institute of Health Research, Biomedical Research Centre, Royal Derby Hospital Centre, School of Medicine, University of Nottingham, Derby DE22 3DT, UK
| | - Catherine L. Boereboom
- MRC-ARUK Centre for Musculoskeletal aging Research and National Institute of Health Research, Biomedical Research Centre, Royal Derby Hospital Centre, School of Medicine, University of Nottingham, Derby DE22 3DT, UK
| | - Haitham Abdulla
- MRC-ARUK Centre for Musculoskeletal aging Research and National Institute of Health Research, Biomedical Research Centre, Royal Derby Hospital Centre, School of Medicine, University of Nottingham, Derby DE22 3DT, UK
| | - Syed S.I. Bukhari
- MRC-ARUK Centre for Musculoskeletal aging Research and National Institute of Health Research, Biomedical Research Centre, Royal Derby Hospital Centre, School of Medicine, University of Nottingham, Derby DE22 3DT, UK
| | - Jonathan N. Lund
- Department of Surgery, Postgraduate Entry Medical School, Royal Derby Hospital Centre, School of Medicine, University of Nottingham, Derby DE22 3DT, UK
| | - John P. Williams
- MRC-ARUK Centre for Musculoskeletal aging Research and National Institute of Health Research, Biomedical Research Centre, Royal Derby Hospital Centre, School of Medicine, University of Nottingham, Derby DE22 3DT, UK
- Department of Surgery, Postgraduate Entry Medical School, Royal Derby Hospital Centre, School of Medicine, University of Nottingham, Derby DE22 3DT, UK
| | - Daniel J. Wilkinson
- MRC-ARUK Centre for Musculoskeletal aging Research and National Institute of Health Research, Biomedical Research Centre, Royal Derby Hospital Centre, School of Medicine, University of Nottingham, Derby DE22 3DT, UK
| | - Kenneth Smith
- MRC-ARUK Centre for Musculoskeletal aging Research and National Institute of Health Research, Biomedical Research Centre, Royal Derby Hospital Centre, School of Medicine, University of Nottingham, Derby DE22 3DT, UK
| | - Fawzi Kadi
- School of Health Sciences, Örebro University, Örebro 70182, Sweden
| | - Nathaniel J. Szewczyk
- MRC-ARUK Centre for Musculoskeletal aging Research and National Institute of Health Research, Biomedical Research Centre, Royal Derby Hospital Centre, School of Medicine, University of Nottingham, Derby DE22 3DT, UK
| | - Philip J. Atherton
- MRC-ARUK Centre for Musculoskeletal aging Research and National Institute of Health Research, Biomedical Research Centre, Royal Derby Hospital Centre, School of Medicine, University of Nottingham, Derby DE22 3DT, UK
| | - Timothy Etheridge
- Department of Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter EX1 2LU, UK
| |
Collapse
|
30
|
Advances in the understanding of the role of type-H vessels in the pathogenesis of osteoporosis. Arch Osteoporos 2020; 15:5. [PMID: 31897773 DOI: 10.1007/s11657-019-0677-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 12/02/2019] [Indexed: 02/03/2023]
Abstract
BACKGROUND Angiogenesis in the bone and its role in bone metabolic plays a fundamental role in the pathology of osteoporosis. Type-H vessels have been reported to exhibit distinct morphological, molecular, and functional properties. This review is aimed to illustrate the relationship between type-H vessels in the bone and bone metabolism. METHODS This manuscript reviews the articles on in vitro and in vivo experiments concerning the topic of type-H vessels and osteoporosis, and other researches in the area published by the author within the time frame from 2014 to 2019. RESULTS Current literatures have demonstrated that age-related loss of type-H vessels plays a critical role in the pathogenesis of osteoporosis. Impaired bone mass can be reserved by enhancing the formation of type-H vessels. Activation of the Notch and Hif-1α signaling pathway in bone tissue and exogenous PDGF-BB treatment increase the number of type-H vessels, along with the restoration of bone mass. The effects of osteoblasts and low-intensity pulsed ultrasound (LIPUS) on type-H vessels remain to be further studied. CONCLUSIONS These studies support unique functions for type-H vessels in the bone that may enable new therapeutic approaches to osteoporosis.
Collapse
|
31
|
Deckelbaum RA, Lobov IB, Cheung E, Halasz G, Rajamani S, Lerner J, Tong C, Li Z, Boland P, Dominguez M, Hughes V, Yancopoulos GD, Murphy AJ, Thurston G, Cao J, Romano C, Gale NW. The potassium channel Kcne3 is a VEGFA-inducible gene selectively expressed by vascular endothelial tip cells. Angiogenesis 2019; 23:179-192. [PMID: 31754927 PMCID: PMC7160073 DOI: 10.1007/s10456-019-09696-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/02/2019] [Indexed: 12/22/2022]
Abstract
Angiogenesis is largely driven by motile endothelial tip-cells capable of invading avascular tissue domains and enabling new vessel formation. Highly responsive to Vascular Endothelial Growth-Factor-A (VEGFA), endothelial tip-cells also suppress angiogenic sprouting in adjacent stalk cells, and thus have been a primary therapeutic focus in addressing neovascular pathologies. Surprisingly, however, there remains a paucity of specific endothelial tip-cell markers. Here, we employ transcriptional profiling and a lacZ reporter allele to identify Kcne3 as an early and selective endothelial tip-cell marker in multiple angiogenic contexts. In development, Kcne3 expression initiates during early phases of angiogenesis (E9) and remains specific to endothelial tip-cells, often adjacent to regions expressing VEGFA. Consistently, Kcne3 activation is highly responsive to exogenous VEGFA but maintains tip-cell specificity throughout normal retinal angiogenesis. We also demonstrate endothelial tip-cell selectivity of Kcne3 in several injury and tumor models. Together, our data show that Kcne3 is a unique marker of sprouting angiogenic tip-cells and offers new opportunities for investigating and targeting this cell type.
Collapse
Affiliation(s)
- Ron A Deckelbaum
- Department of Pre-Therapeutic Target Discovery, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA.
| | | | - Eunice Cheung
- Department of Ophthalmology, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| | - Gabor Halasz
- Department of Bioinformatics, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| | - Saathyaki Rajamani
- Department of Pre-Therapeutic Target Discovery, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| | - Julia Lerner
- Department of Pre-Therapeutic Target Discovery, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| | - Chunxiang Tong
- Department of Pre-Therapeutic Target Discovery, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| | - Zhe Li
- Department of Oncology & Angiogenesis, Regeneron Pharmaceuticals Inc., Tarrytown, NY, 10591, USA
| | - Patricia Boland
- Department of Oncology & Angiogenesis, Regeneron Pharmaceuticals Inc., Tarrytown, NY, 10591, USA
| | - Melissa Dominguez
- Department of Pre-Therapeutic Target Discovery, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| | - Virginia Hughes
- Department of Pre-Therapeutic Target Discovery, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| | - George D Yancopoulos
- Department of Research, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| | - Andrew J Murphy
- Department of Research, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| | - Gavin Thurston
- Department of Oncology & Angiogenesis, Regeneron Pharmaceuticals Inc., Tarrytown, NY, 10591, USA
| | - Jingtai Cao
- Department of Ophthalmology, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| | - Carmelo Romano
- Department of Ophthalmology, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| | - Nicholas W Gale
- Department of Pre-Therapeutic Target Discovery, Regeneron Pharmaceuticals Inc, Tarrytown, NY, 10591, USA
| |
Collapse
|
32
|
Marín-Llera JC, Garciadiego-Cázares D, Chimal-Monroy J. Understanding the Cellular and Molecular Mechanisms That Control Early Cell Fate Decisions During Appendicular Skeletogenesis. Front Genet 2019; 10:977. [PMID: 31681419 PMCID: PMC6797607 DOI: 10.3389/fgene.2019.00977] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 09/13/2019] [Indexed: 12/02/2022] Open
Abstract
The formation of the vertebrate skeleton is orchestrated in time and space by a number of gene regulatory networks that specify and position all skeletal tissues. During embryonic development, bones have two distinct origins: bone tissue differentiates directly from mesenchymal progenitors, whereas most long bones arise from cartilaginous templates through a process known as endochondral ossification. Before endochondral bone development takes place, chondrocytes form a cartilage analgen that will be sequentially segmented to form joints; thus, in the cartilage template, either the cartilage maturation programme or the joint formation programme is activated. Once the cartilage differentiation programme starts, the growth plate begins to form. In contrast, when the joint formation programme is activated, a capsule begins to form that contains special articular cartilage and synovium to generate a functional joint. In this review, we will discuss the mechanisms controlling the earliest molecular events that regulate cell fate during skeletogenesis in long bones. We will explore the initial processes that lead to the recruitment of mesenchymal stem/progenitor cells, the commitment of chondrocyte lineages, and the formation of skeletal elements during morphogenesis. Thereafter, we will review the process of joint specification and joint morphogenesis. We will discuss the links between transcription factor activity, cell–cell interactions, cell–extracellular matrix interactions, growth factor signalling, and other molecular interactions that control mesenchymal stem/progenitor cell fate during embryonic skeletogenesis.
Collapse
Affiliation(s)
- Jessica Cristina Marín-Llera
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | | | - Jesús Chimal-Monroy
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| |
Collapse
|
33
|
Sivan U, De Angelis J, Kusumbe AP. Role of angiocrine signals in bone development, homeostasis and disease. Open Biol 2019; 9:190144. [PMID: 31575330 PMCID: PMC6833221 DOI: 10.1098/rsob.190144] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Skeletal vasculature plays a central role in the maintenance of microenvironments for osteogenesis and haematopoiesis. In addition to supplying oxygen and nutrients, vasculature provides a number of inductive factors termed as angiocrine signals. Blood vessels drive recruitment of osteoblast precursors and bone formation during development. Angiogenesis is indispensable for bone repair and regeneration. Dysregulation of the angiocrine crosstalk is a hallmark of ageing and pathobiological conditions in the skeletal system. The skeletal vascular bed is complex, heterogeneous and characterized by distinct capillary subtypes (type H and type L), which exhibit differential expression of angiocrine factors. Furthermore, distinct blood vessel subtypes with differential angiocrine profiles differentially regulate osteogenesis and haematopoiesis, and drive disease states in the skeletal system. This review provides an overview of the role of angiocrine signals in bone during homeostasis and disease.
Collapse
Affiliation(s)
- Unnikrishnan Sivan
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Jessica De Angelis
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| | - Anjali P Kusumbe
- The Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
| |
Collapse
|
34
|
Cordeiro IR, Kabashima K, Ochi H, Munakata K, Nishimori C, Laslo M, Hanken J, Tanaka M. Environmental Oxygen Exposure Allows for the Evolution of Interdigital Cell Death in Limb Patterning. Dev Cell 2019; 50:155-166.e4. [PMID: 31204171 DOI: 10.1016/j.devcel.2019.05.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 04/01/2019] [Accepted: 05/10/2019] [Indexed: 01/04/2023]
Abstract
Amphibians form fingers without webbing by differential growth between digital and interdigital regions. Amniotes, however, employ interdigital cell death (ICD), an additional mechanism that contributes to a greater variation of limb shapes. Here, we investigate the role of environmental oxygen in the evolution of ICD in tetrapods. While cell death is restricted to the limb margin in amphibians with aquatic tadpoles, Eleutherodactylus coqui, a frog with terrestrial-direct-developing eggs, has cell death in the interdigital region. Chicken requires sufficient oxygen and reactive oxygen species to induce cell death, with the oxygen tension profile itself being distinct between the limbs of chicken and Xenopus laevis frogs. Notably, increasing blood vessel density in X. laevis limbs, as well as incubating tadpoles under high oxygen levels, induces ICD. We propose that the oxygen available to terrestrial eggs was an ecological feature crucial for the evolution of ICD, made possible by conserved autopod-patterning mechanisms.
Collapse
Affiliation(s)
- Ingrid Rosenburg Cordeiro
- School of Life Science and Technology, Tokyo Institute of Technology, B-17, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Kaori Kabashima
- School of Life Science and Technology, Tokyo Institute of Technology, B-17, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Haruki Ochi
- Institute for Promotion of Medical Science Research, Faculty of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata, Yamagata 990-9585, Japan
| | - Keijiro Munakata
- School of Life Science and Technology, Tokyo Institute of Technology, B-17, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Chika Nishimori
- School of Life Science and Technology, Tokyo Institute of Technology, B-17, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan
| | - Mara Laslo
- Department of Organismic and Evolutionary Biology and Museum of Comparative Zoology, Harvard University, 26 Oxford Street, Cambridge, MA 02138, USA
| | - James Hanken
- Department of Organismic and Evolutionary Biology and Museum of Comparative Zoology, Harvard University, 26 Oxford Street, Cambridge, MA 02138, USA
| | - Mikiko Tanaka
- School of Life Science and Technology, Tokyo Institute of Technology, B-17, 4259 Nagatsuta-cho, Midori-ku, Yokohama 226-8501, Japan.
| |
Collapse
|
35
|
Rafipay A, Berg ALR, Erskine L, Vargesson N. Expression analysis of limb element markers during mouse embryonic development. Dev Dyn 2018; 247:1217-1226. [PMID: 30225906 PMCID: PMC6282987 DOI: 10.1002/dvdy.24671] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 08/13/2018] [Accepted: 08/29/2018] [Indexed: 12/18/2022] Open
Abstract
Background: While data regarding expression of limb element and tissue markers during normal mouse limb development exist, few studies show expression patterns in upper and lower limbs throughout key limb development stages. A comparison to normal developmental events is essential when analyzing development of the limb in mutant mice models. Results: Expression patterns of the joint marker Gdf5, tendon and ligament marker Scleraxis, early muscle marker MyoD1, and blood vessel marker Cadherin5 (Cdh5) are presented during the most active phases of embryonic mouse limb patterning. Anti‐neurofilament staining of developing nerves in the fore‐ and hindlimbs and cartilage formation and progression also are described. Conclusions: This study demonstrates and describes a range of key morphological markers and methods that together can be used to assess normal and abnormal limb development. Developmental Dynamics 247:1217–1226, 2018. © 2018 The Authors. Developmental Dynamics published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists Expression patterns of molecular markers throughout both fore‐ and hindlimb development ‐ which can be used to assess normal and abnormal development. Detailled description of innervation during fore‐ and hindlimb development confirming innervation first seen after limb patterning events have begun. Description of cartilage development and progression indicates alizarin red staining not seen until E15.5 in both fore‐ and hindlimbs. Hindlimb lags behind forelimb molecularly and morphologically until E14.5. Detailled description of methods used to study fore‐ and hindlimb development.
Collapse
Affiliation(s)
- Alexandra Rafipay
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen
| | - Amanda L R Berg
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen
| | - Lynda Erskine
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen
| | - Neil Vargesson
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen
| |
Collapse
|
36
|
Cohen-Tayar Y, Cohen H, Mitiagin Y, Abravanel Z, Levy C, Idelson M, Reubinoff B, Itzkovitz S, Raviv S, Kaestner KH, Blinder P, Elkon R, Ashery-Padan R. Pax6 regulation of Sox9 in the mouse retinal pigmented epithelium controls its timely differentiation and choroid vasculature development. Development 2018; 145:dev.163691. [PMID: 29986868 DOI: 10.1242/dev.163691] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 07/02/2018] [Indexed: 01/08/2023]
Abstract
The synchronized differentiation of neuronal and vascular tissues is crucial for normal organ development and function, although there is limited information about the mechanisms regulating the coordinated development of these tissues. The choroid vasculature of the eye serves as the main blood supply to the metabolically active photoreceptors, and develops together with the retinal pigmented epithelium (RPE). Here, we describe a novel regulatory relationship between the RPE transcription factors Pax6 and Sox9 that controls the timing of RPE differentiation and the adjacent choroid maturation. We used a novel machine learning algorithm tool to analyze high resolution imaging of the choroid in Pax6 and Sox9 conditional mutant mice. Additional unbiased transcriptomic analyses in mutant mice and RPE cells generated from human embryonic stem cells, as well as chromatin immunoprecipitation and high-throughput analyses, revealed secreted factors that are regulated by Pax6 and Sox9. These factors might be involved in choroid development and in the pathogenesis of the common blinding disease: age-related macular degeneration (AMD).
Collapse
Affiliation(s)
- Yamit Cohen-Tayar
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Hadar Cohen
- Department of Particle Physics, Raymond and Beverly Sackler School of Physics and Astronomy, Tel-Aviv University, Tel Aviv 69978, Israel
| | - Yulia Mitiagin
- Department of Neurobiology, Biochemistry and Biophysics school, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel Aviv 69978, Israel
| | - Zohar Abravanel
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Carmit Levy
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Masha Idelson
- The Hadassah Human Embryonic Stem Cell Research Center, The Goldyne Savad Institute of Gene Therapy and The Department of Obstetrics and Gynecology, Hadassah-Hebrew University Medical Center, 9112001 Jerusalem, Israel
| | - Benjamin Reubinoff
- The Hadassah Human Embryonic Stem Cell Research Center, The Goldyne Savad Institute of Gene Therapy and The Department of Obstetrics and Gynecology, Hadassah-Hebrew University Medical Center, 9112001 Jerusalem, Israel
| | - Shalev Itzkovitz
- Department of Molecular Cell Biology, Weizmann Institute of Science 76100, Rehovot, Israel
| | - Shaul Raviv
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Klaus H Kaestner
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, 12-126 Smilow Center for Translational Research, 3400 Civic Center Blvd, Philadelphia, PA 19104-6145, USA
| | - Pablo Blinder
- Department of Neurobiology, Biochemistry and Biophysics school, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel Aviv 69978, Israel.,Sagol School for Neuroscience, Tel-Aviv University, Tel Aviv 69978, Israel
| | - Ran Elkon
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv 69978, Israel .,Sagol School for Neuroscience, Tel-Aviv University, Tel Aviv 69978, Israel
| | - Ruth Ashery-Padan
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Sagol School of Neurosciences, Tel Aviv University, Tel Aviv 69978, Israel .,Sagol School for Neuroscience, Tel-Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
37
|
Liu CF, Angelozzi M, Haseeb A, Lefebvre V. SOX9 is dispensable for the initiation of epigenetic remodeling and the activation of marker genes at the onset of chondrogenesis. Development 2018; 145:dev164459. [PMID: 30021842 PMCID: PMC6078338 DOI: 10.1242/dev.164459] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 06/04/2018] [Indexed: 12/16/2022]
Abstract
SOX9 controls cell lineage fate and differentiation in major biological processes. It is known as a potent transcriptional activator of differentiation-specific genes, but its earliest targets and its contribution to priming chromatin for gene activation remain unknown. Here, we address this knowledge gap using chondrogenesis as a model system. By profiling the whole transcriptome and the whole epigenome of wild-type and Sox9-deficient mouse embryo limb buds, we uncover multiple structural and regulatory genes, including Fam101a, Myh14, Sema3c and Sema3d, as specific markers of precartilaginous condensation, and we provide evidence of their direct transactivation by SOX9. Intriguingly, we find that SOX9 helps remove epigenetic signatures of transcriptional repression and establish active-promoter and active-enhancer marks at precartilage- and cartilage-specific loci, but is not absolutely required to initiate these changes and activate transcription. Altogether, these findings widen our current knowledge of SOX9 targets in early chondrogenesis and call for new studies to identify the pioneer and transactivating factors that act upstream of or along with SOX9 to prompt chromatin remodeling and specific gene activation at the onset of chondrogenesis and other processes.
Collapse
Affiliation(s)
- Chia-Feng Liu
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Marco Angelozzi
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Abdul Haseeb
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Véronique Lefebvre
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| |
Collapse
|
38
|
Han Y, You X, Xing W, Zhang Z, Zou W. Paracrine and endocrine actions of bone-the functions of secretory proteins from osteoblasts, osteocytes, and osteoclasts. Bone Res 2018; 6:16. [PMID: 29844945 PMCID: PMC5967329 DOI: 10.1038/s41413-018-0019-6] [Citation(s) in RCA: 323] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 03/21/2018] [Accepted: 04/16/2018] [Indexed: 12/17/2022] Open
Abstract
The skeleton is a dynamic organ that is constantly remodeled. Proteins secreted from bone cells, namely osteoblasts, osteocytes, and osteoclasts exert regulation on osteoblastogenesis, osteclastogenesis, and angiogenesis in a paracrine manner. Osteoblasts secrete a range of different molecules including RANKL/OPG, M-CSF, SEMA3A, WNT5A, and WNT16 that regulate osteoclastogenesis. Osteoblasts also produce VEGFA that stimulates osteoblastogenesis and angiogenesis. Osteocytes produce sclerostin (SOST) that inhibits osteoblast differentiation and promotes osteoclast differentiation. Osteoclasts secrete factors including BMP6, CTHRC1, EFNB2, S1P, WNT10B, SEMA4D, and CT-1 that act on osteoblasts and osteocytes, and thereby influenceaA osteogenesis. Osteoclast precursors produce the angiogenic factor PDGF-BB to promote the formation of Type H vessels, which then stimulate osteoblastogenesis. Besides, the evidences over the past decades show that at least three hormones or "osteokines" from bone cells have endocrine functions. FGF23 is produced by osteoblasts and osteocytes and can regulate phosphate metabolism. Osteocalcin (OCN) secreted by osteoblasts regulates systemic glucose and energy metabolism, reproduction, and cognition. Lipocalin-2 (LCN2) is secreted by osteoblasts and can influence energy metabolism by suppressing appetite in the brain. We review the recent progresses in the paracrine and endocrine functions of the secretory proteins of osteoblasts, osteocytes, and osteoclasts, revealing connections of the skeleton with other tissues and providing added insights into the pathogenesis of degenerative diseases affecting multiple organs and the drug discovery process.
Collapse
Affiliation(s)
- Yujiao Han
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031 China
| | - Xiuling You
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031 China
| | - Wenhui Xing
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031 China
| | - Zhong Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031 China
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031 China
| |
Collapse
|
39
|
Arima Y, Hokimoto S, Tabata N, Nakagawa O, Oshima A, Matsumoto Y, Sato T, Mukunoki T, Otani J, Ishii M, Uchikawa M, Yamamoto E, Izumiya Y, Kaikita K, Ogawa H, Nishiyama K, Tsujita K. Evaluation of Collateral Source Characteristics With 3-Dimensional Analysis Using Micro-X-Ray Computed Tomography. J Am Heart Assoc 2018; 7:JAHA.117.007800. [PMID: 29572323 PMCID: PMC5907550 DOI: 10.1161/jaha.117.007800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Collateral arteries provide an alternative blood supply and protect tissues from ischemic damage in patients with peripheral artery disease. However, the mechanism of collateral artery development is difficult to validate. METHODS AND RESULTS Collateral arteries were visualized using micro-x-ray computed tomography. Developmental characteristics were assessed using confocal microscopy. We conducted a single-center, retrospective, observational study and assessed the dilatation of collateral arteries on ischemic sides. We quantified the vascular volume in both ischemic and nonischemic legs. A prominent increase in vascular volume was observed in the ischemic leg using a murine hind-limb ischemia model. We also performed qualitative assessment and confirmed that the inferior gluteal artery functioned as a major collateral source. Serial analysis of murine hind-limb vessel development revealed that the inferior gluteal artery was a remnant of the ischial artery, which emerged as a representative vessel on the dorsal side during hind-limb organogenesis. We retrospectively analyzed consecutive patients who were admitted for the diagnosis or treatment of peripheral artery disease. The diameter of the inferior gluteal artery on the ischemic side showed significant dilatation compared with that on the nonischemic side. CONCLUSIONS Our findings indicate that an embryonic remnant artery can become a collateral source under ischemic conditions. Flow enhancement in the inferior gluteal artery might become a novel therapeutic approach for patients with peripheral artery disease.
Collapse
Affiliation(s)
- Yuichiro Arima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan .,X-Earth Center, Faculty of Engineering, Kumamoto University, Kumamoto, Japan.,International Research Center for Medical Sciences, Kumamoto, Japan.,Department of Molecular Physiology, National Cerebral and Cardiovascular Research Center Research Institute, Suita, Osaka, Japan
| | - Seiji Hokimoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,X-Earth Center, Faculty of Engineering, Kumamoto University, Kumamoto, Japan.,Kyushu University of Nursing and Social Welfare, Kumamoto, Japan
| | - Noriaki Tabata
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Osamu Nakagawa
- Department of Molecular Physiology, National Cerebral and Cardiovascular Research Center Research Institute, Suita, Osaka, Japan
| | - Asahi Oshima
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yosuke Matsumoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takahiro Sato
- X-Earth Center, Faculty of Engineering, Kumamoto University, Kumamoto, Japan
| | - Toshifumi Mukunoki
- X-Earth Center, Faculty of Engineering, Kumamoto University, Kumamoto, Japan
| | - Jun Otani
- X-Earth Center, Faculty of Engineering, Kumamoto University, Kumamoto, Japan
| | - Masanobu Ishii
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Michie Uchikawa
- International Research Center for Medical Sciences, Kumamoto, Japan
| | - Eiichiro Yamamoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yasuhiro Izumiya
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Koichi Kaikita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hisao Ogawa
- National Cerebral and Cardiovascular Research Center Research Institute, Suita, Osaka, Japan
| | - Koichi Nishiyama
- International Research Center for Medical Sciences, Kumamoto, Japan
| | - Kenichi Tsujita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
40
|
Cheng L, Chen T, Tu Q, Li H, Feng Z, Li Z, Lin D. Naringin improves random skin flap survival in rats. Oncotarget 2017; 8:94142-94150. [PMID: 29212216 PMCID: PMC5706862 DOI: 10.18632/oncotarget.21589] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 09/18/2017] [Indexed: 12/29/2022] Open
Abstract
Background Random-pattern flap transfer is commonly used to treat soft-tissue defects. However, flap necrosis remains a serious problem. Naringin accelerates angiogenesis by activating the expression of vascular endothelial growth factor (VEGF). In the present study, we investigated whether naringin improves the survival of random skin flaps. Results Compared with controls, the naringin-treated groups exhibited significantly larger mean areas of flap survival, significantly increased SOD activity and VEGF expression, and significantly reduced MDA level. Hematoxylin and eosin (HE) staining revealed that naringin promoted angiogenesis and inhibited inflammation. Materials and Methods “McFarlane flap” models were established in 90 male Sprague-Dawley (SD) rats divided into three groups: a 40 mg/kg control group (0.5 % sodium carboxymethylcellulose), a 40 mg/kg naringin-treated group, and an 80 mg/kg naringin-treated group. The extent of necrosis was measured 7 days later, and tissue samples were subjected to histological analysis. Angiogenesis was evaluated via lead oxide–gelatin angiography, immunohistochemistry, and laser Doppler imaging. Inflammation was evaluated by measurement of serum TNF-α (tumor necrosis factor-α) and IL-6 (interleukin-6) levels. Oxidative stress was assessed by measuring superoxide dismutase (SOD) activity and the malondialdehyde (MDA) level. Conclusion Naringin improved random skin flap survival.
Collapse
Affiliation(s)
- Liang Cheng
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Tingxiang Chen
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qiming Tu
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hang Li
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhenghua Feng
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhijie Li
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Dingsheng Lin
- Department of Hand and Plastic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
41
|
Developmental expression of membrane type 4-matrix metalloproteinase (Mt4-mmp/Mmp17) in the mouse embryo. PLoS One 2017; 12:e0184767. [PMID: 28926609 PMCID: PMC5604975 DOI: 10.1371/journal.pone.0184767] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 08/30/2017] [Indexed: 11/19/2022] Open
Abstract
Matrix metalloproteinases (MMPs) constitute a large group of endoproteases that play important functions during embryonic development, tumor metastasis and angiogenesis by degrading components of the extracellular matrix. Within this family, we focused our study on Mt4-mmp (also called Mmp17) that belongs to a distinct subset that is anchored to the cell surface via a glycosylphosphatidylinositol (GPI) moiety and with the catalytic site exposed to the extracellular space. Information about its function and substrates is very limited to date, and little has been reported on its role in the developing embryo. Here, we report a detailed expression analysis of Mt4-mmp during mouse embryonic development by using a LacZ reporter transgenic mouse line. We showed that Mt4-mmp is detected from early stages of development to postnatal stages following a dynamic and restricted pattern of expression. Mt4-mmp was first detected at E8.5 limited to the intersomitic vascularization, the endocardial endothelium and the dorsal aorta. Mt4-mmpLacZ/+ cells were also observed in the neural crest cells, somites, floor plate and notochord at early stages. From E10.5, expression localized in the limb buds and persists during limb development. A strong expression in the brain begins at E12.5 and continues to postnatal stages. Specifically, staining was observed in the olfactory bulb, cerebral cortex, hippocampus, striatum, septum, dorsal thalamus and the spinal cord. In addition, LacZ-positive cells were also detected during eye development, initially at the hyaloid artery and later on located in the lens and the neural retina. Mt4-mmp expression was confirmed by quantitative RT-PCR and western blot analysis in some embryonic tissues. Our data point to distinct functions for this metalloproteinase during embryonic development, particularly during brain formation, angiogenesis and limb development.
Collapse
|
42
|
Abstract
In addition to their conventional role as a conduit system for gases, nutrients, waste products or cells, blood vessels in the skeletal system play active roles in controlling multiple aspects of bone formation and provide niches for hematopoietic stem cells that reside within the bone marrow. In addition, recent studies have highlighted roles for blood vessels during bone healing. Here, we provide an overview of the architecture of the bone vasculature and discuss how blood vessels form within bone, how their formation is modulated, and how they function during development and fracture repair.
Collapse
Affiliation(s)
- Kishor K Sivaraj
- Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis and University of Münster, Faculty of Medicine, Münster D-48149, Germany
| | - Ralf H Adams
- Max-Planck-Institute for Molecular Biomedicine, Department of Tissue Morphogenesis and University of Münster, Faculty of Medicine, Münster D-48149, Germany
| |
Collapse
|
43
|
Bragdon B, Lam S, Aly S, Femia A, Clark A, Hussein A, Morgan EF, Gerstenfeld LC. Earliest phases of chondrogenesis are dependent upon angiogenesis during ectopic bone formation in mice. Bone 2017; 101:49-61. [PMID: 28412469 PMCID: PMC5500242 DOI: 10.1016/j.bone.2017.04.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 04/09/2017] [Accepted: 04/11/2017] [Indexed: 01/12/2023]
Abstract
Endochondral ossification is the process where cartilage forms prior to ossification and in which new bone forms during both fracture healing and ectopic bone formation. Transitioning to ossification is a highly coordinated process between hypertrophic chondrocytes, vascular endothelial cells, osteoblasts and osteoclasts. A critical biological process that is central to the interactions of these various cell types is angiogenesis. Although it is well established that angiogenesis is crucial for fracture repair, less is known pertaining to the role of angiogenesis in ectopic bone formation. Furthermore, fracture repair models are complicated by extensive trauma, subsequent inflammatory responses and concurrent repair processes in multiple tissues. In order to more definitively characterize the relationship between angiogenesis and postnatal endochondral ossification, a model of ectopic bone formation was used. Human demineralized bone matrix (DBM) was implanted in immune-deficient mice (rag null (B6.129S7-Rag1tm1/MOM/J)) to induce ectopic bone. Inhibition of angiogenesis with either a small molecule (TNP-470) or a targeted biological (Vascular Endothelial Growth Factor Receptor type 2 [VEGFR2] blocking antibody) prevented ectopic bone formation by 83% and 77%, respectively. Most striking was that the progression of chondrogenesis was halted during very early phases of chondrocyte differentiation between condensation and prehypertrophy (TNP-470) or the proliferative phase (VEGFR2 blockade) prior to hypertrophy, while osteoclast recruitment and resorption were almost completely inhibited. Our results demonstrate angiogenesis plays a developmental role in endochondral bone formation at a much earlier phase of chondrogenesis than suggested by prior findings.
Collapse
Affiliation(s)
- Beth Bragdon
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Stephanie Lam
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, MA 02118, USA
| | - Sherif Aly
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, MA 02118, USA
| | - Alexandra Femia
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, MA 02118, USA
| | - Abigail Clark
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, MA 02118, USA
| | - Amira Hussein
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, MA 02118, USA
| | - Elise F Morgan
- Department of Mechanical Engineering, Boston University College of Engineering, Boston, MA 02215, USA
| | - Louis C Gerstenfeld
- Department of Orthopaedic Surgery, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
44
|
Mansouri R, Jouan Y, Hay E, Blin-Wakkach C, Frain M, Ostertag A, Le Henaff C, Marty C, Geoffroy V, Marie PJ, Cohen-Solal M, Modrowski D. Osteoblastic heparan sulfate glycosaminoglycans control bone remodeling by regulating Wnt signaling and the crosstalk between bone surface and marrow cells. Cell Death Dis 2017; 8:e2902. [PMID: 28661485 PMCID: PMC5520938 DOI: 10.1038/cddis.2017.287] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 04/21/2017] [Accepted: 05/19/2017] [Indexed: 12/23/2022]
Abstract
Stimulating bone formation is an important challenge for bone anabolism in osteoporotic patients or to repair bone defects. The osteogenic properties of matrix glycosaminoglycans (GAGs) have been explored; however, the functions of GAGs at the surface of bone-forming cells are less documented. Syndecan-2 is a membrane heparan sulfate proteoglycan that is associated with osteoblastic differentiation. We used a transgenic mouse model with high syndecan-2 expression in osteoblasts to enrich the bone surface with cellular GAGs. Bone mass was increased in these transgenic mice. Syndecan-2 overexpression reduced the expression of receptor activator of NF-kB ligand (RANKL) in bone marrow cells and strongly inhibited bone resorption. Osteoblast activity was not modified in the transgenic mice, but bone formation was decreased in 4-month-old transgenic mice because of reduced osteoblast number. Increased proteoglycan expression at the bone surface resulted in decreased osteoblastic and osteoclastic precursors in bone marrow. Indeed, syndecan-2 overexpression increased apoptosis of mesenchymal precursors within the bone marrow. However, syndecan-2 specifically promoted the vasculature characterized by high expression of CD31 and Endomucin in 6-week-old transgenic mice, but this was reduced in 12-week-old transgenic mice. Finally, syndecan-2 functions as an inhibitor of Wnt-β-catenin–T-cell factor signaling pathway, activating glycogen synthase kinase 3 and then decreasing the Wnt-dependent production of Wnt ligands and R-spondin. In conclusion, our results show that GAG supply may improve osteogenesis, but also interfere with the crosstalk between the bone surface and marrow cells, altering the supporting function of osteoblasts.
Collapse
Affiliation(s)
- Rafik Mansouri
- Inserm UMR-1132, BIOSCAR, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Yohann Jouan
- Inserm UMR-1132, BIOSCAR, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Eric Hay
- Inserm UMR-1132, BIOSCAR, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Claudine Blin-Wakkach
- CNRS, UMR 7370, LP2M, Faculté de médecine, 28 avenue de Valombrose, Nice, France.,Université Nice Sophia Antipolis, Parc Valrose, Nice, France
| | | | - Agnès Ostertag
- Inserm UMR-1132, BIOSCAR, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | | | - Caroline Marty
- Inserm UMR-1132, BIOSCAR, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Valérie Geoffroy
- Inserm UMR-1132, BIOSCAR, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Pierre J Marie
- Inserm UMR-1132, BIOSCAR, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Martine Cohen-Solal
- Inserm UMR-1132, BIOSCAR, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Dominique Modrowski
- Inserm UMR-1132, BIOSCAR, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
45
|
Hu K, Olsen BR. Vascular endothelial growth factor control mechanisms in skeletal growth and repair. Dev Dyn 2017; 246:227-234. [PMID: 27750398 PMCID: PMC5354946 DOI: 10.1002/dvdy.24463] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 10/01/2016] [Indexed: 01/04/2023] Open
Abstract
Vascular endothelial growth factor A (VEGF) is a critical regulator of vascular development and postnatal angiogenesis and homeostasis, and it is essential for bone development and repair. Blood vessels serve both as structural templates for bone formation and they provide essential cells, growth factors and minerals needed for synthesis and mineralization, as well as turnover, of the extracellular matrix in bone. Through its regulation of angiogenesis, VEGF contributes to coupling of osteogenesis to angiogenesis, and it directly controls the differentiation and function of osteoblasts and osteoclasts. In this review, we summarize the properties of VEGF and its receptors that are relevant to bone formation and repair; the roles of VEGF during development of endochondral and membranous bones; and the contributions of VEGF to bone healing during different phases of bone repair. Finally, we discuss contributions of altered VEGF function in inherited disorders with bone defects as part of their phenotypes, and we speculate on what will be required before therapeutic strategies based on VEGF modulation can be developed for clinical use to treat patients with bone growth disorders and/or compromised bone repair. Developmental Dynamics 246:227-234, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Kai Hu
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Bjorn R Olsen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts
| |
Collapse
|
46
|
Cell-matrix signals specify bone endothelial cells during developmental osteogenesis. Nat Cell Biol 2017; 19:189-201. [PMID: 28218908 PMCID: PMC5580829 DOI: 10.1038/ncb3476] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 01/18/2017] [Indexed: 02/06/2023]
Abstract
Blood vessels in the mammalian skeletal system control bone formation and support haematopoiesis by generating local niche environments. While a specialized capillary subtype, termed type H, has been recently shown to couple angiogenesis and osteogenesis in adolescent, adult and ageing mice, little is known about the formation of specific endothelial cell populations during early developmental endochondral bone formation. Here, we report that embryonic and early postnatal long bone contains a specialized endothelial cell subtype, termed type E, which strongly supports osteoblast lineage cells and later gives rise to other endothelial cell subpopulations. The differentiation and functional properties of bone endothelial cells require cell-matrix signalling interactions. Loss of endothelial integrin β1 leads to endothelial cell differentiation defects and impaired postnatal bone growth, which is, in part, phenocopied by endothelial cell-specific laminin α5 mutants. Our work outlines fundamental principles of vessel formation and endothelial cell differentiation in the developing skeletal system.
Collapse
|
47
|
Naringin promotes fracture healing through stimulation of angiogenesis by regulating the VEGF/VEGFR-2 signaling pathway in osteoporotic rats. Chem Biol Interact 2017; 261:11-17. [DOI: 10.1016/j.cbi.2016.10.020] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 09/19/2016] [Accepted: 10/25/2016] [Indexed: 12/27/2022]
|
48
|
Ramasamy SK, Kusumbe AP, Schiller M, Zeuschner D, Bixel MG, Milia C, Gamrekelashvili J, Limbourg A, Medvinsky A, Santoro MM, Limbourg FP, Adams RH. Blood flow controls bone vascular function and osteogenesis. Nat Commun 2016; 7:13601. [PMID: 27922003 PMCID: PMC5150650 DOI: 10.1038/ncomms13601] [Citation(s) in RCA: 231] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 10/19/2016] [Indexed: 12/17/2022] Open
Abstract
While blood vessels play important roles in bone homeostasis and repair, fundamental aspects of vascular function in the skeletal system remain poorly understood. Here we show that the long bone vasculature generates a peculiar flow pattern, which is important for proper angiogenesis. Intravital imaging reveals that vessel growth in murine long bone involves the extension and anastomotic fusion of endothelial buds. Impaired blood flow leads to defective angiogenesis and osteogenesis, and downregulation of Notch signalling in endothelial cells. In aged mice, skeletal blood flow and endothelial Notch activity are also reduced leading to decreased angiogenesis and osteogenesis, which is reverted by genetic reactivation of Notch. Blood flow and angiogenesis in aged mice are also enhanced on administration of bisphosphonate, a class of drugs frequently used for the treatment of osteoporosis. We propose that blood flow and endothelial Notch signalling are key factors controlling ageing processes in the skeletal system. Formation of new blood vessels and bone is coupled. Here the authors show that blood flow represents a key regulator of angiogenesis and endothelial Notch signalling in the bone, and that reactivation of Notch signalling in the endothelium of aged mice rejuvenates the bone.
Collapse
Affiliation(s)
- Saravana K Ramasamy
- Faculty of Medicine, Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine and University of Münster, D-48149 Münster, Germany.,Research group Integrative Skeletal Physiology, Institute of Clinical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | - Anjali P Kusumbe
- Faculty of Medicine, Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine and University of Münster, D-48149 Münster, Germany.,Research group Tissue and Tumor Microenvironments, Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7LY, UK
| | - Maria Schiller
- Faculty of Medicine, Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine and University of Münster, D-48149 Münster, Germany
| | - Dagmar Zeuschner
- Electron Microscopy Unit, Max-Planck-Institute for Molecular Biomedicine, D-48149 Münster, Germany
| | - M Gabriele Bixel
- Faculty of Medicine, Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine and University of Münster, D-48149 Münster, Germany
| | - Carlo Milia
- VIB Vesalius Research Center, KU Leuven, 3000 Leuven, Belgium
| | - Jaba Gamrekelashvili
- Department of Nephrology and Hypertension, Hannover Medical School, D-30625 Hannover, Germany
| | - Anne Limbourg
- Department of Plastic and Reconstructive Surgery, Hannover Medical School, D-30625 Hannover, Germany
| | - Alexander Medvinsky
- Research group Ontogeny of Haematopoietic Stem Cells, MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, Scotland
| | - Massimo M Santoro
- VIB Vesalius Research Center, KU Leuven, 3000 Leuven, Belgium.,Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Florian P Limbourg
- Department of Nephrology and Hypertension, Hannover Medical School, D-30625 Hannover, Germany
| | - Ralf H Adams
- Faculty of Medicine, Department of Tissue Morphogenesis, Max-Planck-Institute for Molecular Biomedicine and University of Münster, D-48149 Münster, Germany
| |
Collapse
|
49
|
Uemura M, Nagasawa A, Terai K. Yap/Taz transcriptional activity in endothelial cells promotes intramembranous ossification via the BMP pathway. Sci Rep 2016; 6:27473. [PMID: 27273480 PMCID: PMC4895351 DOI: 10.1038/srep27473] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 05/19/2016] [Indexed: 12/19/2022] Open
Abstract
Osteogenesis is categorized into two groups based on developmental histology, intramembranous and endochondral ossification. The role of blood vessels during endochondral ossification is well known, while their role in intramembranous ossification, especially the intertissue pathway, is poorly understood. Here, we demonstrate endothelial Yap/Taz is a novel regulator of intramembranous ossification in zebrafish. Appropriate blood flow is required for Yap/Taz transcriptional activation in endothelial cells and intramembranous ossification. Additionally, Yap/Taz transcriptional activity in endothelial cells specifically promotes intramembranous ossification. BMP expression by Yap/Taz transactivation in endothelial cells is also identified as a bridging factor between blood vessels and intramembranous ossification. Furthermore, the expression of Runx2 in pre-osteoblast cells is a downstream target of Yap/Taz transcriptional activity in endothelial cells. Our results provide novel insight into the relationship between blood flow and ossification by demonstrating intertissue regulation.
Collapse
Affiliation(s)
- Mami Uemura
- Laboratory of Function and Morphology, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Yayoi 1-1-1 Bunkyo-ku, Tokyo, 113-0032 Japan
| | - Ayumi Nagasawa
- Laboratory of Function and Morphology, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Yayoi 1-1-1 Bunkyo-ku, Tokyo, 113-0032 Japan
| | - Kenta Terai
- Laboratory of Function and Morphology, Institute of Molecular and Cellular Biosciences, The University of Tokyo, Yayoi 1-1-1 Bunkyo-ku, Tokyo, 113-0032 Japan
| |
Collapse
|
50
|
Signaling pathways effecting crosstalk between cartilage and adjacent tissues: Seminars in cell and developmental biology: The biology and pathology of cartilage. Semin Cell Dev Biol 2016; 62:16-33. [PMID: 27180955 DOI: 10.1016/j.semcdb.2016.05.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 05/07/2016] [Indexed: 12/14/2022]
Abstract
Endochondral ossification, the mechanism responsible for the development of the long bones, is dependent on an extremely stringent coordination between the processes of chondrocyte maturation in the growth plate, vascular expansion in the surrounding tissues, and osteoblast differentiation and osteogenesis in the perichondrium and the developing bone center. The synchronization of these processes occurring in adjacent tissues is regulated through vigorous crosstalk between chondrocytes, endothelial cells and osteoblast lineage cells. Our knowledge about the molecular constituents of these bidirectional communications is undoubtedly incomplete, but certainly some signaling pathways effective in cartilage have been recognized to play key roles in steering vascularization and osteogenesis in the perichondrial tissues. These include hypoxia-driven signaling pathways, governed by the hypoxia-inducible factors (HIFs) and vascular endothelial growth factor (VEGF), which are absolutely essential for the survival and functioning of chondrocytes in the avascular growth plate, at least in part by regulating the oxygenation of developing cartilage through the stimulation of angiogenesis in the surrounding tissues. A second coordinating signal emanating from cartilage and regulating developmental processes in the adjacent perichondrium is Indian Hedgehog (IHH). IHH, produced by pre-hypertrophic and early hypertrophic chondrocytes in the growth plate, induces the differentiation of adjacent perichondrial progenitor cells into osteoblasts, thereby harmonizing the site and time of bone formation with the developmental progression of chondrogenesis. Both signaling pathways represent vital mediators of the tightly organized conversion of avascular cartilage into vascularized and mineralized bone during endochondral ossification.
Collapse
|