1
|
Tusnim J, Kutuzov P, Grasman JM. In Vitro Models for Peripheral Nerve Regeneration. Adv Healthc Mater 2024; 13:e2401605. [PMID: 39324286 DOI: 10.1002/adhm.202401605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/14/2024] [Indexed: 09/27/2024]
Abstract
Peripheral nerve injury (PNI) resulting in lesions is highly prevalent clinically, but current therapeutic approaches fail to provide satisfactory outcomes in many patients. While peripheral nerves have intrinsic regenerative capacity, the regenerative capabilities of peripheral nerves are often insufficient to restore full functionality. This highlights an unmet need for developing more effective strategies to repair damaged peripheral nerves and improve regenerative success. Consequently, researchers are actively exploring a variety of therapeutic strategies, encompassing the local delivery of trophic factors or bioactive molecules, the design of advanced biomaterials that interact with regenerating axons, and augmentation with nerve guidance conduits or complex prostheses. However, clinical translation of these technologies remains limited, emphasizing the need for continued research on peripheral nerve regeneration modalities that can enhance functional restoration. Experimental models that accurately recapitulate key aspects of peripheral nerve injury and repair biology can accelerate therapeutic development by enabling systematic testing of new techniques. Advancing regenerative therapies for PNI requires bridging the gap between basic science discoveries and clinical application. This review discusses different in vitro models of peripheral nerve injury and repair, including their advantages, limitations, and potential applications.
Collapse
Affiliation(s)
- Jarin Tusnim
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Peter Kutuzov
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Jonathan M Grasman
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| |
Collapse
|
2
|
Smith PO, Huang G, Devries K, Nazhat SN, Phillips JB. Automated production of nerve repair constructs containing endothelial cell tube-like structures. Biofabrication 2024; 17:015024. [PMID: 39500048 DOI: 10.1088/1758-5090/ad8efd] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/05/2024] [Indexed: 11/21/2024]
Abstract
Engineered neural tissue (EngNT) is a stabilised aligned cellular hydrogel that offers a potential alternative to the nerve autograft for the treatment of severe peripheral nerve injury. This work aimed to automate the production of EngNT, to improve the feasibility of scalable manufacture for clinical translation. Endothelial cells were used as the cellular component of the EngNT, with the formation of endothelial cell tube-like structures mimicking the polarised vascular structures formed early on in the natural regenerative process. Gel aspiration-ejection for the production of EngNT was automated by integrating a syringe pump with a robotic positioning system, using software coded in Python to control both devices. Having established the production method and tested mechanical properties, the EngNT containing human umbilical vein endothelial cells (EngNT-HUVEC) was characterised in terms of viability and alignment, compatibility with neurite outgrowth from rat dorsal root ganglion neurons and formation of endothelial cell networksin vitro. EngNT-HUVEC manufactured using the automated system contained viable and aligned endothelial cells, which developed into a network of multinucleated endothelial cell tube-like structures inside the constructs and an outer layer of endothelialisation. The EngNT-HUVEC constructs were made in various sizes within minutes. Constructs provided support and guidance to regenerating neuritesin vitro. This work automated the formation of EngNT, facilitating high throughput manufacture at scale. The formation of endothelial cell tube-like structures within stabilised hydrogels provides an engineered tissue with potential for use in nerve repair.
Collapse
Affiliation(s)
- Poppy O Smith
- UCL Centre for Nerve Engineering, Department of Pharmacology, UCL School of Pharmacy, University College London, London, United Kingdom
| | - Guanbingxue Huang
- Department of Mining and Materials Engineering, McGill University, Montreal, Quebec, Canada
| | - Kate Devries
- Department of Mining and Materials Engineering, McGill University, Montreal, Quebec, Canada
| | - Showan N Nazhat
- Department of Mining and Materials Engineering, McGill University, Montreal, Quebec, Canada
| | - James B Phillips
- UCL Centre for Nerve Engineering, Department of Pharmacology, UCL School of Pharmacy, University College London, London, United Kingdom
| |
Collapse
|
3
|
Muangsanit P, Smith P. Harnessing endothelial cells and vascularization strategies for nerve regeneration. Neural Regen Res 2024; 19:2337-2338. [PMID: 38526263 PMCID: PMC11090433 DOI: 10.4103/nrr.nrr-d-23-01840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/25/2023] [Accepted: 01/05/2024] [Indexed: 03/26/2024] Open
Affiliation(s)
- Papon Muangsanit
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
| | - Poppy Smith
- UCL Centre for Nerve Engineering, UCL, London, UK; Department of Pharmacology, UCL School of Pharmacy, UCL, London, UK
| |
Collapse
|
4
|
He J, Qian L, Li Z, Wang Y, Liu K, Wei H, Sun Y, He J, Yao K, Weng J, Hu X, Zhang D, He Y. A tissue bandage for pelvic ganglia injury. Nat Commun 2024; 15:8972. [PMID: 39419980 PMCID: PMC11487282 DOI: 10.1038/s41467-024-53302-5] [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: 10/21/2023] [Accepted: 10/08/2024] [Indexed: 10/19/2024] Open
Abstract
Neurogenic bladder often occurs after pelvic ganglia injury. Its symptoms, like severe urinary retention and incontinence, have a significant impact on individuals' quality of life. Unfortunately, there are currently no effective treatments available for this type of injury. Here, we designed a fiber-enhanced tissue bandage for injured pelvic ganglia. Tight junctions formed in tissue bandages create a mini tissue structure that enhances resistance in an in vivo environment and delivers growth factors to support the healing of ganglia. Strength fibers are similar to clinical bandages and guarantee ease of handling. Furthermore, tissue bandages can be stored at low temperatures over 5 months without compromising cell viability, meeting the requirements for clinical products. A tissue bandage was applied to a male rat with a bilateral major pelvic ganglia crush injury. Compared to the severe neurogenic bladder symptoms observed in the injury and scaffold groups, tissue bandages significantly improved bladder function. We found that tissue bandage increases resistance to mechanical injury by boosting the expression of cytoskeletal proteins within the major pelvic ganglia. Overall, tissue bandages show promise as a practical therapeutic approach for ganglia repair, offering hope for developing more effective treatments for this thorny condition.
Collapse
Affiliation(s)
- Jing He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
| | - Lin Qian
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhuang Li
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yanpeng Wang
- Center for Reproductive Medicine, Department of Gynecology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Kai Liu
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Haibin Wei
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yuan Sun
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jiaoyan He
- Department of Postgraduate Education, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Ke Yao
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jiahao Weng
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xuanhan Hu
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Dahong Zhang
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China.
| |
Collapse
|
5
|
Tusnim J, Budharaju K, Grasman JM. Fabrication of ECM protein coated hollow collagen channels to study peripheral nerve regeneration. Sci Rep 2024; 14:16096. [PMID: 38997331 PMCID: PMC11245515 DOI: 10.1038/s41598-024-67046-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 07/08/2024] [Indexed: 07/14/2024] Open
Abstract
Peripheral nerve injury is a prevalent clinical problem that often leads to lifelong disability and reduced quality of life. Although peripheral nerves can regenerate, recovery after severe injury is slow and incomplete. The current gold standard treatment, autologous nerve transplantation, has limitations including donor site morbidity and poor functional outcomes, highlighting the need for improved repair strategies. We developed a reproducible in vitro hollow channel collagen gel construct to investigate peripheral nerve regeneration (PNR) by exploring the influence of key extracellular matrix (ECM) proteins on axonal growth and regeneration. Channels were coated with ECM proteins: collagen IV, laminin, or fibronectin and seeded with dorsal root ganglia (DRG) collected from E16 rat embryos to compare the ability of the ECM proteins to enhance axonal growth. Robust axonal extension and Schwann cell (SC) infiltration were observed in fibronectin-coated channels, suggesting its superiority over other ECM proteins. Differential effects of ECM proteins on axons and SCs indicated direct growth stimulation beyond SC-mediated guidance. In vitro laceration injury modeling further confirmed fibronectin's superior pro-regenerative effects, showcasing its potential in enhancing axonal regrowth post-injury. Advancing in vitro modeling that closely replicates native microenvironments will accelerate progress in overcoming the limitations of current nerve repair approaches.
Collapse
Affiliation(s)
- Jarin Tusnim
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Karthik Budharaju
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Jonathan M Grasman
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA.
| |
Collapse
|
6
|
Testa L, Dotta S, Vercelli A, Marvaldi L. Communicating pain: emerging axonal signaling in peripheral neuropathic pain. Front Neuroanat 2024; 18:1398400. [PMID: 39045347 PMCID: PMC11265228 DOI: 10.3389/fnana.2024.1398400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 05/21/2024] [Indexed: 07/25/2024] Open
Abstract
Peripheral nerve damage often leads to the onset of neuropathic pain (NeuP). This condition afflicts millions of people, significantly burdening healthcare systems and putting strain on families' financial well-being. Here, we will focus on the role of peripheral sensory neurons, specifically the Dorsal Root Ganglia neurons (DRG neurons) in the development of NeuP. After axotomy, DRG neurons activate regenerative signals of axons-soma communication to promote a gene program that activates an axonal branching and elongation processes. The results of a neuronal morphological cytoskeleton change are not always associated with functional recovery. Moreover, any axonal miss-targeting may contribute to NeuP development. In this review, we will explore the epidemiology of NeuP and its molecular causes at the level of the peripheral nervous system and the target organs, with major focus on the neuronal cross-talk between intrinsic and extrinsic factors. Specifically, we will describe how failures in the neuronal regenerative program can exacerbate NeuP.
Collapse
Affiliation(s)
- Livia Testa
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Torino), Torino, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, Torino, Italy
| | - Sofia Dotta
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Torino), Torino, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, Torino, Italy
| | - Alessandro Vercelli
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Torino), Torino, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, Torino, Italy
| | - Letizia Marvaldi
- Neuroscience Institute Cavalieri Ottolenghi, Orbassano (Torino), Torino, Italy
- Department of Neuroscience “Rita Levi-Montalcini”, Torino, Italy
| |
Collapse
|
7
|
Mandal A, Moneme C, Tewari BP, Goldstein AM, Sontheimer H, Cheng L, Moore SR, Levin D. A novel method for culturing enteric neurons generates neurospheres containing functional myenteric neuronal subtypes. J Neurosci Methods 2024; 407:110144. [PMID: 38670535 PMCID: PMC11144385 DOI: 10.1016/j.jneumeth.2024.110144] [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: 12/23/2023] [Revised: 03/04/2024] [Accepted: 04/20/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND The enteric nervous system (ENS) is comprised of neurons, glia, and neural progenitor cells that regulate essential gastrointestinal functions. Advances in high-efficiency enteric neuron culture would facilitate discoveries surrounding ENS regulatory processes, pathophysiology, and therapeutics. NEW METHOD Development of a simple, robust, one-step method to culture murine enteric neurospheres in a 3D matrix that supports neural growth and differentiation. RESULTS Myenteric plexus cells isolated from the entire length of adult murine small intestine formed ≥3000 neurospheres within 7 days. Matrigel-embedded neurospheres exhibited abundant neural stem and progenitor cells expressing Sox2, Sox10 and Msi1 by day 4. By day 5, neural progenitor cell marker Nestin appeared in the periphery of neurospheres prior to differentiation. Neurospheres produced extensive neurons and neurites, confirmed by Tubulin beta III, PGP9.5, HuD/C, and NeuN immunofluorescence, including neural subtypes Calretinin, ChAT, and nNOS following 8 days of differentiation. Individual neurons within and external to neurospheres generated depolarization induced action potentials which were inhibited in the presence of sodium channel blocker, Tetrodotoxin. Differentiated neurospheres also contained a limited number of glia and endothelial cells. COMPARISON WITH EXISTING METHODS This novel one-step neurosphere growth and differentiation culture system, in 3D format (in the presence of GDNF, EGF, and FGF2), allows for ∼2-fold increase in neurosphere count in the derivation of enteric neurons with measurable action potentials. CONCLUSION Our method describes a novel, robust 3D culture of electrophysiologically active enteric neurons from adult myenteric neural stem and progenitor cells.
Collapse
Affiliation(s)
- Arabinda Mandal
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Chioma Moneme
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Bhanu P Tewari
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Harald Sontheimer
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Lily Cheng
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Sean R Moore
- Department of Pediatrics, Division of Pediatric Gastroenterology Hepatology, and Nutrition, University of Virginia, Charlottesville, VA, USA.
| | - Daniel Levin
- Department of Surgery, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
8
|
Blaszkiewicz M, Tao T, Mensah-Arhin K, Willows JW, Bates R, Huang W, Cao L, Smith RL, Townsend KL. Gene therapy approaches for obesity-induced adipose neuropathy: Device-targeted AAV-mediated neurotrophic factor delivery to adipocytes in subcutaneous adipose. Mol Ther 2024; 32:1407-1424. [PMID: 38429927 PMCID: PMC11081869 DOI: 10.1016/j.ymthe.2024.02.035] [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: 08/07/2023] [Revised: 12/15/2023] [Accepted: 02/27/2024] [Indexed: 03/03/2024] Open
Abstract
Maintaining functional adipose innervation is critical for metabolic health. We found that subcutaneous white adipose tissue (scWAT) undergoes peripheral neuropathy (PN) with obesity, diabetes, and aging (reduced small-fiber innervation and nerve/synaptic/growth-cone/vesicle markers, altered nerve activity). Unlike with nerve injuries, peripheral nerves do not regenerate with PN, and therefore new therapies are needed for treatment of this condition affecting 20-30 million Americans. Here, we validated a gene therapy approach using an adipocyte-tropic adeno-associated virus (AAV; serotype Rec2) to deliver neurotrophic factors (brain-derived neurotrophic factor [BDNF] and nerve growth factor [NGF]) directly to scWAT to improve tissue-specific PN as a proof-of-concept approach. AAVRec2-BDNF intra-adipose delivery improved tissue innervation in obese/diabetic mice with PN, but after longer periods of dietary obesity there was reduced efficacy, revealing a key time window for therapies. AAVRec2-NGF also increased scWAT innervation in obese mice and was more effective than BDNF, likely because Rec2 targeted adipocytes, the tissue's endogenous NGF source. AAVRec2-NGF also worked well even after 25 weeks of dietary obesity, unlike BDNF, which likely needs a vector that targets its physiological cellular source (stromal vascular fraction cells). Given the differing effects of AAVs carrying NGF versus BDNF, a combined therapy may be ideal for PN.
Collapse
Affiliation(s)
| | - Tianyi Tao
- Department of Neurological Surgery, The Ohio State University, Columbus, OH 43210, USA
| | - Kofi Mensah-Arhin
- Department of Neurological Surgery, The Ohio State University, Columbus, OH 43210, USA
| | - Jake W Willows
- Department of Neurological Surgery, The Ohio State University, Columbus, OH 43210, USA
| | - Rhiannon Bates
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH 43210, USA
| | - Wei Huang
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH 43210, USA
| | - Lei Cao
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH 43210, USA
| | - Rosemary L Smith
- College of Engineering, University of Maine, Orono, ME 04469, USA
| | - Kristy L Townsend
- Department of Neurological Surgery, The Ohio State University, Columbus, OH 43210, USA; College of Engineering, University of Maine, Orono, ME 04469, USA.
| |
Collapse
|
9
|
Palmisano B, Farinacci G, Campolo F, Tavanti C, Stefano A, Donsante S, Ippolito E, Giannicola G, Venneri MA, Corsi A, Riminucci M. A pathogenic role for brain-derived neurotrophic factor (BDNF) in fibrous dysplasia of bone. Bone 2024; 181:117047. [PMID: 38331308 DOI: 10.1016/j.bone.2024.117047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/10/2024] [Accepted: 02/06/2024] [Indexed: 02/10/2024]
Abstract
Brain derived neurotrophic factor (BDNF) is a neurotrophin, expressed in the central nervous system and in peripheral tissues, that is regulated by the Gsα/cAMP pathway. In bone, it regulates osteogenesis and stimulates RANKL secretion and osteoclast formation in osteolytic tumors such as Multiple Myeloma. Fibrous dysplasia (FD) of bone is a rare genetic disease of the skeleton caused by gain-of-function mutations of the Gsα gene in which RANKL-dependent enhanced bone resorption is a major cause of bone fragility and clinical morbidity. We observed that BDNF transcripts are expressed in human FD lesions. Specifically, immunolocalization studies performed on biopsies obtained from FD patients revealed the expression of BDNF in osteoblasts and, to a lower extent, in the spindle-shaped cells within the fibrous tissue. Therefore, we hypothesized that BDNF can play a role in the pathogenesis of FD by stimulating RANKL secretion and bone resorption. To test this hypothesis, we used the EF1α-GsαR201C mouse model of the human disease (FD mice). Western blot analysis revealed a higher expression of BDNF in bone segments of FD mice compared to WT mice and the immunolabeling pattern within mouse FD lesions was similar to that observed in human FD. Treatment of FD mice with a monoclonal antibody against BDNF reduced the fibrous tissue along with the number of osteoclasts and osteoblasts within femoral lesions. These results reveal BDNF as a new player in the pathogenesis of FD and a potential molecular mechanism by which osteoclastogenesis may be nourished within FD bone lesions. They also suggest that BDNF inhibition may be a new approach to reduce abnormal bone remodeling in FD.
Collapse
Affiliation(s)
- Biagio Palmisano
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Giorgia Farinacci
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Federica Campolo
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Chiara Tavanti
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Alessia Stefano
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Samantha Donsante
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Ernesto Ippolito
- Department of Orthopaedic Surgery, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Giuseppe Giannicola
- Department of Anatomical, Histological, Medico Legal and Orthopaedic Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Alessandro Corsi
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Mara Riminucci
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy.
| |
Collapse
|
10
|
Stadlmayr S, Peter K, Millesi F, Rad A, Wolf S, Mero S, Zehl M, Mentler A, Gusenbauer C, Konnerth J, Schniepp HC, Lichtenegger H, Naghilou A, Radtke C. Comparative Analysis of Various Spider Silks in Regard to Nerve Regeneration: Material Properties and Schwann Cell Response. Adv Healthc Mater 2024; 13:e2302968. [PMID: 38079208 PMCID: PMC11468126 DOI: 10.1002/adhm.202302968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/20/2023] [Indexed: 12/26/2023]
Abstract
Peripheral nerve reconstruction through the employment of nerve guidance conduits with Trichonephila dragline silk as a luminal filling has emerged as an outstanding preclinical alternative to avoid nerve autografts. Yet, it remains unknown whether the outcome is similar for silk fibers harvested from other spider species. This study compares the regenerative potential of dragline silk from two orb-weaving spiders, Trichonephila inaurata and Nuctenea umbratica, as well as the silk of the jumping spider Phidippus regius. Proliferation, migration, and transcriptomic state of Schwann cells seeded on these silks are investigated. In addition, fiber morphology, primary protein structure, and mechanical properties are studied. The results demonstrate that the increased velocity of Schwann cells on Phidippus regius fibers can be primarily attributed to the interplay between the silk's primary protein structure and its mechanical properties. Furthermore, the capacity of silk fibers to trigger cells toward a gene expression profile of a myelinating Schwann cell phenotype is shown. The findings for the first time allow an in-depth comparison of the specific cellular response to various native spider silks and a correlation with the fibers' material properties. This knowledge is essential to open up possibilities for targeted manufacturing of synthetic nervous tissue replacement.
Collapse
Affiliation(s)
- Sarah Stadlmayr
- Department of PlasticReconstructive and Aesthetic SurgeryMedical University of ViennaVienna1090Austria
- Austrian Cluster for Tissue RegenerationViennaAustria
| | - Karolina Peter
- Institute for Physics and Materials ScienceUniversity of Natural Resources and Life SciencesVienna1190Austria
| | - Flavia Millesi
- Department of PlasticReconstructive and Aesthetic SurgeryMedical University of ViennaVienna1090Austria
- Austrian Cluster for Tissue RegenerationViennaAustria
| | - Anda Rad
- Department of PlasticReconstructive and Aesthetic SurgeryMedical University of ViennaVienna1090Austria
| | - Sonja Wolf
- Department of PlasticReconstructive and Aesthetic SurgeryMedical University of ViennaVienna1090Austria
| | - Sascha Mero
- Department of PlasticReconstructive and Aesthetic SurgeryMedical University of ViennaVienna1090Austria
| | - Martin Zehl
- Department of Analytical ChemistryFaculty of ChemistryUniversity of ViennaVienna1090Austria
| | - Axel Mentler
- Institute of Soil ResearchUniversity of Natural Resources and Life SciencesVienna1190Austria
| | - Claudia Gusenbauer
- Institute of Wood Technology and Renewable MaterialsUniversity of Natural Resources and Life SciencesTulln an der Donau3430Austria
| | - Johannes Konnerth
- Institute of Wood Technology and Renewable MaterialsUniversity of Natural Resources and Life SciencesTulln an der Donau3430Austria
| | | | - Helga Lichtenegger
- Institute for Physics and Materials ScienceUniversity of Natural Resources and Life SciencesVienna1190Austria
| | - Aida Naghilou
- Department of PlasticReconstructive and Aesthetic SurgeryMedical University of ViennaVienna1090Austria
- Austrian Cluster for Tissue RegenerationViennaAustria
- Medical Systems Biophysics and BioengineeringLeiden Academic Centre for Drug ResearchLeiden UniversityLeiden2333The Netherlands
| | - Christine Radtke
- Department of PlasticReconstructive and Aesthetic SurgeryMedical University of ViennaVienna1090Austria
- Austrian Cluster for Tissue RegenerationViennaAustria
| |
Collapse
|
11
|
Li Q, Liu S, Zheng T, Li M, Qi B, Zhou L, Liu B, Ma D, Zhao C, Chen Z. Grafted human-induced pluripotent stem cells-derived oligodendrocyte progenitor cells combined with human umbilical vein endothelial cells contribute to functional recovery following spinal cord injury. Stem Cell Res Ther 2024; 15:35. [PMID: 38321505 PMCID: PMC10848469 DOI: 10.1186/s13287-024-03651-1] [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: 05/19/2023] [Accepted: 01/29/2024] [Indexed: 02/08/2024] Open
Abstract
BACKGROUND Spinal cord injury (SCI) is a devastating disease that causes extensive damage to oligodendrocytes and neurons leading to demyelination and axonal degeneration. In this study, we co-transplanted cell grafts containing oligodendrocyte progenitor cells (OPCs) derived from human-induced pluripotent stem cells (iPSCs) combined with human umbilical vein endothelial cells (HUVECs), which were reported to promote OPCs survival and migration, into rat contusion models to promote functional recovery after SCI. METHODS OPCs were derived from iPSCs and identified by immunofluorescence at different time points. Functional assays in vitro were performed to evaluate the effect of HUVECs on the proliferation, migration, and survival of OPCs by co-culture and migration assay, as well as on the neuronal axonal growth. A combination of OPCs and HUVECs was transplanted into the rat contusive model. Upon 8 weeks, immunofluorescence staining was performed to test the safety of transplanted cells and to observe the neuronal repairment, myelination, and neural circuit reconstruction at the injured area; also, the functional recovery was assessed by Basso, Beattie, and Bresnahan open-field scale, Ladder climb, SEP, and MEP. Furthermore, the effect of HUVECs on grafts was also determined in vivo. RESULTS Data showed that HUVECs promote the proliferation, migration, and survival of OPCs both in vitro and in vivo. Furthermore, 8 weeks upon engraftment, the rats with OPCs and HUVECs co-transplantation noticeably facilitated remyelination, enhanced functional connection between the grafts and the host and promoted functional recovery. In addition, compared with the OPCs-alone transplantation, the co-transplantation generated more sensory neurons at the lesion border and significantly improved the sensory functional recovery. CONCLUSIONS Our study demonstrates that transplantation of OPCs combined with HUVECs significantly enhances both motor and sensory functional recovery after SCI. No significance was observed between OPCs combined with HUVECs group and OPCs-alone group in motor function recovery, while the sensory function recovery was significantly promoted in OPCs combined with HUVECs groups compared with the other two groups. These findings provide novel insights into the field of SCI research.
Collapse
Affiliation(s)
- Qian Li
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Sumei Liu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Tianqi Zheng
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Mo Li
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Boling Qi
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Liping Zhou
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Bochao Liu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Dan Ma
- Translational Medicine Research Group (TMRG), Aston Medical School, Aston University, Birmingham, B4 7ET, UK
| | - Chao Zhao
- Department of Clinical Neurosciences, Wellcome Trust-Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Zhiguo Chen
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China.
- Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing, 100069, China.
- Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Beijing, 100069, China.
| |
Collapse
|
12
|
Ma D, Zhang H, Yin L, Xu H, Wu L, Shaji R, Rezai F, Mulla A, Kaur S, Tan S, Kysela B, Wang Y, Chen Z, Zhao C, Gu Y. Human iPSC-derived endothelial cells promote CNS remyelination via BDNF and mTORC1 pathway. Glia 2024; 72:133-155. [PMID: 37675625 DOI: 10.1002/glia.24466] [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: 09/01/2022] [Revised: 08/06/2023] [Accepted: 08/08/2023] [Indexed: 09/08/2023]
Abstract
Damage of myelin is a component of many diseases in the central nervous system (CNS). The activation and maturation of the quiescent oligodendrocyte progenitor cells (OPCs) are the crucial cellular processes for CNS remyelination, which is influenced by neuroinflammation in the lesion microenvironment. Endothelial cells derived from human induced pluripotent stem cells (hiPSC-ECs) have shown promise in restoring function in various preclinical animal models. Here we ask whether and whether transplantation of hiPSC-ECs could benefit remyelination in a mouse model of CNS demyelination. Our results show that in vitro, hiPSC-ECs increase OPC proliferation, migration and differentiation via secreted soluble factors including brain-derived neurotrophic factor (BDNF). hiPSC-ECs also promote the survival of oligodendrocyte lineage cells in vitro and in vivo. Transplantation of hiPSC-ECs into a toxin-induced demyelination lesion in mouse corpus callosum (CC) leads to increased density of oligodendrocyte lineage cells and level of myelin in demyelinated area, correlated with a decreased neuroinflammation and an increased proportion of pro-regenerative M2 phenotype in microglia/macrophages. The hiPSC-EC-exposed oligodendrocyte lineage cells showed significant increase in the level of phosphorylated S6 ribosomal protein (pS6) both in vitro and in vivo, indicating an involvement of mTORC1 pathway. These results suggest that hiPSC-ECs may benefit myelin protection and regeneration which providing a potential source of cell therapy for a wide range of diseases and injuries associated with myelin damage.
Collapse
Affiliation(s)
- Dan Ma
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Translational Medicine Research Group (TMRG), Aston Medical School, Aston University, Birmingham, UK
| | - Huiyuan Zhang
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Molecular Pharmacology Laboratory, Institute of Molecular Medicine, Peking University, Beijing, China
- ALLIFE Medical Science and Technology Co. Ltd, Beijing, China
| | - Le Yin
- Translational Medicine Research Group (TMRG), Aston Medical School, Aston University, Birmingham, UK
- Molecular Pharmacology Laboratory, Institute of Molecular Medicine, Peking University, Beijing, China
- ALLIFE Medical Science and Technology Co. Ltd, Beijing, China
| | - Hao Xu
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Molecular Pharmacology Laboratory, Institute of Molecular Medicine, Peking University, Beijing, China
- ALLIFE Medical Science and Technology Co. Ltd, Beijing, China
| | - Lida Wu
- Translational Medicine Research Group (TMRG), Aston Medical School, Aston University, Birmingham, UK
- Molecular Pharmacology Laboratory, Institute of Molecular Medicine, Peking University, Beijing, China
- ALLIFE Medical Science and Technology Co. Ltd, Beijing, China
| | - Rahul Shaji
- Translational Medicine Research Group (TMRG), Aston Medical School, Aston University, Birmingham, UK
| | - Fatema Rezai
- Translational Medicine Research Group (TMRG), Aston Medical School, Aston University, Birmingham, UK
| | - Ayesha Mulla
- Translational Medicine Research Group (TMRG), Aston Medical School, Aston University, Birmingham, UK
| | - Sukhteerath Kaur
- Translational Medicine Research Group (TMRG), Aston Medical School, Aston University, Birmingham, UK
| | - Shengjiang Tan
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, Cambridge, UK
| | - Boris Kysela
- Translational Medicine Research Group (TMRG), Aston Medical School, Aston University, Birmingham, UK
| | - Yilong Wang
- Department of Neurology, Tiantan Hospital Capital Medical University, National Center and National Clinical Research Center for Neurological Diseases, Advanced Innovation Center for Human Brain Protection, Chinese Institute for Brain Research, Beijing, China
| | - Zhiguo Chen
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Chao Zhao
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Yuchun Gu
- Translational Medicine Research Group (TMRG), Aston Medical School, Aston University, Birmingham, UK
- Molecular Pharmacology Laboratory, Institute of Molecular Medicine, Peking University, Beijing, China
- ALLIFE Medical Science and Technology Co. Ltd, Beijing, China
| |
Collapse
|
13
|
Shin M, Ha T, Lim J, An J, Beak G, Choi J, Melvin AA, Yoon J, Choi J. Human Motor System-Based Biohybrid Robot-On-a-Chip for Drug Evaluation of Neurodegenerative Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305371. [PMID: 38036423 PMCID: PMC10811491 DOI: 10.1002/advs.202305371] [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: 08/03/2023] [Revised: 10/21/2023] [Indexed: 12/02/2023]
Abstract
Biohybrid robots have been developed for biomedical applications and industrial robotics. However, the biohybrid robots have limitations to be applied in neurodegenerative disease research due to the absence of a central nervous system. In addition, the organoids-on-a-chip has not yet been able to replicate the physiological function of muscle movement in the human motor system, which is essential for evaluating the accuracy of the drugs used for treating neurodegenerative diseases. Here, a human motor system-based biohybrid robot-on-a-chip composed of a brain organoid, multi-motor neuron spheroids, and muscle bundle on solid substrateis proposed to evaluate the drug effect on neurodegenerative diseases for the first time. The electrophysiological signals from the cerebral organoid induced the muscle bundle movement through motor neuron spheroids. To evaluate the drug effect on Parkinson's disease (PD), a patient-derived midbrain organoid is generated and incorporated into a biohybrid robot-on-a-chip. The drug effect on PD is successfully evaluated by measuring muscle bundle movement. The muscle bundle movement of PD patient-derived midbrain organoid-based biohybrid robot-on-a-chip is increased from 4.5 ± 0.99 µm to 18.67 ± 2.25 µm in response to levodopa. The proposed human motor system-based biohybrid robot-on-a-chip can serve as a standard biohybrid robot model for drug evaluation.
Collapse
Affiliation(s)
- Minkyu Shin
- Department of Chemical & Biomolecular EngineeringSogang University35 Baekbeom‐ro, Mapo‐guSeoul04107Republic of Korea
| | - Taehyeong Ha
- Department of Chemical & Biomolecular EngineeringSogang University35 Baekbeom‐ro, Mapo‐guSeoul04107Republic of Korea
| | - Joungpyo Lim
- Department of Chemical & Biomolecular EngineeringSogang University35 Baekbeom‐ro, Mapo‐guSeoul04107Republic of Korea
| | - Joohyun An
- Department of Chemical & Biomolecular EngineeringSogang University35 Baekbeom‐ro, Mapo‐guSeoul04107Republic of Korea
| | - Geunyoung Beak
- Department of Chemical & Biomolecular EngineeringSogang University35 Baekbeom‐ro, Mapo‐guSeoul04107Republic of Korea
| | - Jin‐Ha Choi
- School of Chemical EngineeringJeonbuk National University567 Baekje‐daero, Deokjin‐guJeonju‐siJeollabuk‐do54896Republic of Korea
| | - Ambrose Ashwin Melvin
- Department of Chemical & Biomolecular EngineeringSogang University35 Baekbeom‐ro, Mapo‐guSeoul04107Republic of Korea
| | - Jinho Yoon
- Department of Biomedical‐Chemical EngineeringThe Catholic University of Korea43 Jibong‐ro, Wonmi‐guBucheon‐siGyeonggi‐do14662Republic of Korea
- Department of BiotechnologyThe Catholic University of Korea43 Jibong‐ro, Wonmi‐guBucheon‐siGyeonggi‐do14662Republic ofKorea
| | - Jeong‐Woo Choi
- Department of Chemical & Biomolecular EngineeringSogang University35 Baekbeom‐ro, Mapo‐guSeoul04107Republic of Korea
| |
Collapse
|
14
|
Wei SY, Chen PY, Hsieh CC, Chen YS, Chen TH, Yu YS, Tsai MC, Xie RH, Chen GY, Yin GC, Melero-Martin JM, Chen YC. Engineering large and geometrically controlled vascularized nerve tissue in collagen hydrogels to restore large-sized volumetric muscle loss. Biomaterials 2023; 303:122402. [PMID: 37988898 PMCID: PMC11606314 DOI: 10.1016/j.biomaterials.2023.122402] [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: 08/04/2023] [Revised: 09/29/2023] [Accepted: 11/13/2023] [Indexed: 11/23/2023]
Abstract
Developing scalable vascularized and innervated tissue is a critical challenge for the successful clinical application of tissue-engineered constructs. Collagen hydrogels are extensively utilized in cell-mediated vascular network formation because of their naturally excellent biological properties. However, the substantial increase in hydrogel contraction induced by populated cells limits their long-term use. Previous studies attempted to mitigate this issue by concentrating collagen pre-polymer solutions or synthesizing covalently crosslinked collagen hydrogels. However, these methods only partially reduce hydrogel contraction while hindering blood vessel formation within the hydrogels. To address this challenge, we introduced additional support in the form of a supportive spacer to counteract the contraction forces of populated cells and prevent hydrogel contraction. This approach was found to promote cell spreading, resist hydrogel contraction, control hydrogel/tissue geometry, and even facilitate the engineering of functional blood vessels and host nerve growth in just one week. Subsequently, implanting these engineered tissues into muscle defect sites resulted in timely anastomosis with the host vasculature, leading to enhanced myogenesis, increased muscle innervation, and the restoration of injured muscle functionality. Overall, this innovative strategy expands the applicability of collagen hydrogels in fabricating large vascularized nerve tissue constructs for repairing volumetric muscle loss (∼63 %) and restoring muscle function.
Collapse
Affiliation(s)
- Shih-Yen Wei
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Po-Yu Chen
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Chia-Chang Hsieh
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Yu-Shan Chen
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Tzu-Hsuan Chen
- Department of Materials Science and Engineering, Carnegie Mellon University, PA, USA
| | - Yu-Shan Yu
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Min-Chun Tsai
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan
| | - Ren-Hao Xie
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Guan-Yu Chen
- Department of Electrical and Computer Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Institute of Biomedical Engineering, College of Electrical and Computer Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Gung-Chian Yin
- National Synchrotron Radiation Research Center, Hsinchu, Taiwan
| | - Juan M Melero-Martin
- Department of Cardiac Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA; Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Ying-Chieh Chen
- Department of Materials Science and Engineering, National Tsing-Hua University, Hsinchu, Taiwan.
| |
Collapse
|
15
|
Liu Q, Yu M, Liao M, Ran Z, Tang X, Hu J, Su B, Fu G, Wu Q. The ratio of alpha-calcitonin gene-related peptide to substance P is associated with the transition of bone metabolic states during aging and healing. J Mol Histol 2023; 54:689-702. [PMID: 37857924 DOI: 10.1007/s10735-023-10167-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 09/30/2023] [Indexed: 10/21/2023]
Abstract
Alpha-calcitonin gene-related peptide (αCGRP) and substance P (SP) are functionally correlated sensory neuropeptides deeply involved in bone homeostasis. However, they are usually studied individually rather than as an organic whole. To figure out whether they are interdependent, we firstly recorded the real-time αCGRP and SP levels in aging bone and healing fracture, which revealed a moderate to high level of αCGRP coupled with a low αCGRP/SP ratio in an anabolic state, and a high level of αCGRP coupled with a high αCGRP/SP ratio in a catabolic state, suggesting the importance of αCGRP/SP ratio in driving aging and healing scenarios. During facture healing, increase in αCGRP/SP ratio by adding αCGRP led to better callus formation and faster callus remodeling, while simultaneous addition of αCGRP and SP resulted in hypertrophic callus and delayed remodeling. The characteristics in inflammation and osteoclast activation further confirmed the importance of high αCGRP/SP ratio during catabolic bone remodeling. In vitro assays using different mixtures of αCGRP-SP proved that the osteogenic potential of the mixtures depended mostly on αCGRP, while their effects on osteoclasts and neutrophils relied on both peptides. These results demonstrated that αCGRP and SP were spatiotemporally interdependent. The αCGRP/SP ratio may be more important than the dose of a single neuropeptide in managing age-related and trauma-related bone diseases.
Collapse
Affiliation(s)
- Qianzi Liu
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, 400015, China
| | - Minxuan Yu
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, 400015, China
| | - Menglin Liao
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, 400015, China
| | - Zhiyue Ran
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, 400015, China
| | - Xiaofeng Tang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, 400015, China
| | - Jun Hu
- Department of Stomatology, Qijiang District People's Hospital, Chongqing, 401420, China
| | - Beiju Su
- Chongqing Dazu District Hospital of Traditional Chinese Medicine, Chongqing, 402360, China
| | - Gang Fu
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, 400015, China.
- Department of Oral Implantology, Stomatological Hospital of Chongqing Medical University, Chongqing, 400015, China.
| | - Qingqing Wu
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, 400015, China.
- Department of Oral Implantology, Stomatological Hospital of Chongqing Medical University, Chongqing, 400015, China.
| |
Collapse
|
16
|
Isosaari L, Vuorenpää H, Yrjänäinen A, Kapucu FE, Kelloniemi M, Pakarinen TK, Miettinen S, Narkilahti S. Simultaneous induction of vasculature and neuronal network formation on a chip reveals a dynamic interrelationship between cell types. Cell Commun Signal 2023; 21:132. [PMID: 37316873 DOI: 10.1186/s12964-023-01159-4] [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: 03/14/2023] [Accepted: 05/06/2023] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND Neuronal networks receive and deliver information to regulate bodily functions while the vascular network provides oxygen, nutrients, and signaling molecules to tissues. Neurovascular interactions are vital for both tissue development and maintaining homeostasis in adulthood; these two network systems align and reciprocally communicate with one another. Although communication between network systems has been acknowledged, the lack of relevant in vitro models has hindered research at the mechanistic level. For example, the current used in vitro neurovascular models are typically established to be short-term (≤ 7 days) culture models, and they miss the supporting vascular mural cells. METHODS In this study, we utilized human induced pluripotent stem cell (hiPSC) -derived neurons, fluorescence tagged human umbilical vein endothelial cells (HUVECs), and either human bone marrow or adipose stem/stromal cells (BMSCs or ASCs) as the mural cell types to create a novel 3D neurovascular network-on-a-chip model. Collagen 1-fibrin matrix was used to establish long-term (≥ 14 days) 3D cell culture in a perfusable microphysiological environment. RESULTS Aprotinin-supplemented endothelial cell growth medium-2 (EGM-2) supported the simultaneous formation of neuronal networks, vascular structures, mural cell differentiation, and the stability of the 3D matrix. The formed neuronal and vascular networks were morphologically and functionally characterized. Neuronal networks supported vasculature formation based on direct cell contacts and by dramatically increasing the secretion of angiogenesis-related factors in multicultures in contrast to cocultures without neurons. Both utilized mural cell types supported the formation of neurovascular networks; however, the BMSCs seemed to boost neurovascular networks to greater extent. CONCLUSIONS Overall, our study provides a novel human neurovascular network model that is applicable for creating in vivo-like tissue models with intrinsic neurovascular interactions. The 3D neurovascular network model on chip forms an initial platform for the development of vascularized and innervated organ-on-chip and further body-on-chip concepts and offers the possibility for mechanistic studies on neurovascular communication both under healthy and in disease conditions. Video Abstract.
Collapse
Affiliation(s)
- Lotta Isosaari
- NeuroGroup, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Hanna Vuorenpää
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Alma Yrjänäinen
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Fikret Emre Kapucu
- NeuroGroup, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Minna Kelloniemi
- Department of Plastic and Reconstructive Surgery, Tampere University Hospital, Tampere, Finland
| | | | - Susanna Miettinen
- Adult Stem Cell Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Susanna Narkilahti
- NeuroGroup, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| |
Collapse
|
17
|
Redigolo L, Sanfilippo V, La Mendola D, Forte G, Satriano C. Bioinspired Nanoplatforms Based on Graphene Oxide and Neurotrophin-Mimicking Peptides. MEMBRANES 2023; 13:membranes13050489. [PMID: 37233550 DOI: 10.3390/membranes13050489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/22/2023] [Accepted: 04/26/2023] [Indexed: 05/27/2023]
Abstract
Neurotrophins (NTs), which are crucial for the functioning of the nervous system, are also known to regulate vascularization. Graphene-based materials may drive neural growth and differentiation, and, thus, have great potential in regenerative medicine. In this work, we scrutinized the nano-biointerface between the cell membrane and hybrids made of neurotrophin-mimicking peptides and graphene oxide (GO) assemblies (pep-GO), to exploit their potential in theranostics (i.e., therapy and imaging/diagnostics) for targeting neurodegenerative diseases (ND) as well as angiogenesis. The pep-GO systems were assembled via spontaneous physisorption onto GO nanosheets of the peptide sequences BDNF(1-12), NT3(1-13), and NGF(1-14), mimicking the brain-derived neurotrophic factor (BDNF), the neurotrophin 3 (NT3), and the nerve growth factor (NGF), respectively. The interaction of pep-GO nanoplatforms at the biointerface with artificial cell membranes was scrutinized both in 3D and 2D by utilizing model phospholipids self-assembled as small unilamellar vesicles (SUVs) or planar-supported lipid bilayers (SLBs), respectively. The experimental studies were paralleled via molecular dynamics (MD) computational analyses. Proof-of-work in vitro cellular experiments with undifferentiated neuroblastoma (SH-SY5Y), neuron-like, differentiated neuroblastoma (dSH-SY5Y), and human umbilical vein endothelial cells (HUVECs) were carried out to shed light on the capability of the pep-GO nanoplatforms to stimulate the neurite outgrowth as well as tubulogenesis and cell migration.
Collapse
Affiliation(s)
- Luigi Redigolo
- Nano Hybrid Biointerfaces Lab (NHBIL), Department of Chemical Sciences, University of Catania, Viale Andrea Doria, 6, 95125 Catania, Italy
| | - Vanessa Sanfilippo
- Nano Hybrid Biointerfaces Lab (NHBIL), Department of Chemical Sciences, University of Catania, Viale Andrea Doria, 6, 95125 Catania, Italy
| | - Diego La Mendola
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano, 6, 56126 Pisa, Italy
| | - Giuseppe Forte
- Department of Drug and Health Science, University of Catania, Viale Andrea Doria, 6, 95125 Catania, Italy
| | - Cristina Satriano
- Nano Hybrid Biointerfaces Lab (NHBIL), Department of Chemical Sciences, University of Catania, Viale Andrea Doria, 6, 95125 Catania, Italy
| |
Collapse
|
18
|
Kumar V, Kingsley D, Perikamana SM, Mogha P, Goodwin CR, Varghese S. Self-assembled innervated vasculature-on-a-chip to study nociception. Biofabrication 2023; 15:10.1088/1758-5090/acc904. [PMID: 36996841 PMCID: PMC10152403 DOI: 10.1088/1758-5090/acc904] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 03/30/2023] [Indexed: 04/01/2023]
Abstract
Nociceptor sensory neurons play a key role in eliciting pain. An active crosstalk between nociceptor neurons and the vascular system at the molecular and cellular level is required to sense and respond to noxious stimuli. Besides nociception, interaction between nociceptor neurons and vasculature also contributes to neurogenesis and angiogenesis.In vitromodels of innervated vasculature can greatly help delineate these roles while facilitating disease modeling and drug screening. Herein, we report the development of a microfluidic-assisted tissue model of nociception in the presence of microvasculature. The self-assembled innervated microvasculature was engineered using endothelial cells and primary dorsal root ganglion (DRG) neurons. The sensory neurons and the endothelial cells displayed distinct morphologies in presence of each other. The neurons exhibited an elevated response to capsaicin in the presence of vasculature. Concomitantly, increased transient receptor potential cation channel subfamily V member 1 (TRPV1) receptor expression was observed in the DRG neurons in presence of vascularization. Finally, we demonstrated the applicability of this platform for modeling nociception associated with tissue acidosis. While not demonstrated here, this platform could also serve as a tool to study pain resulting from vascular disorders while also paving the way towards the development of innervated microphysiological models.
Collapse
Affiliation(s)
- Vardhman Kumar
- Department of Biomedical Engineering, Duke University, Durham NC
| | - David Kingsley
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham NC
| | | | - Pankaj Mogha
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham NC
| | - C Rory Goodwin
- Department of Neurosurgery, Spine Division, Duke University Medical Center, Durham, NC
| | - Shyni Varghese
- Department of Biomedical Engineering, Duke University, Durham NC
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham NC
- Department of Mechanical Engineering and Material Science, Duke University, Durham NC
| |
Collapse
|
19
|
Singh S, Fereshetyan K, Shorter S, Paliokha R, Dremencov E, Yenkoyan K, Ovsepian SV. Brain-derived neurotrophic factor (BDNF) in perinatal depression: Side show or pivotal factor? Drug Discov Today 2023; 28:103467. [PMID: 36528281 DOI: 10.1016/j.drudis.2022.103467] [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: 07/21/2022] [Revised: 12/03/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022]
Abstract
Perinatal depression is the most common psychiatric complication of pregnancy, with its detrimental effects on maternal and infant health widely underrated. There is a pressing need for specific molecular biomarkers, with pregnancy-related decline in brain-derived neurotrophic factor (BDNF) in the blood and downregulation of TrkB receptor in the brain reported in clinical and preclinical studies. In this review, we explore the emerging role of BDNF in reproductive biology and discuss evidence suggesting its deficiency as a risk factor for perinatal depression. With the increasing evidence for restoration of serum BDNF levels by antidepressant therapy, the strengthening association of perinatal depression with deficiency of BDNF supports its potential as a surrogate endpoint for preclinical and clinical studies.
Collapse
Affiliation(s)
- Saumya Singh
- Faculty of Science and Engineering, University of Greenwich London, Chatham Maritime, Kent ME4 4TB, UK
| | - Katarine Fereshetyan
- Neuroscience Laboratory, Cobrain Center, Yerevan State Medical University of M. Heratsi, 0025, Yerevan, Armenia
| | - Susan Shorter
- Faculty of Science and Engineering, University of Greenwich London, Chatham Maritime, Kent ME4 4TB, UK
| | - Ruslan Paliokha
- Centre of Biosciences, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Eliyahu Dremencov
- Centre of Biosciences, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Konstantin Yenkoyan
- Neuroscience Laboratory, Cobrain Center, Yerevan State Medical University of M. Heratsi, 0025, Yerevan, Armenia
| | - Saak V Ovsepian
- Faculty of Science and Engineering, University of Greenwich London, Chatham Maritime, Kent ME4 4TB, UK.
| |
Collapse
|
20
|
Bonaterra GA, Schmitt J, Schneider K, Schwarzbach H, Aziz-Kalbhenn H, Kelber O, Müller J, Kinscherf R. Phytohustil ® and root extract of Althaea officinalis L. exert anti-inflammatory and anti-oxidative properties and improve the migratory capacity of endothelial cells in vitro. Front Pharmacol 2022; 13:948248. [PMID: 36569306 PMCID: PMC9773075 DOI: 10.3389/fphar.2022.948248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022] Open
Abstract
Introduction: Althaea officinalis L.'s root extract (REA) has been used as a medicinal plant since ancient times to treat a cough. Applying REA leads to a protective film that induces a faster regeneration of the lesioned laryngopharyngeal mucosa caused by dry coughs. The buccopharyngeal mucosa is a highly vascularized tissue. In this regard, anti-inflammatory/-oxidant phytochemicals that improve the repair of the lesion site, e.g., neovascularization in the wound, are critical for promoting healing. For this reason, it is essential to investigate the effects of Phytohustil® and REA on different cellular components of the mucosa under conditions similar to those found in the injured mucosa. Thus, this in vitro study investigated the anti-inflammatory/oxidative and pro-migratory properties of Phytohustil® cough syrup on vascular endothelial cells. Methods: Human umbilical vein endothelial cells (HUVEC) were pretreated (24 h) with Phytohustil®, its excipients, or REA, followed by incubation with hydrogen peroxide (H2O2; 1 h; pro-oxidative) or with lipopolysaccharides (LPS; 3 h; pro-inflammatory). Viability and cytotoxicity were measured by PrestoBlue® assay. Intracellular reactive oxygen species (ROS) were quantified with 20-70-dichlorofluorescein diacetate (DCFDA). The release of interleukin 6 (IL6) was determined by enzyme-linked immunosorbent assay (ELISA). The migratory capacity of HUVEC was measured using a scratch assay. Results: Our results show that Phytohustil®, its excipients and REA were not cytotoxic. Pretreatment of HUVEC (24 h) with Phytohustil® or REA inhibited the LPS-activated IL6 release. Phytohustil® or REA inhibited the H2O2-induced cytotoxicity and intracellular ROS production. Phytohustil® and REA significantly stimulated wound closure compared to the control. Conclusion: Our data show that Phytohustil® and REA have anti-inflammatory/-oxidant properties and improve the migratory capacity of vascular endothelial cells. These properties may contribute to the healing characteristics of Phytohustil® and support the benefit of Phytohustil® in patient's treatment of irritated oral mucosa.
Collapse
Affiliation(s)
- Gabriel A. Bonaterra
- Department of Medical Cell Biology, Anatomy and Cell Biology, University of Marburg, Marburg, Germany,*Correspondence: Gabriel A. Bonaterra,
| | - Johanna Schmitt
- Department of Medical Cell Biology, Anatomy and Cell Biology, University of Marburg, Marburg, Germany
| | - Kim Schneider
- Department of Medical Cell Biology, Anatomy and Cell Biology, University of Marburg, Marburg, Germany
| | - Hans Schwarzbach
- Department of Medical Cell Biology, Anatomy and Cell Biology, University of Marburg, Marburg, Germany
| | - Heba Aziz-Kalbhenn
- Bayer Consumer Health Division, Phytomedicines Supply and Development Center, Steigerwald Arzneimittelwerk GmbH, Darmstadt, Germany
| | - Olaf Kelber
- Bayer Consumer Health Division, Phytomedicines Supply and Development Center, Steigerwald Arzneimittelwerk GmbH, Darmstadt, Germany
| | - Jürgen Müller
- Bayer Consumer Health Division, Phytomedicines Supply and Development Center, Steigerwald Arzneimittelwerk GmbH, Darmstadt, Germany
| | - Ralf Kinscherf
- Department of Medical Cell Biology, Anatomy and Cell Biology, University of Marburg, Marburg, Germany
| |
Collapse
|
21
|
Łuczkowska K, Kulig P, Baumert B, Machaliński B. Brain-derived neurotrophic factor: focus on the pathogenesis of multiple myeloma and the development of treatment-induced peripheral neuropathy. Leuk Lymphoma 2022; 63:3044-3051. [PMID: 35999712 DOI: 10.1080/10428194.2022.2113535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
For many years, intensive research has been carried out on the in-depth understanding of the pathogenesis of multiple myeloma (MM). Nevertheless, the multifactorial nature of the disease, the development of drug resistance, and the side effects of therapy, make it difficult to effectively treat patients. One of the many factors involved in the pathogenesis of MM is brain-derived neurotrophic factor (BDNF). This factor is widely described as a neuroregenerative and neuroprotective agent, but it also regulates non-neuronal cell functions, such as proliferation, apoptosis, and viability. Therefore, BDNF appears to be a good therapeutic target in MM. On the other hand, its decreased concentration during treatment closely correlates with the development of peripheral neuropathy (PN). BDNF dualism requires a detailed understanding of its action on individual molecular mechanisms. Perhaps the optimization of the BDNF level will contribute to the improvement of MM treatment and the reduction of chemotherapy side effects.
Collapse
Affiliation(s)
- Karolina Łuczkowska
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Piotr Kulig
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Bartłomiej Baumert
- Department of Bone Marrow Transplantation, Pomeranian Medical University, Szczecin, Poland
| | - Bogusław Machaliński
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland.,Department of Bone Marrow Transplantation, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
22
|
Sefiani A, Rusyn I, Geoffroy CG. Novel adult cortical neuron processing and screening method illustrates sex- and age-dependent effects of pharmaceutical compounds. Sci Rep 2022; 12:13125. [PMID: 35908049 PMCID: PMC9338961 DOI: 10.1038/s41598-022-17389-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 07/25/2022] [Indexed: 11/22/2022] Open
Abstract
Neurodegenerative diseases and neurotraumatic injuries are typically age-associated disorders that can reduce neuron survival, neurite outgrowth, and synaptic plasticity leading to loss of cognitive capacity, executive function, and motor control. In pursuit of reducing the loss of said neurological functions, novel compounds are sought that promote neuron viability, neuritogenesis, and/or synaptic plasticity. Current high content in vitro screenings typically use cells that are iPSC-derived, embryonic, or originate from post-natal tissues; however, most patients suffering from neurodegenerative diseases and neurotrauma are of middle-age and older. The chasm in maturity between the neurons used in drug screens and those in a target population is a barrier for translational success of in vitro results. It has been historically challenging to culture adult neurons let alone conduct screenings; therefore, age-appropriate drug screenings have previously not been plausible. We have modified Miltenyi's protocol to increase neuronal yield, neuron purity, and neural viability at a reduced cost to expand our capacity to screen compounds directly in primary adult neurons. To our knowledge, we developed the first morphology-based screening system using adult cortical neurons and the first to incorporate age and sex as biological variables in a screen using adult cortical neurons. By using primary adult cortical neurons from mice that were 4 to 48 weeks old for screening pharmaceutical agents, we have demonstrated age- and sex-dependent effects on neuritogenesis and neuron survival in vitro. Utilizing age- and sex-appropriate in vitro models to find novel compounds increasing neuron survival and neurite outgrowth, made possible by our modified adult neuron processing method, will greatly increase the relevance of in vitro screening for finding neuroprotective compounds.
Collapse
Affiliation(s)
- Arthur Sefiani
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University, Bryan, TX, 77807, USA
| | - Ivan Rusyn
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Cédric G Geoffroy
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University, Bryan, TX, 77807, USA.
| |
Collapse
|
23
|
Malheiro A, Seijas-Gamardo A, Harichandan A, Mota C, Wieringa P, Moroni L. Development of an In Vitro Biomimetic Peripheral Neurovascular Platform. ACS APPLIED MATERIALS & INTERFACES 2022; 14:31567-31585. [PMID: 35815638 PMCID: PMC9305708 DOI: 10.1021/acsami.2c03861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Nerves and blood vessels are present in most organs and are indispensable for their function and homeostasis. Within these organs, neurovascular (NV) tissue forms congruent patterns and establishes vital interactions. Several human pathologies, including diabetes type II, produce NV disruptions with serious consequences that are complicated to study using animal models. Complex in vitro organ platforms, with neural and vascular supply, allow the investigation of such interactions, whether in a normal or pathological context, in an affordable, simple, and direct manner. To date, a few in vitro models contain NV tissue, and most strategies report models with nonbiomimetic representations of the native environment. To this end, we have established here an NV platform that contains mature vasculature and neural tissue, composed of human microvascular endothelial cells (HMVECs), induced pluripotent stem cell (iPSCs)-derived sensory neurons, and primary rat Schwann cells (SCs) within a fibrin-embedded polymeric scaffold. First, we show that SCs can induce the formation of and stabilize vascular networks to the same degree as the traditional and more thoroughly studied human dermal fibroblasts (HDFs). We also show that through SC prepatterning, we are able to control vessel orientation. Using our NV platform, we demonstrate the concomitant formation of three-dimensional neural and vascular tissue, and the influence of different medium formulations and cell types on the NV tissue outcome. Finally, we propose a protocol to form mature NV tissue, via the integration of independent neural and vascular constituents. The platform described here provides a versatile and advanced model for in vitro research of the NV axis.
Collapse
Affiliation(s)
- Afonso Malheiro
- Complex Tissue Regeneration
Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ET Maastricht, The Netherlands
| | - Adrián Seijas-Gamardo
- Complex Tissue Regeneration
Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ET Maastricht, The Netherlands
| | - Abhishek Harichandan
- Complex Tissue Regeneration
Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ET Maastricht, The Netherlands
| | - Carlos Mota
- Complex Tissue Regeneration
Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ET Maastricht, The Netherlands
| | - Paul Wieringa
- Complex Tissue Regeneration
Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ET Maastricht, The Netherlands
| | - Lorenzo Moroni
- Complex Tissue Regeneration
Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ET Maastricht, The Netherlands
| |
Collapse
|
24
|
Neto E, Monteiro AC, Leite Pereira C, Simões M, Conde JP, Chu V, Sarmento B, Lamghari M. Micropathological Chip Modeling the Neurovascular Unit Response to Inflammatory Bone Condition. Adv Healthc Mater 2022; 11:e2102305. [PMID: 35158409 PMCID: PMC11468530 DOI: 10.1002/adhm.202102305] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/12/2022] [Indexed: 12/17/2022]
Abstract
Organ-on-a-chip in vitro platforms accurately mimic complex microenvironments offering the ability to recapitulate and dissect mechanisms of physiological and pathological settings, revealing their major importance to develop new therapeutic targets. Bone diseases, such as osteoarthritis, are extremely complex, comprising of the action of inflammatory mediators leading to unbalanced bone homeostasis and de-regulation of sensory innervation and angiogenesis. Although there are models to mimic bone vascularization or innervation, in vitro platforms merging the complexity of bone, vasculature, innervation, and inflammation are missing. Therefore, in this study a microfluidic-based neuro-vascularized bone chip (NVB chip) is proposed to 1) model the mechanistic interactions between innervation and angiogenesis in the inflammatory bone niche, and 2) explore, as a screening tool, novel strategies targeting inflammatory diseases, using a nano-based drug delivery system. It is possible to set the design of the platform and achieve the optimized conditions to address the neurovascular network under inflammation. Moreover, this system is validated by delivering anti-inflammatory drug-loaded nanoparticles to counteract the neuronal growth associated with pain perception. This reliable in vitro tool will allow understanding the bone neurovascular system, enlightening novel mechanisms behind the inflammatory bone diseases, bone destruction, and pain opening new avenues for new therapies discovery.
Collapse
Affiliation(s)
- Estrela Neto
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
- INEB – Instituto Nacional de Engenharia BiomédicaUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
| | - Ana Carolina Monteiro
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
- INEB – Instituto Nacional de Engenharia BiomédicaUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
| | - Catarina Leite Pereira
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
- INEB – Instituto Nacional de Engenharia BiomédicaUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
| | - Miguel Simões
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
- INEB – Instituto Nacional de Engenharia BiomédicaUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
| | - João Pedro Conde
- Instituto de Engenharia de Sistemas e Computadores (INESC)Microsystems and NanotechnologiesRua Alves Redol, 91000‐029LisboaPortugal
| | - Virginia Chu
- Instituto de Engenharia de Sistemas e Computadores (INESC)Microsystems and NanotechnologiesRua Alves Redol, 91000‐029LisboaPortugal
| | - Bruno Sarmento
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
- INEB – Instituto Nacional de Engenharia BiomédicaUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
- CESPUInstituto de Investigação e Formação Avançada em Ciências e Tecnologias da SaúdeRua Central da Gandra, 137Gandra4585‐116Portugal
| | - Meriem Lamghari
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
- INEB – Instituto Nacional de Engenharia BiomédicaUniversidade do PortoRua Alfredo Allen, 208Porto4200‐135Portugal
| |
Collapse
|
25
|
Mikhailova MM, Sydoruk KV, Davydova LI, Yastremsky EV, Chvalun SN, Debabov VG, Bogush VG, Panteleyev AA. Nonwoven spidroin materials as scaffolds for ex vivo cultivation of aortic fragments and dorsal root ganglia. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2022; 33:1685-1703. [PMID: 35499451 DOI: 10.1080/09205063.2022.2073426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Recombinant spidroins (RS; the analogues of silk proteins of spider's web) have multiple properties beneficial for bioengineering, including their suitability for electrospinning and thus, for production of materials with oriented fibers. This makes RS-based matrices potentially effective in stimulating regeneration of peripheral nerves. The restoration of injured nerves also depends on prompt regrowth of blood vessels. Therefore, prospective scaffold materials for neuro-regenerative therapy should positively affect both the nerves and the blood vessels. Currently, the experimental models suitable for culturing and quantitative assessment of the vascular and neuronal cells on the same material are lacking. Here, we assessed the suitability of electrospun RS-based matrices for cultivation of the mouse aorta and dorsal root ganglia (DRG) explants. We also quantified the effects of matrix topography upon both types of tissues. The RS-based materials have effectively supported aortic explants survival and sprouting. The cumulative length of endothelial sprouts on rS1/9-coated inserts was significantly higher as compared to type I collagen coatings, suggesting stimulatory effects on angiogenesis in vitro. In contrast to matrices with random fibers, on matrices with parallel fibers the migration of both smooth muscle and endothelial cells was highly oriented. Furthermore, alignment of RS fibers effectively directs the growth of axons and the migration of Schwann cells from DRGs. Thus, the electrospun RS matrices are highly suitable to culture both, the DRGs and aortic explants and to study the effects of matrix topography on cell migration. This model has a high potential for further endeavor into interactions of nerve and vascular cells and tissues.
Collapse
Affiliation(s)
| | - Konstantin V Sydoruk
- National Research Centre «Kurchatov Institute», Moscow, Russia.,National Research Centre «Kurchatov Institute» - GosNIIGenetika, Moscow, Russia
| | - Lubov I Davydova
- National Research Centre «Kurchatov Institute», Moscow, Russia.,National Research Centre «Kurchatov Institute» - GosNIIGenetika, Moscow, Russia
| | - Evgeniy V Yastremsky
- National Research Centre «Kurchatov Institute», Moscow, Russia.,Shubnikov Institute of Crystallography of FSRC "Crystallography and Photonics" RAS, Moscow, Russia
| | | | - Vladimir G Debabov
- National Research Centre «Kurchatov Institute», Moscow, Russia.,National Research Centre «Kurchatov Institute» - GosNIIGenetika, Moscow, Russia
| | - Vladimir G Bogush
- National Research Centre «Kurchatov Institute», Moscow, Russia.,National Research Centre «Kurchatov Institute» - GosNIIGenetika, Moscow, Russia
| | | |
Collapse
|
26
|
Roca FG, Santos LG, Roig MM, Medina LM, Martínez-Ramos C, Pradas MM. Novel Tissue-Engineered Multimodular Hyaluronic Acid-Polylactic Acid Conduits for the Regeneration of Sciatic Nerve Defect. Biomedicines 2022; 10:biomedicines10050963. [PMID: 35625700 PMCID: PMC9138968 DOI: 10.3390/biomedicines10050963] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/14/2022] [Accepted: 04/18/2022] [Indexed: 02/01/2023] Open
Abstract
The gold standard for the treatment of peripheral nerve injuries, the autograft, presents several drawbacks, and engineered constructs are currently suitable only for short gaps or small diameter nerves. Here, we study a novel tissue-engineered multimodular nerve guidance conduit for the treatment of large nerve damages based in a polylactic acid (PLA) microfibrillar structure inserted inside several co-linear hyaluronic acid (HA) conduits. The highly aligned PLA microfibers provide a topographical cue that guides axonal growth, and the HA conduits play the role of an epineurium and retain the pre-seeded auxiliary cells. The multimodular design increases the flexibility of the device. Its performance for the regeneration of a critical-size (15 mm) rabbit sciatic nerve defect was studied and, after six months, very good nerve regeneration was observed. The multimodular approach contributed to a better vascularization through the micrometrical gaps between HA conduits, and the pre-seeded Schwann cells increased axonal growth. Six months after surgery, a cross-sectional available area occupied by myelinated nerve fibers above 65% at the central and distal portions was obtained when the multimodular device with pre-seeded Schwann cells was employed. The results validate the multi-module approach for the regeneration of large nerve defects and open new possibilities for surgical solutions in this field.
Collapse
Affiliation(s)
- Fernando Gisbert Roca
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, 46022 Valencia, Spain; (F.G.R.); (L.G.S.); (C.M.-R.)
| | - Luis Gil Santos
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, 46022 Valencia, Spain; (F.G.R.); (L.G.S.); (C.M.-R.)
| | - Manuel Mata Roig
- Department of Pathology, Faculty of Medicine and Dentistry, Universitat de València, 46010 Valencia, Spain; (M.M.R.); (L.M.M.)
| | - Lara Milian Medina
- Department of Pathology, Faculty of Medicine and Dentistry, Universitat de València, 46010 Valencia, Spain; (M.M.R.); (L.M.M.)
| | - Cristina Martínez-Ramos
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, 46022 Valencia, Spain; (F.G.R.); (L.G.S.); (C.M.-R.)
- Unitat Predepartamental de Medicina, Universitat Jaume I, 12071 Castellón de la Plana, Spain
| | - Manuel Monleón Pradas
- Center for Biomaterials and Tissue Engineering, Universitat Politècnica de València, 46022 Valencia, Spain; (F.G.R.); (L.G.S.); (C.M.-R.)
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-963-877000
| |
Collapse
|
27
|
Endothelial Cell Metabolism in Vascular Functions. Cancers (Basel) 2022; 14:cancers14081929. [PMID: 35454836 PMCID: PMC9031281 DOI: 10.3390/cancers14081929] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Recent findings in the field of vascular biology are nourishing the idea that targeting the endothelial cell metabolism may be an alternative strategy to antiangiogenic therapy, as well as a novel therapeutic approach for cardiovascular disease. Deepening the molecular mechanisms regulating how ECs re-adapt their metabolic status in response to the changeable conditions of the tissue microenvironment may be beneficial to develop novel innovative treatments to counteract the aberrant growth of vasculature. Abstract The endothelium is the innermost layer of all blood and lymphatic vessels composed of a monolayer of specialized endothelial cells (ECs). It is regarded as a dynamic and multifunctional endocrine organ that takes part in essential processes, such as the control of blood fluidity, the modulation of vascular tone, the regulation of immune response and leukocyte trafficking into perivascular tissues, and angiogenesis. The inability of ECs to perform their normal biological functions, known as endothelial dysfunction, is multi-factorial; for instance, it implicates the failure of ECs to support the normal antithrombotic and anti-inflammatory status, resulting in the onset of unfavorable cardiovascular conditions such as atherosclerosis, coronary artery disease, hypertension, heart problems, and other vascular pathologies. Notably, it is emerging that the ability of ECs to adapt their metabolic status to persistent changes of the tissue microenvironment could be vital for the maintenance of vascular functions and to prevent adverse vascular events. The main purpose of the present article is to shed light on the unique metabolic plasticity of ECs as a prospective therapeutic target; this may lead to the development of novel strategies for cardiovascular diseases and cancer.
Collapse
|
28
|
Li Y, Fraser D, Mereness J, Van Hove A, Basu S, Newman M, Benoit DSW. Tissue Engineered Neurovascularization Strategies for Craniofacial Tissue Regeneration. ACS APPLIED BIO MATERIALS 2022; 5:20-39. [PMID: 35014834 PMCID: PMC9016342 DOI: 10.1021/acsabm.1c00979] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Craniofacial tissue injuries, diseases, and defects, including those within bone, dental, and periodontal tissues and salivary glands, impact an estimated 1 billion patients globally. Craniofacial tissue dysfunction significantly reduces quality of life, and successful repair of damaged tissues remains a significant challenge. Blood vessels and nerves are colocalized within craniofacial tissues and act synergistically during tissue regeneration. Therefore, the success of craniofacial regenerative approaches is predicated on successful recruitment, regeneration, or integration of both vascularization and innervation. Tissue engineering strategies have been widely used to encourage vascularization and, more recently, to improve innervation through host tissue recruitment or prevascularization/innervation of engineered tissues. However, current scaffold designs and cell or growth factor delivery approaches often fail to synergistically coordinate both vascularization and innervation to orchestrate successful tissue regeneration. Additionally, tissue engineering approaches are typically investigated separately for vascularization and innervation. Since both tissues act in concert to improve craniofacial tissue regeneration outcomes, a revised approach for development of engineered materials is required. This review aims to provide an overview of neurovascularization in craniofacial tissues and strategies to target either process thus far. Finally, key design principles are described for engineering approaches that will support both vascularization and innervation for successful craniofacial tissue regeneration.
Collapse
Affiliation(s)
- Yiming Li
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - David Fraser
- Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States.,Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, New York 14620, United States.,Translational Biomedical Sciences Program, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Jared Mereness
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States.,Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Amy Van Hove
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Sayantani Basu
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Maureen Newman
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Danielle S W Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Orthopaedics and Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York 14642, United States.,Eastman Institute for Oral Health, University of Rochester Medical Center, Rochester, New York 14620, United States.,Translational Biomedical Sciences Program, University of Rochester Medical Center, Rochester, New York 14642, United States.,Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York 14642, United States.,Materials Science Program, University of Rochester, Rochester, New York 14627, United States.,Department of Chemical Engineering, University of Rochester, Rochester, New York 14627, United States.,Department of Biomedical Genetics and Center for Oral Biology, University of Rochester Medical Center, Rochester, New York 14642, United States
| |
Collapse
|
29
|
Zheng T, Wu L, Sun S, Xu J, Han Q, Liu Y, Wu R, Li G. Co-culture of Schwann cells and endothelial cells for synergistically regulating dorsal root ganglion behavior on chitosan-based anisotropic topology for peripheral nerve regeneration. BURNS & TRAUMA 2022; 10:tkac030. [PMID: 36071954 PMCID: PMC9444262 DOI: 10.1093/burnst/tkac030] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/20/2022] [Accepted: 05/06/2022] [Indexed: 11/29/2022]
Abstract
Background Anisotropic topologies are known to regulate cell-oriented growth and induce cell differentiation, which is conducive to accelerating nerve regeneration, while co-culture of endothelial cells (ECs) and Schwann cells (SCs) can significantly promote the axon growth of dorsal root ganglion (DRG). However, the synergistic regulation of EC and SC co-culture of DRG behavior on anisotropic topologies is still rarely reported. The study aims to investigate the effect of anisotropic topology co-cultured with Schwann cells and endothelial cells on dorsal root ganglion behavior for promoting peripheral nerve regeneration. Methods Chitosan/artemisia sphaerocephala (CS/AS) scaffolds with anisotropic topology were first prepared using micro-molding technology, and then the surface was modified with dopamine to facilitate cell adhesion and growth. The physical and chemical properties of the scaffolds were characterized through morphology, wettability, surface roughness and component variation. SCs and ECs were co-cultured with DRG cells on anisotropic topology scaffolds to evaluate the axon growth behavior. Results Dopamine-modified topological CS/AS scaffolds had good hydrophilicity and provided an appropriate environment for cell growth. Cellular immunofluorescence showed that in contrast to DRG growth alone, co-culture of SCs and ECs could not only promote the growth of DRG axons, but also offered a stronger guidance for orientation growth of neurons, which could effectively prevent axons from tangling and knotting, and thus may significantly inhibit neurofibroma formation. Moreover, the co-culture of SCs and ECs could promote the release of nerve growth factor and vascular endothelial growth factor, and up-regulate genes relevant to cell proliferation, myelination and skeletal development via the PI3K-Akt, MAPK and cytokine and receptor chemokine pathways. Conclusions The co-culture of SCs and ECs significantly improved the growth behavior of DRG on anisotropic topological scaffolds, which may provide an important basis for the development of nerve grafts in peripheral nerve regeneration.
Collapse
Affiliation(s)
- Tiantian Zheng
- Key laboratory of Neuroregeneration of Jiangsu and Ministry of Education , Co-innovation Center of Neuroregeneration, NMPA Key Lab for Research and Evaluation of Tissue Engineering Technology Products, , Nantong , P. R. China
- Nantong University. 226001 , Co-innovation Center of Neuroregeneration, NMPA Key Lab for Research and Evaluation of Tissue Engineering Technology Products, , Nantong , P. R. China
| | - Linliang Wu
- Key laboratory of Neuroregeneration of Jiangsu and Ministry of Education , Co-innovation Center of Neuroregeneration, NMPA Key Lab for Research and Evaluation of Tissue Engineering Technology Products, , Nantong , P. R. China
- Nantong University. 226001 , Co-innovation Center of Neuroregeneration, NMPA Key Lab for Research and Evaluation of Tissue Engineering Technology Products, , Nantong , P. R. China
| | - Shaolan Sun
- Key laboratory of Neuroregeneration of Jiangsu and Ministry of Education , Co-innovation Center of Neuroregeneration, NMPA Key Lab for Research and Evaluation of Tissue Engineering Technology Products, , Nantong , P. R. China
- Nantong University. 226001 , Co-innovation Center of Neuroregeneration, NMPA Key Lab for Research and Evaluation of Tissue Engineering Technology Products, , Nantong , P. R. China
| | - Jiawei Xu
- Key laboratory of Neuroregeneration of Jiangsu and Ministry of Education , Co-innovation Center of Neuroregeneration, NMPA Key Lab for Research and Evaluation of Tissue Engineering Technology Products, , Nantong , P. R. China
- Nantong University. 226001 , Co-innovation Center of Neuroregeneration, NMPA Key Lab for Research and Evaluation of Tissue Engineering Technology Products, , Nantong , P. R. China
| | - Qi Han
- Key laboratory of Neuroregeneration of Jiangsu and Ministry of Education , Co-innovation Center of Neuroregeneration, NMPA Key Lab for Research and Evaluation of Tissue Engineering Technology Products, , Nantong , P. R. China
- Nantong University. 226001 , Co-innovation Center of Neuroregeneration, NMPA Key Lab for Research and Evaluation of Tissue Engineering Technology Products, , Nantong , P. R. China
| | - Yifan Liu
- School of Medicine, Nantong University. 226001 , Nantong , P. R. China
| | - Ronghua Wu
- Key laboratory of Neuroregeneration of Jiangsu and Ministry of Education , Co-innovation Center of Neuroregeneration, NMPA Key Lab for Research and Evaluation of Tissue Engineering Technology Products, , Nantong , P. R. China
- Nantong University. 226001 , Co-innovation Center of Neuroregeneration, NMPA Key Lab for Research and Evaluation of Tissue Engineering Technology Products, , Nantong , P. R. China
| | - Guicai Li
- Key laboratory of Neuroregeneration of Jiangsu and Ministry of Education , Co-innovation Center of Neuroregeneration, NMPA Key Lab for Research and Evaluation of Tissue Engineering Technology Products, , Nantong , P. R. China
- Nantong University. 226001 , Co-innovation Center of Neuroregeneration, NMPA Key Lab for Research and Evaluation of Tissue Engineering Technology Products, , Nantong , P. R. China
- School of Medicine, Nantong University. 226001 , Nantong , P. R. China
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University , 530021, Nanning , P.R.China
- National Engineering Laboratory for Modern Silk, Soochow University , Suzhou 215123 , China
| |
Collapse
|
30
|
Elorza Ridaura I, Sorrentino S, Moroni L. Parallels between the Developing Vascular and Neural Systems: Signaling Pathways and Future Perspectives for Regenerative Medicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101837. [PMID: 34693660 PMCID: PMC8655224 DOI: 10.1002/advs.202101837] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/23/2021] [Indexed: 05/10/2023]
Abstract
Neurovascular disorders, which involve the vascular and nervous systems, are common. Research on such disorders usually focuses on either vascular or nervous components, without looking at how they interact. Adopting a neurovascular perspective is essential to improve current treatments. Therefore, comparing molecular processes known to be involved in both systems separately can provide insight into promising areas of future research. Since development and regeneration share many mechanisms, comparing signaling molecules involved in both the developing vascular and nervous systems and shedding light to those that they have in common can reveal processes, which have not yet been studied from a regenerative perspective, yet hold great potential. Hence, this review discusses and compares processes involved in the development of the vascular and nervous systems, in order to provide an overview of the molecular mechanisms, which are most promising with regards to treatment for neurovascular disorders. Vascular endothelial growth factor, semaphorins, and ephrins are found to hold the most potential, while fibroblast growth factor, bone morphogenic protein, slits, and sonic hedgehog are shown to participate in both the developing vascular and nervous systems, yet have not been studied at the neurovascular level, therefore being of special interest for future research.
Collapse
Affiliation(s)
- Idoia Elorza Ridaura
- Complex Tissue Regeneration DepartmentMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
| | - Stefano Sorrentino
- CNR Nanotec – Institute of NanotechnologyCampus Ecotekne, via MonteroniLecce73100Italy
| | - Lorenzo Moroni
- Complex Tissue Regeneration DepartmentMERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityUniversiteitssingel 40Maastricht6229ERThe Netherlands
- CNR Nanotec – Institute of NanotechnologyCampus Ecotekne, via MonteroniLecce73100Italy
| |
Collapse
|
31
|
Fralish Z, Lotz EM, Chavez T, Khodabukus A, Bursac N. Neuromuscular Development and Disease: Learning From in vitro and in vivo Models. Front Cell Dev Biol 2021; 9:764732. [PMID: 34778273 PMCID: PMC8579029 DOI: 10.3389/fcell.2021.764732] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/06/2021] [Indexed: 01/02/2023] Open
Abstract
The neuromuscular junction (NMJ) is a specialized cholinergic synaptic interface between a motor neuron and a skeletal muscle fiber that translates presynaptic electrical impulses into motor function. NMJ formation and maintenance require tightly regulated signaling and cellular communication among motor neurons, myogenic cells, and Schwann cells. Neuromuscular diseases (NMDs) can result in loss of NMJ function and motor input leading to paralysis or even death. Although small animal models have been instrumental in advancing our understanding of the NMJ structure and function, the complexities of studying this multi-tissue system in vivo and poor clinical outcomes of candidate therapies developed in small animal models has driven the need for in vitro models of functional human NMJ to complement animal studies. In this review, we discuss prevailing models of NMDs and highlight the current progress and ongoing challenges in developing human iPSC-derived (hiPSC) 3D cell culture models of functional NMJs. We first review in vivo development of motor neurons, skeletal muscle, Schwann cells, and the NMJ alongside current methods for directing the differentiation of relevant cell types from hiPSCs. We further compare the efficacy of modeling NMDs in animals and human cell culture systems in the context of five NMDs: amyotrophic lateral sclerosis, myasthenia gravis, Duchenne muscular dystrophy, myotonic dystrophy, and Pompe disease. Finally, we discuss further work necessary for hiPSC-derived NMJ models to function as effective personalized NMD platforms.
Collapse
Affiliation(s)
| | | | | | | | - Nenad Bursac
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, United States
| |
Collapse
|
32
|
Yoneda T, Hiasa M, Okui T, Hata K. Sensory nerves: A driver of the vicious cycle in bone metastasis? J Bone Oncol 2021; 30:100387. [PMID: 34504741 PMCID: PMC8411232 DOI: 10.1016/j.jbo.2021.100387] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 12/04/2022] Open
Abstract
Bone is one of the preferential target organs of cancer metastasis. Bone metastasis is associated with various complications, of which bone pain is most common and debilitating. The cancer-associated bone pain (CABP) is induced as a consequence of increased neurogenesis, reprogramming and axonogenesis of sensory nerves (SNs) in harmony with sensitization and excitation of SNs in response to the tumor microenvironment created in bone. Importantly, CABP is associated with increased mortality, of which precise cellular and molecular mechanism remains poorly understood. Bone is densely innervated by autonomic nerves (ANs) (sympathetic and parasympathetic nerves) and SNs. Recent studies have shown that the nerves innervating the tumor microenvironment establish intimate communications with tumors, producing various stimuli for tumors to progress and disseminate. In this review, our current understanding of the role of SNs innervating bone in the pathophysiology of CABP will be overviewed. Then the hypothesis that SNs facilitate cancer progression in bone will be discussed in conjunction with our recent findings that SNs play an important role not only in the induction of CABP but also the progression of bone metastasis using a preclinical model of CABP. It is suggested that SNs are a critical component of the bone microenvironment that drives the vicious cycle between bone and cancer to progress bone metastasis. Suppression of the activity of bone-innervating SNs may have potential therapeutic effects on the progression of bone metastasis and induction of CABP.
Collapse
Key Words
- AN, autonomic nerve
- BDNF, brain-derived neurotrophic factor
- BMP, bone morphogenetic protein
- BMSC, bone marrow stromal cells
- Bone microenvironment
- CABP, cancer-associated bone pain
- CALCRL, calcitonin receptor-like receptor
- CAP, cancer-associated pain
- CCL2, C–C motif chemokine 2
- CGRP, calcitonin gene-related peptide
- CNS, central nervous system
- COX, cyclooxygenase
- CREB, cyclic AMP-responsive element-binding protein
- CRPC, castration-resistant prostate cancer
- CXCL1, C-X-C Motif Chemokine Ligand 1
- CXCL2, C-X-C Motif Chemokine Ligand 2
- Cancer-associated bone pain
- DRG, dorsal root ganglion
- ERK1/2, extracellular receptor kinase ½
- G-CSF, granulocyte colony-stimulating factor
- GDNF, glial-derived neurotrophic factor
- HGF, hepatocyte growth factor
- HIF-1α, hypoxia-inducible transcription factor-1α
- HMGB-1, high mobility group box-1
- HSCs, hematopoietic stem cells
- HUVECs, human umbilical vein endothelial cells
- IL-1β, interleukin 1β
- MM, multiple myeloma
- MOR, mu-opioid receptor
- NE, norepinephrine
- NGF, nerve growth factor
- NI, nerve invasion
- NPY, neuropeptide Y
- NSAIDs, nonsteroidal anti-inflammatory drugs
- Nociceptors
- OA, osteoarthritis
- OPG, osteoprotegerin
- PACAP, pituitary adenylate cyclase-activating peptide
- PD-1, programmed cell death-1
- PD-L1, programmed death-ligand 1
- PDAC, pancreatic ductal adenocarcinoma
- PGE2, prostaglandin E2
- PNI, perineural invasion
- PanIN, pancreatic intraepithelial neoplasia
- Perineural invasion
- RAGE, receptor for advanced glycation end products
- RAMP1, receptor activity modifying protein 1
- RANKL, receptor activator of NF-κB ligand
- RTX, resiniferatoxin
- SN, sensory nerves
- SP, substance P
- SRE, skeletal-related event
- Sensory nerves
- TGFβ, transforming growth factor β
- TNFα, tumor necrosis factor α
- TRPV1
- TrkA, tyrosine kinase receptor type 1
- VEGF, vascular endothelial growth factor
- VIP, vasoactive intestinal peptide
- a3V-H+-ATPase, a3 isoform vacuolar proton pump
Collapse
Affiliation(s)
- Toshiyuki Yoneda
- Department of Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Masahiro Hiasa
- Department of Biomaterials and Bioengineerings, University of Tokushima Graduate School of Dentistry, Tokushima, Japan
| | - Tatsuo Okui
- Department of Oral and Maxillofacial Surgery and Biopathology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Science, Okayama, Japan
| | - Kenji Hata
- Department of Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| |
Collapse
|
33
|
Hang Y, Ma X, Liu C, Li S, Zhang S, Feng R, Shang Q, Liu Q, Ding Z, Zhang X, Yu L, Lu Q, Shao C, Chen H, Shi Y, He J, Kaplan DL. Blastocyst-Inspired Hydrogels to Maintain Undifferentiation of Mouse Embryonic Stem Cells. ACS NANO 2021; 15:14162-14173. [PMID: 34516077 DOI: 10.1021/acsnano.0c10468] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Stem cell fate is determined by specific niches that provide multiple physical, chemical, and biological cues. However, the hierarchy or cascade of impact of these cues remains elusive due to their spatiotemporal complexity. Here, anisotropic silk protein nanofiber-based hydrogels with suitable cell adhesion capacity are developed to mimic the physical microenvironment inside the blastocele. The hydrogels enable mouse embryonic stem cells (mESCs) to maintain stemness in vitro in the absence of both leukemia inhibitory factor (LIF) and mouse embryonic fibroblasts (MEFs), two critical factors in the standard protocol for mESC maintenance. The mESCs on hydrogels can achieve superior pluripotency, genetic stability, developmental capacity, and germline transmission to those cultured with the standard protocol. Such biomaterials establish an improved dynamic niche through stimulating the secretion of autocrine factors and are sufficient to maintain the pluripotency and propagation of ESCs. The mESCs on hydrogels are distinct in their expression profiles and more resemble ESCs in vivo. The physical cues can thus initiate a self-sustaining stemness-maintaining program. In addition to providing a relatively simple and low-cost option for expansion and utility of ESCs in biological research and therapeutic applications, this biomimetic material helps gain more insights into the underpinnings of early mammalian embryogenesis.
Collapse
Affiliation(s)
- Yingjie Hang
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People's Republic of China
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Xiaoliang Ma
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Chunxiao Liu
- State Key Laboratory of Radiation Medicine and Radiation Protection, Institutes for Translational Medicine, Soochow University, Suzhou 215123, People's Republic of China
| | - Siyuan Li
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People's Republic of China
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Sixuan Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Ruyan Feng
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Qianwen Shang
- State Key Laboratory of Radiation Medicine and Radiation Protection, Institutes for Translational Medicine, Soochow University, Suzhou 215123, People's Republic of China
| | - Qi Liu
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Zhaozhao Ding
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People's Republic of China
| | - Xiaoyi Zhang
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People's Republic of China
| | - Liyin Yu
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Qiang Lu
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People's Republic of China
| | - Changshun Shao
- State Key Laboratory of Radiation Medicine and Radiation Protection, Institutes for Translational Medicine, Soochow University, Suzhou 215123, People's Republic of China
| | - Hong Chen
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Yufang Shi
- State Key Laboratory of Radiation Medicine and Radiation Protection, Institutes for Translational Medicine, Soochow University, Suzhou 215123, People's Republic of China
| | - Jiuyang He
- CAS Engineering Laboratory for Nanozyme, Key Laboratory of Protein and Peptide Pharmaceutical, Institute Academy of Science, Beijing 100101, People's Republic of China
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
34
|
Malheiro A, Wieringa P, Moroni L. Peripheral neurovascular link: an overview of interactions and in vitro models. Trends Endocrinol Metab 2021; 32:623-638. [PMID: 34127366 DOI: 10.1016/j.tem.2021.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 04/23/2021] [Accepted: 05/10/2021] [Indexed: 12/26/2022]
Abstract
Nerves and blood vessels (BVs) establish extensive arborized networks to innervate tissues and deliver oxygen/metabolic support. Developmental cues direct the formation of these intricate and often overlapping patterns, which reflect close interactions within the peripheral neurovascular system. Besides the mutual dependence to survive and function, nerves and BVs share several receptors and ligands, as well as principles of differentiation, growth and pathfinding. Neurovascular (NV) interactions are maintained in adult life and are essential for certain regenerative mechanisms, such as wound healing. In pathological situations (e.g., type 2 diabetes mellitus), the NV system can be severely perturbed and become dysfunctional. Unwanted neural growth and vascularization are also associated with the progression of some pathologies, such as cancer and endometriosis. In this review, we describe the fundamental NV interactions in development, highlighting the similarities between both networks and wiring mechanisms. We also describe the NV contribution to regenerative processes and potential pathological dysfunctions. Finally, we provide an overview of current in vitro models used to replicate and investigate the NV ecosystem, addressing present limitations and future perspectives.
Collapse
Affiliation(s)
- Afonso Malheiro
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, 6229ER Maastricht, The Netherlands
| | - Paul Wieringa
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, 6229ER Maastricht, The Netherlands
| | - Lorenzo Moroni
- Complex Tissue Regeneration Department, MERLN Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, 6229ER Maastricht, The Netherlands.
| |
Collapse
|
35
|
Azoulay D, Horowitz NA. Brain-derived neurotrophic factor in hematological malignancies: From detrimental to potentially beneficial. Blood Rev 2021; 51:100871. [PMID: 34344590 DOI: 10.1016/j.blre.2021.100871] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/13/2021] [Accepted: 07/21/2021] [Indexed: 12/20/2022]
Abstract
Emerging studies have highlighted brain-derived neurotrophic factor (BDNF), a neuronal growth factor abundant in the peripheral blood, and its tyrosine kinase receptor TRKB, as onco-genes and proteins that support the survival of malignant hematological cells. In contrast, other researchers reported on a favorable association between BDNF blood levels and prognosis, chemotherapy response and neurological side effects in patients with hematological malignancies. Here, we review the accumulated data regarding the expression of BDNF and its receptors in normal hematopoietic and lymphatic cells and tissue. In addition, in-vitro experiments, animal models and human sample studies that investigated the role of BDNF and its receptors in hematological malignancies are discussed. Finally, directions for future research aimed at revealing the mechanisms underlying the protective effect of BDNF in patients with these diseases are suggested.
Collapse
Affiliation(s)
- David Azoulay
- Hematology Unit and Laboratories, Galilee Medical Center, Naharia, Israel; Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel.
| | - Netanel A Horowitz
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel; The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
36
|
Peper S, Vo T, Ahuja N, Awad K, Mikos AG, Varanasi V. Bioprinted nanocomposite hydrogels: A proposed approach to functional restoration of skeletal muscle and vascular tissue following volumetric muscle loss. Curr Opin Pharmacol 2021; 58:35-43. [PMID: 33853025 PMCID: PMC8718378 DOI: 10.1016/j.coph.2021.03.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/27/2021] [Accepted: 03/11/2021] [Indexed: 01/03/2023]
Abstract
Musculoskeletal conditions are the highest contributor to global disability, accounting for 16% of all ages lived with disability. Volumetric muscle loss (VML) is classified as significant damage to skeletal muscle compartments and motor units, leading to significant tissue loss, functional deficits, and long-term disability. In this review, the current tissue engineering approaches in terms of fabrication techniques, materials, cell sources, and growth factors for enhanced angiogenesis and neuromuscular junction (NMJ) in VML repair, are discussed. Review of results recently published in the literature suggested that bioprinted nanocomposite hydrogels (NC gels) seeded with adult muscle progenitor cells that promote secretion of endogenous vascular growth factors have potential applications in promoting skeletal muscle regeneration, revascularization, and NMJ repair (Figure 1). Despite recent advancements, future research is needed on NC gels and the complex processes underlying vascular infiltration and NMJ repair in VML injuries.
Collapse
Affiliation(s)
- Sara Peper
- Bone Muscle Research Center, College of Nursing & Health Innovation, The University of Texas at Arlington, 701 South Nedderman Drive, Arlington, TX, 76019, USA; Department of Bioengineering, College of Engineering, The University of Texas at Arlington, 701 South Nedderman Drive, Box 19138, Arlington, TX, 76019, USA
| | - Thy Vo
- Bone Muscle Research Center, College of Nursing & Health Innovation, The University of Texas at Arlington, 701 South Nedderman Drive, Arlington, TX, 76019, USA; Department of Kinesiology, College of Nursing & Health Innovation, The University of Texas at Arlington, 411 South Nedderman Drive, Box 19407, Arlington, TX, 76019, USA
| | - Neelam Ahuja
- Bone Muscle Research Center, College of Nursing & Health Innovation, The University of Texas at Arlington, 701 South Nedderman Drive, Arlington, TX, 76019, USA; Department of Kinesiology, College of Nursing & Health Innovation, The University of Texas at Arlington, 411 South Nedderman Drive, Box 19407, Arlington, TX, 76019, USA
| | - Kamal Awad
- Bone Muscle Research Center, College of Nursing & Health Innovation, The University of Texas at Arlington, 701 South Nedderman Drive, Arlington, TX, 76019, USA; Department of Materials Science & Engineering, College of Engineering, The University of Texas at Arlington, 701 South Nedderman Drive, Box 19138, Arlington, TX, 76019 & National Research Center, 12622, Egypt
| | - Antonios G Mikos
- Center for Engineering Complex Tissues, Center for Excellence in Tissue Engineering, J.W. Cox Laboratory for Biomedical Engineering, Rice University, P.O. Box 1892, Houston, TX, 77251, USA
| | - Venu Varanasi
- Bone Muscle Research Center, College of Nursing & Health Innovation, The University of Texas at Arlington, 701 South Nedderman Drive, Arlington, TX, 76019, USA; Department of Nursing, College of Nursing & Health Innovation, The University of Texas at Arlington, 411 South Nedderman Drive Box 19407, Arlington, TX, 76019, USA.
| |
Collapse
|
37
|
Muangsanit P, Roberton V, Costa E, Phillips JB. Engineered aligned endothelial cell structures in tethered collagen hydrogels promote peripheral nerve regeneration. Acta Biomater 2021; 126:224-237. [PMID: 33766800 DOI: 10.1016/j.actbio.2021.03.039] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 03/01/2021] [Accepted: 03/16/2021] [Indexed: 02/07/2023]
Abstract
Vascularisation is important in nerve tissue engineering to provide blood supply and nutrients for long-term survival of implanted cells. Furthermore, blood vessels in regenerating nerves have been shown to serve as tracks for Schwann cells to migrate along and thus form Bands of Büngner which promote axonal regeneration. In this study, we have developed tissue-engineered constructs containing aligned endothelial cells, or co-cultures of both endothelial cells and Schwann cells to test whether these structures could promote regeneration across peripheral nerve gaps. Type I rat tail collagen gels containing HUVECs (Human Umbilical Vein Endothelial Cells, 4 × 106 cells/ml) were cast in perforated tethering silicone conduits to facilitate cellular self-alignment and tube formation for 4 days of culture. For co-culture constructs, optimal tube formation and cellular alignment was achieved with a ratio of 4:0.5 × 106 cells/ml (HUVECs:Schwann cells). An in vivo test of the engineered constructs to bridge a 10 mm gap in rat sciatic nerves for 4 weeks revealed that constructs containing only HUVECs significantly promoted axonal regeneration and vascularisation across the gap, as compared to conventional aligned Schwann cell constructs and those containing co-cultured HUVECs and Schwann cells. Our results suggest that tissue-engineered constructs containing aligned endothelial cells within collagen matrix could be good candidates to treat peripheral nerve injury. STATEMENT OF SIGNIFICANCE: Nerve tissue engineering provides a potential way to overcome the limitations associated with current clinical grafting techniques for the repair of severe peripheral nerve injuries. However, the therapeutic cells within engineered nerve tissue require effective vascularisation in order to survive. This work therefore aimed to develop engineered nerve constructs containing aligned tube-like structures made from endothelial cells. Not only did this provide a method to improve vascularisation, it demonstrated for the first time that aligned endothelial cells can outperform Schwann cells in promoting nerve regeneration in the rat sciatic nerve model. This has introduced the concept of developing pre-vascularised engineered nerve tissues, and indicated the potential usefulness of endothelial cell structures in tissue engineering for peripheral nerve repair.
Collapse
Affiliation(s)
- Papon Muangsanit
- Department of Biomaterials and Tissue Engineering, Eastman Dental Institute, University College London, 256 Grays Inn Rd, London WC1X8LD, United Kingdom; UCL Centre for Nerve Engineering, University College London, London, United Kingdom; Department of Pharmacology, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, Bloomsbury, London WC1N 1AX, United Kingdom.
| | - Victoria Roberton
- UCL Centre for Nerve Engineering, University College London, London, United Kingdom; Department of Pharmacology, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, Bloomsbury, London WC1N 1AX, United Kingdom
| | - Eleni Costa
- Department of Pharmacology, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, Bloomsbury, London WC1N 1AX, United Kingdom
| | - James B Phillips
- UCL Centre for Nerve Engineering, University College London, London, United Kingdom; Department of Pharmacology, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, Bloomsbury, London WC1N 1AX, United Kingdom
| |
Collapse
|
38
|
Kannan S, Lee M, Muthusamy S, Blasiak A, Sriram G, Cao T. Peripheral sensory neurons promote angiogenesis in neurovascular models derived from hESCs. Stem Cell Res 2021; 52:102231. [PMID: 33601097 DOI: 10.1016/j.scr.2021.102231] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/10/2021] [Accepted: 02/03/2021] [Indexed: 01/06/2023] Open
Abstract
In the adult tissues, blood vessels traverse the body with neurons side by side; and share common signaling molecules. Developmental studies on animal models have shown that peripheral sensory neurons (PSNs) secrete angiogenic factors and endothelial cells (ECs) secrete neurotrophic factors which contribute to their coexistence, thereby forming the peripheral neurovascular (PNV) unit. Despite the large number of studies showing that innervation and vascularization complement each other, the interaction between human PSNs and ECs is still largely unknown. To study this interaction and to evaluate if PSNs affect angiogenesis, we derived both PSNs and ECs from human embryonic stem cells (hESCs) and developed a co-culture system. Seeding the two cell types together showed that PSNs induced endothelial morphogenesis with formation of vessel-like structures (VLSs). The PSN precursors, neural crest stem cells also induced VLS formation in the co-culture system; however, to a lesser extent. This sheds new light on the in vitro angiogenic potential of these cell types. PSNs derived from hESCs are powerful tools for studying development and disease as human PSNs are inaccessible for in vitro assays. Our novel approach, with optimized media condition allowed for integrating hESC-derived PSNs with hESC-derived ECs in three-dimensional (3D) collagen gel for creating a completely humanised PNV model. This preliminary model showed that innervation improves the development of vascularized channels in vitro, and provides insight to the development of innervated 3D models in future.
Collapse
Affiliation(s)
- Sathya Kannan
- Faculty of Dentistry, National University of Singapore, Singapore
| | - Marcus Lee
- Faculty of Dentistry, National University of Singapore, Singapore
| | | | - Agata Blasiak
- The N.1 Institute for Health, National University of Singapore, Singapore
| | - Gopu Sriram
- Faculty of Dentistry, National University of Singapore, Singapore; NUS Centre for Additive Manufacturing (AM.NUS), National University of Singapore, Singapore.
| | - Tong Cao
- Faculty of Dentistry, National University of Singapore, Singapore.
| |
Collapse
|
39
|
Dömer P, Kayal J, Janssen-Bienhold U, Kewitz B, Kretschmer T, Heinen C. Rapid and efficient immunomagnetic isolation of endothelial cells from human peripheral nerves. Sci Rep 2021; 11:1951. [PMID: 33479384 PMCID: PMC7820485 DOI: 10.1038/s41598-021-81361-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 01/04/2021] [Indexed: 01/18/2023] Open
Abstract
Endothelial cells (ECs) have gained an increased scientific focus since they were reported to provide guidance for Schwann cells and subsequently following axons after nerve injuries. However, previous protocols for the isolation of nerve-derived ECs from human nerves are ineffective regarding time and yield. Therefore, we established a novel and efficient protocol for the isolation of ECs from human peripheral nerves by means of immunomagnetic CD31-antibody conjugated Dynabeads and assessed the purity of the isolated cells. The easy-to-follow and time-effective isolation method allows the isolation of > 95% pure ECs. The isolated ECs were shown to express highly specific EC marker proteins and revealed functional properties by formation of CD31 and VE-cadherin positive adherens junctions, as well as ZO-1 positive tight-junctions. Moreover, the formation of capillary EC-tubes was observed in-vitro. The novel protocol for the isolation of human nerve-derived ECs allows and simplifies the usage of ECs in research of the human blood-nerve-barrier and peripheral nerve regeneration. Additionally, a potential experimental application of patient-derived nerve ECs in the in-vitro vascularization of artificial nerve grafts is feasible.
Collapse
Affiliation(s)
- Patrick Dömer
- Department of Neuroscience, Carl Von Ossietzky University Oldenburg, Carl von Ossietzky Str. 9-11, Oldenburg, Germany.
- Department of Neurosurgery, Evangelisches Krankenhaus, Campus Carl von Ossietzky University Oldenburg, Oldenburg, Germany.
| | - Janine Kayal
- Department of Neuroscience, Carl Von Ossietzky University Oldenburg, Carl von Ossietzky Str. 9-11, Oldenburg, Germany
| | - Ulrike Janssen-Bienhold
- Department of Neuroscience, Carl Von Ossietzky University Oldenburg, Carl von Ossietzky Str. 9-11, Oldenburg, Germany
- Research Center Neurosensory Science, Carl Von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Bettina Kewitz
- Department of Neuroscience, Carl Von Ossietzky University Oldenburg, Carl von Ossietzky Str. 9-11, Oldenburg, Germany
- Department of Neurosurgery, Evangelisches Krankenhaus, Campus Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Thomas Kretschmer
- Department of Neurosurgery and Neurorestauration, Klinikum Klagenfurt Am Wörthersee, Klagenfurt, Austria
| | - Christian Heinen
- Department of Neurosurgery, Evangelisches Krankenhaus, Campus Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| |
Collapse
|
40
|
Chouaib B, Collart-Dutilleul PY, Blanc-Sylvestre N, Younes R, Gergely C, Raoul C, Scamps F, Cuisinier F, Romieu O. Identification of secreted factors in dental pulp cell-conditioned medium optimized for neuronal growth. Neurochem Int 2021; 144:104961. [PMID: 33465470 DOI: 10.1016/j.neuint.2021.104961] [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: 09/19/2020] [Revised: 01/04/2021] [Accepted: 01/11/2021] [Indexed: 02/05/2023]
Abstract
With their potent regenerative and protective capacities, stem cell-derived conditioned media emerged as an effective alternative to cell therapy, and have a prospect to be manufactured as pharmaceutical products for tissue regeneration applications. Our study investigates the neuroregenerative potential of human dental pulp cells (DPCs) conditioned medium (CM) and defines an optimization strategy of DPC-CM for enhanced neuronal outgrowth. Primary sensory neurons from mouse dorsal root ganglia were cultured with or without DPC-CM, and the lengths of βIII-tubulin positive neurites were measured. The impacts of several manufacturing features as the duration of cell conditioning, CM storage, and preconditioning of DPCs with some factors on CM functional activity were assessed on neurite length. We observed that DPC-CM significantly enhanced neurites outgrowth of sensory neurons in a concentration-dependent manner. The frozen storage of DPC-CM had no impact on experimental outcomes and 48 h of DPC conditioning is optimal for an effective activity of CM. To further understand the regenerative feature of DPC-CM, we studied DPC secretome by human growth factor antibody array analysis and revealed the presence of several factors involved in either neurogenesis, neuroprotection, angiogenesis, and osteogenesis. The conditioning of DPCs with the B-27 supplement enhanced significantly the neuroregenerative effect of their secretome by changing its composition in growth factors. Here, we show that DPC-CM significantly stimulate neurite outgrowth in primary sensory neurons. Moreover, we identified secreted protein candidates that can potentially promote this promising regenerative feature of DPC-CM.
Collapse
Affiliation(s)
| | | | | | - Richard Younes
- LBN, Univ Montpellier, Montpellier, France; The Neuroscience Institute of Montpellier, Inserm UMR1051, Univ Montpellier, Saint Eloi Hospital, Montpellier, France
| | | | - Cédric Raoul
- The Neuroscience Institute of Montpellier, Inserm UMR1051, Univ Montpellier, Saint Eloi Hospital, Montpellier, France
| | - Frédérique Scamps
- The Neuroscience Institute of Montpellier, Inserm UMR1051, Univ Montpellier, Saint Eloi Hospital, Montpellier, France
| | | | | |
Collapse
|
41
|
Hagen KM, Ousman SS. The Neuroimmunology of Guillain-Barré Syndrome and the Potential Role of an Aging Immune System. Front Aging Neurosci 2021; 12:613628. [PMID: 33584245 PMCID: PMC7873882 DOI: 10.3389/fnagi.2020.613628] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/11/2020] [Indexed: 12/15/2022] Open
Abstract
Guillain-Barré syndrome (GBS) is a paralyzing autoimmune condition affecting the peripheral nervous system (PNS). Within GBS there are several variants affecting different aspects of the peripheral nerve. In general, there appears to be a role for T cells, macrophages, B cells, and complement in initiating and perpetuating attacks on gangliosides of Schwann cells and axons. Of note, GBS has an increased prevalence and severity with increasing age. In addition, there are alterations in immune cell functioning that may play a role in differences in GBS with age alongside general age-related declines in reparative processes (e.g., delayed de-differentiation of Schwann cells and decline in phagocytic ability of macrophages). The present review will explore the immune response in GBS as well as in animal models of several variants of the disorder. In addition, the potential involvement of an aging immune system in contributing to the increased prevalence and severity of GBS with age will be theorized.
Collapse
Affiliation(s)
- Kathleen M. Hagen
- Department of Neuroscience, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Shalina S. Ousman
- Departments of Clinical Neurosciences and Cell Biology and Anatomy, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
42
|
Shah Mohammadi M, Buchen JT, Pasquina PF, Niklason LE, Alvarez LM, Jariwala SH. Critical Considerations for Regeneration of Vascularized Composite Tissues. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:366-381. [PMID: 33115331 DOI: 10.1089/ten.teb.2020.0223] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Effective vascularization is vital for survival and functionality of complex tissue-engineered organs. The formation of the microvasculature, composed of endothelial cells (ECs) alone, has been mostly used to restore the vascular networks in organs. However, recent heterocellular studies demonstrate that co-culturing is a more effective approach in revascularization of engineered organs. This review presents key considerations for manufacturing of artificial vascularized composite tissues. We summarize the importance of co-cultures and the multicellular interactions with ECs, as well as design and use of bioreactors, as critical considerations for tissue vascularization. In addition, as an emerging scaffolding technique, this review also highlights the current caveats and hurdles associated with three-dimensional bioprinting and discusses recent developments in bioprinting strategies such as four-dimensional bioprinting and its future outlook for manufacturing of vascularized tissue constructs. Finally, the review concludes with addressing the critical challenges in the regulatory pathway and clinical translation of artificial composite tissue grafts. Impact statement Regeneration of composite tissues is critical as biophysical and biochemical characteristics differ between various types of tissues. Engineering a vascularized composite tissue has remained unresolved and requires additional evaluations along with optimization of methodologies and standard operating procedures. To this end, the main hurdle is creating a viable vascular endothelium that remains functional for a longer duration postimplantation, and can be manufactured using clinically appropriate source of cell lines that are scalable in vitro for the fabrication of human-scale organs. This review presents key considerations for regeneration and manufacturing of vascularized composite tissues as the field advances.
Collapse
Affiliation(s)
- Maziar Shah Mohammadi
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA.,Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| | - Jack T Buchen
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA.,Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| | - Paul F Pasquina
- Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA.,Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Laura E Niklason
- Department of Anesthesia and Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Luis M Alvarez
- Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA.,Lung Biotechnology PBC, Silver Spring, Maryland, USA
| | - Shailly H Jariwala
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA.,Department of Physical Medicine and Rehabilitation, The Center for Rehabilitation Sciences Research, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| |
Collapse
|
43
|
Leroux A, Paiva Dos Santos B, Leng J, Oliveira H, Amédée J. Sensory neurons from dorsal root ganglia regulate endothelial cell function in extracellular matrix remodelling. Cell Commun Signal 2020; 18:162. [PMID: 33076927 PMCID: PMC7574530 DOI: 10.1186/s12964-020-00656-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 09/06/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Recent physiological and experimental data highlight the role of the sensory nervous system in bone repair, but its precise role on angiogenesis in a bone regeneration context is still unknown. Our previous work demonstrated that sensory neurons (SNs) induce the osteoblastic differentiation of mesenchymal stem cells, but the influence of SNs on endothelial cells (ECs) was not studied. METHODS Here, in order to study in vitro the interplay between SNs and ECs, we used microfluidic devices as an indirect co-culture model. Gene expression analysis of angiogenic markers, as well as measurements of metalloproteinases protein levels and enzymatic activity, were performed. RESULTS We were able to demonstrate that two sensory neuropeptides, calcitonin gene-related peptide (CGRP) and substance P (SP), were involved in the transcriptional upregulation of angiogenic markers (vascular endothelial growth factor, angiopoietin 1, type 4 collagen, matrix metalloproteinase 2) in ECs. Co-cultures of ECs with SNs also increased the protein level and enzymatic activity of matrix metalloproteinases 2 and 9 (MMP2/MMP9) in ECs. CONCLUSIONS Our results suggest a role of sensory neurons, and more specifically of CGRP and SP, in the remodelling of endothelial cells extracellular matrix, thus supporting and enhancing the angiogenesis process. Video abstract.
Collapse
Affiliation(s)
- Alice Leroux
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000, Bordeaux, France.
| | | | - Jacques Leng
- Univ. Bordeaux, CNRS, Solvay, LOF, UMR 5258, F-33006, Pessac, France
| | - Hugo Oliveira
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000, Bordeaux, France
| | - Joëlle Amédée
- Univ. Bordeaux, INSERM, BIOTIS, U1026, F-33000, Bordeaux, France
| |
Collapse
|
44
|
Cucchi DGJ, Groen RWJ, Janssen JJWM, Cloos J. Ex vivo cultures and drug testing of primary acute myeloid leukemia samples: Current techniques and implications for experimental design and outcome. Drug Resist Updat 2020; 53:100730. [PMID: 33096284 DOI: 10.1016/j.drup.2020.100730] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 09/03/2020] [Accepted: 09/29/2020] [Indexed: 12/11/2022]
Abstract
New treatment options of acute myeloid leukemia (AML) are rapidly emerging. Pre-clinical models such as ex vivo cultures are extensively used towards the development of novel drugs and to study synergistic drug combinations, as well as to discover biomarkers for both drug response and anti-cancer drug resistance. Although these approaches empower efficient investigation of multiple drugs in a multitude of primary AML samples, their translational value and reproducibility are hampered by the lack of standardized methodologies and by culture system-specific behavior of AML cells and chemotherapeutic drugs. Moreover, distinct research questions require specific methods which rely on specific technical knowledge and skills. To address these aspects, we herein review commonly used culture techniques in light of diverse research questions. In addition, culture-dependent effects on drug resistance towards commonly used drugs in the treatment of AML are summarized including several pitfalls that may arise because of culture technique artifacts. The primary aim of the current review is to provide practical guidelines for ex vivo primary AML culture experimental design.
Collapse
Affiliation(s)
- D G J Cucchi
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands
| | - R W J Groen
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands
| | - J J W M Janssen
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands
| | - J Cloos
- Department of Hematology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, the Netherlands.
| |
Collapse
|
45
|
Azoulay D, Horowitz NA. Brain-derived neurotrophic factor as a potential biomarker of chemotherapy-induced peripheral neuropathy and prognosis in haematological malignancies; what we have learned, the challenges and a need for global standardization. Br J Haematol 2020; 191:17-18. [PMID: 32557540 DOI: 10.1111/bjh.16893] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 12/29/2022]
Affiliation(s)
- David Azoulay
- Hematology Unit and Laboratories, Galilee Medical Center, Naharia, Israel.,Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Netanel A Horowitz
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel.,The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
46
|
Sutherland TC, Geoffroy CG. The Influence of Neuron-Extrinsic Factors and Aging on Injury Progression and Axonal Repair in the Central Nervous System. Front Cell Dev Biol 2020; 8:190. [PMID: 32269994 PMCID: PMC7109259 DOI: 10.3389/fcell.2020.00190] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/06/2020] [Indexed: 12/21/2022] Open
Abstract
In the aging western population, the average age of incidence for spinal cord injury (SCI) has increased, as has the length of survival of SCI patients. This places great importance on understanding SCI in middle-aged and aging patients. Axon regeneration after injury is an area of study that has received substantial attention and made important experimental progress, however, our understanding of how aging affects this process, and any therapeutic effort to modulate repair, is incomplete. The growth and regeneration of axons is mediated by both neuron intrinsic and extrinsic factors. In this review we explore some of the key extrinsic influences on axon regeneration in the literature, focusing on inflammation and astrogliosis, other cellular responses, components of the extracellular matrix, and myelin proteins. We will describe how each element supports the contention that axonal growth after injury in the central nervous system shows an age-dependent decline, and how this may affect outcomes after a SCI.
Collapse
Affiliation(s)
- Theresa C Sutherland
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX, United States
| | - Cédric G Geoffroy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX, United States
| |
Collapse
|
47
|
Farmer K, Abd-Elrahman KS, Derksen A, Rowe EM, Thompson AM, Rudyk CA, Prowse NA, Dwyer Z, Bureau SC, Fortin T, Ferguson SSG, Hayley S. mGluR5 Allosteric Modulation Promotes Neurorecovery in a 6-OHDA-Toxicant Model of Parkinson's Disease. Mol Neurobiol 2019; 57:1418-1431. [PMID: 31754998 DOI: 10.1007/s12035-019-01818-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 10/14/2019] [Indexed: 10/25/2022]
Abstract
Parkinson's disease is a neurodegenerative disease characterized by a loss of dopaminergic substantia nigra neurons and depletion of dopamine. To date, current therapeutic approaches focus on managing motor symptoms and trying to slow neurodegeneration, with minimal capacity to promote neurorecovery. mGluR5 plays a key role in neuroplasticity, and altered mGluR5 signaling contributes to synucleinopathy and dyskinesia in patients with Parkinson's disease. Here, we tested whether the mGluR5-negative allosteric modulator, (2-chloro-4-[2[2,5-dimethyl-1-[4-(trifluoromethoxy) phenyl] imidazol-4-yl] ethynyl] pyridine (CTEP), would be effective in improving motor deficits and promoting neural recovery in a 6-hydroxydopamine (6-OHDA) mouse model. Lesions were induced by 6-ODHA striatal infusion, and 30 days later treatment with CTEP (2 mg/kg) or vehicle commenced for either 1 or 12 weeks. Animals were subjected to behavioral, pathological, and molecular analyses. We also assessed how long the effects of CTEP persisted, and finally, using rapamycin, determined the role of the mTOR pathway. CTEP treatment induced a duration-dependent improvement in apomorphine-induced rotation and performance on rotarod in lesioned mice. Moreover, CTEP promoted a recovery of striatal tyrosine hydroxylase-positive fibers and normalized FosB levels in lesioned mice. The beneficial effects of CTEP were paralleled by an activation of mammalian target of rapamycin (mTOR) pathway and elevated brain-derived neurotrophic factor levels in the striatum of lesioned mice. The mTOR inhibitor, rapamycin (sirolimus), abolished CTEP-induced neurorecovery and rescue of motor deficits. Our findings indicate that mTOR pathway is a useful target to promote recovery and that mGluR5 allosteric regulators may potentially be repurposed to selectively target this pathway to enhance neuroplasticity in patients with Parkinson's disease.
Collapse
Affiliation(s)
- Kyle Farmer
- Department of Neuroscience, Carleton University, Ottawa, Ontario, K1S 5B6, Canada
| | - Khaled S Abd-Elrahman
- University of Ottawa Brain and Mind Institute, Ottawa, Ontario, K1H 8M5, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada
| | - Alexa Derksen
- Department of Neuroscience, Carleton University, Ottawa, Ontario, K1S 5B6, Canada
| | - Elyn M Rowe
- Department of Neuroscience, Carleton University, Ottawa, Ontario, K1S 5B6, Canada
| | - Ashley M Thompson
- Department of Neuroscience, Carleton University, Ottawa, Ontario, K1S 5B6, Canada
| | - Christopher A Rudyk
- Department of Neuroscience, Carleton University, Ottawa, Ontario, K1S 5B6, Canada
| | - Natalie A Prowse
- Department of Neuroscience, Carleton University, Ottawa, Ontario, K1S 5B6, Canada
| | - Zachary Dwyer
- Department of Neuroscience, Carleton University, Ottawa, Ontario, K1S 5B6, Canada
| | - Samantha C Bureau
- Department of Neuroscience, Carleton University, Ottawa, Ontario, K1S 5B6, Canada
| | - Teresa Fortin
- Department of Neuroscience, Carleton University, Ottawa, Ontario, K1S 5B6, Canada
| | - Stephen S G Ferguson
- University of Ottawa Brain and Mind Institute, Ottawa, Ontario, K1H 8M5, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada
| | - Shawn Hayley
- Department of Neuroscience, Carleton University, Ottawa, Ontario, K1S 5B6, Canada.
| |
Collapse
|
48
|
Grasman JM, Williams MD, Razis CG, Bonzanni M, Golding AS, Cairns DM, Levin M, Kaplan DL. Hyperosmolar potassium inhibits myofibroblast conversion and reduces scar tissue formation. ACS Biomater Sci Eng 2019; 5:5327-5336. [PMID: 32440531 PMCID: PMC7241611 DOI: 10.1021/acsbiomaterials.9b00810] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Scar formation is a natural result of almost all wound healing in adult mammals. Unfortunately, scarring disrupts normal tissue function and can cause significant physical and psychological distress. In addition to improving surgical techniques to limit scar formation, several therapies are under development towards the same goal. Many of these treatments aim to disrupt transforming growth factor β1 (TGFβ1) signaling, as this is a critical control point for fibroblast differentiation into myofibroblasts; a contractile cell that organizes synthesized collagen fibrils into scar tissue. The present study aimed to examine the role of hyperosmolar potassium gluconate (KGluc) on fibroblast function in skin repair. KGluc was first determined to negatively regulate fibroblast proliferation, metabolism, and migration in a dose-dependent manner in vitro. Increasing concentrations of KGluc also inhibited differentiation into myofibroblasts, suggesting that local KGluc treatment might reduce fibrosis. KGluc delivery was confirmed via loading into collagen hydrogels and used to treat a full thickness skin wound in mice. KGluc qualitatively slowed initial closure of the wounds and resulted in tissue that more closely resembled mature, healthy skin (epidermal thickness and dermal-epidermal morphology) when compared to unloaded collagen hydrogels. KGluc treatment significantly reduced the number of myofibroblasts within the dermis while upregulated blood vessel density with respect to unloaded hydrogels, likely a result of disruption of TGFβ1 signaling. Taken together, these data demonstrate the effectiveness of KGluc treatment on skin wound healing and suggest that this may be an efficient treatment to limit scar formation.
Collapse
Affiliation(s)
- Jonathan M. Grasman
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155
| | - Marisa D. Williams
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155
| | - Constantine G. Razis
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155
| | - Mattia Bonzanni
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155
- Allen Discovery Center, Tufts University, Medford, Massachusetts 02155
| | - Anne S. Golding
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts 02155
| | - Dana M. Cairns
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155
| | - Michael Levin
- Department of Biology, Tufts University, Medford, Massachusetts 02155
- Allen Discovery Center, Tufts University, Medford, Massachusetts 02155
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155
- Allen Discovery Center, Tufts University, Medford, Massachusetts 02155
| |
Collapse
|
49
|
Nguyen NM, Song KM, Choi MJ, Ghatak K, Limanjaya A, Kwon MH, Chung DY, Ock J, Yin GN, Park CS, Suh JK, Ryu JK. Three-Dimensional Reconstruction of Neurovascular Network in Whole Mount Preparations and Thick-Cut Transverse Sections of Mouse Urinary Bladder. World J Mens Health 2019; 39:131-138. [PMID: 31496149 PMCID: PMC7752506 DOI: 10.5534/wjmh.190089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 08/01/2019] [Indexed: 11/15/2022] Open
Abstract
PURPOSE Proper functional and structural integrity of nervous and vascular system in urinary bladder plays an important role in normal bladder function and the disruption of these structures is known to be related to lower urinary tract symptoms. Here, we present an immunohistochemical staining method that delineates neurovascular structures in the mouse urinary bladder by using immunohistochemical staining with three-dimensional reconstruction. MATERIALS AND METHODS The urinary bladder was harvested from 8-week-old C57BL/6 male mouse. Lamina propria and detrusor muscle layer were dissected for whole mount staining, and thick-cut (60-μm) sections were prepared for full-thickness bladder staining. Immunofluorescent staining of bladder tissue was performed with antibodies against CD31 (an endothelial cell marker), smooth muscle α-actin (a smooth muscle cell marker), NG2 (a pericyte marker), and βIII-tubulin (a neuronal marker). We reconstructed three-dimensional images of bladder neurovascular system from stacks of two-dimensional images. RESULTS Three-dimensional images obtained from thick-cut sections clearly provided good anatomic information about neurovascular structures in the three layers of bladder, such as urothelium, lamina propria, and detrusor muscle layer. Whole mount images of lamina propria and detrusor muscle layer also clearly delineated spatial relationship between nervous and vascular systems. The microvessel density was higher in the lamina propria than in the detrusor muscle layer. Nerve fibers were evenly innervated into the lamina propria and detrusor muscle. CONCLUSIONS This study provides comprehensive insight into three-dimensional neurovascular structures of mouse urinary bladder. Our technique may constitute a standard tool to evaluate pathologic changes in a variety of urinary bladder diseases.
Collapse
Affiliation(s)
- Nhat Minh Nguyen
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Kang Moon Song
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Min Ji Choi
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Kalyan Ghatak
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Anita Limanjaya
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Mi Hye Kwon
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Doo Yong Chung
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Jiyeon Ock
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Guo Nan Yin
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Korea
| | - Chang Shin Park
- Department of Pharmacology and Medicinal Toxicology Research Center, Inha University School of Medicine, Incheon, Korea
| | - Jun Kyu Suh
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Korea.
| | - Ji Kan Ryu
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Korea.
| |
Collapse
|
50
|
Abstract
Regulating the intrinsic interactions between blood vessels and nerve cells has the potential to enhance repair and regeneration of the central nervous system. Here, we evaluate the efficacy of aligned microvessels to induce and control directional axon growth from neural progenitor cells in vitro and host axons in a rat spinal cord injury model. Interstitial fluid flow aligned microvessels generated from co-cultures of cerebral-derived endothelial cells and pericytes in a three-dimensional scaffold. The endothelial barrier function was evaluated by immunostaining for tight junction proteins and quantifying the permeability coefficient (~10−7 cm/s). Addition of neural progenitor cells to the co-culture resulted in the extension of Tuj-positive axons in the direction of the microvessels. To validate these findings in vivo, scaffolds were transplanted into an acute spinal cord hemisection injury with microvessels aligned with the rostral-caudal direction. At three weeks post-surgery, sagittal sections indicated close alignment between the host axons and the transplanted microvessels. Overall, this work demonstrates the efficacy of exploiting neurovascular interaction to direct axon growth in the injured spinal cord and the potential to use this strategy to facilitate central nervous system regeneration.
Collapse
|