1
|
Midekssa FS, Davidson CD, Wieger ME, Kamen JL, Hanna KM, Jayco DKP, Hu MM, Friend NE, Putnam AJ, Helms AS, Shikanov A, Baker BM. Semi-synthetic fibrous fibrin composites promote 3D microvascular assembly, survival, and host integration of endothelial cells without mesenchymal cell support. Bioact Mater 2025; 49:652-669. [PMID: 40235652 PMCID: PMC11999628 DOI: 10.1016/j.bioactmat.2025.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 01/14/2025] [Accepted: 02/19/2025] [Indexed: 04/17/2025] Open
Abstract
Vasculogenic assembly of 3D capillary networks remains a promising approach to vascularizing tissue-engineered grafts, a significant outstanding challenge in tissue engineering and regenerative medicine. Current approaches for vasculogenic assembly rely on the inclusion of supporting mesenchymal cells alongside endothelial cells, co-encapsulated within vasculo-conducive materials such as low-density fibrin hydrogels. Here, we established a material-based approach to circumvent the need for supporting mesenchymal cells and report that the inclusion of synthetic matrix fibers in dense (>3 mg mL-1) 3D fibrin hydrogels can enhance vasculogenic assembly in endothelial cell monocultures. Surprisingly, we found that the addition of non-cell-adhesive synthetic matrix fibers compared to cell-adhesive synthetic fibers best encouraged vasculogenic assembly, proliferation, lumenogenesis, a vasculogenic transcriptional program, and additionally promoted cell-matrix interactions and intercellular force transmission. Implanting fiber-reinforced prevascularized constructs to assess graft-host vascular integration, we demonstrate additive effects of enhanced vascular network assembly during in vitro pre-culture, fiber-mediated improvements in endothelial cell survival and vascular maintenance post-implantation, and enhanced host cell infiltration that collectively enabled graft vessel integration with host circulation. This work establishes synthetic matrix fibers as an inexpensive alternative to sourcing and expanding secondary supporting cell types for the prevascularization of tissue constructs.
Collapse
Affiliation(s)
- Firaol S. Midekssa
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Christopher D. Davidson
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Megan E. Wieger
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Jordan L. Kamen
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Kaylin M. Hanna
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Danica Kristen P. Jayco
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Michael M. Hu
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Nicole E. Friend
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Andrew J. Putnam
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| | - Adam S. Helms
- Division of Cardiovascular Medicine, University of Michigan Ann Arbor, MI 48109, United States
| | - Ariella Shikanov
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, 48109, United States
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Brendon M. Baker
- Department of Biomedical Engineering, University of Michigan Ann Arbor, MI, 48109, United States
| |
Collapse
|
2
|
Lang A, Eastburn EA, Younesi M, Nijsure MP, Siciliano C, Pranatharthi Haran A, Panebianco CJ, Seidl E, Tang R, Alsberg E, Willett NJ, Gottardi R, Huh D, Boerckel JD. CYR61 delivery promotes angiogenesis during bone fracture repair. NPJ Regen Med 2025; 10:20. [PMID: 40263309 PMCID: PMC12015299 DOI: 10.1038/s41536-025-00398-y] [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: 05/09/2024] [Accepted: 01/31/2025] [Indexed: 04/24/2025] Open
Abstract
Compromised vascular supply and insufficient neovascularization impede bone repair, increasing risk of non-union. CYR61, Cysteine-rich angiogenic inducer of 61kD (also known as CCN1), is a matricellular growth factor that has been implicated in fracture repair. Here, we map the distribution of endogenous CYR61 during bone repair and evaluate the effects of recombinant CYR61 delivery on vascularized bone regeneration. In vitro, CYR61 treatment did not alter chondrogenesis or osteogenic gene expression, but significantly enhanced angiogenesis. In a mouse femoral fracture model, CYR61 delivery did not alter cartilage or bone formation, but accelerated neovascularization during fracture repair. Early initiation of ambulatory mechanical loading disrupted CYR61-induced neovascularization. Together, these data indicate that CYR61 delivery can enhance angiogenesis during bone repair, particularly for fractures with stable fixation, and may have therapeutic potential for fractures with limited blood vessel supply.
Collapse
Affiliation(s)
- Annemarie Lang
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Emily A Eastburn
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Mousa Younesi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Madhura P Nijsure
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Carly Siciliano
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Annapurna Pranatharthi Haran
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Elizabeth Seidl
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Rui Tang
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Eben Alsberg
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, USA
- Jesse Brown Veterans Affairs Medical Center (JBVAMC), Chicago, IL, USA
| | - Nick J Willett
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, USA
- The Veterans Affairs Portland Health Care System, Portland, OR, USA
| | - Riccardo Gottardi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Joel D Boerckel
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Akan I, Bacaksiz T, Maden M, Avci A, Tuncez M, Kazimoglu C. Does preoperative muscle biopsy predict the outcome of lower extremity amputation in diabetic patients? a prospective observational study. BMC Musculoskelet Disord 2025; 26:208. [PMID: 40022079 PMCID: PMC11869632 DOI: 10.1186/s12891-025-08448-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 02/18/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Determining the most appropriate level of amputation in patients with diabetes mellitus has not been well established. The purpose of this study is to determine whether muscle biopsy reveals predictive information about the success rate of patients undergoing diabetic major lower limb amputation. METHODS A prospective observational study was conducted among diabetic patients who underwent below-knee amputation. Skin-subcutaneous and muscle biopsy samples were obtained during the operation from 62 patients who undergo major limb amputation. Depending on the complications after surgery, patients were assigned into three groups: Group 1 consisted of patients with adequate wound healing without any complications; Group 2 included patients with prolonged wound healing requiring additional interventions like debridement; and Group 3 consisted of patients who underwent reamputation at a more proximal level. Biopsy samples of the groups were compared regarding degenerative cells, inflammatory cells, and the presence of infection. RESULTS There was a significant difference between Groups 1 and 3 regarding the presence of abscess formation and infection (p < 001). Comparison of Groups 1 and 3 revealed significant differences regarding inflammatory cell count, respectively (p < 001). According to the results of the ROC analysis performed for histopathologic cellular evaluation, 15% for inflammatory cell ratio in muscle samples and 25% for degenerative cell ratio both in muscle and skin samples were determined as cut-off values. CONCLUSIONS The presence of increased degenerative cell count and infection in muscle biopsy areassociated with higher rates of reoperation. The present study revealed that preoperative muscle biopsy has predictive value in patients undergoing major limb amputation. LEVEL OF EVIDENCE Level II, Prospective observational study.
Collapse
Affiliation(s)
- Ihsan Akan
- Department of Orthopaedics and Traumatology, Izmir Katip Celebi University, Izmir, Turkey.
| | - Tayfun Bacaksiz
- Department of Orthopaedics and Traumatology, Izmir Katip Celebi University, Izmir, Turkey
| | - Mehmet Maden
- Department of Orthopaedics and Traumatology, Izmir Ataturk Training and Research Hospital, Izmir, Turkey
| | - Arzu Avci
- Department of Pathology, Izmir Tınaztepe University, Izmir, Turkey
| | - Mahmut Tuncez
- Department of Orthopaedics and Traumatology, Izmir Ataturk Training and Research Hospital, Izmir, Turkey
| | - Cemal Kazimoglu
- Department of Orthopaedics and Traumatology, Izmir Katip Celebi University, Izmir, Turkey
| |
Collapse
|
4
|
Williams KE, Harrer JA, LaBelle SA, Leguineche K, Kaiser J, Karipott S, Lin A, Vongphachanh A, Fulton T, Walker Rosenthal J, Muhib F, Ong KG, Weiss JA, Willett NJ, Guldberg RE. Early resistance rehabilitation improves functional regeneration following segmental bone defect injury. NPJ Regen Med 2024; 9:38. [PMID: 39668145 PMCID: PMC11638264 DOI: 10.1038/s41536-024-00377-9] [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: 08/04/2023] [Accepted: 11/01/2024] [Indexed: 12/14/2024] Open
Abstract
Many studies have explored different loading and rehabilitation strategies, yet rehabilitation intensity and its impact on the local strain environment and bone healing have largely not been investigated. This study combined implantable strain sensors and subject-specific finite element models in a 2 mm rodent segmental bone defect model. After injury animals were underwent high or low intensity rehabilitation. High intensity rehabilitation increased local strains within the regenerative niche by an average of 44% compared to the low intensity rehabilitation. Finite element modeling demonstrated that resistance rehabilitation significantly increased compressive strain by a factor of 2.0 at week 2 and 4.45 after 4 weeks of rehabilitation. Animals that underwent resistance running had the greatest bone volume and improved functional recovery with regenerated femurs that matched intact failure torque and torsional stiffness values. These results demonstrate the potential for early resistance rehabilitation to improve bone healing.
Collapse
Affiliation(s)
- Kylie E Williams
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR, 97403, USA
| | - Julia Andraca Harrer
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR, 97403, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Atlanta Veteran's Affairs Medical Center, Atlanta, GA, USA
| | - Steven A LaBelle
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 841123, USA
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT, 841126, USA
| | - Kelly Leguineche
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR, 97403, USA
| | - Jarred Kaiser
- Atlanta Veteran's Affairs Medical Center, Atlanta, GA, USA
- Emory University, Atlanta, GA, USA
| | - Salil Karipott
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR, 97403, USA
| | - Angela Lin
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR, 97403, USA
| | - Alyssa Vongphachanh
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR, 97403, USA
| | - Travis Fulton
- Atlanta Veteran's Affairs Medical Center, Atlanta, GA, USA
| | - J Walker Rosenthal
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR, 97403, USA
| | - Farhan Muhib
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 841123, USA
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT, 841126, USA
| | - Keat Ghee Ong
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR, 97403, USA
| | - Jeffrey A Weiss
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 841123, USA
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT, 841126, USA
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, 841123, USA
| | - Nick J Willett
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR, 97403, USA.
| | - Robert E Guldberg
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR, 97403, USA.
| |
Collapse
|
5
|
Li M, Wu H, Gao K, Wang Y, Hu J, Guo Z, Hu R, Zhang M, Pang X, Guo M, Liu Y, Zhao L, He W, Ding S, Li W, Cheng W. Smart Implantable Hydrogel for Large Segmental Bone Regeneration. Adv Healthc Mater 2024; 13:e2402916. [PMID: 39344873 DOI: 10.1002/adhm.202402916] [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: 08/06/2024] [Revised: 09/16/2024] [Indexed: 10/01/2024]
Abstract
Large segmental bone defects often lead to nonunion and dysfunction, posing a significant challenge for clinicians. Inspired by the intrinsic bone defect repair logic of "vascularization and then osteogenesis", this study originally reports a smart implantable hydrogel (PDS-DC) with high mechanical properties, controllable scaffold degradation, and timing drug release that can proactively match different bone healing cycles to efficiently promote bone regeneration. The main scaffold of PDS-DC consists of polyacrylamide, polydopamine, and silk fibroin, which endows it with superior interfacial adhesion, structural toughness, and mechanical stiffness. In particular, the adjustment of scaffold cross-linking agent mixing ratio can effectively regulate the in vivo degradation rate of PDS-DC and intelligently satisfy the requirements of different bone defect healing cycles. Ultimately, PDS hydrogel loaded with free desferrioxamine (DFO) and CaCO3 mineralized ZIF-90 loaded bone morphogenetic protein-2 (BMP-2) to stimulate efficient angiogenesis and osteogenesis. Notably, DFO is released rapidly by free diffusion, whereas BMP-2 is released slowly by pH-dependent layer-by-layer disintegration, resulting in a significant difference in release time, thus matching the intrinsic logic of bone defect repair. In vivo and in vitro results confirm that PDS-DC can effectively realize high-quality bone generation and intelligently regulate to adapt to different demands of bone defects.
Collapse
Affiliation(s)
- Menghan Li
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Haiping Wu
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, P. R. China
| | - Ke Gao
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Yubo Wang
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, 401147, P. R. China
| | - Jiaqi Hu
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, 401147, P. R. China
| | - Ziling Guo
- Department of Forensic Medicine, Faculty of Basic Medical Sciences, Chongqing Engineering Research Center for Criminal Investigation Technology, Chongqing Key Laboratory of Forensic Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Ruiwei Hu
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Mengxuan Zhang
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Xiaoxiao Pang
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, 401147, P. R. China
| | - Minghui Guo
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Yuanjie Liu
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Lina Zhao
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Wen He
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Shijia Ding
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Wenyang Li
- Stomatological Hospital of Chongqing Medical University, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing Medical University, Chongqing, 401147, P. R. China
| | - Wei Cheng
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| |
Collapse
|
6
|
Zheng Z, Chen X, Wang Y, Wen P, Duan Q, Zhang P, Shan L, Ni Z, Feng Y, Xue Y, Li X, Zhang L, Liu J. Self-Growing Hydrogel Bioadhesives for Chronic Wound Management. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2408538. [PMID: 39149779 DOI: 10.1002/adma.202408538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/21/2024] [Indexed: 08/17/2024]
Abstract
Hydrogel bioadhesives have emerged as a promising alternative to wound dressings for chronic wound management. However, many existing bioadhesives do not meet the functional requirements for efficient wound management through dynamically mechanical modulation, due to the reduced wound contractibility, frequent wound recurrence, incapability to actively adapt to external microenvironment variation, especially for those gradually-expanded chronic wounds. Here, a self-growing hydrogel bioadhesive (sGHB) patch that exhibits instant adhesion to biological tissues but also a gradual increase in mechanical strength and interfacial adhesive strength within a 120-h application is presented. The gradually increased mechanics of the sGHB patch could effectively mitigate the stress concentration at the wound edge, and also resist the wound expansion at various stages, thus mechanically contracting the chronic wounds in a programmable manner. The self-growing hydrogel patch demonstrated enhanced wound healing efficacy in a mouse diabetic wound model, by regulating the inflammatory response, promoting the faster re-epithelialization and angiogenesis through mechanical modulation. Such kind of self-growing hydrogel bioadhesives have potential clinical utility for a variety of wound management where dynamic mechanical modulation is indispensable.
Collapse
Affiliation(s)
- Ziman Zheng
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
- Engineering Research Center for Nanomaterials, Henan University, Kaifeng, 475004, China
| | - Xingmei Chen
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yafei Wang
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Ping Wen
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Qingfang Duan
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Pei Zhang
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Liangjie Shan
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Zhipeng Ni
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yinghui Feng
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yu Xue
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Xing Li
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Lin Zhang
- Engineering Research Center for Nanomaterials, Henan University, Kaifeng, 475004, China
| | - Ji Liu
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
- Shenzhen Key Laboratory of Intelligent Robotics and Flexible Manufacturing Systems, Southern University of Science and Technology, Shenzhen, 518055, China
| |
Collapse
|
7
|
Libby JR, Royce H, Walker SR, Li L. The role of extracellular matrix in angiogenesis: Beyond adhesion and structure. BIOMATERIALS AND BIOSYSTEMS 2024; 15:100097. [PMID: 39129826 PMCID: PMC11315062 DOI: 10.1016/j.bbiosy.2024.100097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 06/19/2024] [Accepted: 07/06/2024] [Indexed: 08/13/2024] Open
Abstract
While the extracellular matrix (ECM) has long been recognized for its structural contributions, anchoring cells for adhesion, providing mechanical support, and maintaining tissue integrity, recent efforts have elucidated its dynamic, reciprocal, and diverse properties on angiogenesis. The ECM modulates angiogenic signaling and mechanical transduction, influences the extent and degree of receptor activation, controls cellular behaviors, and serves as a reservoir for bioactive macromolecules. Collectively, these factors guide the formation, maturation, and stabilization of a functional vascular network. This review aims to shed light on the versatile roles of the ECM in angiogenesis, transcending its traditional functions as a mere structural material. We will explore its engagement and synergy in signaling modulation, interactions with various angiogenic factors, and highlight its importance in both health and disease. By capturing the essence of the ECM's diverse functionalities, we highlight the significance in the broader context of vascular biology, enabling the design of novel biomaterials to engineer vascularized tissues and their potential therapeutic implications.
Collapse
Affiliation(s)
- Jaxson R. Libby
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Haley Royce
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, NH, USA
| | - Sarah R. Walker
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Linqing Li
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, NH, USA
- Department of Chemistry, University of New Hampshire, Durham, NH, USA
| |
Collapse
|
8
|
Zhong J, Huang W, Ahmad R, Chen J, Wu C, Hu J, Zheng K, Swain MV, Li Q. A Soft-Tissue Driven Bone Remodeling Algorithm for Mandibular Residual Ridge Resorption Based on Patient CT Image Data. Adv Healthc Mater 2024; 13:e2400091. [PMID: 38722148 DOI: 10.1002/adhm.202400091] [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: 01/09/2024] [Revised: 05/04/2024] [Indexed: 09/03/2024]
Abstract
The role of the biomechanical stimulation generated from soft tissue has not been well quantified or separated from the self-regulated hard tissue remodeling governed by Wolff's Law. Prosthodontic overdentures, commonly used to restore masticatory functions, can cause localized ischemia and inflammation as they often compress patients' oral mucosa and impede local circulation. This biomechanical stimulus in mucosa is found to accelerate the self-regulated residual ridge resorption (RRR), posing ongoing clinical challenges. Based on the dedicated long-term clinical datasets, this work develops an in-silico framework with a combination of techniques, including advanced image post-processing, patient-specific finite element models and unsupervised machine learning Self-Organizing map algorithm, to identify the soft tissue induced RRR and quantitatively elucidate the governing relationship between the RRR and hydrostatic pressure in mucosa. The proposed governing equation has not only enabled a predictive simulation for RRR as showcased in this study, providing a biomechanical basis for optimizing prosthodontic treatments, but also extended the understanding of the mechanobiological responses in the soft-hard tissue interfaces and the role in bone remodeling.
Collapse
Affiliation(s)
- Jingxiao Zhong
- School of Aerospace, Mechanical and Mechatronic Engineering, the University of Sydney, Sydney, 2006, Australia
| | - Wenwei Huang
- School of Aerospace, Mechanical and Mechatronic Engineering, the University of Sydney, Sydney, 2006, Australia
| | - Rohana Ahmad
- Faculty of Dentistry and Integrative Pharmacogenomics Institute, Universiti Teknologi MARA, Selangor, 40450, Malaysia
| | - Junning Chen
- College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, EX4 4QJ, UK
| | - Chi Wu
- School of Aerospace, Mechanical and Mechatronic Engineering, the University of Sydney, Sydney, 2006, Australia
| | - Jingrui Hu
- College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, EX4 4QJ, UK
| | - Keke Zheng
- Institute for Mechanical, Process and Energy Engineering, School of Engineering and Physical Sciences, Heriot Watt University, Edinburgh, EH14 4AS, UK
| | - Michael V Swain
- School of Aerospace, Mechanical and Mechatronic Engineering, the University of Sydney, Sydney, 2006, Australia
| | - Qing Li
- School of Aerospace, Mechanical and Mechatronic Engineering, the University of Sydney, Sydney, 2006, Australia
| |
Collapse
|
9
|
Joyce JD, Moore GA, Goswami P, Harrell TL, Taylor TM, Hawks SA, Green JC, Jia M, Irwin MD, Leslie E, Duggal NK, Thompson CK, Bertke AS. SARS-CoV-2 Rapidly Infects Peripheral Sensory and Autonomic Neurons, Contributing to Central Nervous System Neuroinvasion before Viremia. Int J Mol Sci 2024; 25:8245. [PMID: 39125815 PMCID: PMC11311394 DOI: 10.3390/ijms25158245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Neurological symptoms associated with COVID-19, acute and long term, suggest SARS-CoV-2 affects both the peripheral and central nervous systems (PNS/CNS). Although studies have shown olfactory and hematogenous invasion into the CNS, coinciding with neuroinflammation, little attention has been paid to susceptibility of the PNS to infection or to its contribution to CNS invasion. Here we show that sensory and autonomic neurons in the PNS are susceptible to productive infection with SARS-CoV-2 and outline physiological and molecular mechanisms mediating neuroinvasion. Our infection of K18-hACE2 mice, wild-type mice, and golden Syrian hamsters, as well as primary peripheral sensory and autonomic neuronal cultures, show viral RNA, proteins, and infectious virus in PNS neurons, satellite glial cells, and functionally connected CNS tissues. Additionally, we demonstrate, in vitro, that neuropilin-1 facilitates SARS-CoV-2 neuronal entry. SARS-CoV-2 rapidly invades the PNS prior to viremia, establishes a productive infection in peripheral neurons, and results in sensory symptoms often reported by COVID-19 patients.
Collapse
Affiliation(s)
- Jonathan D. Joyce
- Translational Biology, Medicine, and Health, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA; (J.D.J.)
- Center for Emerging Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Greyson A. Moore
- Biomedical and Veterinary Science, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Poorna Goswami
- Translational Biology, Medicine, and Health, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA; (J.D.J.)
| | - Telvin L. Harrell
- Biomedical and Veterinary Science, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Tina M. Taylor
- Population Health Sciences, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Seth A. Hawks
- Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Jillian C. Green
- Biomedical and Veterinary Science, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Mo Jia
- Population Health Sciences, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Matthew D. Irwin
- Biomedical and Veterinary Science, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Emma Leslie
- Translational Biology, Medicine, and Health, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA; (J.D.J.)
| | - Nisha K. Duggal
- Center for Emerging Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
- Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Christopher K. Thompson
- School of Neuroscience, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| | - Andrea S. Bertke
- Center for Emerging Zoonotic and Arthropod-borne Pathogens, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
- Population Health Sciences, Virginia Maryland College of Veterinary Medicine, Virginia Polytechnic Institute & State University, Blacksburg, VA 24060, USA
| |
Collapse
|
10
|
Kang Y, Na J, Karima G, Amirthalingam S, Hwang NS, Kim HD. Mesenchymal Stem Cell Spheroids: A Promising Tool for Vascularized Tissue Regeneration. Tissue Eng Regen Med 2024; 21:673-693. [PMID: 38578424 PMCID: PMC11187036 DOI: 10.1007/s13770-024-00636-2] [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: 01/05/2024] [Revised: 02/29/2024] [Accepted: 03/05/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are undifferentiated cells that can differentiate into specific cell lineages when exposed to the right conditions. The ability of MSCs to differentiate into particular cells is considered very important in biological research and clinical applications. MSC spheroids are clusters of MSCs cultured in three dimensions, which play an important role in enhancing the proliferation and differentiation of MSCs. MSCs can also participate in vascular formation by differentiating into endothelial cells and secreting paracrine factors. Vascularization ability is essential in impaired tissue repair and function recovery. Therefore, the vascularization ability of MSCs, which enhances angiogenesis and accelerates tissue healing has made MSCs a promising tool for tissue regeneration. However, MSC spheroids are a relatively new research field, and more research is needed to understand their full potential. METHODS In this review, we highlight the importance of MSC spheroids' vascularization ability in tissue engineering and regenerative medicine while providing the current status of studies on the MSC spheroids' vascularization and suggesting potential future research directions for MSC spheroids. RESULTS Studies both in vivo and in vitro have demonstrated MSC spheroids' capacity to develop into endothelial cells and stimulate vasculogenesis. CONCLUSION MSC spheroids show potential to enhance vascularization ability in tissue regeneration. Yet, further research is required to comprehensively understand the relationship between MSC spheroids and vascularization mechanisms.
Collapse
Affiliation(s)
- Yoonjoo Kang
- Department of IT Convergence (Brain Korea Plus 21), Korea National University of Transportation, Chungju, 27469, Republic of Korea
| | - Jinwoo Na
- Department of Polymer Science and Engineering, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
| | - Gul Karima
- Department of Polymer Science and Engineering, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea
| | - Sivashanmugam Amirthalingam
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Engineering Research, Seoul National University, Seoul, 08826, Republic of Korea
| | - Nathaniel S Hwang
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Engineering Research, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hwan D Kim
- Department of IT Convergence (Brain Korea Plus 21), Korea National University of Transportation, Chungju, 27469, Republic of Korea.
- Department of Polymer Science and Engineering, Korea National University of Transportation, 50 Daehak-ro, Chungju, 27469, Republic of Korea.
- Department of Biomedical Engineering, Korea National University of Transportation, Chungju, 27469, Republic of Korea.
| |
Collapse
|
11
|
Liu Y, Wang Y, Lin M, Liu H, Pan Y, Wu J, Guo Z, Li J, Yan B, Zhou H, Fan Y, Hu G, Liang H, Zhang S, Siu MFF, Wu Y, Bai J, Liu C. Cellular Scale Curvature in Bioceramic Scaffolds Enhanced Bone Regeneration by Regulating Skeletal Stem Cells and Vascularization. Adv Healthc Mater 2024:e2401667. [PMID: 38923234 DOI: 10.1002/adhm.202401667] [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: 06/13/2024] [Revised: 06/20/2024] [Indexed: 06/28/2024]
Abstract
Critical-sized segmental bone defects cannot heal spontaneously, leading to disability and significant increase in mortality. However, current treatments utilizing bone grafts face a variety of challenges from donor availability to poor osseointegration. Drugs such as growth factors increase cancer risk and are very costly. Here, a porous bioceramic scaffold that promotes bone regeneration via solely mechanobiological design is reported. Two types of scaffolds with high versus low pore curvatures are created using high-precision 3D printing technology to fabricate pore curvatures radius in the 100s of micrometers. While both are able to support bone formation, the high-curvature pores induce higher ectopic bone formation and increased vessel invasion. Scaffolds with high-curvature pores also promote faster regeneration of critical-sized segmental bone defects by activating mechanosensitive pathways. High-curvature pore recruits skeletal stem cells and type H vessels from both the periosteum and the marrow during the early phase of repair. High-curvature pores have increased survival of transplanted GFP-labeled skeletal stem cells (SSCs) and recruit more host SSCs. Taken together, the bioceramic scaffolds with defined micrometer-scale pore curvatures demonstrate a mechanobiological approach for orthopedic scaffold design.
Collapse
Affiliation(s)
- Yang Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Yue Wang
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
| | - Minmin Lin
- Department of Biomedical Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Hongzhi Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Yonghao Pan
- Department of Biomedical Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Jianqun Wu
- College of Medicine, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
| | - Ziyu Guo
- Department of Biomedical Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Jiawei Li
- Department of Biomedical Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Bingtong Yan
- Department of Biomedical Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Hang Zhou
- Department of Biomedical Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Yuanhao Fan
- Department of Biomedical Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, 518055, P. R. China
| | - Ganqing Hu
- Department of Biomedical Engineering, Cornell University, Ithaca, NY, 14850, USA
| | - Haowen Liang
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
| | - Shibo Zhang
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
| | - Ming-Fung Francis Siu
- Department of Building and Real Estate, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, 999077, China
| | - Yongbo Wu
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
| | - Jiaming Bai
- Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
| | - Chao Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Nanshan District, Shenzhen, 518055, P. R. China
| |
Collapse
|
12
|
Lim J, Fang HW, Bupphathong S, Sung PC, Yeh CE, Huang W, Lin CH. The Edifice of Vasculature-On-Chips: A Focused Review on the Key Elements and Assembly of Angiogenesis Models. ACS Biomater Sci Eng 2024; 10:3548-3567. [PMID: 38712543 PMCID: PMC11167599 DOI: 10.1021/acsbiomaterials.3c01978] [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: 12/29/2023] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 05/08/2024]
Abstract
The conception of vascularized organ-on-a-chip models provides researchers with the ability to supply controlled biological and physical cues that simulate the in vivo dynamic microphysiological environment of native blood vessels. The intention of this niche research area is to improve our understanding of the role of the vasculature in health or disease progression in vitro by allowing researchers to monitor angiogenic responses and cell-cell or cell-matrix interactions in real time. This review offers a comprehensive overview of the essential elements, including cells, biomaterials, microenvironmental factors, microfluidic chip design, and standard validation procedures that currently govern angiogenesis-on-a-chip assemblies. In addition, we emphasize the importance of incorporating a microvasculature component into organ-on-chip devices in critical biomedical research areas, such as tissue engineering, drug discovery, and disease modeling. Ultimately, advances in this area of research could provide innovative solutions and a personalized approach to ongoing medical challenges.
Collapse
Affiliation(s)
- Joshua Lim
- Graduate
Institute of Nanomedicine and Medical Engineering, College of Biomedical
Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsu-Wei Fang
- High-value
Biomaterials Research and Commercialization Center, National Taipei University of Technology, Taipei 10608, Taiwan
- Department
of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei 10608, Taiwan
- Institute
of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Sasinan Bupphathong
- Graduate
Institute of Nanomedicine and Medical Engineering, College of Biomedical
Engineering, Taipei Medical University, Taipei 11031, Taiwan
- High-value
Biomaterials Research and Commercialization Center, National Taipei University of Technology, Taipei 10608, Taiwan
| | - Po-Chan Sung
- School
of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Chen-En Yeh
- School
of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Wei Huang
- Department
of Orthodontics, Rutgers School of Dental
Medicine, Newark, New Jersey 07103, United States
| | - Chih-Hsin Lin
- Graduate
Institute of Nanomedicine and Medical Engineering, College of Biomedical
Engineering, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
13
|
Zhang Z, Yao P, Fan S. Advances in regenerative rehabilitation in the rehabilitation of musculoskeletal injuries. Regen Med 2024; 19:345-354. [PMID: 38860852 PMCID: PMC11346529 DOI: 10.1080/17460751.2024.2357956] [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: 01/15/2024] [Accepted: 05/17/2024] [Indexed: 06/12/2024] Open
Abstract
In the rapidly advancing field of regenerative medicine, relying solely on cell transplantation alone may be insufficient for achieving functional recovery, and rehabilitation before and after transplantation is crucial. Regenerative rehabilitation functions by synergizing the therapeutic effects of regeneration and rehabilitation to maximize tissue regeneration and patient function. We used the keywords "regenerative rehabilitation" to search across the database for published works; this review discusses the development of regenerative rehabilitation for the treatment of musculoskeletal injuries. Rehabilitation has become a crucial component of regenerative medicine because it can enhance patients' functional activity and facilitate their early return to society. Experimental data increasingly demonstrates that rehabilitation interventions support the regeneration of transplanted tissues.
Collapse
Affiliation(s)
- Zirui Zhang
- University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8571, Japan
- Department of Rehabilitation Medicine, Chinese People's Liberation Army Joint Logistics Force 940 Hospital, 818, Anning East Road, Lanzhou, Gansu, 730000, PR China
| | - Pengfei Yao
- Department of Rehabilitation Medicine, Chinese People's Liberation Army Joint Logistics Force 940 Hospital, 818, Anning East Road, Lanzhou, Gansu, 730000, PR China
| | - Shuai Fan
- Department of Rehabilitation Medicine, The Ninth People's Hospital of Shanghai, Jiao Tong University, 639, Manufacturing Bureau Road, Huangpu District, Shanghai, 200000, PR China
| |
Collapse
|
14
|
Bala V, Patel V, Sewell-Loftin MK. Cadherin Expression Is Regulated by Mechanical Phenotypes of Fibroblasts in the Perivascular Matrix. Cells Tissues Organs 2024; 213:446-463. [PMID: 38768571 PMCID: PMC11576492 DOI: 10.1159/000539319] [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: 12/01/2023] [Accepted: 05/08/2024] [Indexed: 05/22/2024] Open
Abstract
INTRODUCTION The influence of mechanical forces generated by stromal cells in the perivascular matrix is thought to be a key regulator in controlling blood vessel growth. Cadherins are mechanosensors that facilitate and maintain cell-cell interactions and blood vessel integrity, but little is known about how stromal cells regulate cadherin signaling in the vasculature. Our objective was to investigate the relationship between mechanical phenotypes of stromal cells with cadherin expression in 3D tissue engineering models of vascular growth. METHODS Stromal cell lines were subjected to a bead displacement assay to track matrix distortions and characterize mechanical phenotypes in 3D microtissue models. These cells included human ventricular cardiac (NHCF), dermal (NHDF), lung (NHLF), breast cancer-associated (CAF), and normal breast fibroblasts (NBF). Cells were embedded in a fibrin matrix (10 mg/mL) with fluorescent tracker beads; images were collected every 30 min. We also studied endothelial cells (ECs) in co-culture with mechanically active or inactive stromal cells and quantified N-Cad, OB-Cad, and VE-Cad expression using immunofluorescence. RESULTS Bead displacement studies identified mechanically active stromal cells (CAFs, NHCFs, NHDFs) that generate matrix distortions and mechanically inactive cells (NHLFs, NBFs). CAFs, NHCFs, and NHDFs displaced the matrix with an average magnitude of 3.17 ± 0.11 μm, 3.13 ± 0.06 μm, and 2.76 ± 0.05 μm, respectively, while NHLFs and NBFs displaced the matrix with an average of 1.82 ± 0.05 μm and 2.66 ± 0.06 μm in fibrin gels. Compared to ECs only, CAFs + ECs as well as NBFs + ECs in 3D co-culture significantly decreased expression of VE-Cad; in addition, Pearson's Correlation Coefficient for N-Cad and VE-Cad showed a strong correlation (>0.7), suggesting cadherin colocalization. Using a microtissue model, we demonstrated that mechanical phenotypes associated with increased matrix deformations correspond to enhanced angiogenic growth. The results could suggest a mechanism to control tight junction regulation in developing vascular beds for tissue engineering scaffolds or understanding vascular growth during developmental processes. CONCLUSION Our studies provide novel data for how mechanical phenotype of stromal cells in combination with secreted factor profiles is related to cadherin regulation, localization, and vascularization potential in 3D microtissue models.
Collapse
Affiliation(s)
- Vaishali Bala
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Vidhi Patel
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Mary Kathryn Sewell-Loftin
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham AL 35233
| |
Collapse
|
15
|
Guerra A, Belinha J, Salgado C, Monteiro FJ, Natal Jorge R. Computational Insights into the Interplay of Mechanical Forces in Angiogenesis. Biomedicines 2024; 12:1045. [PMID: 38791007 PMCID: PMC11117778 DOI: 10.3390/biomedicines12051045] [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: 03/18/2024] [Revised: 04/15/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
This study employs a meshless computational model to investigate the impacts of compression and traction on angiogenesis, exploring their effects on vascular endothelial growth factor (VEGF) diffusion and subsequent capillary network formation. Three distinct initial domain geometries were defined to simulate variations in endothelial cell sprouting and VEGF release. Compression and traction were applied, and the ensuing effects on VEGF diffusion coefficients were analysed. Compression promoted angiogenesis, increasing capillary network density. The reduction in the VEGF diffusion coefficient under compression altered VEGF concentration, impacting endothelial cell migration patterns. The findings were consistent across diverse simulation scenarios, demonstrating the robust influence of compression on angiogenesis. This computational study enhances our understanding of the intricate interplay between mechanical forces and angiogenesis. Compression emerges as an effective mediator of angiogenesis, influencing VEGF diffusion and vascular pattern. These insights may contribute to innovative therapeutic strategies for angiogenesis-related disorders, fostering tissue regeneration and addressing diseases where angiogenesis is crucial.
Collapse
Affiliation(s)
- Ana Guerra
- INEGI—Instituto de Ciência e Inovação em Engenharia Mecânica e Engenharia Industrial, 4200-465 Porto, Portugal
| | - Jorge Belinha
- ISEP—Instituto Superior de Engenharia do Porto, Departamento de Engenharia Mecânica, Politécnico do Porto, Rua Dr. António Bernardino de Almeida, 431, 4249-015 Porto, Portugal;
| | - Christiane Salgado
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (C.S.); (F.J.M.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Fernando Jorge Monteiro
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (C.S.); (F.J.M.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Renato Natal Jorge
- LAETA—Laboratório Associado de Energia, Transportes e Aeronáutica, Universidade do Porto, 4200-165 Porto, Portugal;
- FEUP—Faculdade de Engenharia, Departamento de Engenharia Mecânica, Universidade do Porto, 4200-165 Porto, Portugal
| |
Collapse
|
16
|
Kou Z, Liu C, Zhang W, Sun C, Liu L, Zhang Q. Heterogeneity of primary and metastatic CAFs: From differential treatment outcomes to treatment opportunities (Review). Int J Oncol 2024; 64:54. [PMID: 38577950 PMCID: PMC11015919 DOI: 10.3892/ijo.2024.5642] [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: 11/09/2023] [Accepted: 03/13/2024] [Indexed: 04/06/2024] Open
Abstract
Compared with primary tumor sites, metastatic sites appear more resistant to treatments and respond differently to the treatment regimen. It may be due to the heterogeneity in the microenvironment between metastatic sites and primary tumors. Cancer‑associated fibroblasts (CAFs) are widely present in the tumor stroma as key components of the tumor microenvironment. Primary tumor CAFs (pCAFs) and metastatic CAFs (mCAFs) are heterogeneous in terms of source, activation mode, markers and functional phenotypes. They can shape the tumor microenvironment according to organ, showing heterogeneity between primary tumors and metastases, which may affect the sensitivity of these sites to treatment. It was hypothesized that understanding the heterogeneity between pCAFs and mCAFs can provide a glimpse into the difference in treatment outcomes, providing new ideas for improving the rate of metastasis control in various cancers.
Collapse
Affiliation(s)
- Zixing Kou
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
| | - Cun Liu
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Wenfeng Zhang
- State Key Laboratory of Quality Research in Chinese Medicine and Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa Island 999078, Macau SAR, P.R. China
| | - Changgang Sun
- College of Traditional Chinese Medicine, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 621000, P.R. China
| | - Lijuan Liu
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, Shandong 621000, P.R. China
| | - Qiming Zhang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, P.R. China
- Department of Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100007, P.R. China
| |
Collapse
|
17
|
Lang A, Eastburn EA, Younesi M, Nijsure M, Siciliano C, Haran AP, Panebianco CJ, Seidl E, Tang R, Alsberg E, Willett NJ, Gottardi R, Huh D, Boerckel JD. Cyr61 delivery promotes angiogenesis during bone fracture repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.05.588239. [PMID: 38617208 PMCID: PMC11014620 DOI: 10.1101/2024.04.05.588239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Compromised vascular supply and insufficient neovascularization impede bone repair, increasing risk of non-union. Cyr61, Cysteine-rich angiogenic inducer of 61kD (also known as CCN1), is a matricellular growth factor that is regulated by mechanical cues during fracture repair. Here, we map the distribution of endogenous Cyr61 during bone repair and evaluate the effects of recombinant Cyr61 delivery on vascularized bone regeneration. In vitro, Cyr61 treatment did not alter chondrogenesis or osteogenic gene expression, but significantly enhanced angiogenesis. In a mouse femoral fracture model, Cyr61 delivery did not alter cartilage or bone formation, but accelerated neovascularization during fracture repair. Early initiation of ambulatory mechanical loading disrupted Cyr61-induced neovascularization. Together, these data indicate that Cyr61 delivery can enhance angiogenesis during bone repair, particularly for fractures with stable fixation, and may have therapeutic potential for fractures with limited blood vessel supply.
Collapse
Affiliation(s)
- Annemarie Lang
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, United States
| | - Emily A. Eastburn
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Mousa Younesi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Madhura Nijsure
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Carly Siciliano
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Annapurna Pranatharthi Haran
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | | | - Elizabeth Seidl
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Rui Tang
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, United States
| | - Eben Alsberg
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, United States
| | - Nick J. Willett
- Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States
- The Veterans Affairs Portland Health Care System, Portland, OR, United States
| | - Riccardo Gottardi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
- Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Dongeun Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Joel D. Boerckel
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, United States
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
18
|
Collins JM, Lang A, Parisi C, Moharrer Y, Nijsure MP, Thomas Kim JH, Ahmed S, Szeto GL, Qin L, Gottardi R, Dyment NA, Nowlan NC, Boerckel JD. YAP and TAZ couple osteoblast precursor mobilization to angiogenesis and mechanoregulation in murine bone development. Dev Cell 2024; 59:211-227.e5. [PMID: 38141609 PMCID: PMC10843704 DOI: 10.1016/j.devcel.2023.11.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/07/2023] [Accepted: 11/30/2023] [Indexed: 12/25/2023]
Abstract
Fetal bone development occurs through the conversion of avascular cartilage to vascularized bone at the growth plate. This requires coordinated mobilization of osteoblast precursors with blood vessels. In adult bone, vessel-adjacent osteoblast precursors are maintained by mechanical stimuli; however, the mechanisms by which these cells mobilize and respond to mechanical cues during embryonic development are unknown. Here, we show that the mechanoresponsive transcriptional regulators Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) spatially couple osteoblast precursor mobilization to angiogenesis, regulate vascular morphogenesis to control cartilage remodeling, and mediate mechanoregulation of embryonic murine osteogenesis. Mechanistically, YAP and TAZ regulate a subset of osteoblast-lineage cells, identified by single-cell RNA sequencing as vessel-associated osteoblast precursors, which regulate transcriptional programs that direct blood vessel invasion through collagen-integrin interactions and Cxcl12. Functionally, in 3D human cell co-culture, CXCL12 treatment rescues angiogenesis impaired by stromal cell YAP/TAZ depletion. Together, these data establish functions of the vessel-associated osteoblast precursors in bone development.
Collapse
Affiliation(s)
- Joseph M Collins
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Annemarie Lang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cristian Parisi
- Department of Bioengineering, Imperial College London, London, UK
| | - Yasaman Moharrer
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Department of Mechanical Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Madhura P Nijsure
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jong Hyun Thomas Kim
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Saima Ahmed
- Department of Bioengineering, Imperial College London, London, UK
| | | | - Ling Qin
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Riccardo Gottardi
- Department of Pediatrics, Division of Pulmonary Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nathaniel A Dyment
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Niamh C Nowlan
- Department of Bioengineering, Imperial College London, London, UK; School of Mechanical and Materials Engineering, University College Dublin, Dublin, Ireland; UCD Conway Institute, University College Dublin, Dublin, Ireland
| | - Joel D Boerckel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA; Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
19
|
Kendall JJ, Ledoux C, Marques FC, Boaretti D, Schulte FA, Morgan EF, Müller R. An in silico micro-multiphysics agent-based approach for simulating bone regeneration in a mouse femur defect model. Front Bioeng Biotechnol 2023; 11:1289127. [PMID: 38164405 PMCID: PMC10757951 DOI: 10.3389/fbioe.2023.1289127] [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: 09/05/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024] Open
Abstract
Bone defects represent a challenging clinical problem as they can lead to non-union. In silico models are well suited to study bone regeneration under varying conditions by linking both cellular and systems scales. This paper presents an in silico micro-multiphysics agent-based (micro-MPA) model for bone regeneration following an osteotomy. The model includes vasculature, bone, and immune cells, as well as their interaction with the local environment. The model was calibrated by time-lapsed micro-computed tomography data of femoral osteotomies in C57Bl/6J mice, and the differences between predicted bone volume fractions and the longitudinal in vivo measurements were quantitatively evaluated using root mean square error (RMSE). The model performed well in simulating bone regeneration across the osteotomy gap, with no difference (5.5% RMSE, p = 0.68) between the in silico and in vivo groups for the 5-week healing period - from the inflammatory phase to the remodelling phase - in the volume spanning the osteotomy gap. Overall, the proposed micro-MPA model was able to simulate the influence of the local mechanical environment on bone regeneration, and both this environment and cytokine concentrations were found to be key factors in promoting bone regeneration. Further, the validated model matched clinical observations that larger gap sizes correlate with worse healing outcomes and ultimately simulated non-union. This model could help design and guide future experimental studies in bone repair, by identifying which are the most critical in vivo experiments to perform.
Collapse
Affiliation(s)
- Jack J. Kendall
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
- Center for Multiscale and Translational Mechanobiology, Boston University, Boston, MA, United States
| | - Charles Ledoux
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | | | | | | | - Elise F. Morgan
- Center for Multiscale and Translational Mechanobiology, Boston University, Boston, MA, United States
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
20
|
Zhao LM, Da LC, Wang R, Wang L, Jiang YL, Zhang XZ, Li YX, Lei XX, Song YT, Zou CY, Huang LP, Zhang WQ, Zhang QY, Li QJ, Nie R, Zhang Y, Liang Y, Li-Ling J, Xie HQ. Promotion of uterine reconstruction by a tissue-engineered uterus with biomimetic structure and extracellular matrix microenvironment. SCIENCE ADVANCES 2023; 9:eadi6488. [PMID: 37967178 PMCID: PMC10651121 DOI: 10.1126/sciadv.adi6488] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 10/13/2023] [Indexed: 11/17/2023]
Abstract
The recurrence rate for severe intrauterine adhesions is as high as 60%, and there is still lack of effective prevention and treatment. Inspired by the nature of uterus, we have developed a bilayer scaffold (ECM-SPS) with biomimetic heterogeneous features and extracellular matrix (ECM) microenvironment of the uterus. As proved by subtotal uterine reconstruction experiments, the mechanical and antiadhesion properties of the bilayer scaffold could meet the requirement for uterine repair. With the modification with tissue-specific cell-derived ECM, the ECM-SPS had the ECM microenvironment signatures of both the endometrium and myometrium and exhibited the property of inducing stem cell-directed differentiation. Furthermore, the ECM-SPS has recruited more endogenous stem cells to promote endometrial regeneration at the initial stage of repair, which was accompanied by more smooth muscle regeneration and a higher pregnancy rate. The reconstructed uterus could also sustain normal pregnancy and live birth. The ECM-SPS may thereby provide a potential treatment for women with severe intrauterine adhesions.
Collapse
Affiliation(s)
- Long-mei Zhao
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan 610212, China
| | - Lin-cui Da
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Reproductive Center of Fujian Maternity and Child Health Care Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian 350001, China
| | - Rui Wang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Long Wang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yan-lin Jiang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiu-zhen Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ya-xing Li
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiong-xin Lei
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yu-ting Song
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chen-yu Zou
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Li-ping Huang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wen-qian Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qing-yi Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qian-jin Li
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Rong Nie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yi Zhang
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yan Liang
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jesse Li-Ling
- Center of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hui-qi Xie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
- Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan 610212, China
| |
Collapse
|
21
|
Dazzi C, Mehl J, Benamar M, Gerhardt H, Knaus P, Duda GN, Checa S. External mechanical loading overrules cell-cell mechanical communication in sprouting angiogenesis during early bone regeneration. PLoS Comput Biol 2023; 19:e1011647. [PMID: 37956208 PMCID: PMC10681321 DOI: 10.1371/journal.pcbi.1011647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 11/27/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
Sprouting angiogenesis plays a key role during bone regeneration. For example, insufficient early revascularization of the injured site can lead to delayed or non-healing. During sprouting, endothelial cells are known to be mechano-sensitive and respond to local mechanical stimuli. Endothelial cells interact and communicate mechanically with their surroundings, such as outer-vascular stromal cells, through cell-induced traction forces. In addition, external physiological loads act at the healing site, resulting in tissue deformations and impacting cellular arrangements. How these two distinct mechanical cues (cell-induced and external) impact angiogenesis and sprout patterning in early bone healing remains however largely unknown. Therefore, the aim of this study was to investigate the relative role of externally applied and cell-induced mechanical signals in driving sprout patterning at the onset of bone healing. To investigate cellular self-organisation in early bone healing, an in silico model accounting for the mechano-regulation of sprouting angiogenesis and stromal cell organization was developed. Computer model predictions were compared to in vivo experiments of a mouse osteotomy model stabilized with a rigid or a semirigid fixation system. We found that the magnitude and orientation of principal strains within the healing region can explain experimentally observed sprout patterning, under both fixation conditions. Furthermore, upon simulating the selective inhibition of either cell-induced or externally applied mechanical cues, external mechanical signals appear to overrule the mechanical communication acting on a cell-cell interaction level. Such findings illustrate the relevance of external mechanical signals over the local cell-mediated mechanical cues and could be used in the design of fracture treatment strategies for bone regeneration.
Collapse
Affiliation(s)
- Chiara Dazzi
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Julia Mehl
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Mounir Benamar
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Holger Gerhardt
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Georg N. Duda
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health Centre for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Sara Checa
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
22
|
Williams KE, Andraca Harrer J, LaBelle SA, Leguineche K, Kaiser J, Karipott S, Lin A, Vongphachanh A, Fulton T, Rosenthal JW, Muhib F, Ong KG, Weiss JA, Willett NJ, Guldberg RE. Early Resistance Rehabilitation Improves Functional Regeneration Following Segmental Bone Defect Injury. RESEARCH SQUARE 2023:rs.3.rs-3236150. [PMID: 37886569 PMCID: PMC10602073 DOI: 10.21203/rs.3.rs-3236150/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Mechanical loading is integral to bone development and repair. The application of mechanical loads through rehabilitation are regularly prescribed as a clinical aide following severe bone injuries. However, current rehabilitation regimens typically involve long periods of non-loading and rely on subjective patient feedback, leading to muscle atrophy and soft tissue fibrosis. While many pre-clinical studies have focused on unloading, ambulatory loading, or direct mechanical compression, rehabilitation intensity and its impact on the local strain environment and subsequent bone healing have largely not been investigated. This study combines implantable strain sensors and subject-specific finite element models in a pre-clinical rodent model with a defect size on the cusp of critically-sized. Animals were enrolled in either high or low intensity rehabilitation one week post injury to investigate how rehabilitation intensity affects the local mechanical environment and subsequent functional bone regeneration. The high intensity rehabilitation animals were given free access to running wheels with resistance, which increased local strains within the regenerative niche by an average of 44% compared to the low intensity (no-resistance) group. Finite element modeling demonstrated that resistance rehabilitation significantly increased compressive strain by a factor of 2.0 at week 1 and 4.45 after 4 weeks of rehabilitation. The resistance rehabilitation group had significantly increased regenerated bone volume and higher bone bridging rates than its sedentary counterpart (bone volume: 22.00 mm3 ± 4.26 resistance rehabilitation vs 8.00 mm3 ± 2.27 sedentary; bridging rates: 90% resistance rehabilitation vs 50% sedentary). In addition, animals that underwent resistance running had femurs with improved mechanical properties compared to those left in sedentary conditions, with failure torque and torsional stiffness values matching their contralateral, intact femurs (stiffness: 0.036 Nm/deg ± 0.006 resistance rehabilitation vs 0.008 Nm/deg ± 0.006 sedentary). Running on a wheel with no resistance rehabilitation also increased bridging rates (100% no resistance rehabilitation vs 50% sedentary). Analysis of bone volume and von Frey suggest no-resistance rehabilitation may improve bone regeneration and hindlimb functionality. These results demonstrate the potential for early resistance rehabilitation as a rehabilitation regimen to improve bone regeneration and functional recovery.
Collapse
Affiliation(s)
- Kylie E. Williams
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR 97403
| | - Julia Andraca Harrer
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR 97403
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA
- Atlanta Veteran’s Affairs Medical Center, Decatur, GA
| | - Steven A. LaBelle
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 841123
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT 84112
| | - Kelly Leguineche
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR 97403
| | - Jarred Kaiser
- Atlanta Veteran’s Affairs Medical Center, Decatur, GA
- Emory University, Decatur, GA
| | - Salil Karipott
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR 97403
| | - Angela Lin
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR 97403
| | - Alyssa Vongphachanh
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR 97403
| | - Travis Fulton
- Atlanta Veteran’s Affairs Medical Center, Decatur, GA
| | - J. Walker Rosenthal
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR 97403
| | - Farhan Muhib
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 841123
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT 84112
| | - Keat Ghee Ong
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR 97403
| | - Jeffrey A. Weiss
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 841123
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 841123
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, UT 84112
| | - Nick J. Willett
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR 97403
| | - Robert E. Guldberg
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR 97403
| |
Collapse
|
23
|
LaBelle SA, Poulson AM, Maas SA, Rauff A, Ateshian GA, Weiss JA. Spatial Configurations of 3D Extracellular Matrix Collagen Density and Anisotropy Simultaneously Guide Angiogenesis. PLoS Comput Biol 2023; 19:e1011553. [PMID: 37871113 PMCID: PMC10621972 DOI: 10.1371/journal.pcbi.1011553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 11/02/2023] [Accepted: 09/29/2023] [Indexed: 10/25/2023] Open
Abstract
Extracellular matrix (ECM) collagen density and fibril anisotropy are thought to affect the development of new vasculatures during pathologic and homeostatic angiogenesis. Computational simulation is emerging as a tool to investigate the role of matrix structural configurations on cell guidance. However, prior computational models have only considered the orientation of collagen as a model input. Recent experimental evidence indicates that cell guidance is simultaneously influenced by the direction and intensity of alignment (i.e., degree of anisotropy) as well as the local collagen density. The objective of this study was to explore the role of ECM collagen anisotropy and density during sprouting angiogenesis through simulation in the AngioFE and FEBio modeling frameworks. AngioFE is a plugin for FEBio (Finite Elements for Biomechanics) that simulates cell-matrix interactions during sprouting angiogenesis. We extended AngioFE to represent ECM collagen as deformable 3D ellipsoidal fibril distributions (EFDs). The rate and direction of microvessel growth were modified to depend simultaneously on the ECM collagen anisotropy (orientation and degree of anisotropy) and density. The sensitivity of growing neovessels to these stimuli was adjusted so that AngioFE could reproduce the growth and guidance observed in experiments where microvessels were cultured in collagen gels of varying anisotropy and density. We then compared outcomes from simulations using EFDs to simulations that used AngioFE's prior vector field representation of collagen anisotropy. We found that EFD simulations were more accurate than vector field simulations in predicting experimentally observed microvessel guidance. Predictive simulations demonstrated the ability of anisotropy gradients to recruit microvessels across short and long distances relevant to wound healing. Further, simulations predicted that collagen alignment could enable microvessels to overcome dense tissue interfaces such as tumor-associated collagen structures (TACS) found in desmoplasia and tumor-stroma interfaces. This approach can be generalized to other mechanobiological relationships during cell guidance phenomena in computational settings.
Collapse
Affiliation(s)
- Steven A. LaBelle
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, United States of America
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - A. Marsh Poulson
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Steve A. Maas
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, United States of America
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Adam Rauff
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, United States of America
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Gerard A. Ateshian
- Department of Mechanical Engineering, Columbia University, New York, New York, United States of America
| | - Jeffrey A. Weiss
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, United States of America
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah, United States of America
| |
Collapse
|
24
|
Whisler J, Shahreza S, Schlegelmilch K, Ege N, Javanmardi Y, Malandrino A, Agrawal A, Fantin A, Serwinski B, Azizgolshani H, Park C, Shone V, Demuren OO, Del Rosario A, Butty VL, Holroyd N, Domart MC, Hooper S, Szita N, Boyer LA, Walker-Samuel S, Djordjevic B, Sheridan GK, Collinson L, Calvo F, Ruhrberg C, Sahai E, Kamm R, Moeendarbary E. Emergent mechanical control of vascular morphogenesis. SCIENCE ADVANCES 2023; 9:eadg9781. [PMID: 37566656 PMCID: PMC10421067 DOI: 10.1126/sciadv.adg9781] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 07/13/2023] [Indexed: 08/13/2023]
Abstract
Vascularization is driven by morphogen signals and mechanical cues that coordinately regulate cellular force generation, migration, and shape change to sculpt the developing vascular network. However, it remains unclear whether developing vasculature actively regulates its own mechanical properties to achieve effective vascularization. We engineered tissue constructs containing endothelial cells and fibroblasts to investigate the mechanics of vascularization. Tissue stiffness increases during vascular morphogenesis resulting from emergent interactions between endothelial cells, fibroblasts, and ECM and correlates with enhanced vascular function. Contractile cellular forces are key to emergent tissue stiffening and synergize with ECM mechanical properties to modulate the mechanics of vascularization. Emergent tissue stiffening and vascular function rely on mechanotransduction signaling within fibroblasts, mediated by YAP1. Mouse embryos lacking YAP1 in fibroblasts exhibit both reduced tissue stiffness and develop lethal vascular defects. Translating our findings through biology-inspired vascular tissue engineering approaches will have substantial implications in regenerative medicine.
Collapse
Affiliation(s)
- Jordan Whisler
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Somayeh Shahreza
- Department of Mechanical Engineering, University College London, London, UK
| | | | - Nil Ege
- Tumour Cell Biology Laboratory, Francis Crick Institute, London, UK
- Mnemo Therapeutics, 101 Boulevard Murat, 75016 Paris, France
| | - Yousef Javanmardi
- Department of Mechanical Engineering, University College London, London, UK
| | - Andrea Malandrino
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Engineering and Research Center for Biomedical Engineering, Universitat Politècnica de Catalunya (UPC), Av. Eduard Maristany, 10-14 08019 Barcelona, Spain
| | - Ayushi Agrawal
- Department of Mechanical Engineering, University College London, London, UK
| | - Alessandro Fantin
- UCL Institute of Ophthalmology, University College London, London, UK
- Department of Biosciences, University of Milan, Via G. Celoria 26, 20133 Milan, Italy
| | - Bianca Serwinski
- Department of Mechanical Engineering, University College London, London, UK
- 199 Biotechnologies Ltd., Gloucester Road, London W2 6LD, UK
- Northeastern University London, London, E1W 1LP, UK
| | - Hesham Azizgolshani
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Clara Park
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Victoria Shone
- Experimental Histopathology Laboratory, Francis Crick Institute, London, UK
| | - Olukunle O. Demuren
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Amanda Del Rosario
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Vincent L. Butty
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Natalie Holroyd
- UCL Centre for Advanced Biomedical Imaging, Paul O'Gorman Building, 72 Huntley Street, London, UK
| | | | - Steven Hooper
- Tumour Cell Biology Laboratory, Francis Crick Institute, London, UK
| | - Nicolas Szita
- Department of Biochemical Engineering, University College London, London, UK
| | - Laurie A. Boyer
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Simon Walker-Samuel
- UCL Centre for Advanced Biomedical Imaging, Paul O'Gorman Building, 72 Huntley Street, London, UK
| | - Boris Djordjevic
- Department of Mechanical Engineering, University College London, London, UK
- 199 Biotechnologies Ltd., Gloucester Road, London W2 6LD, UK
| | - Graham K. Sheridan
- School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham, UK
| | - Lucy Collinson
- Electron Microscopy Laboratory, Francis Crick Institute, London, UK
| | - Fernando Calvo
- Instituto de Biomedicina y Biotecnología de Cantabria (Consejo Superior de Investigaciones Científicas, Universidad de Cantabria), Santander, Spain
| | | | - Erik Sahai
- Tumour Cell Biology Laboratory, Francis Crick Institute, London, UK
| | - Roger Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Emad Moeendarbary
- Department of Mechanical Engineering, University College London, London, UK
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- 199 Biotechnologies Ltd., Gloucester Road, London W2 6LD, UK
| |
Collapse
|
25
|
Wei Q, Zhu X, Wang L, Zhang W, Yang X, Wei W. Extracellular matrix in synovium development, homeostasis and arthritis disease. Int Immunopharmacol 2023; 121:110453. [PMID: 37331300 DOI: 10.1016/j.intimp.2023.110453] [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: 03/14/2023] [Revised: 05/27/2023] [Accepted: 06/02/2023] [Indexed: 06/20/2023]
Abstract
Extracellular matrix (ECM) is a three-dimensional network entity composed of extracellular macromolecules. ECM in synovium not only supports the structural integrity of synovium, but also plays a crucial role in regulating homeostasis and damage repair response in synovium. Obvious disorders in the composition, behavior and function of synovial ECM will lead to the occurrence and development of arthritis diseases such as rheumatoid arthritis (RA), osteoarthritis (OA) and psoriatic arthritis (PsA). Based on the importance of synovial ECM, targeted regulation of the composition and structure of ECM is considered to be an effective measure for the treatment of arthritis disease. This paper reviews the current research status of synovial ECM biology, discusses the role and mechanism of synovial ECM in physiological status and arthritis disease, and summarizes the current strategies for targeting synovial ECM to provide information for the pathogenesis, diagnosis and treatment of arthritis disease.
Collapse
Affiliation(s)
- Qi Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Xuemin Zhu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Luping Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Wankang Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China
| | - Xuezhi Yang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei 230032, China.
| |
Collapse
|
26
|
Raskov H, Gaggar S, Tajik A, Orhan A, Gögenur I. The Matrix Reloaded-The Role of the Extracellular Matrix in Cancer. Cancers (Basel) 2023; 15:2057. [PMID: 37046716 PMCID: PMC10093330 DOI: 10.3390/cancers15072057] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
As the core component of all organs, the extracellular matrix (ECM) is an interlocking macromolecular meshwork of proteins, glycoproteins, and proteoglycans that provides mechanical support to cells and tissues. In cancer, the ECM can be remodelled in response to environmental cues, and it controls a plethora of cellular functions, including metabolism, cell polarity, migration, and proliferation, to sustain and support oncogenesis. The biophysical and biochemical properties of the ECM, such as its structural arrangement and being a reservoir for bioactive molecules, control several intra- and intercellular signalling pathways and induce cytoskeletal changes that alter cell shapes, behaviour, and viability. Desmoplasia is a major component of solid tumours. The abnormal deposition and composition of the tumour matrix lead to biochemical and biomechanical alterations that determine disease development and resistance to treatment. This review summarises the complex roles of ECM in cancer and highlights the possible therapeutic targets and how to potentially remodel the dysregulated ECM in the future. Furthering our understanding of the ECM in cancer is important as the modification of the ECM will probably become an important tool in the characterisation of individual tumours and personalised treatment options.
Collapse
Affiliation(s)
- Hans Raskov
- Center for Surgical Science, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark
| | - Shruti Gaggar
- Center for Surgical Science, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark
| | - Asma Tajik
- Center for Surgical Science, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark
| | - Adile Orhan
- Center for Surgical Science, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Clinical Oncology, Zealand University Hospital, 4000 Roskilde, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
27
|
Wu P, Asada H, Hakamada M, Mabuchi M. Bioengineering of High Cell Density Tissues with Hierarchical Vascular Networks for Ex Vivo Whole Organs. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2209149. [PMID: 36545785 DOI: 10.1002/adma.202209149] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/25/2022] [Indexed: 06/17/2023]
Abstract
The development of tissue-like structures such as cell sheets, spheroids, and organoids has contributed to progress in regenerative medicine. Simultaneous achievement of scale up and high cell density of these tissues is challenging because sufficient oxygen cannot be supplied to the inside of large, high cell density tissues. Here, in vitro fabrication of vessels to supply oxygen to the inside of millimeter-sized scaffold-free tissues whose cell density is ≈200 million cells mL-1 , corresponding to those of native tissues, is shown. Hierarchical vascular networks by anastomosis of capillaries and a large vessel are essential for oxygen supply, whereas a large vessel or capillary networks alone make negligible contributions to the supply. The hierarchical vascular networks are formed by a top-down approach, which offers a new option for ex vivo whole organs without decellularization and 3D-bioprinting.
Collapse
Affiliation(s)
- Peizheng Wu
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto, 606-8501, Japan
| | - Hiroki Asada
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto, 606-8501, Japan
| | - Masataka Hakamada
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto, 606-8501, Japan
| | - Mamoru Mabuchi
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto, 606-8501, Japan
| |
Collapse
|
28
|
Li L, Yao ZC, Parian A, Yang YH, Chao J, Yin J, Salimian KJ, Reddy SK, Zaheer A, Gearhart SL, Mao HQ, Selaru FM. A nanofiber-hydrogel composite improves tissue repair in a rat model of Crohn's disease perianal fistulas. SCIENCE ADVANCES 2023; 9:eade1067. [PMID: 36598982 PMCID: PMC9812382 DOI: 10.1126/sciadv.ade1067] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 11/23/2022] [Indexed: 06/17/2023]
Abstract
Perianal fistulas (PAFs) represent a severe complication of Crohn's disease (CD). Despite the advent of biologic and small-molecule therapeutics for luminal disease, PAFs in CD (CD-PAF) are relatively resistant to treatment, with less than 50% responding to any therapy. We report an injectable, biodegradable, mechanically fragmented nanofiber-hydrogel composite (mfNHC) loaded with adipose-derived stem cells (ADSCs) for the treatment of fistulas in a rat model of CD-PAF. The ADSC-loaded mfNHC results in a higher degree of healing when compared to surgical treatment of fistulas, which is a standard treatment. The volume of fistulas treated with mfNHC is decreased sixfold compared to the surgical treatment control. Molecular studies reveal that utilization of mfNHC reduced local inflammation and improved tissue regeneration. This study demonstrates that ADSC-loaded mfNHC is a promising therapy for CD-PAF, and warrants further studies to advance mfNHC toward clinical translation.
Collapse
Affiliation(s)
- Ling Li
- Division of Gastroenterology and Hepatology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Zhi-Cheng Yao
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alyssa Parian
- Division of Gastroenterology and Hepatology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Yueh-Hsun Yang
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jeffrey Chao
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Public Health Studies, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Jason Yin
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kevan J. Salimian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sashank K. Reddy
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Atif Zaheer
- Division of Radiology and Radiological Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Susan L. Gearhart
- Division of Colorectal Surgery, Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hai-Quan Mao
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Florin M. Selaru
- Division of Gastroenterology and Hepatology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
29
|
Chavez T, Gerecht S. Engineering of the microenvironment to accelerate vascular regeneration. Trends Mol Med 2023; 29:35-47. [PMID: 36371337 PMCID: PMC9742290 DOI: 10.1016/j.molmed.2022.10.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 11/11/2022]
Abstract
Blood vessels are crucial for tissue development, functionality, and homeostasis and are typically a determinant in the progression of healing and regeneration. The tissue microenvironment provides physicochemical cues that affect cellular function, and the study of the microenvironment can be accelerated by the engineering of approaches capable of mimicking various aspects of the microenvironment. In this review, we introduce the major components of the vascular niche and focus on the roles of oxygen and the extracellular matrix (ECM). We demonstrate how vascular engineering approaches enhance our understanding of the microenvironment's impact on the vasculature towards vascular regeneration and describe the current limitations and future directions towards clinical utilization.
Collapse
Affiliation(s)
- Taylor Chavez
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Sharon Gerecht
- Department of Biomedical Engineering, Duke University, Durham, NC, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
30
|
Barrasa-Ramos S, Dessalles CA, Hautefeuille M, Barakat AI. Mechanical regulation of the early stages of angiogenesis. J R Soc Interface 2022; 19:20220360. [PMID: 36475392 PMCID: PMC9727679 DOI: 10.1098/rsif.2022.0360] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Favouring or thwarting the development of a vascular network is essential in fields as diverse as oncology, cardiovascular disease or tissue engineering. As a result, understanding and controlling angiogenesis has become a major scientific challenge. Mechanical factors play a fundamental role in angiogenesis and can potentially be exploited for optimizing the architecture of the resulting vascular network. Largely focusing on in vitro systems but also supported by some in vivo evidence, the aim of this Highlight Review is dual. First, we describe the current knowledge with particular focus on the effects of fluid and solid mechanical stimuli on the early stages of the angiogenic process, most notably the destabilization of existing vessels and the initiation and elongation of new vessels. Second, we explore inherent difficulties in the field and propose future perspectives on the use of in vitro and physics-based modelling to overcome these difficulties.
Collapse
Affiliation(s)
- Sara Barrasa-Ramos
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Claire A. Dessalles
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Mathieu Hautefeuille
- Laboratoire de Biologie du Développement (UMR7622), Institut de Biologie Paris Seine, Sorbonne Université, Paris, France,Facultad de Ciencias, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Abdul I. Barakat
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| |
Collapse
|
31
|
Yang C, Liu Y, Wang Z, Lin M, Liu C. Controlled mechanical loading improves bone regeneration by regulating type H vessels in a S1Pr1-dependent manner. FASEB J 2022; 36:e22530. [PMID: 36063128 DOI: 10.1096/fj.202200339rrr] [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: 03/01/2022] [Revised: 08/04/2022] [Accepted: 08/22/2022] [Indexed: 11/11/2022]
Abstract
Despite the best treatment, approximately 10% of fractures still face undesirable repair and result in delayed unions or non-unions. Dynamic mechanical stimulation promotes bone formation, when applied at the correct time frame, with optimal loading magnitude, frequency, and repetition. Controlled mechanical loading significantly increases osteogenic cells during the matrix deposition phase of bone repair. In the bone defect, the blood vessel network guides the initial bone formation activities. A unique blood vessel subtype (Type H) exists in bone, which expresses high levels of CD31 and endomucin, and functions to couple angiogenesis and osteogenesis. However, how this form of controlled mechanical loading regulates the Type H vessels and promotes bone formation is still not clear. Sphingosine 1-phosphate (S1P) participates in the bone anabolic process and is a key regulator of the blood vessel. Its receptor, sphingosine 1-phosphate receptor 1 (S1Pr1), is a mechanosensitive protein that regulates vascular integrity. Therefore, we hypothesis that controlled anabolic mechanical loading promotes bone repair by acting on Type H vessels. To study the effect of S1Pr1 on loading induced-bone repair, we utilized a stabilized tibial defect model, which allows for the application of anabolic mechanical loading. Mechanical loading upregulated S1Pr1 within the entire defect, with up to 80% expressed in blood vessels, as observed by deep tissue imaging. Additionally, S1Pr1 antagonism by W146 inhibited the anabolic effects of mechanical loading. We showed that mechanical loading or activating S1Pr1 could induce YAP nuclear translocation, a key regulator in the cell's mechanical response, in endothelial cells (ECs) in vitro. Inhibition of S1Pr1 in endothelial cells by siRNA reduced loading-induced YAP nuclear translocation and expressions of angiogenic genes. In vivo, YAP nuclear translocation in Type H vessels was up-regulated after mechanical loading but was inhibited by antagonizing S1Pr1. S1Pr1 agonist, FTY720, increased bone volume and Type H vessel volume, similar to that of mechanical stimulation. In conclusion, controlled anabolic mechanical loading enhanced bone formation mainly through Type H vessels in a S1Pr1-dependent manner.
Collapse
Affiliation(s)
- Chengyu Yang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Yang Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Ziyan Wang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Minmin Lin
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| | - Chao Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China.,Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
32
|
Bera K, Kiepas A, Zhang Y, Sun SX, Konstantopoulos K. The interplay between physical cues and mechanosensitive ion channels in cancer metastasis. Front Cell Dev Biol 2022; 10:954099. [PMID: 36158191 PMCID: PMC9490090 DOI: 10.3389/fcell.2022.954099] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Physical cues have emerged as critical influencers of cell function during physiological processes, like development and organogenesis, and throughout pathological abnormalities, including cancer progression and fibrosis. While ion channels have been implicated in maintaining cellular homeostasis, their cell surface localization often places them among the first few molecules to sense external cues. Mechanosensitive ion channels (MICs) are especially important transducers of physical stimuli into biochemical signals. In this review, we describe how physical cues in the tumor microenvironment are sensed by MICs and contribute to cancer metastasis. First, we highlight mechanical perturbations, by both solid and fluid surroundings typically found in the tumor microenvironment and during critical stages of cancer cell dissemination from the primary tumor. Next, we describe how Piezo1/2 and transient receptor potential (TRP) channels respond to these physical cues to regulate cancer cell behavior during different stages of metastasis. We conclude by proposing alternative mechanisms of MIC activation that work in tandem with cytoskeletal components and other ion channels to bestow cells with the capacity to sense, respond and navigate through the surrounding microenvironment. Collectively, this review provides a perspective for devising treatment strategies against cancer by targeting MICs that sense aberrant physical characteristics during metastasis, the most lethal aspect of cancer.
Collapse
Affiliation(s)
- Kaustav Bera
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
| | - Alexander Kiepas
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
- *Correspondence: Alexander Kiepas, ; Konstantinos Konstantopoulos,
| | - Yuqi Zhang
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
| | - Sean X. Sun
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Department of Mechanical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Konstantinos Konstantopoulos
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Department of Oncology, The Johns Hopkins University, Baltimore, MD, United States
- *Correspondence: Alexander Kiepas, ; Konstantinos Konstantopoulos,
| |
Collapse
|
33
|
Li Y, Wong IY, Guo M. Reciprocity of Cell Mechanics with Extracellular Stimuli: Emerging Opportunities for Translational Medicine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2107305. [PMID: 35319155 PMCID: PMC9463119 DOI: 10.1002/smll.202107305] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/20/2022] [Indexed: 06/14/2023]
Abstract
Human cells encounter dynamic mechanical cues in healthy and diseased tissues, which regulate their molecular and biophysical phenotype, including intracellular mechanics as well as force generation. Recent developments in bio/nanomaterials and microfluidics permit exquisitely sensitive measurements of cell mechanics, as well as spatiotemporal control over external mechanical stimuli to regulate cell behavior. In this review, the mechanobiology of cells interacting bidirectionally with their surrounding microenvironment, and the potential relevance for translational medicine are considered. Key fundamental concepts underlying the mechanics of living cells as well as the extracelluar matrix are first introduced. Then the authors consider case studies based on 1) microfluidic measurements of nonadherent cell deformability, 2) cell migration on micro/nano-topographies, 3) traction measurements of cells in three-dimensional (3D) matrix, 4) mechanical programming of organoid morphogenesis, as well as 5) active mechanical stimuli for potential therapeutics. These examples highlight the promise of disease diagnosis using mechanical measurements, a systems-level understanding linking molecular with biophysical phenotype, as well as therapies based on mechanical perturbations. This review concludes with a critical discussion of these emerging technologies and future directions at the interface of engineering, biology, and medicine.
Collapse
Affiliation(s)
- Yiwei Li
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan, Hubei, 430074, China
| | - Ian Y Wong
- School of Engineering, Center for Biomedical Engineering, Joint Program in Cancer Biology, Brown University, 184 Hope St Box D, Providence, RI, 02912, USA
| | - Ming Guo
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
34
|
Yao ZC, Yang YH, Kong J, Zhu Y, Li L, Chang C, Zhang C, Yin J, Chao J, Selaru FM, Reddy SK, Mao HQ. Biostimulatory Micro-Fragmented Nanofiber-Hydrogel Composite Improves Mesenchymal Stem Cell Delivery and Soft Tissue Remodeling. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2202309. [PMID: 35948487 PMCID: PMC9994419 DOI: 10.1002/smll.202202309] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/24/2022] [Indexed: 06/15/2023]
Abstract
Functional microgels are preferred stem cell carriers due to the ease of delivery through minimally invasive injection and seamless integration with the surrounding host tissue. A biostimulatory nanofiber-hydrogel composite (NHC) has been previously developed through covalently crosslinking a hyaluronic acid hydrogel network with surface-functionalized poly (ε-caprolactone) nanofiber fragments. The NHC mimics the microarchitecture of native soft tissue matrix, showing enhanced cell infiltration, immunomodulation, and proangiogenic properties. Here, injectability of the pre-formed NHC is improved by mechanical fragmentation, making it into micro-fragmented NHC (mfNHC) in a granular gel form as a stem cell carrier to deliver mesenchymal stem cells (MSCs) for soft tissue remodeling. The mfNHC shows a similar storage modulus but a significantly reduced injection force, as compared with the corresponding bulk NHC. When injected subcutaneously in a rat model, mfNHC-MSC constructs initiate an elevated level of host macrophage infiltration, more pro-regenerative polarization, and subsequently, improved angiogenesis and adipogenesis response when compared to mfNHC alone. A similar trend of host cell infiltration and pro-angiogenic response is detected in a swine model with a larger volume injection. These results suggest a strong potential for use of the mfNHC as an injectable carrier for cell delivery and soft tissue remodeling.
Collapse
Affiliation(s)
- Zhi-Cheng Yao
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21213, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Yueh-Hsun Yang
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21213, USA
- Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
| | - Jiayuan Kong
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21213, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Yining Zhu
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21213, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Ling Li
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Calvin Chang
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21213, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Chi Zhang
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21213, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Jason Yin
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21213, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Jeffrey Chao
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21213, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Public Health Studies, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Florin M Selaru
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Sashank K Reddy
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, MD, 21287, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Hai-Quan Mao
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21213, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| |
Collapse
|
35
|
Zarubova J, Hasani-Sadrabadi MM, Ardehali R, Li S. Immunoengineering strategies to enhance vascularization and tissue regeneration. Adv Drug Deliv Rev 2022; 184:114233. [PMID: 35304171 PMCID: PMC10726003 DOI: 10.1016/j.addr.2022.114233] [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] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 03/07/2022] [Accepted: 03/11/2022] [Indexed: 12/11/2022]
Abstract
Immune cells have emerged as powerful regulators of regenerative as well as pathological processes. The vast majority of regenerative immunoengineering efforts have focused on macrophages; however, growing evidence suggests that other cells of both the innate and adaptive immune system are as important for successful revascularization and tissue repair. Moreover, spatiotemporal regulation of immune cells and their signaling have a significant impact on the regeneration speed and the extent of functional recovery. In this review, we summarize the contribution of different types of immune cells to the healing process and discuss ways to manipulate and control immune cells in favor of vascularization and tissue regeneration. In addition to cell delivery and cell-free therapies using extracellular vesicles, we discuss in situ strategies and engineering approaches to attract specific types of immune cells and modulate their phenotypes. This field is making advances to uncover the extraordinary potential of immune cells and their secretome in the regulation of vascularization and tissue remodeling. Understanding the principles of immunoregulation will help us design advanced immunoengineering platforms to harness their power for tissue regeneration.
Collapse
Affiliation(s)
- Jana Zarubova
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Prague 14220, Czech Republic
| | | | - Reza Ardehali
- Division of Cardiology, Department of Internal Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Song Li
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; Eli and Edythe Broad Stem Cell Research Center, University of California, Los Angeles, CA 90095, USA; Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
36
|
LaBelle SA, Dinkins SS, Hoying JB, Budko EV, Rauff A, Strobel HA, Lin AH, Weiss JA. Matrix anisotropy promotes angiogenesis in a density-dependent manner. Am J Physiol Heart Circ Physiol 2022; 322:H806-H818. [PMID: 35333118 PMCID: PMC8993529 DOI: 10.1152/ajpheart.00072.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/16/2022] [Accepted: 03/16/2022] [Indexed: 11/22/2022]
Abstract
Angiogenesis is necessary for wound healing, tumorigenesis, implant inosculation, and homeostasis. In each situation, matrix structure and mechanics play a role in determining whether new vasculatures can establish transport to new or hypoxic tissues. Neovessel growth and directional guidance are sensitive to three-dimensional (3-D) matrix anisotropy and density, although the individual and integrated roles of these matrix features have not been fully recapitulated in vitro. We developed a tension-based method to align 3-D collagen constructs seeded with microvessel fragments in matrices of three levels of collagen fibril anisotropy and two levels of collagen density. The extent and direction of neovessel growth from the parent microvessel fragments increased with matrix anisotropy and decreased with density. The proangiogenic effects of anisotropy were attenuated at higher matrix densities. We also examined the impact of matrix anisotropy in an experimental model of neovessel invasion across a tissue interface. Matrix density was found to dictate the success of interface crossing, whereas interface curvature and fibril alignment were found to control directional guidance. Our findings indicate that complex configurations of matrix density and alignment can facilitate or complicate the establishment or maintenance of vascular networks in pathological and homeostatic angiogenesis. Furthermore, we extend preexisting methods for tuning collagen anisotropy in thick constructs. This approach addresses gaps in tissue engineering and cell culture by supporting the inclusion of large multicellular structures in prealigned constructs.NEW & NOTEWORTHY Matrix anisotropy and density have a considerable effect on angiogenic vessel growth and directional guidance. However, the current literature relies on 2-D and simplified models of angiogenesis (e.g., tubulogenesis and vasculogenesis). We present a method to align 3-D collagen scaffolds embedded with microvessel fragments to different levels of anisotropy. Neovessel growth increases with anisotropy and decreases with density, which may guide angiogenic neovessels across tissue interfaces such as during implant inosculation and tumorigenesis.
Collapse
Affiliation(s)
- Steven A LaBelle
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah
| | - Shad S Dinkins
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah
| | - James B Hoying
- Advanced Solutions Life Sciences, Manchester, New Hampshire
| | - Elena V Budko
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah
| | - Adam Rauff
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah
| | | | - Allen H Lin
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah
| | - Jeffrey A Weiss
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah
| |
Collapse
|
37
|
Fu M, Peng D, Lan T, Wei Y, Wei X. Multifunctional regulatory protein connective tissue growth factor (CTGF): A potential therapeutic target for diverse diseases. Acta Pharm Sin B 2022; 12:1740-1760. [PMID: 35847511 PMCID: PMC9279711 DOI: 10.1016/j.apsb.2022.01.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/22/2021] [Accepted: 12/16/2021] [Indexed: 12/24/2022] Open
Abstract
Connective tissue growth factor (CTGF), a multifunctional protein of the CCN family, regulates cell proliferation, differentiation, adhesion, and a variety of other biological processes. It is involved in the disease-related pathways such as the Hippo pathway, p53 and nuclear factor kappa-B (NF-κB) pathways and thus contributes to the developments of inflammation, fibrosis, cancer and other diseases as a downstream effector. Therefore, CTGF might be a potential therapeutic target for treating various diseases. In recent years, the research on the potential of CTGF in the treatment of diseases has also been paid more attention. Several drugs targeting CTGF (monoclonal antibodies FG3149 and FG3019) are being assessed by clinical or preclinical trials and have shown promising outcomes. In this review, the cellular events regulated by CTGF, and the relationships between CTGF and pathogenesis of diseases are systematically summarized. In addition, we highlight the current researches, focusing on the preclinical and clinical trials concerned with CTGF as the therapeutic target.
Collapse
|
38
|
Moss SM, Ortiz-Hernandez M, Levin D, Richburg CA, Gerton T, Cook M, Houlton JJ, Rizvi ZH, Goodwin PC, Golway M, Ripley B, Hoying JB. A Biofabrication Strategy for a Custom-Shaped, Non-Synthetic Bone Graft Precursor with a Prevascularized Tissue Shell. Front Bioeng Biotechnol 2022; 10:838415. [PMID: 35356783 PMCID: PMC8959609 DOI: 10.3389/fbioe.2022.838415] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/08/2022] [Indexed: 11/13/2022] Open
Abstract
Critical-sized defects of irregular bones requiring bone grafting, such as in craniofacial reconstruction, are particularly challenging to repair. With bone-grafting procedures growing in number annually, there is a reciprocal growing interest in bone graft substitutes to meet the demand. Autogenous osteo(myo)cutaneous grafts harvested from a secondary surgical site are the gold standard for reconstruction but are associated with donor-site morbidity and are in limited supply. We developed a bone graft strategy for irregular bone-involved reconstruction that is customizable to defect geometry and patient anatomy, is free of synthetic materials, is cellularized, and has an outer pre-vascularized tissue layer to enhance engraftment and promote osteogenesis. The graft, comprised of bioprinted human-derived demineralized bone matrix blended with native matrix proteins containing human mesenchymal stromal cells and encased in a simple tissue shell containing isolated, human adipose microvessels, ossifies when implanted in rats. Ossification follows robust vascularization within and around the graft, including the formation of a vascular leash, and develops mechanical strength. These results demonstrate an early feasibility animal study of a biofabrication strategy to manufacture a 3D printed patient-matched, osteoconductive, tissue-banked, bone graft without synthetic materials for use in craniofacial reconstruction. The bone fabrication workflow is designed to be performed within the hospital near the Point of Care.
Collapse
Affiliation(s)
- Sarah M. Moss
- Advanced Solutions Life Sciences, Louisville, KY, United States
| | - Monica Ortiz-Hernandez
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, United States
| | - Dmitry Levin
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, United States
| | - Chris A. Richburg
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
| | - Thomas Gerton
- Advanced Solutions Life Sciences, Louisville, KY, United States
| | - Madison Cook
- Advanced Solutions Life Sciences, Louisville, KY, United States
| | - Jeffrey J. Houlton
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, United States
| | - Zain H. Rizvi
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, United States
| | | | - Michael Golway
- Advanced Solutions Life Sciences, Louisville, KY, United States
| | - Beth Ripley
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, United States
| | - James B. Hoying
- Advanced Solutions Life Sciences, Louisville, KY, United States
| |
Collapse
|
39
|
Owen-Woods C, Kusumbe A. Fundamentals of bone vasculature: Specialization, interactions and functions. Semin Cell Dev Biol 2022; 123:36-47. [PMID: 34281770 DOI: 10.1016/j.semcdb.2021.06.025] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 02/06/2023]
Abstract
Angiogenesis, hematopoiesis and osteogenesis are fundamental processes mediating complex and essential biological functions. In the bone marrow, endothelial cells (ECs) are a principal mediator of regulatory signals that govern hematopoietic and mesenchymal stem cells. EC and osteoblast interactions and niche functions of ECs are fundamental in maintaining bone health and coordinating repair and regeneration following injury. These cellular interactions are subject to dysregulation and deterioration under stress, aging, chronic disease states and malignancy. Thus, the prospect of manipulating the bone vasculature has tremendous potential to advance therapeutic interventions for the management of bone diseases. This review discusses the current state of vascular-skeletal tissue interactions focusing on osteoblast and hematopoietic stem cells interaction with ECs.
Collapse
Affiliation(s)
- Charlotte Owen-Woods
- Tissue and Tumor Microenvironments Group, NDORMS, University of Oxford, Oxford OX3 7FY, UK
| | - Anjali Kusumbe
- Tissue and Tumor Microenvironments Group, NDORMS, University of Oxford, Oxford OX3 7FY, UK.
| |
Collapse
|
40
|
Shokrani H, Shokrani A, Sajadi SM, Seidi F, Mashhadzadeh AH, Rabiee N, Saeb MR, Aminabhavi T, Webster TJ. Cell-Seeded Biomaterial Scaffolds: The Urgent Need for Unanswered Accelerated Angiogenesis. Int J Nanomedicine 2022; 17:1035-1068. [PMID: 35309965 PMCID: PMC8927652 DOI: 10.2147/ijn.s353062] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/22/2022] [Indexed: 12/12/2022] Open
Abstract
One of the most arduous challenges in tissue engineering is neovascularization, without which there is a lack of nutrients delivered to a target tissue. Angiogenesis should be completed at an optimal density and within an appropriate period of time to prevent cell necrosis. Failure to meet this challenge brings about poor functionality for the tissue in comparison with the native tissue, extensively reducing cell viability. Prior studies devoted to angiogenesis have provided researchers with some biomaterial scaffolds and cell choices for angiogenesis. For example, while most current angiogenesis approaches require a variety of stimulatory factors ranging from biomechanical to biomolecular to cellular, some other promising stimulatory factors have been underdeveloped (such as electrical, topographical, and magnetic). When it comes to choosing biomaterial scaffolds in tissue engineering for angiogenesis, key traits rush to mind including biocompatibility, appropriate physical and mechanical properties (adhesion strength, shear stress, and malleability), as well as identifying the appropriate biomaterial in terms of stability and degradation profile, all of which may leave essential trace materials behind adversely influencing angiogenesis. Nevertheless, the selection of the best biomaterial and cells still remains an area of hot dispute as such previous studies have not sufficiently classified, integrated, or compared approaches. To address the aforementioned need, this review article summarizes a variety of natural and synthetic scaffolds including hydrogels that support angiogenesis. Furthermore, we review a variety of cell sources utilized for cell seeding and influential factors used for angiogenesis with a concentrated focus on biomechanical factors, with unique stimulatory factors. Lastly, we provide a bottom-to-up overview of angiogenic biomaterials and cell selection, highlighting parameters that need to be addressed in future studies.
Collapse
Affiliation(s)
- Hanieh Shokrani
- Department of Chemical Engineering, Sharif University of Technology, Tehran, Iran
| | - Amirhossein Shokrani
- Department of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| | - S Mohammad Sajadi
- Department of Nutrition, Cihan University-Erbil, Erbil, 625, Iraq
- Department of Phytochemistry, SRC, Soran University, Soran, KRG, 624, Iraq
- Correspondence: S Mohammad Sajadi; Navid Rabiee, Email ; ;
| | - Farzad Seidi
- Jiangsu Co–Innovation Center for Efficient Processing and Utilization of Forest Resources and International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing, 210037, People’s Republic of China
| | - Amin Hamed Mashhadzadeh
- Mechanical and Aerospace Engineering, School of Engineering and Digital Sciences, Nazarbayev University, Nur-Sultan, 010000, Kazakhstan
| | - Navid Rabiee
- Department of Physics, Sharif University of Technology, Tehran, Iran
- School of Engineering, Macquarie University, Sydney, New South Wales, 2109, Australia
| | - Mohammad Reza Saeb
- Department of Polymer Technology, Faculty of Chemistry, Gdańsk University of Technology, Gdańsk, Poland
| | - Tejraj Aminabhavi
- School of Advanced Sciences, KLE Technological University, Hubballi, Karnataka, 580 031, India
- Department of Chemistry, Karnatak University, Dharwad, 580 003, India
| | - Thomas J Webster
- School of Health Sciences and Biomedical Engineering, Hebei University, Tianjin, People’s Republic of China
- Center for Biomaterials, Vellore Institute of Technology, Vellore, India
| |
Collapse
|
41
|
Yi X, Liu F, Gao K, Chen F, Wang Y, Li H, Wang X, Huang Y, Fu H, Zhou W, Fan JB, Wang S, Gao Y. Reconstructable Uterus-Derived Materials for Uterus Recovery toward Efficient Live Births. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2106510. [PMID: 34854148 DOI: 10.1002/adma.202106510] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/17/2021] [Indexed: 06/13/2023]
Abstract
Uterine factor infertility is increasingly common in modern society and has severely affected human life and health. However, the existing biomaterial scaffold-mediated systems remain limited in efficient uterus recovery, leading to low pregnancy rate and live births. Here, reconstructable uterus-derived materials (RUMs) are demonstrated by combining uterus-derived extracellular matrix and seeded chorionic villi mesenchymal stem cells for uterus recovery, achieving highly efficient live births in rats with severe uterine injury. The RUMs can be designed into different states (such as, liquid RUMs and solid RUMs) and shapes (such as, cuboid, triangular-prism, and cube) in terms of requirements. The RUMs can effectively prevent intrauterine adhesion, and promote endometrial regeneration and muscle collagen reconstruction, as well as, accelerate wound healing by constructing a physical barrier and secreting cytokines, allowing efficient uterus recovery. The injured uterus nearly achieves complete recovery after treating with the RUMs and has normal pregnancies for supporting fetal development and live births, similar to the normal rats. The study provides a regenerative medicine therapeutics for uterine factor infertility.
Collapse
Affiliation(s)
- Xiao Yi
- Research Centre for Artificial Organ and Tissue Engineering & Institute of Regenerative Medicine, Department of Hepatobiliary Surgery II, Zhujiang Hospital, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, 510280, P. R. China
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, P. R. China
| | - Fan Liu
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, P. R. China
| | - Kunjie Gao
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, P. R. China
| | - Feng Chen
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, P. R. China
| | - Yifeng Wang
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, P. R. China
| | - Huayan Li
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, P. R. China
| | - Xuefeng Wang
- Department of Gynecology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Yi Huang
- Department of Gynecology, Nanhai People's Hospital, Southern Medical University, Guangzhou, Guangdong Province, P. R. China
| | - Huijiao Fu
- Department of Gynecology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Weijie Zhou
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Jun-Bing Fan
- Research Centre for Artificial Organ and Tissue Engineering & Institute of Regenerative Medicine, Department of Hepatobiliary Surgery II, Zhujiang Hospital, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, 510280, P. R. China
| | - Shutao Wang
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, CAS Center for Excellence in Nanoscience, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Yi Gao
- Research Centre for Artificial Organ and Tissue Engineering & Institute of Regenerative Medicine, Department of Hepatobiliary Surgery II, Zhujiang Hospital, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, 510280, P. R. China
| |
Collapse
|
42
|
Yao G, Mo X, Yin C, Lou W, Wang Q, Huang S, Mao L, Chen S, Zhao K, Pan T, Huang L, Lin Y. A programmable and skin temperature-activated electromechanical synergistic dressing for effective wound healing. SCIENCE ADVANCES 2022; 8:eabl8379. [PMID: 35080981 PMCID: PMC8791608 DOI: 10.1126/sciadv.abl8379] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Mechanical regulation and electric stimulation hold great promise in skin tissue engineering for manipulating wound healing. However, the complexity of equipment operation and stimulation implementation remains an ongoing challenge in clinical applications. Here, we propose a programmable and skin temperature-activated electromechanical synergistic wound dressing composed of a shape memory alloy-based mechanical metamaterial for wound contraction and an antibacterial electret thin film for electric field generation. This strategy is successfully demonstrated on rats to achieve effective wound healing in as short as 4 and 8 days for linear and circular wounds, respectively, with a statistically significant over 50% improvement in wound closure rate versus the blank control group. The optimally designed electromechanical synergistic stimulation could regulate the wound microenvironment to accelerate healing metabolism, promote wound closure, and inhibit infection. This work provided an effective wound healing strategy in the context of a programmable temperature-responsive, battery-free electromechanical synergistic biomedical device.
Collapse
Affiliation(s)
- Guang Yao
- School of Materials and Energy, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
- State Key Laboratory of Electronic Thin films and Integrated Devices, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
- Medico-Engineering Cooperation on Applied Medicine Research Center, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
- Corresponding author. (G.Y.); (L.H.); (Y.L.)
| | - Xiaoyi Mo
- School of Materials and Energy, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
| | - Chenhui Yin
- School of Materials and Energy, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
| | - Wenhao Lou
- School of Materials and Energy, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
| | - Qian Wang
- School of Materials and Energy, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
| | - Sirong Huang
- School of Materials and Energy, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
| | - Linna Mao
- School of Materials and Energy, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
| | - Sihong Chen
- School of Materials and Energy, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
| | - Kangning Zhao
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, International School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, Hubei, China
| | - Taisong Pan
- School of Materials and Energy, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
- State Key Laboratory of Electronic Thin films and Integrated Devices, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
- Medico-Engineering Cooperation on Applied Medicine Research Center, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
| | - Lin Huang
- School of Electronic Science and Engineering, the Center for Information in Medicine, University of Electronic Science and Technology of China, Chengdu 611731, Sichuan, China
- Corresponding author. (G.Y.); (L.H.); (Y.L.)
| | - Yuan Lin
- School of Materials and Energy, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
- State Key Laboratory of Electronic Thin films and Integrated Devices, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
- Medico-Engineering Cooperation on Applied Medicine Research Center, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
- Corresponding author. (G.Y.); (L.H.); (Y.L.)
| |
Collapse
|
43
|
Sun Y, Wan B, Wang R, Zhang B, Luo P, Wang D, Nie JJ, Chen D, Wu X. Mechanical Stimulation on Mesenchymal Stem Cells and Surrounding Microenvironments in Bone Regeneration: Regulations and Applications. Front Cell Dev Biol 2022; 10:808303. [PMID: 35127684 PMCID: PMC8815029 DOI: 10.3389/fcell.2022.808303] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/03/2022] [Indexed: 01/15/2023] Open
Abstract
Treatment of bone defects remains a challenge in the clinic. Artificial bone grafts are the most promising alternative to autologous bone grafting. However, one of the limiting factors of artificial bone grafts is the limited means of regulating stem cell differentiation during bone regeneration. As a weight-bearing organ, bone is in a continuous mechanical environment. External mechanical force, a type of biophysical stimulation, plays an essential role in bone regeneration. It is generally accepted that osteocytes are mechanosensitive cells in bone. However, recent studies have shown that mesenchymal stem cells (MSCs) can also respond to mechanical signals. This article reviews the mechanotransduction mechanisms of MSCs, the regulation of mechanical stimulation on microenvironments surrounding MSCs by modulating the immune response, angiogenesis and osteogenesis, and the application of mechanical stimulation of MSCs in bone regeneration. The review provides a deep and extensive understanding of mechanical stimulation mechanisms, and prospects feasible designs of biomaterials for bone regeneration and the potential clinical applications of mechanical stimulation.
Collapse
Affiliation(s)
- Yuyang Sun
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing, China
| | - Ben Wan
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing, China
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Center for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam (VU), Amsterdam Movement Science (AMS), Amsterdam, Netherlands
| | - Renxian Wang
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing, China
| | - Bowen Zhang
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing, China
| | - Peng Luo
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing, China
| | - Diaodiao Wang
- Department of Joint Surgery, Peking University Ninth School of Clinical Medicine, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Jing-Jun Nie
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing, China
- *Correspondence: Jing-Jun Nie, ; Dafu Chen,
| | - Dafu Chen
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing, China
- *Correspondence: Jing-Jun Nie, ; Dafu Chen,
| | - Xinbao Wu
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing, China
| |
Collapse
|
44
|
Abstract
Fracture healing is a complex, multistep process that is highly sensitive to mechanical signaling. To optimize repair, surgeons prescribe immediate weight-bearing as-tolerated within 24 hours after surgical fixation; however, this recommendation is based on anecdotal evidence and assessment of bulk healing outcomes (e.g., callus size, bone volume, etc.). Given challenges in accurately characterizing the mechanical environment and the ever-changing properties of the regenerate, the principles governing mechanical regulation of repair, including their cell and molecular basis, are not yet well defined. However, the use of mechanobiological rodent models, and their relatively large genetic toolbox, combined with recent advances in imaging approaches and single-cell analyses is improving our understanding of the bone microenvironment in response to loading. This review describes the identification and characterization of distinct cell populations involved in bone healing and highlights the most recent findings on mechanical regulation of bone homeostasis and repair with an emphasis on osteo-angio coupling. A discussion on aging and its impact on bone mechanoresponsiveness emphasizes the need for novel mechanotherapeutics that can re-sensitize skeletal stem and progenitor cells to physical rehabilitation protocols.
Collapse
Affiliation(s)
- Tareq Anani
- Department of Orthopedic Surgery, New York University Langone Health, New York, NY 10010, USA
| | - Alesha B Castillo
- Department of Orthopedic Surgery, New York University Langone Health, New York, NY 10010, USA; Department of Biomedical Engineering, Tandon School of Engineering, New York University, New York, NY 11201, USA; Department of Veterans Affairs, New York Harbor Healthcare System, Manhattan Campus, New York, NY 10010, USA.
| |
Collapse
|
45
|
Wang Z, Liu Y, Zhang J, Lin M, Xiao C, Bai H, Liu C. Mechanical loading alleviated the inhibition of β2-adrenergic receptor agonist terbutaline on bone regeneration. FASEB J 2021; 35:e22033. [PMID: 34739146 DOI: 10.1096/fj.202101045rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 10/19/2021] [Accepted: 10/22/2021] [Indexed: 12/14/2022]
Abstract
The long-term use of adrenergic medication in treating various conditions, such as asthma, increases the chances of bone fracture. Dynamic mechanical loading at a specific time is a method for improving bone quality and promoting healing. Therefore, we hypothesized that precisely controlling the mechanical environment can contribute to the alleviation of the negative effects of chronic treatment with the common asthma drug terbutaline, which is a β2-adrenergic receptor agonist that facilitates bone homeostasis and defect repair through its anabolic effect on osteogenic cells. Our in vitro results showed that terbutaline can directly inhibit osteogenesis by impairing osteogenic differentiation and mineralization. Chronic treatment in vivo was simulated by administering terbutaline to C57BL/6J mice for 4 weeks before bone defect surgery and mechanical loading. We utilized a stabilized tibial defect model, which allowed the application of anabolic mechanical loading. During homeostasis, chronic terbutaline treatment reduced the bone formation rate, the fracture toughness of long bones, and the concentrations of bone formation markers in the sera. During defect repair, terbutaline decreased the bone volume, type H vessel, and total blood vessel volume. Terbutaline treatment reduced the number of osteogenic cells. Periostin, which was secreted mainly by Prrx1+ osteoprogenitors and F4/80+ macrophages, was inhibited by treating the bone defect with terbutaline. Interestingly, controlled mechanical loading facilitated the recovery of bone volume and periostin expression and the number of osteogenic cells within the defect. In conclusion, mechanical loading can rescue negative effects on new bone accrual and repair induced by chronic terbutaline treatment.
Collapse
Affiliation(s)
- Ziyan Wang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Yang Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Jianing Zhang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Minmin Lin
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Chufan Xiao
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Haoying Bai
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Chao Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
46
|
Marsico G, Martin‐Saldaña S, Pandit A. Therapeutic Biomaterial Approaches to Alleviate Chronic Limb Threatening Ischemia. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003119. [PMID: 33854887 PMCID: PMC8025020 DOI: 10.1002/advs.202003119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/24/2020] [Indexed: 05/14/2023]
Abstract
Chronic limb threatening ischemia (CLTI) is a severe condition defined by the blockage of arteries in the lower extremities that leads to the degeneration of blood vessels and is characterized by the formation of non-healing ulcers and necrosis. The gold standard therapies such as bypass and endovascular surgery aim at the removal of the blockage. These therapies are not suitable for the so-called "no option patients" which present multiple artery occlusions with a likelihood of significant limb amputation. Therefore, CLTI represents a significant clinical challenge, and the efforts of developing new treatments have been focused on stimulating angiogenesis in the ischemic muscle. The delivery of pro-angiogenic nucleic acid, protein, and stem cell-based interventions have limited efficacy due to their short survival. Engineered biomaterials have emerged as a promising method to improve the effectiveness of these latter strategies. Several synthetic and natural biomaterials are tested in different formulations aiming to incorporate nucleic acid, proteins, stem cells, macrophages, or endothelial cells in supportive matrices. In this review, an overview of the biomaterials used alone and in combination with growth factors, nucleic acid, and cells in preclinical models is provided and their potential to induce revascularization and regeneration for CLTI applications is discussed.
Collapse
Affiliation(s)
- Grazia Marsico
- CÚRAM SFI Research Centre for Medical DevicesNational University of IrelandGalwayIreland
| | - Sergio Martin‐Saldaña
- CÚRAM SFI Research Centre for Medical DevicesNational University of IrelandGalwayIreland
| | - Abhay Pandit
- CÚRAM SFI Research Centre for Medical DevicesNational University of IrelandGalwayIreland
| |
Collapse
|
47
|
Zhang P, Liu X, Guo P, Li X, He Z, Li Z, Stoddart MJ, Grad S, Tian W, Chen D, Zou X, Zhou Z, Liu S. Effect of cyclic mechanical loading on immunoinflammatory microenvironment in biofabricating hydroxyapatite scaffold for bone regeneration. Bioact Mater 2021; 6:3097-3108. [PMID: 33778191 PMCID: PMC7960680 DOI: 10.1016/j.bioactmat.2021.02.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 02/18/2021] [Accepted: 02/18/2021] [Indexed: 12/19/2022] Open
Abstract
It has been proven that the mechanical microenvironment can impact the differentiation of mesenchymal stem cells (MSCs). However, the effect of mechanical stimuli in biofabricating hydroxyapatite scaffolds on the inflammatory response of MSCs remains unclear. This study aimed to investigate the effect of mechanical loading on the inflammatory response of MSCs seeded on scaffolds. Cyclic mechanical loading was applied to biofabricate the cell-scaffold composite for 15 min/day over 7, 14, or 21 days. At the predetermined time points, culture supernatant was collected for inflammatory mediator detection, and gene expression was analyzed by qRT-PCR. The results showed that the expression of inflammatory mediators (IL1B and IL8) was downregulated (p < 0.05) and the expression of ALP (p < 0.01) and COL1A1 (p < 0.05) was upregulated under mechanical loading. The cell-scaffold composites biofabricated with or without mechanical loading were freeze-dried to prepare extracellular matrix-based scaffolds (ECM-based scaffolds). Murine macrophages were seeded on the ECM-based scaffolds to evaluate their polarization. The ECM-based scaffolds that were biofabricated with mechanical loading before freeze-drying enhanced the expression of M2 polarization-related biomarkers (Arginase 1 and Mrc1, p < 0.05) of macrophages in vitro and increased bone volume/total volume ratio in vivo. Overall, these findings demonstrated that mechanical loading could dually modulate the inflammatory responses and osteogenic differentiation of MSCs. Besides, the ECM-based scaffolds that were biofabricated with mechanical loading before freeze-drying facilitated the M2 polarization of macrophages in vitro and bone regeneration in vivo. Mechanical loading may be a promising biofabrication strategy for bone biomaterials. Compressive mechanical loading is applied to biofabricate the MSCs-hydroxyapatite composites for bone regeneration. Mechanical loading can modulate the inflammatory responses and osteogenic differentiation of MSCs seeded on scaffold. ECM-based scaffolds from initially loading biofabrication facilitated the M2 polarization of macrophages and bone repair. Mechanical loading may be a promising biofabrication strategy for bone biomaterials.
Collapse
Affiliation(s)
- Penghui Zhang
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopaedic Research Institute /Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Xizhe Liu
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopaedic Research Institute /Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Peng Guo
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopaedic Research Institute /Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Xianlong Li
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopaedic Research Institute /Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhongyuan He
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopaedic Research Institute /Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhen Li
- AO Research Institute Davos, Clavadelerstrasse 8, Davos, 7270, Switzerland
| | - Martin J Stoddart
- AO Research Institute Davos, Clavadelerstrasse 8, Davos, 7270, Switzerland
| | - Sibylle Grad
- AO Research Institute Davos, Clavadelerstrasse 8, Davos, 7270, Switzerland
| | - Wei Tian
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Orthopaedics and Traumatology, Beijing JiShuiTan Hospital, Beijing, 100035, China
| | - Dafu Chen
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Orthopaedics and Traumatology, Beijing JiShuiTan Hospital, Beijing, 100035, China
| | - Xuenong Zou
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopaedic Research Institute /Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhiyu Zhou
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopaedic Research Institute /Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Shaoyu Liu
- Innovation Platform of Regeneration and Repair of Spinal Cord and Nerve Injury, Department of Orthopaedic Surgery, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, China.,Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Orthopaedic Research Institute /Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| |
Collapse
|
48
|
Abstract
Supramolecular biopolymers (SBPs) are those polymeric units derived from macromolecules that can assemble with each other by noncovalent interactions. Macromolecular structures are commonly found in living systems such as proteins, DNA/RNA, and polysaccharides. Bioorganic chemistry allows the generation of sequence-specific supramolecular units like SBPs that can be tailored for novel applications in tissue engineering (TE). SBPs hold advantages over other conventional polymers previously used for TE; these materials can be easily functionalized; they are self-healing, biodegradable, stimuli-responsive, and nonimmunogenic. These characteristics are vital for the further development of current trends in TE, such as the use of pluripotent cells for organoid generation, cell-free scaffolds for tissue regeneration, patient-derived organ models, and controlled delivery systems of small molecules. In this review, we will analyse the 3 subtypes of SBPs: peptide-, nucleic acid-, and oligosaccharide-derived. Then, we will discuss the role that SBPs will be playing in TE as dynamic scaffolds, therapeutic scaffolds, and bioinks. Finally, we will describe possible outlooks of SBPs for TE.
Collapse
|
49
|
Cheuy V, Picciolini S, Bedoni M. Progressing the field of Regenerative Rehabilitation through novel interdisciplinary interaction. NPJ Regen Med 2020; 5:16. [PMID: 33042583 PMCID: PMC7511907 DOI: 10.1038/s41536-020-00102-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 08/31/2020] [Indexed: 02/06/2023] Open
Abstract
The synergy between biological and bioengineering advances is critical to developing novel and impactful translational therapies. However, there currently are few opportunities for regenerative scientists to be exposed to the methodologies commonly employed in the clinic by rehabilitation professionals, and most rehabilitation scientists and clinicians are not exposed to the many advances of regenerative medicine. This disconnect has impeded the pace of progress in the field. The Eighth Annual International Symposium on Regenerative Rehabilitation brought together basic scientists, engineers, and rehabilitation clinicians to present scientifically rigorous and cutting-edge research and clinical management, focusing on new and innovative approaches that combine discoveries in tissue engineering, medical devices, and cellular therapies with rehabilitative protocols.
Collapse
Affiliation(s)
- Victor Cheuy
- Department of Physical Therapy and Rehabilitation Science, University of California San Francisco, San Francisco, CA USA.,Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA USA
| | | | | |
Collapse
|