1
|
Michór P, Renardson L, Li S, Boltze J. Neurorestorative Approaches for Ischemic StrokeChallenges, Opportunities, and Recent Advances. Neuroscience 2024; 550:69-78. [PMID: 38763225 DOI: 10.1016/j.neuroscience.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/21/2024]
Abstract
Despite recent advances in acute stroke management, most patients experiencing a stroke will suffer from residual brain damage and functional impairment. Addressing those residual deficits would require neurorestoration, i.e., rebuilding brain tissue to repair the structural brain damage caused by stroke. However, there are major pathobiological, anatomical and technological hurdles making neurorestorative approaches remarkably challenging, and true neurorestoration after larger ischemic lesions could not yet be achieved. On the other hand, there has been steady advancement in our understanding of the limits of tissue regeneration in the adult mammalian brain as well as of the fundamental organization of brain tissue growth during embryo- and ontogenesis. This has been paralleled by the development of novel animal models to study stroke, advancement of biomaterials that can be used to support neurorestoration, and in stem cell technologies. This review gives a detailed explanation of the major hurdles so far preventing the achievement of neurorestoration after stroke. It will also describe novel concepts and advancements in biomaterial science, brain organoid culturing, and animal modeling that may enable the investigation of post-stroke neurorestorative approaches in translationally relevant setups. Finally, there will be a review of recent achievements in experimental studies that have the potential to be the starting point of research and development activities that may eventually bring post-stroke neurorestoration within reach.
Collapse
Affiliation(s)
- Paulina Michór
- University of Warwick, School of Life Sciences, Coventry CV4 7AL, United Kingdom
| | - Lydia Renardson
- University of Warwick, Warwick Medical School, Coventry CV4 7AL, United Kingdom
| | - Shen Li
- Department of Neurology and Psychiatry, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Johannes Boltze
- University of Warwick, School of Life Sciences, Coventry CV4 7AL, United Kingdom.
| |
Collapse
|
2
|
Wang Z, Huang C, Shi Z, Liu H, Han X, Chen Z, Li S, Wang Z, Huang J. A taurine-based hydrogel with the neuroprotective effect and the ability to promote neural stem cell proliferation. BIOMATERIALS ADVANCES 2024; 161:213895. [PMID: 38795474 DOI: 10.1016/j.bioadv.2024.213895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 05/05/2024] [Accepted: 05/14/2024] [Indexed: 05/28/2024]
Abstract
Ischemic stroke, a cerebrovascular disease caused by arterial occlusion in the brain, can lead to brain impairment and even death. Stem cell therapies have shown positive advantages to treat ischemic stroke because of their extended time window, but the cell viability is poor when transplanted into the brain directly. Therefore, a new hydrogel GelMA-T was developed by introducing taurine on GelMA to transplant neural stem cells. The GelMA-T displayed the desired photocuring ability, micropore structure, and cytocompatibility. Its compressive modulus was more similar to neural tissue compared to that of GelMA. The GelMA-T could protect SH-SY5Y cells from injury induced by OGD/R. Furthermore, the NE-4C cells showed better proliferation performance in GelMA-T than that in GelMA during both 2D and 3D cultures. All results demonstrate that GelMA-T possesses a neuroprotective effect for ischemia/reperfusion injury against ischemic stroke and plays a positive role in promoting NSC proliferation. The novel hydrogel is anticipated to function as cell vehicles for the transplantation of neural stem cells into the stroke cavity, aiming to treat ischemic stroke.
Collapse
Affiliation(s)
- Zhichao Wang
- Centre for Advanced Jet Engineering Technology (CaJET), Key Laboratory of High-efficiency and Clean Mechanical Manufacture (Ministry of Education), National Experimental Teaching Demonstration Center for Mechanical Engineering (Shandong University), School of Mechanical Engineering, Shandong University, Jinan 250061, China
| | - Chuanzhen Huang
- School of Mechanical Engineering, Yanshan University, Qinhuangdao 066004, China.
| | - Zhenyu Shi
- School of Mechanical Engineering, Hebei University of Technology, Tianjin 300401, China.
| | - Hanlian Liu
- Centre for Advanced Jet Engineering Technology (CaJET), Key Laboratory of High-efficiency and Clean Mechanical Manufacture (Ministry of Education), National Experimental Teaching Demonstration Center for Mechanical Engineering (Shandong University), School of Mechanical Engineering, Shandong University, Jinan 250061, China.
| | - Xu Han
- Centre for Advanced Jet Engineering Technology (CaJET), Key Laboratory of High-efficiency and Clean Mechanical Manufacture (Ministry of Education), National Experimental Teaching Demonstration Center for Mechanical Engineering (Shandong University), School of Mechanical Engineering, Shandong University, Jinan 250061, China
| | - Zhuang Chen
- Centre for Advanced Jet Engineering Technology (CaJET), Key Laboratory of High-efficiency and Clean Mechanical Manufacture (Ministry of Education), National Experimental Teaching Demonstration Center for Mechanical Engineering (Shandong University), School of Mechanical Engineering, Shandong University, Jinan 250061, China
| | - Shuying Li
- Centre for Advanced Jet Engineering Technology (CaJET), Key Laboratory of High-efficiency and Clean Mechanical Manufacture (Ministry of Education), National Experimental Teaching Demonstration Center for Mechanical Engineering (Shandong University), School of Mechanical Engineering, Shandong University, Jinan 250061, China
| | - Zhen Wang
- School of Mechanical Engineering, Yanshan University, Qinhuangdao 066004, China
| | - Jun Huang
- Centre for Advanced Jet Engineering Technology (CaJET), Key Laboratory of High-efficiency and Clean Mechanical Manufacture (Ministry of Education), National Experimental Teaching Demonstration Center for Mechanical Engineering (Shandong University), School of Mechanical Engineering, Shandong University, Jinan 250061, China
| |
Collapse
|
3
|
Zhang J, Chen Z, Chen Q. Advanced Nano-Drug Delivery Systems in the Treatment of Ischemic Stroke. Molecules 2024; 29:1848. [PMID: 38675668 PMCID: PMC11054753 DOI: 10.3390/molecules29081848] [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: 03/04/2024] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
In recent years, the frequency of strokes has been on the rise year by year and has become the second leading cause of death around the world, which is characterized by a high mortality rate, high recurrence rate, and high disability rate. Ischemic strokes account for a large percentage of strokes. A reperfusion injury in ischemic strokes is a complex cascade of oxidative stress, neuroinflammation, immune infiltration, and mitochondrial damage. Conventional treatments are ineffective, and the presence of the blood-brain barrier (BBB) leads to inefficient drug delivery utilization, so researchers are turning their attention to nano-drug delivery systems. Functionalized nano-drug delivery systems have been widely studied and applied to the study of cerebral ischemic diseases due to their favorable biocompatibility, high efficiency, strong specificity, and specific targeting ability. In this paper, we briefly describe the pathological process of reperfusion injuries in strokes and focus on the therapeutic research progress of nano-drug delivery systems in ischemic strokes, aiming to provide certain references to understand the progress of research on nano-drug delivery systems (NDDSs).
Collapse
Affiliation(s)
- Jiajie Zhang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (J.Z.); (Z.C.)
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; (J.Z.); (Z.C.)
| | - Qi Chen
- Interdisciplinary Institute for Medical Engineering, Fuzhou University, Fuzhou 350108, China
| |
Collapse
|
4
|
Wu X, Zhang T, Jia J, Chen Y, Zhang Y, Fang Z, Zhang C, Bai Y, Li Z, Li Y. Perspective insights into versatile hydrogels for stroke: From molecular mechanisms to functional applications. Biomed Pharmacother 2024; 173:116309. [PMID: 38479180 DOI: 10.1016/j.biopha.2024.116309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 02/13/2024] [Accepted: 02/17/2024] [Indexed: 03/27/2024] Open
Abstract
As the leading killer of life and health, stroke leads to limb paralysis, speech disorder, dysphagia, cognitive impairment, mental depression and other symptoms, which entail a significant financial burden to society and families. At present, physiology, clinical medicine, engineering, and materials science, advanced biomaterials standing on the foothold of these interdisciplinary disciplines provide new opportunities and possibilities for the cure of stroke. Among them, hydrogels have been endowed with more possibilities. It is well-known that hydrogels can be employed as potential biosensors, medication delivery vectors, and cell transporters or matrices in tissue engineering in tissue engineering, and outperform many traditional therapeutic drugs, surgery, and materials. Therefore, hydrogels become a popular scaffolding treatment option for stroke. Diverse synthetic hydrogels were designed according to different pathophysiological mechanisms from the recently reported literature will be thoroughly explored. The biological uses of several types of hydrogels will be highlighted, including pro-angiogenesis, pro-neurogenesis, anti-oxidation, anti-inflammation and anti-apoptosis. Finally, considerations and challenges of using hydrogels in the treatment of stroke are summarized.
Collapse
Affiliation(s)
- Xinghan Wu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Tiejun Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jing Jia
- Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Yining Chen
- Key laboratory for Leather Chemistry and Engineering of the Education Ministry, Sichuan University, Chengdu, Sichuan 610065, China
| | - Ying Zhang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenwei Fang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chenyu Zhang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yang Bai
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhengjun Li
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Yuwen Li
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
5
|
Khaire OT, Mhaske A, Prasad AG, Almalki WH, Srivastava N, Kesharwani P, Shukla R. State-of-the-art drug delivery system to target the lymphatics. J Drug Target 2024; 32:347-364. [PMID: 38253594 DOI: 10.1080/1061186x.2024.2309671] [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: 09/08/2023] [Accepted: 01/07/2024] [Indexed: 01/24/2024]
Abstract
PRIMARY OBJECTIVE The primary objective of the review is to assess the potential of lymphatic-targeted drug delivery systems, with a particular emphasis on their role in tumour therapy and vaccination efficacy. REASON FOR LYMPHATIC TARGETING The lymphatic system's crucial functions in maintaining bodily equilibrium, regulating metabolism, and orchestrating immune responses make it an ideal target for drug delivery. Lymph nodes, being primary sites for tumour metastasis, underscore the importance of targeting the lymphatic system for effective treatment. OUTCOME Nanotechnologies and innovative biomaterials have facilitated the development of lymphatic-targeted drug carriers, leveraging endogenous macromolecules to enhance drug delivery efficiency. Various systems such as liposomes, micelles, inorganic nanomaterials, hydrogels, and nano-capsules demonstrate significant potential for delivering drugs to the lymphatic system. CONCLUSION Understanding the physiological functions of the lymphatic system and its involvement in diseases underscores the promise of targeted drug delivery in improving treatment outcomes. The strategic targeting of the lymphatic system presents opportunities to enhance patient prognosis and advance therapeutic interventions across various medical contexts, indicating the importance of ongoing research and development in this area.
Collapse
Affiliation(s)
- Omkar T Khaire
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, UP, India
| | - Akshada Mhaske
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, UP, India
| | - Aprameya Ganesh Prasad
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Nidhi Srivastava
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, UP, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, New Delhi, India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, UP, India
| |
Collapse
|
6
|
Gao Y, Zhang TL, Zhang HJ, Gao J, Yang PF. A Promising Application of Injectable Hydrogels in Nerve Repair and Regeneration for Ischemic Stroke. Int J Nanomedicine 2024; 19:327-345. [PMID: 38229707 PMCID: PMC10790665 DOI: 10.2147/ijn.s442304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 12/13/2023] [Indexed: 01/18/2024] Open
Abstract
Ischemic stroke, a condition that often leads to severe nerve damage, induces complex pathological and physiological changes in nerve tissue. The mature central nervous system (CNS) lacks intrinsic regenerative capacity, resulting in a poor prognosis and long-term neurological impairments. There is no available therapy that can fully restore CNS functionality. However, the utilization of injectable hydrogels has emerged as a promising strategy for nerve repair and regeneration. Injectable hydrogels possess exceptional properties, such as biocompatibility, tunable mechanical properties, and the ability to provide a supportive environment for cell growth and tissue regeneration. Recently, various hydrogel-based tissue engineering approaches, including cell encapsulation, controlled release of therapeutic factors, and incorporation of bioactive molecules, have demonstrated great potential in the treatment of CNS injuries caused by ischemic stroke. This article aims to provide a comprehensive review of the application and development of injectable hydrogels for the treatment of ischemic stroke-induced CNS injuries, shedding light on their therapeutic prospects, challenges, recent advancements, and future directions. Additionally, it will discuss the underlying mechanisms involved in hydrogel-mediated nerve repair and regeneration, as well as the need for further preclinical and clinical studies to validate their efficacy and safety.
Collapse
Affiliation(s)
- Yuan Gao
- Oriental Pan-Vascular Devices Innovation College, University of Shanghai for Science and Technology, Shanghai, People's Republic of China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, People’s Republic of China
| | - Ting-Lin Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Hong-Jian Zhang
- Oriental Pan-Vascular Devices Innovation College, University of Shanghai for Science and Technology, Shanghai, People's Republic of China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, People’s Republic of China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| | - Peng-Fei Yang
- Oriental Pan-Vascular Devices Innovation College, University of Shanghai for Science and Technology, Shanghai, People's Republic of China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, People’s Republic of China
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, People’s Republic of China
| |
Collapse
|
7
|
Mu J, Hao P, Duan H, Zhao W, Wang Z, Yang Z, Li X. Non-human primate models of focal cortical ischemia for neuronal replacement therapy. J Cereb Blood Flow Metab 2023; 43:1456-1474. [PMID: 37254891 PMCID: PMC10414004 DOI: 10.1177/0271678x231179544] [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: 10/02/2022] [Revised: 03/13/2023] [Accepted: 04/26/2023] [Indexed: 06/01/2023]
Abstract
Despite the high prevalence, stroke remains incurable due to the limited regeneration capacity in the central nervous system. Neuronal replacement strategies are highly diverse biomedical fields that attempt to replace lost neurons by utilizing exogenous stem cell transplants, biomaterials, and direct neuronal reprogramming. Although these approaches have achieved encouraging outcomes mostly in the rodent stroke model, further preclinical validation in non-human primates (NHP) is still needed prior to clinical trials. In this paper, we briefly review the recent progress of promising neuronal replacement therapy in NHP stroke studies. Moreover, we summarize the key characteristics of the NHP as highly valuable translational tools and discuss (1) NHP species and their advantages in terms of genetics, physiology, neuroanatomy, immunology, and behavior; (2) various methods for establishing NHP focal ischemic models to study the regenerative and plastic changes associated with motor functional recovery; and (3) a comprehensive analysis of experimentally and clinically accessible outcomes and a potential adaptive mechanism. Our review specifically aims to facilitate the selection of the appropriate NHP cortical ischemic models and efficient prognostic evaluation methods in preclinical stroke research design of neuronal replacement strategies.
Collapse
Affiliation(s)
- Jiao Mu
- Beijing Key Laboratory for Biomaterials and Neural Regeneration, School of Engineering Medicine, Beihang University, Beijing, China
| | - Peng Hao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Hongmei Duan
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Wen Zhao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zijue Wang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zhaoyang Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiaoguang Li
- Beijing Key Laboratory for Biomaterials and Neural Regeneration, School of Engineering Medicine, Beihang University, Beijing, China
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
8
|
Liu H, Wei T, Huang Q, Liu W, Yang Y, Jin Y, Wu D, Yuan K, Zhang P. The roles, mechanism, and mobilization strategy of endogenous neural stem cells in brain injury. Front Aging Neurosci 2022; 14:924262. [PMID: 36062152 PMCID: PMC9428262 DOI: 10.3389/fnagi.2022.924262] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Brain injury poses a heavy disease burden in the world, resulting in chronic deficits. Therapies for brain injuries have been focused on pharmacologic, small molecule, endocrine and cell-based therapies. Endogenous neural stem cells (eNSCs) are a group of stem cells which can be activated in vivo by damage, neurotrophic factors, physical factor stimulation, and physical exercise. The activated eNSCs can proliferate, migrate and differentiate into neuron, oligodendrocyte and astrocyte, and play an important role in brain injury repair and neural plasticity. The roles of eNSCs in the repair of brain injury include but are not limited to ameliorating cognitive function, improving learning and memory function, and promoting functional gait behaviors. The activation and mobilization of eNSCs is important to the repair of injured brain. In this review we describe the current knowledge of the common character of brain injury, the roles and mechanism of eNSCs in brain injury. And then we discuss the current mobilization strategy of eNSCs following brain injury. We hope that a comprehensive awareness of the roles and mobilization strategy of eNSCs in the repair of cerebral ischemia may help to find some new therapeutic targets and strategy for treatment of stroke.
Collapse
Affiliation(s)
- Haijing Liu
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Tao Wei
- Library, Kunming Medical University, Kunming, China
- School of Continuing Education, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Qin Huang
- Department of Teaching Affairs and Administration, Kunming Medical University, Kunming, China
| | - Wei Liu
- School of Public Health, Kunming Medical University, Kunming, China
| | - Yaopeng Yang
- Department of Pulmonary and Critical Care Medicine, The Sixth Affiliated Hospital of Kunming Medical University, Yuxi, China
| | - Yaju Jin
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Danli Wu
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Kai Yuan
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
- *Correspondence: Kai Yuan,
| | - Pengyue Zhang
- Key Laboratory of Acupuncture and Massage for Treatment of Encephalopathy, College of Acupuncture, Tuina and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
- Pengyue Zhang,
| |
Collapse
|
9
|
Nebie O, Buée L, Blum D, Burnouf T. Can the administration of platelet lysates to the brain help treat neurological disorders? Cell Mol Life Sci 2022; 79:379. [PMID: 35750991 PMCID: PMC9243829 DOI: 10.1007/s00018-022-04397-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/09/2022] [Accepted: 05/23/2022] [Indexed: 11/03/2022]
Abstract
Neurodegenerative disorders of the central nervous system (CNS) and brain traumatic insults are characterized by complex overlapping pathophysiological alterations encompassing neuroinflammation, alterations of synaptic functions, oxidative stress, and progressive neurodegeneration that eventually lead to irreversible motor and cognitive dysfunctions. A single pharmacological approach is unlikely to provide a complementary set of molecular therapeutic actions suitable to resolve these complex pathologies. Recent preclinical data are providing evidence-based scientific rationales to support biotherapies based on administering neurotrophic factors and extracellular vesicles present in the lysates of human platelets collected from healthy donors to the brain. Here, we present the most recent findings on the composition of the platelet proteome that can activate complementary signaling pathways in vivo to trigger neuroprotection, synapse protection, anti-inflammation, antioxidation, and neurorestoration. We also report experimental data where the administration of human platelet lysates (HPL) was safe and resulted in beneficial neuroprotective effects in established rodent models of neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, traumatic brain injury, and stroke. Platelet-based biotherapies, prepared from collected platelet concentrates (PC), are emerging as a novel pragmatic and accessible translational therapeutic strategy for treating neurological diseases. Based on this assumption, we further elaborated on various clinical, manufacturing, and regulatory issues that need to be addressed to ensure the ethical supply, quality, and safety of HPL preparations for treating neurodegenerative and traumatic pathologies of the CNS. HPL made from PC may become a unique approach for scientifically based treatments of neurological disorders readily accessible in low-, middle-, and high-income countries.
Collapse
Affiliation(s)
- Ouada Nebie
- College of Biomedical Engineering, Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience and Cognition, 59045, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, LiCEND, 59000, Lille, France
| | - Luc Buée
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience and Cognition, 59045, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, LiCEND, 59000, Lille, France
- NeuroTMULille International Laboratory, Univ. Lille, Lille, France
| | - David Blum
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience and Cognition, 59045, Lille, France.
- Alzheimer and Tauopathies, LabEx DISTALZ, LiCEND, 59000, Lille, France.
- NeuroTMULille International Laboratory, Univ. Lille, Lille, France.
- NeuroTMULille International Laboratory, Taipei Medical University, Taipei, 11031, Taiwan.
| | - Thierry Burnouf
- College of Biomedical Engineering, Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan.
- NeuroTMULille International Laboratory, Taipei Medical University, Taipei, 11031, Taiwan.
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan.
- International PhD Program in Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- Brain and Consciousness Research Centre, Taipei Medical University Shuang-Ho Hospital, New Taipei City, 23561, Taiwan.
- Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
10
|
El-Husseiny HM, Mady EA, Hamabe L, Abugomaa A, Shimada K, Yoshida T, Tanaka T, Yokoi A, Elbadawy M, Tanaka R. Smart/stimuli-responsive hydrogels: Cutting-edge platforms for tissue engineering and other biomedical applications. Mater Today Bio 2022; 13:100186. [PMID: 34917924 PMCID: PMC8669385 DOI: 10.1016/j.mtbio.2021.100186] [Citation(s) in RCA: 124] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/14/2021] [Accepted: 12/08/2021] [Indexed: 02/07/2023] Open
Abstract
Recently, biomedicine and tissue regeneration have emerged as great advances that impacted the spectrum of healthcare. This left the door open for further improvement of their applications to revitalize the impaired tissues. Hence, restoring their functions. The implementation of therapeutic protocols that merge biomimetic scaffolds, bioactive molecules, and cells plays a pivotal role in this track. Smart/stimuli-responsive hydrogels are remarkable three-dimensional (3D) bioscaffolds intended for tissue engineering and other biomedical purposes. They can simulate the physicochemical, mechanical, and biological characters of the innate tissues. Also, they provide the aqueous conditions for cell growth, support 3D conformation, provide mechanical stability for the cells, and serve as potent delivery matrices for bioactive molecules. Many natural and artificial polymers were broadly utilized to design these intelligent platforms with novel advanced characteristics and tailored functionalities that fit such applications. In the present review, we highlighted the different types of smart/stimuli-responsive hydrogels with emphasis on their synthesis scheme. Besides, the mechanisms of their responsiveness to different stimuli were elaborated. Their potential for tissue engineering applications was discussed. Furthermore, their exploitation in other biomedical applications as targeted drug delivery, smart biosensors, actuators, 3D and 4D printing, and 3D cell culture were outlined. In addition, we threw light on smart self-healing hydrogels and their applications in biomedicine. Eventually, we presented their future perceptions in biomedical and tissue regeneration applications. Conclusively, current progress in the design of smart/stimuli-responsive hydrogels enhances their prospective to function as intelligent, and sophisticated systems in different biomedical applications.
Collapse
Affiliation(s)
- Hussein M. El-Husseiny
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo, 1838509, Japan
- Department of Surgery, Anesthesiology, and Radiology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya, 13736, Egypt
| | - Eman A. Mady
- Department of Animal Hygiene, Behavior and Management, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya, 13736, Egypt
| | - Lina Hamabe
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo, 1838509, Japan
| | - Amira Abugomaa
- Faculty of Veterinary Medicine, Mansoura University, Mansoura, Dakahliya, 35516, Egypt
| | - Kazumi Shimada
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo, 1838509, Japan
- Division of Research Animal Laboratory and Translational Medicine, Research and Development Center, Osaka Medical College, 2-7 Daigaku-machi, Takatsuki City, Osaka, 569-8686, Japan
| | - Tomohiko Yoshida
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo, 1838509, Japan
| | - Takashi Tanaka
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo, 1838509, Japan
| | - Aimi Yokoi
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo, 1838509, Japan
| | - Mohamed Elbadawy
- Department of Pharmacology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Elqaliobiya, 13736, Egypt
| | - Ryou Tanaka
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai Cho, Fuchu-shi, Tokyo, 1838509, Japan
| |
Collapse
|
11
|
Totten JD, Alhadrami HA, Jiffri EH, McMullen CJ, Seib FP, Carswell HVO. Towards clinical translation of 'second-generation' regenerative stroke therapies: hydrogels as game changers? Trends Biotechnol 2021; 40:708-720. [PMID: 34815101 DOI: 10.1016/j.tibtech.2021.10.009] [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: 08/09/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 12/19/2022]
Abstract
Stroke is an unmet clinical need with a paucity of treatments, at least in part because chronic stroke pathologies are prohibitive to 'first-generation' stem cell-based therapies. Hydrogels can remodel the hostile stroke microenvironment to aid endogenous and exogenous regenerative repair processes. However, no clinical trials have yet been successfully commissioned for these 'second-generation' hydrogel-based therapies for chronic ischaemic stroke regeneration. This review recommends a path forward to improve hydrogel technology for future clinical translation for stroke. Specifically, we suggest that a better understanding of human host stroke tissue-hydrogel interactions in addition to the effects of scaling up hydrogel volume to human-sized cavities would help guide translation of these second-generation regenerative stroke therapies.
Collapse
Affiliation(s)
- John D Totten
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Hani A Alhadrami
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Essam H Jiffri
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Calum J McMullen
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - F Philipp Seib
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK; EPSRC Future Manufacturing Research Hub for Continuous Manufacturing and Advanced Crystallisation (CMAC), University of Strathclyde, Technology and Innovation Centre, Glasgow G1 1RD, UK
| | - Hilary V O Carswell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK.
| |
Collapse
|
12
|
Rial-Hermida MI, Rey-Rico A, Blanco-Fernandez B, Carballo-Pedrares N, Byrne EM, Mano JF. Recent Progress on Polysaccharide-Based Hydrogels for Controlled Delivery of Therapeutic Biomolecules. ACS Biomater Sci Eng 2021; 7:4102-4127. [PMID: 34137581 PMCID: PMC8919265 DOI: 10.1021/acsbiomaterials.0c01784] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 06/02/2021] [Indexed: 12/24/2022]
Abstract
A plethora of applications using polysaccharides have been developed in recent years due to their availability as well as their frequent nontoxicity and biodegradability. These polymers are usually obtained from renewable sources or are byproducts of industrial processes, thus, their use is collaborative in waste management and shows promise for an enhanced sustainable circular economy. Regarding the development of novel delivery systems for biotherapeutics, the potential of polysaccharides is attractive for the previously mentioned properties and also for the possibility of chemical modification of their structures, their ability to form matrixes of diverse architectures and mechanical properties, as well as for their ability to maintain bioactivity following incorporation of the biomolecules into the matrix. Biotherapeutics, such as proteins, growth factors, gene vectors, enzymes, hormones, DNA/RNA, and antibodies are currently in use as major therapeutics in a wide range of pathologies. In the present review, we summarize recent progress in the development of polysaccharide-based hydrogels of diverse nature, alone or in combination with other polymers or drug delivery systems, which have been implemented in the delivery of biotherapeutics in the pharmaceutical and biomedical fields.
Collapse
Affiliation(s)
- M. Isabel Rial-Hermida
- Department
of Chemistry, CICECO−Aveiro Institute of Materials, University of Aveiro 3810-193 Aveiro, Portugal
| | - Ana Rey-Rico
- Cell
Therapy and Regenerative Medicine
Unit, Centro de Investigacións Científicas Avanzadas
(CICA), Universidade da Coruña, 15071 A Coruña, Spain
| | - Barbara Blanco-Fernandez
- Institute
for Bioengineering of Catalonia (IBEC), The Barcelona Institute of
Science and Technology, 08028 Barcelona, Spain
- CIBER
en Bioingeniería, Biomateriales y
Nanomedicina, CIBER-BBN, 28029 Madrid, Spain
| | - Natalia Carballo-Pedrares
- Cell
Therapy and Regenerative Medicine
Unit, Centro de Investigacións Científicas Avanzadas
(CICA), Universidade da Coruña, 15071 A Coruña, Spain
| | - Eimear M. Byrne
- Wellcome-Wolfson
Institute For Experimental Medicine, Queen’s
University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - João F. Mano
- Department
of Chemistry, CICECO−Aveiro Institute of Materials, University of Aveiro 3810-193 Aveiro, Portugal
| |
Collapse
|
13
|
Echeverria Molina MI, Malollari KG, Komvopoulos K. Design Challenges in Polymeric Scaffolds for Tissue Engineering. Front Bioeng Biotechnol 2021; 9:617141. [PMID: 34195178 PMCID: PMC8236583 DOI: 10.3389/fbioe.2021.617141] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 03/08/2021] [Indexed: 12/11/2022] Open
Abstract
Numerous surgical procedures are daily performed worldwide to replace and repair damaged tissue. Tissue engineering is the field devoted to the regeneration of damaged tissue through the incorporation of cells in biocompatible and biodegradable porous constructs, known as scaffolds. The scaffolds act as host biomaterials of the incubating cells, guiding their attachment, growth, differentiation, proliferation, phenotype, and migration for the development of new tissue. Furthermore, cellular behavior and fate are bound to the biodegradation of the scaffold during tissue generation. This article provides a critical appraisal of how key biomaterial scaffold parameters, such as structure architecture, biochemistry, mechanical behavior, and biodegradability, impart the needed morphological, structural, and biochemical cues for eliciting cell behavior in various tissue engineering applications. Particular emphasis is given on specific scaffold attributes pertaining to skin and brain tissue generation, where further progress is needed (skin) or the research is at a relatively primitive stage (brain), and the enumeration of some of the most important challenges regarding scaffold constructs for tissue engineering.
Collapse
Affiliation(s)
- Maria I Echeverria Molina
- Department of Mechanical Engineering, University of California, Berkeley, Berkeley, CA, United States
| | - Katerina G Malollari
- Department of Mechanical Engineering, University of California, Berkeley, Berkeley, CA, United States
| | - Kyriakos Komvopoulos
- Department of Mechanical Engineering, University of California, Berkeley, Berkeley, CA, United States
| |
Collapse
|
14
|
Asgharzade S, Talaei A, Farkhondeh T, Forouzanfar F. Combining Growth Factor and Stem Cell Therapy for Stroke Rehabilitation, A Review. Curr Drug Targets 2021; 21:781-791. [PMID: 31914912 DOI: 10.2174/1389450121666200107100747] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/28/2019] [Accepted: 12/18/2019] [Indexed: 12/12/2022]
Abstract
Stroke is a serious, life-threatening condition demanding vigorous search for new therapies. Recent research has focused on stem cell-based therapies as a viable choice following ischemic stroke, based on studies displaying that stem cells transplanted to the brain not only survive but also cause functional recovery. Growth factors defined as polypeptides that regulate the growth and differentiation of many cell types. Many studies have demonstrated that combined use of growth factors may increase results by the stimulation of endogenous neurogenesis, anti-inflammatory, neuroprotection properties, and enhancement of stem cell survival rates and so may be more effective than a single stem cell therapy. This paper reviews and discusses the most promising new stroke recovery research, including combination treatment.
Collapse
Affiliation(s)
- Samira Asgharzade
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Andisheh Talaei
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Fatemeh Forouzanfar
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
15
|
Ucar B. Natural biomaterials in brain repair: A focus on collagen. Neurochem Int 2021; 146:105033. [PMID: 33785419 DOI: 10.1016/j.neuint.2021.105033] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 03/07/2021] [Accepted: 03/22/2021] [Indexed: 12/16/2022]
Abstract
Biomaterials derived from natural resources have increasingly been used for versatile applications in the central nervous system (CNS). Thanks to their biocompatibility and biodegradability, natural biomaterials offer vast possibilities for future clinical repair strategies for the CNS. These materials can be used for diverse applications such as hydrogels to fill the tissue cavities, microparticles to deliver drugs across the blood-brain barrier, and scaffolds to transplant stem cells. In this review, various uses of prominent protein and polysaccharide biomaterials, with a special focus on collagen, in repair and regenerative applications for the brain are summarized together with their individual advantages and disadvantages.
Collapse
Affiliation(s)
- Buket Ucar
- Laboratory of Psychiatry and Experimental Alzheimer's Research, Medical University of Innsbruck, Austria.
| |
Collapse
|
16
|
Carvalho IC, Mansur HS, Leonel AG, Mansur AAP, Lobato ZIP. Soft matter polysaccharide-based hydrogels as versatile bioengineered platforms for brain tissue repair and regeneration. Int J Biol Macromol 2021; 182:1091-1111. [PMID: 33892028 DOI: 10.1016/j.ijbiomac.2021.04.116] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/12/2021] [Accepted: 04/17/2021] [Indexed: 01/08/2023]
Abstract
Acute or chronic brain injuries promote deaths and the life-long debilitating neurological status where, despite advances in therapeutic strategies, clinical outcome hardly achieves total patient recovery. In recent decades, brain tissue engineering emerged as an encouraging area of research for helping in damaged central nervous system (CNS) recovery. Polysaccharides are abundant naturally occurring biomacromolecules with a great potential enhancement of advanced technologies in brain tissue repair and regeneration (BTRR). Besides carrying rich biological information, polysaccharides can interact and communicate with biomolecules, including glycosaminoglycans present in cell membranes and many signaling moieties, growth factors, chemokines, and axon guidance molecules. This review includes a comprehensive investigation of the current progress on designing and developing polysaccharide-based soft matter biomaterials for BTRR. Although few interesting reviews concerning BTRR have been reported, this is the first report specifically focusing on covering multiple polysaccharides and polysaccharide-based functionalized biomacromolecules in this emerging and intriguing field of multidisciplinary knowledge. This review aims to cover the state of art challenges and prospects of this fascinating field while presenting the richness of possibilities of using these natural biomacromolecules for advanced biomaterials in prospective neural tissue engineering applications.
Collapse
Affiliation(s)
- Isadora C Carvalho
- Center of Nanoscience, Nanotechnology and Innovation - CeNano(2)I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 Belo Horizonte/M.G., Brazil
| | - Herman S Mansur
- Center of Nanoscience, Nanotechnology and Innovation - CeNano(2)I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 Belo Horizonte/M.G., Brazil.
| | - Alice G Leonel
- Center of Nanoscience, Nanotechnology and Innovation - CeNano(2)I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 Belo Horizonte/M.G., Brazil
| | - Alexandra A P Mansur
- Center of Nanoscience, Nanotechnology and Innovation - CeNano(2)I, Department of Metallurgical and Materials Engineering, Federal University of Minas Gerais - UFMG, Av. Antônio Carlos, 6627 Belo Horizonte/M.G., Brazil
| | - Zelia I P Lobato
- Department of Preventive Veterinary Medicine, Veterinary School, Federal University of Minas Gerais - UFMG, Brazil
| |
Collapse
|
17
|
Ibeanu N, Egbu R, Onyekuru L, Javaheri H, Tee Khaw P, R. Williams G, Brocchini S, Awwad S. Injectables and Depots to Prolong Drug Action of Proteins and Peptides. Pharmaceutics 2020; 12:E999. [PMID: 33096803 PMCID: PMC7589296 DOI: 10.3390/pharmaceutics12100999] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/29/2020] [Accepted: 10/12/2020] [Indexed: 12/30/2022] Open
Abstract
Proteins and peptides have emerged in recent years to treat a wide range of multifaceted diseases such as cancer, diabetes and inflammation. The emergence of polypeptides has yielded advancements in the fields of biopharmaceutical production and formulation. Polypeptides often display poor pharmacokinetics, limited permeability across biological barriers, suboptimal biodistribution, and some proclivity for immunogenicity. Frequent administration of polypeptides is generally required to maintain adequate therapeutic levels, which can limit efficacy and compliance while increasing adverse reactions. Many strategies to increase the duration of action of therapeutic polypeptides have been described with many clinical products having been developed. This review describes approaches to optimise polypeptide delivery organised by the commonly used routes of administration. Future innovations in formulation may hold the key to the continued successful development of proteins and peptides with optimal clinical properties.
Collapse
Affiliation(s)
- Nkiruka Ibeanu
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1V 9EL, UK;
| | - Raphael Egbu
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
| | - Lesley Onyekuru
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
| | - Hoda Javaheri
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
| | - Peng Tee Khaw
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1V 9EL, UK;
| | - Gareth R. Williams
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
| | - Steve Brocchini
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1V 9EL, UK;
| | - Sahar Awwad
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1V 9EL, UK;
| |
Collapse
|
18
|
An Injectable Hyaluronan-Methylcellulose (HAMC) Hydrogel Combined with Wharton's Jelly-Derived Mesenchymal Stromal Cells (WJ-MSCs) Promotes Degenerative Disc Repair. Int J Mol Sci 2020; 21:ijms21197391. [PMID: 33036383 PMCID: PMC7582266 DOI: 10.3390/ijms21197391] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 09/25/2020] [Accepted: 10/02/2020] [Indexed: 02/06/2023] Open
Abstract
Intervertebral disc (IVD) degeneration is one of the predominant causes of chronic low back pain (LBP), which is a leading cause of disability worldwide. Despite substantial progress in cell therapy for the treatment of IVD degeneration, significant challenges remain for clinical application. Here, we investigated the effectiveness of hyaluronan-methylcellulose (HAMC) hydrogels loaded with Wharton's Jelly-derived mesenchymal stromal cell (WJ-MSCs) in vitro and in a rat coccygeal IVD degeneration model. Following induction of injury-induced IVD degeneration, female Sprague-Dawley rats were randomized into four groups to undergo a single intradiscal injection of the following: (1) phosphate buffered saline (PBS) vehicle, (2) HAMC, (3) WJ-MSCs (2 × 104 cells), and (4) WJ-MSCs-loaded HAMC (WJ-MSCs/HAMC) (n = 10/each group). Coccygeal discs were removed following sacrifice 6 weeks after implantation for radiologic and histologic analysis. We confirmed previous findings that encapsulation in HAMC increases the viability of WJ-MSCs for disc repair. The HAMC gel maintained significant cell viability in vitro. In addition, combined implantation of WJ-MSCs and HAMC significantly promoted degenerative disc repair compared to WJ-MSCs alone, presumably by improving nucleus pulposus cells viability and decreasing extracellular matrix degradation. Our results suggest that WJ-MSCs-loaded HAMC promotes IVD repair more effectively than cell injection alone and supports the potential clinical use of HAMC for cell delivery to arrest IVD degeneration or to promote IVD regeneration.
Collapse
|
19
|
Jensen G, Holloway JL, Stabenfeldt SE. Hyaluronic Acid Biomaterials for Central Nervous System Regenerative Medicine. Cells 2020; 9:E2113. [PMID: 32957463 PMCID: PMC7565873 DOI: 10.3390/cells9092113] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/09/2020] [Accepted: 09/11/2020] [Indexed: 12/16/2022] Open
Abstract
Hyaluronic acid (HA) is a primary component of the brain extracellular matrix and functions through cellular receptors to regulate cell behavior within the central nervous system (CNS). These behaviors, such as migration, proliferation, differentiation, and inflammation contribute to maintenance and homeostasis of the CNS. However, such equilibrium is disrupted following injury or disease leading to significantly altered extracellular matrix milieu and cell functions. This imbalance thereby inhibits inherent homeostatic processes that support critical tissue health and functionality in the CNS. To mitigate the damage sustained by injury/disease, HA-based tissue engineering constructs have been investigated for CNS regenerative medicine applications. HA's effectiveness in tissue healing and regeneration is primarily attributed to its impact on cell signaling and the ease of customizing chemical and mechanical properties. This review focuses on recent findings to highlight the applications of HA-based materials in CNS regenerative medicine.
Collapse
Affiliation(s)
- Gregory Jensen
- Chemical Engineering, School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ 85224, USA;
| | - Julianne L. Holloway
- Chemical Engineering, School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ 85224, USA;
| | - Sarah E. Stabenfeldt
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
20
|
Suda S, Nito C, Yokobori S, Sakamoto Y, Nakajima M, Sowa K, Obinata H, Sasaki K, Savitz SI, Kimura K. Recent Advances in Cell-Based Therapies for Ischemic Stroke. Int J Mol Sci 2020; 21:ijms21186718. [PMID: 32937754 PMCID: PMC7555943 DOI: 10.3390/ijms21186718] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/09/2020] [Accepted: 09/10/2020] [Indexed: 12/14/2022] Open
Abstract
Stroke is the most prevalent cardiovascular disease worldwide, and is still one of the leading causes of death and disability. Stem cell-based therapy is actively being investigated as a new potential treatment for certain neurological disorders, including stroke. Various types of cells, including bone marrow mononuclear cells, bone marrow mesenchymal stem cells, dental pulp stem cells, neural stem cells, inducible pluripotent stem cells, and genetically modified stem cells have been found to improve neurological outcomes in animal models of stroke, and there are some ongoing clinical trials assessing their efficacy in humans. In this review, we aim to summarize the recent advances in cell-based therapies to treat stroke.
Collapse
Affiliation(s)
- Satoshi Suda
- Department of Neurology, Nippon Medical School, Tokyo 113-8602, Japan; (C.N.); (Y.S.); (M.N.); (K.S.); (K.K.)
- Correspondence: ; Tel.: +81-3-3822-2131; Fax: +81-3-3822-4865
| | - Chikako Nito
- Department of Neurology, Nippon Medical School, Tokyo 113-8602, Japan; (C.N.); (Y.S.); (M.N.); (K.S.); (K.K.)
| | - Shoji Yokobori
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8602, Japan; (S.Y.); (H.O.); (K.S.)
| | - Yuki Sakamoto
- Department of Neurology, Nippon Medical School, Tokyo 113-8602, Japan; (C.N.); (Y.S.); (M.N.); (K.S.); (K.K.)
| | - Masataka Nakajima
- Department of Neurology, Nippon Medical School, Tokyo 113-8602, Japan; (C.N.); (Y.S.); (M.N.); (K.S.); (K.K.)
| | - Kota Sowa
- Department of Neurology, Nippon Medical School, Tokyo 113-8602, Japan; (C.N.); (Y.S.); (M.N.); (K.S.); (K.K.)
| | - Hirofumi Obinata
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8602, Japan; (S.Y.); (H.O.); (K.S.)
| | - Kazuma Sasaki
- Department of Emergency and Critical Care Medicine, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8602, Japan; (S.Y.); (H.O.); (K.S.)
| | - Sean I. Savitz
- Institute for Stroke and Cerebrovascular Disease, UTHealth, Houston, TX 77030, USA;
| | - Kazumi Kimura
- Department of Neurology, Nippon Medical School, Tokyo 113-8602, Japan; (C.N.); (Y.S.); (M.N.); (K.S.); (K.K.)
| |
Collapse
|
21
|
Rey F, Barzaghini B, Nardini A, Bordoni M, Zuccotti GV, Cereda C, Raimondi MT, Carelli S. Advances in Tissue Engineering and Innovative Fabrication Techniques for 3-D-Structures: Translational Applications in Neurodegenerative Diseases. Cells 2020; 9:cells9071636. [PMID: 32646008 PMCID: PMC7407518 DOI: 10.3390/cells9071636] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/01/2020] [Accepted: 07/06/2020] [Indexed: 12/11/2022] Open
Abstract
In the field of regenerative medicine applied to neurodegenerative diseases, one of the most important challenges is the obtainment of innovative scaffolds aimed at improving the development of new frontiers in stem-cell therapy. In recent years, additive manufacturing techniques have gained more and more relevance proving the great potential of the fabrication of precision 3-D scaffolds. In this review, recent advances in additive manufacturing techniques are presented and discussed, with an overview on stimulus-triggered approaches, such as 3-D Printing and laser-based techniques, and deposition-based approaches. Innovative 3-D bioprinting techniques, which allow the production of cell/molecule-laden scaffolds, are becoming a promising frontier in disease modelling and therapy. In this context, the specific biomaterial, stiffness, precise geometrical patterns, and structural properties are to be considered of great relevance for their subsequent translational applications. Moreover, this work reports numerous recent advances in neural diseases modelling and specifically focuses on pre-clinical and clinical translation for scaffolding technology in multiple neurodegenerative diseases.
Collapse
Affiliation(s)
- Federica Rey
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, Via Grassi 74, 20157 Milan, Italy; (F.R.); (G.V.Z.)
- Pediatric Clinical Research Center Fondazione “Romeo ed Enrica Invernizzi”, University of Milano, Via Grassi 74, 20157 Milano, Italy
| | - Bianca Barzaghini
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milano, Italy; (B.B.); (A.N.)
| | - Alessandra Nardini
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milano, Italy; (B.B.); (A.N.)
| | - Matteo Bordoni
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Centro di Eccellenza sulle Malattie Neurodegenerative, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy;
| | - Gian Vincenzo Zuccotti
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, Via Grassi 74, 20157 Milan, Italy; (F.R.); (G.V.Z.)
- Pediatric Clinical Research Center Fondazione “Romeo ed Enrica Invernizzi”, University of Milano, Via Grassi 74, 20157 Milano, Italy
| | - Cristina Cereda
- Genomic and post-Genomic Center, IRCCS Mondino Foundation, Via Mondino 2, 27100 Pavia, Italy;
| | - Manuela Teresa Raimondi
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milano, Italy; (B.B.); (A.N.)
- Correspondence: (M.T.R.); (S.C.); Tel.: +390-223-994-306 (M.T.R.); +390-250-319-825 (S.C.)
| | - Stephana Carelli
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, Via Grassi 74, 20157 Milan, Italy; (F.R.); (G.V.Z.)
- Pediatric Clinical Research Center Fondazione “Romeo ed Enrica Invernizzi”, University of Milano, Via Grassi 74, 20157 Milano, Italy
- Correspondence: (M.T.R.); (S.C.); Tel.: +390-223-994-306 (M.T.R.); +390-250-319-825 (S.C.)
| |
Collapse
|
22
|
Design and evaluation of a biosynthesized cellulose drug releasing duraplasty. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 110:110677. [DOI: 10.1016/j.msec.2020.110677] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 12/20/2019] [Accepted: 01/18/2020] [Indexed: 12/16/2022]
|
23
|
Purvis EM, O'Donnell JC, Chen HI, Cullen DK. Tissue Engineering and Biomaterial Strategies to Elicit Endogenous Neuronal Replacement in the Brain. Front Neurol 2020; 11:344. [PMID: 32411087 PMCID: PMC7199479 DOI: 10.3389/fneur.2020.00344] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 04/07/2020] [Indexed: 12/19/2022] Open
Abstract
Neurogenesis in the postnatal mammalian brain is known to occur in the dentate gyrus of the hippocampus and the subventricular zone. These neurogenic niches serve as endogenous sources of neural precursor cells that could potentially replace neurons that have been lost or damaged throughout the brain. As an example, manipulation of the subventricular zone to augment neurogenesis has become a popular strategy for attempting to replace neurons that have been lost due to acute brain injury or neurodegenerative disease. In this review article, we describe current experimental strategies to enhance the regenerative potential of endogenous neural precursor cell sources by enhancing cell proliferation in neurogenic regions and/or redirecting migration, including pharmacological, biomaterial, and tissue engineering strategies. In particular, we discuss a novel replacement strategy based on exogenously biofabricated "living scaffolds" that could enhance and redirect endogenous neuroblast migration from the subventricular zone to specified regions throughout the brain. This approach utilizes the first implantable, biomimetic tissue-engineered rostral migratory stream, thereby leveraging the brain's natural mechanism for sustained neuronal replacement by replicating the structure and function of the native rostral migratory stream. Across all these strategies, we discuss several challenges that need to be overcome to successfully harness endogenous neural precursor cells to promote nervous system repair and functional restoration. With further development, the diverse and innovative tissue engineering and biomaterial strategies explored in this review have the potential to facilitate functional neuronal replacement to mitigate neurological and psychiatric symptoms caused by injury, developmental disorders, or neurodegenerative disease.
Collapse
Affiliation(s)
- Erin M. Purvis
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - John C. O'Donnell
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - H. Isaac Chen
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
| | - D. Kacy Cullen
- Center for Brain Injury & Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, United States
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
24
|
Injectable hydrogel enables local and sustained co-delivery to the brain: Two clinically approved biomolecules, cyclosporine and erythropoietin, accelerate functional recovery in rat model of stroke. Biomaterials 2020; 235:119794. [DOI: 10.1016/j.biomaterials.2020.119794] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 01/06/2020] [Accepted: 01/15/2020] [Indexed: 12/20/2022]
|
25
|
Madhusudanan P, Raju G, Shankarappa S. Hydrogel systems and their role in neural tissue engineering. J R Soc Interface 2020; 17:20190505. [PMID: 31910776 PMCID: PMC7014813 DOI: 10.1098/rsif.2019.0505] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 12/02/2019] [Indexed: 12/27/2022] Open
Abstract
Neural tissue engineering (NTE) is a rapidly progressing field that promises to address several serious neurological conditions that are currently difficult to treat. Selecting the right scaffolding material to promote neural and non-neural cell differentiation as well as axonal growth is essential for the overall design strategy for NTE. Among the varieties of scaffolds, hydrogels have proved to be excellent candidates for culturing and differentiating cells of neural origin. Considering the intrinsic resistance of the nervous system against regeneration, hydrogels have been abundantly used in applications that involve the release of neurotrophic factors, antagonists of neural growth inhibitors and other neural growth-promoting agents. Recent developments in the field include the utilization of encapsulating hydrogels in neural cell therapy for providing localized trophic support and shielding neural cells from immune activity. In this review, we categorize and discuss the various hydrogel-based strategies that have been examined for neural-specific applications and also highlight their strengths and weaknesses. We also discuss future prospects and challenges ahead for the utilization of hydrogels in NTE.
Collapse
Affiliation(s)
| | | | - Sahadev Shankarappa
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham, Kochi 682041, India
| |
Collapse
|
26
|
Growth factor delivery: Defining the next generation platforms for tissue engineering. J Control Release 2019; 306:40-58. [DOI: 10.1016/j.jconrel.2019.05.028] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/15/2019] [Accepted: 05/17/2019] [Indexed: 12/14/2022]
|
27
|
Guo T, Guo Y, Gong Y, Ji J, Hao S, Deng J, Wang B. An enhanced charge-driven intranasal delivery of nicardipine attenuates brain injury after intracerebral hemorrhage. Int J Pharm 2019; 566:46-56. [PMID: 31121211 DOI: 10.1016/j.ijpharm.2019.05.050] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/30/2019] [Accepted: 05/19/2019] [Indexed: 12/20/2022]
Abstract
Intranasal drug delivery provided an alternative and effective approach for the intervention of an intracerebral hemorrhage (ICH). However, the short retention time at the absorption site and slow drug transport in intranasal gel influence the drug bioavailability and outcome of ICH. Herein, we fabricated a novel intranasal gel with oriented drug migration utilizing a charge-driven strategy to attenuate brain injury after ICH. Nicardipine hydrochloride (NCD) was entrapped in chitosan nanoparticles (CS NPs) and dispersed in an HAMC gel. Subsequently, one side of the gel was coated with a positively charged film. The oriented migration of CS NPs in the HAMC gel was determined, and the drug bioavailability was also enhanced. Furthermore, a blood-induced ICH rat model was established to evaluate the therapeutic effect of CS NPs + HAMC composites. Intranasal administration of the CS NPs + HAMC (+) composite showed a stronger neuroprotective effect in terms of brain edema reduction and neural apoptosis inhibition compared to the CS NPs + HAMC composite. These results suggested that the oriented and rapid drug transport from nose to brain can be achieved using the charge-driven strategy, and this intranasal drug delivery system has the potential to provide a new therapeutic strategy for the treatment of ICH.
Collapse
Affiliation(s)
- Tingwang Guo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China; College of Environment and Resources, Chongqing Technology and Business University, Chongqing 400067, China
| | - Yuanyuan Guo
- College of Chemistry and Chemical Engineering, Chongqing University, Chongqing 400030, China
| | - Yuhua Gong
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Jingou Ji
- College of Chemistry and Chemical Engineering, Chongqing University, Chongqing 400030, China
| | - Shilei Hao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China.
| | - Jia Deng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China; College of Environment and Resources, Chongqing Technology and Business University, Chongqing 400067, China.
| | - Bochu Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China.
| |
Collapse
|
28
|
Tang JD, Mura C, Lampe KJ. Stimuli-Responsive, Pentapeptide, Nanofiber Hydrogel for Tissue Engineering. J Am Chem Soc 2019; 141:4886-4899. [PMID: 30830776 DOI: 10.1021/jacs.8b13363] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Short peptides are uniquely versatile building blocks for self-assembly. Supramolecular peptide assemblies can be used to construct functional hydrogel biomaterials-an attractive approach for neural tissue engineering. Here, we report a new class of short, five-residue peptides that form hydrogels with nanofiber structures. Using rheology and spectroscopy, we describe how sequence variations, pH, and peptide concentration alter the mechanical properties of our pentapeptide hydrogels. We find that this class of seven unmodified peptides forms robust hydrogels from 0.2-20 kPa at low weight percent (less than 3 wt %) in cell culture media and undergoes shear-thinning and rapid self-healing. The peptides self-assemble into long fibrils with sequence-dependent fibrillar morphologies. These fibrils exhibit a unique twisted ribbon shape, as visualized by transmission electron microscopy (TEM) and Cryo-EM imaging, with diameters in the low tens of nanometers and periodicities similar to amyloid fibrils. Experimental gelation behavior corroborates our molecular dynamics simulations, which demonstrate peptide assembly behavior, an increase in β-sheet content, and patterns of variation in solvent accessibility. Our rapidly assembling pentapeptides for injectable delivery (RAPID) hydrogels are syringe-injectable and support cytocompatible encapsulation of oligodendrocyte progenitor cells (OPCs), as well as their proliferation and three-dimensional process extension. Furthermore, RAPID gels protect OPCs from mechanical membrane disruption and acute loss of viability when ejected from a syringe needle, highlighting the protective capability of the hydrogel as potential cell carriers for transplantation therapies. The tunable mechanical and structural properties of these supramolecular assemblies are shown to be permissive to cell expansion and remodeling, making this hydrogel system suitable as an injectable material for cell delivery and tissue engineering applications.
Collapse
|
29
|
Bruggeman KF, Moriarty N, Dowd E, Nisbet DR, Parish CL. Harnessing stem cells and biomaterials to promote neural repair. Br J Pharmacol 2019; 176:355-368. [PMID: 30444942 PMCID: PMC6329623 DOI: 10.1111/bph.14545] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 10/16/2018] [Accepted: 10/22/2018] [Indexed: 01/06/2023] Open
Abstract
With the limited capacity for self-repair in the adult CNS, efforts to stimulate quiescent stem cell populations within discrete brain regions, as well as harness the potential of stem cell transplants, offer significant hope for neural repair. These new cells are capable of providing trophic cues to support residual host populations and/or replace those cells lost to the primary insult. However, issues with low-level adult neurogenesis, cell survival, directed differentiation and inadequate reinnervation of host tissue have impeded the full potential of these therapeutic approaches and their clinical advancement. Biomaterials offer novel approaches to stimulate endogenous neurogenesis, as well as for the delivery and support of neural progenitor transplants, providing a tissue-appropriate physical and trophic milieu for the newly integrating cells. In this review, we will discuss the various approaches by which bioengineered scaffolds may improve stem cell-based therapies for repair of the CNS.
Collapse
Affiliation(s)
- K F Bruggeman
- Laboratory of Advanced Biomaterials, Research School of EngineeringThe Australian National UniversityCanberraACTAustralia
| | - N Moriarty
- Pharmacology and Therapeutics and Galway Neuroscience CentreNational University of Ireland GalwayGalwayIreland
| | - E Dowd
- Pharmacology and Therapeutics and Galway Neuroscience CentreNational University of Ireland GalwayGalwayIreland
| | - D R Nisbet
- Laboratory of Advanced Biomaterials, Research School of EngineeringThe Australian National UniversityCanberraACTAustralia
| | - C L Parish
- The Florey Institute of Neuroscience and Mental HealthThe University of MelbourneParkvilleVICAustralia
| |
Collapse
|
30
|
Liao LY, Lau BWM, Sánchez-Vidaña DI, Gao Q. Exogenous neural stem cell transplantation for cerebral ischemia. Neural Regen Res 2019; 14:1129-1137. [PMID: 30804235 PMCID: PMC6425845 DOI: 10.4103/1673-5374.251188] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Cerebral ischemic injury is the main manifestation of stroke, and its incidence in stroke patients is 70–80%. Although ischemic stroke can be treated with tissue-type plasminogen activator, its time window of effectiveness is narrow. Therefore, the incidence of paralysis, hypoesthesia, aphasia, dysphagia, and cognitive impairment caused by cerebral ischemia is high. Nerve tissue regeneration can promote the recovery of the aforementioned dysfunction. Neural stem cells can participate in the reconstruction of the damaged nervous system and promote the recovery of nervous function during self-repair of damaged brain tissue. Neural stem cell transplantation for ischemic stroke has been a hot topic for more than 10 years. This review discusses the treatment of ischemic stroke with neural stem cells, as well as the mechanisms of their involvement in stroke treatment.
Collapse
Affiliation(s)
- Ling-Yi Liao
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Benson Wui-Man Lau
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Dalinda Isabel Sánchez-Vidaña
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Qiang Gao
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan Province; Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| |
Collapse
|
31
|
Ravina K, Briggs DI, Kislal S, Warraich Z, Nguyen T, Lam RK, Zarembinski TI, Shamloo M. Intracerebral Delivery of Brain-Derived Neurotrophic Factor Using HyStem ®-C Hydrogel Implants Improves Functional Recovery and Reduces Neuroinflammation in a Rat Model of Ischemic Stroke. Int J Mol Sci 2018; 19:ijms19123782. [PMID: 30486515 PMCID: PMC6321015 DOI: 10.3390/ijms19123782] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 11/19/2018] [Indexed: 01/01/2023] Open
Abstract
Ischemic stroke is a leading cause of death and disability worldwide. Potential therapeutics aimed at neural repair and functional recovery are limited in their blood-brain barrier permeability and may exert systemic or off-target effects. We examined the effects of brain-derived neurotrophic factor (BDNF), delivered via an extended release HyStem®-C hydrogel implant or vehicle, on sensorimotor function, infarct volume, and neuroinflammation, following permanent distal middle cerebral artery occlusion (dMCAo) in rats. Eight days following dMCAo or sham surgery, treatments were implanted directly into the infarction site. Rats received either vehicle, BDNF-only (0.167 µg/µL), hydrogel-only, hydrogel impregnated with 0.057 µg/µL of BDNF (hydrogel + BDNFLOW), or hydrogel impregnated with 0.167 µg/µL of BDNF (hydrogel + BDNFHIGH). The adhesive removal test (ART) and 28-point Neuroscore (28-PN) were used to evaluate sensorimotor function up to two months post-ischemia. The hydrogel + BDNFHIGH group showed significant improvements on the ART six to eight weeks following treatment and their behavioral performance was consistently greater on the 28-PN. Infarct volume was reduced in rats treated with hydrogel + BDNFHIGH as were levels of microglial, phagocyte, and astrocyte marker immunoexpression in the corpus striatum. These data suggest that targeted intracerebral delivery of BDNF using hydrogels may mitigate ischemic brain injury and restore functional deficits by reducing neuroinflammation.
Collapse
Affiliation(s)
- Kristine Ravina
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, CA 94304-1334, USA.
| | - Denise I Briggs
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, CA 94304-1334, USA.
| | - Sezen Kislal
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, CA 94304-1334, USA.
| | - Zuha Warraich
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, CA 94304-1334, USA.
| | - Tiffany Nguyen
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, CA 94304-1334, USA.
| | - Rachel K Lam
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, CA 94304-1334, USA.
| | | | - Mehrdad Shamloo
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, CA 94304-1334, USA.
| |
Collapse
|
32
|
Kornev VA, Grebenik EA, Solovieva AB, Dmitriev RI, Timashev PS. Hydrogel-assisted neuroregeneration approaches towards brain injury therapy: A state-of-the-art review. Comput Struct Biotechnol J 2018; 16:488-502. [PMID: 30455858 PMCID: PMC6232648 DOI: 10.1016/j.csbj.2018.10.011] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/18/2018] [Accepted: 10/19/2018] [Indexed: 12/16/2022] Open
Abstract
Recent years have witnessed the development of an enormous variety of hydrogel-based systems for neuroregeneration. Formed from hydrophilic polymers and comprised of up to 90% of water, these three-dimensional networks are promising tools for brain tissue regeneration. They can assist structural and functional restoration of damaged tissues by providing mechanical support and navigating cell fate. Hydrogels also show the potential for brain injury therapy due to their broadly tunable physical, chemical, and biological properties. Hydrogel polymers, which have been extensively implemented in recent brain injury repair studies, include hyaluronic acid, collagen type I, alginate, chitosan, methylcellulose, Matrigel, fibrin, gellan gum, self-assembling peptides and proteins, poly(ethylene glycol), methacrylates, and methacrylamides. When viewed as tools for neuroregeneration, hydrogels can be divided into: (1) hydrogels suitable for brain injury therapy, (2) hydrogels that do not meet basic therapeutic requirements and (3) promising hydrogels which meet the criteria for further investigations. Our analysis shows that fibrin, collagen I and self-assembling peptide-based hydrogels display very attractive properties for neuroregeneration.
Collapse
Affiliation(s)
- Vladimir A. Kornev
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya st., Moscow 119991, Russian Federation
| | - Ekaterina A. Grebenik
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya st., Moscow 119991, Russian Federation
| | - Anna B. Solovieva
- N. N. Semenov Institute of Chemical Physics, Russian Academy of Sciences, 4 Kosygina st., Moscow 117977, Russian Federation
| | - Ruslan I. Dmitriev
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya st., Moscow 119991, Russian Federation
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Peter S. Timashev
- Institute for Regenerative Medicine, Sechenov University, 8-2 Trubetskaya st., Moscow 119991, Russian Federation
- N. N. Semenov Institute of Chemical Physics, Russian Academy of Sciences, 4 Kosygina st., Moscow 117977, Russian Federation
- Institute of Photonic Technologies, Research Center “Crystallography and Photonics” Russian Academy of Sciences, 2 Pionerskaya st., Troitsk, Moscow 108840, Russian Federation
| |
Collapse
|
33
|
Hydrogel Scaffolds: Towards Restitution of Ischemic Stroke-Injured Brain. Transl Stroke Res 2018; 10:1-18. [DOI: 10.1007/s12975-018-0655-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 05/17/2018] [Accepted: 08/19/2018] [Indexed: 12/27/2022]
|
34
|
Hassanzadeh P, Atyabi F, Dinarvand R. Tissue engineering: Still facing a long way ahead. J Control Release 2018; 279:181-197. [DOI: 10.1016/j.jconrel.2018.04.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/09/2018] [Accepted: 04/11/2018] [Indexed: 02/07/2023]
|
35
|
Modo MM, Jolkkonen J, Zille M, Boltze J. Future of Animal Modeling for Poststroke Tissue Repair. Stroke 2018; 49:1099-1106. [PMID: 29669872 PMCID: PMC6013070 DOI: 10.1161/strokeaha.117.018293] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/13/2017] [Accepted: 12/14/2017] [Indexed: 02/06/2023]
Affiliation(s)
- Michel M Modo
- From the Departments of Radiology and Bioengineering, McGowan Institute for Regenerative Medicine, University of Pittsburgh, PA (M.M.M.)
| | - Jukka Jolkkonen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio (J.J.)
- Neurocenter, Kuopio University Hospital, Finland (J.J.)
| | - Marietta Zille
- Department of Translational Medicine and Cell Technology, Fraunhofer Research Institution for Marine Biotechnology and Institute for Medical and Marine Biotechnology, University of Lübeck, Mönkhofer Weg, Germany (M.Z., J.B.)
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Ratzeburger Allee, Germany (M.Z.)
| | - Johannes Boltze
- Department of Translational Medicine and Cell Technology, Fraunhofer Research Institution for Marine Biotechnology and Institute for Medical and Marine Biotechnology, University of Lübeck, Mönkhofer Weg, Germany (M.Z., J.B.)
| |
Collapse
|
36
|
Hickey K, Stabenfeldt SE. Using biomaterials to modulate chemotactic signaling for central nervous system repair. Biomed Mater 2018; 13:044106. [PMID: 29411713 PMCID: PMC5991092 DOI: 10.1088/1748-605x/aaad82] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Chemotaxis enables cellular communication and movement within the body. This review focuses on exploiting chemotaxis as a tool for repair of the central nervous system (CNS) damaged from injury and/or degenerative diseases. Chemokines and factors alone may initiate repair following CNS injury/disease, but exogenous administration may enhance repair and promote regeneration. This review will discuss critical chemotactic molecules and factors that may promote neural regeneration. Additionally, this review highlights how biomaterials can impact the presentation and delivery of chemokines and growth factors to alter the regenerative response.
Collapse
Affiliation(s)
- Kassondra Hickey
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States of America
| | | |
Collapse
|
37
|
Xu Z, Wang W, Ren Y, Zhang W, Fang P, Huang L, Wang X, Shi P. Regeneration of cortical tissue from brain injury by implantation of defined molecular gradient of semaphorin 3A. Biomaterials 2018; 157:125-135. [DOI: 10.1016/j.biomaterials.2017.12.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 12/04/2017] [Accepted: 12/11/2017] [Indexed: 10/18/2022]
|
38
|
Characterisation of hyaluronic acid methylcellulose hydrogels for 3D bioprinting. J Mech Behav Biomed Mater 2017; 77:389-399. [PMID: 29017117 DOI: 10.1016/j.jmbbm.2017.09.031] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 09/23/2017] [Accepted: 09/25/2017] [Indexed: 01/05/2023]
Abstract
Hydrogels containing hyaluronic acid (HA) and methylcellulose (MC) have shown promising results for three dimensional (3D) bioprinting applications. However, several parameters influence the applicability bioprinting and there is scarce data in the literature characterising HAMC. We assessed eight concentrations of HAMC for printability, swelling and stability over time, rheological and structural behaviour, and viability of mesenchymal stem cells. We show that HAMC blends behave as viscous solutions at 4°C and have faster gelation times at higher temperatures, typically gelling upon reaching 37°C. We found the storage, loss and compressive moduli to be dependent on HAMC concentration and incubation time at 37°C, and show the compressive modulus to be strain-rate dependent. Swelling and stability was influenced by time, more so than pH environment. We demonstrated that mesenchymal stem cell viability was above 75% in bioprinted structures and cells remain viable for at least one week after 3D bioprinting. The mechanical properties of HAMC are highly tuneable and we show that higher concentrations of HAMC are particularly suited to cell-encapsulated 3D bioprinting applications that require scaffold structure and delivery of cells.
Collapse
|
39
|
Dutta D, Hickey K, Salifu M, Fauer C, Willingham C, Stabenfeldt SE. Spatiotemporal presentation of exogenous SDF-1 with PLGA nanoparticles modulates SDF-1/CXCR4 signaling axis in the rodent cortex. Biomater Sci 2017; 5:1640-1651. [PMID: 28703822 PMCID: PMC5588897 DOI: 10.1039/c7bm00489c] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Stromal cell-derived factor-1 (SDF-1) and its key receptor CXCR4 have been implicated in directing cellular recruitment for several pathological/disease conditions thus also gained considerable attention for regenerative medicine. One regenerative approach includes sustained release of SDF-1 to stimulate prolonged stem cell recruitment. However, the impact of SDF-1 sustained release on the endogenous SDF-1/CXCR4 signaling axis is largely unknown as auto-regulatory mechanisms typically dictate cytokine/receptor signaling. We hypothesize that spatiotemporal presentation of exogenous SDF-1 is a key factor in achieving long-term manipulation of endogenous SDF-1/CXCR4 signaling. Here in the present study, we sought to probe our hypothesis using a transgenic mouse model to contrast the spatial activation of endogenous SDF-1 and CXCR4 in response to exogenous SDF-1 injected in bolus or controlled release (PLGA nanoparticles) form in the adult rodent cortex. Our data suggests that the manner of SDF-1 presentation significantly affected initial CXCR4 cellular activation/recruitment despite having similar protein payloads over the first 24 h (∼30 ng for both bolus and sustained release groups). Yet, one week post-injection, this response was negligible. Therefore, the transient nature CXCR4 recruitment/activation in response to bolus or controlled release SDF-1 indicated that cytokine/receptor auto-regulatory mechanisms may demand more complex release profiles (i.e. delayed and/or pulsed release) to achieve sustained cellular response.
Collapse
Affiliation(s)
- D Dutta
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA.
| | - K Hickey
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA.
| | - M Salifu
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA.
| | - C Fauer
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA.
| | - C Willingham
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA.
| | - S E Stabenfeldt
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
40
|
Cook DJ, Nguyen C, Chun HN, L Llorente I, Chiu AS, Machnicki M, Zarembinski TI, Carmichael ST. Hydrogel-delivered brain-derived neurotrophic factor promotes tissue repair and recovery after stroke. J Cereb Blood Flow Metab 2017; 37:1030-1045. [PMID: 27174996 PMCID: PMC5363479 DOI: 10.1177/0271678x16649964] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/20/2015] [Accepted: 03/20/2016] [Indexed: 11/15/2022]
Abstract
Stroke is the leading cause of adult disability. Systemic delivery of candidate neural repair therapies is limited by the blood-brain barrier and off-target effects. We tested a bioengineering approach for local depot release of BDNF from the infarct cavity for neural repair in chronic periods after stroke. The brain release levels of a hyaluronic acid hydrogel + BDNF were tested in several stroke models in mouse (strains C57Bl/6, DBA) and non-human primate ( Macaca fascicularis) and tracked with MRI. The behavioral recovery effects of hydrogel + BDNF and the effects on tissue repair outcomes were determined. Hydrogel-delivered BDNF diffuses from the stroke cavity into peri-infarct tissue over 3 weeks in two mouse stroke models, compared with 1 week for direct BDNF injection. Hydrogel delivery of BDNF promotes recovery of motor function. Mapping of motor system connections indicates that hydrogel-BDNF induces axonal sprouting within existing cortical and cortico-striatal systems. Pharmacogenetic studies show that hydrogel-BDNF induces the initial migration of immature neurons into the peri-infarct cortex and their long-term survival. In chronic stroke in the non-human primate, hydrogel-released BDNF can be detected up to 2 cm from the infarct, a distance relevant to human functional recovery in stroke. The hydrogel can be tracked by MRI in mouse and primate.
Collapse
Affiliation(s)
- Douglas J Cook
- Department of Surgery, Division of Neurosurgery, Kingston General Hospital, Kingston, Canada
| | - Cynthia Nguyen
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Hyun N Chun
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Irene L Llorente
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Abraham S Chiu
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Michal Machnicki
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | | | - S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, USA
| |
Collapse
|
41
|
Chan HH, Wathen CA, Ni M, Zhuo S. Stem cell therapies for ischemic stroke: current animal models, clinical trials and biomaterials. RSC Adv 2017. [DOI: 10.1039/c7ra00336f] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
We report the facilitation of stem cell therapy in stroke by tissue engineering and applications of biomaterials.
Collapse
Affiliation(s)
- Hugh H. Chan
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education & Fujian Provincial Key Laboratory of Photonics Technology
- Fujian Normal University
- Fuzhou 350007
- P. R. China
- Department of Neuroscience
| | | | - Ming Ni
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education & Fujian Provincial Key Laboratory of Photonics Technology
- Fujian Normal University
- Fuzhou 350007
- P. R. China
| | - Shuangmu Zhuo
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education & Fujian Provincial Key Laboratory of Photonics Technology
- Fujian Normal University
- Fuzhou 350007
- P. R. China
| |
Collapse
|
42
|
Duan H, Li X, Wang C, Hao P, Song W, Li M, Zhao W, Gao Y, Yang Z. Functional hyaluronate collagen scaffolds induce NSCs differentiation into functional neurons in repairing the traumatic brain injury. Acta Biomater 2016; 45:182-195. [PMID: 27562609 DOI: 10.1016/j.actbio.2016.08.043] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 08/14/2016] [Accepted: 08/20/2016] [Indexed: 12/14/2022]
Abstract
The traumatic brain injury (TBI) usually causes brain tissue defects, including neuronal death or loss, which ultimately results in dysfunction in some degree. The cell replacement therapy is now one of the most promising methods for such injury. There are currently various methods to induce the differentiation of stem cells into neurons, but all extremely complex, slow and unstable. Here we report that the sodium hyaluronate collagen scaffold loaded with bFGF (bFGF-controlled releasing system, bFGF-CRS) can induce neural stem cells (NSCs) to differentiate into multi-type and mature functional neurons at a high percentage of 82±1.528% in two weeks. The quantitative real-time (QRT) PCR results reveal that a long-term activation of bFGF receptors could up-regulate ERK/MAPK signal pathways, thus facilitating the formation of presynaptic and postsynaptic structure among the induced neuronal cells (iN cells). The functional synaptic connections established among iN cells were detected by the planar multielectrode dish system. When jointly transplanting the bFGF-CRS and NSCs into the CA1 zone of the rat TBI area, the results suggested that bFGF-CRS provided an optimal microenvironment, which promoted survival, neuronal differentiation of transplanted NSCs and functional synapse formation not only among iN cells but also between iN cells and the host brain tissue in TBI rats, consequently leading to the cognitive function recovery of TBI rats. These findings in vitro and in vivo may lay a foundation for the application of bFGF-CRS and shed light on the delivery of exogenous cells or nutrients to the CNS injury or disease area. STATEMENT OF SIGNIFICANCE A sodium hyaluronate collagen scaffold was specifically functionalized with nutrient-bFGF which can induce the differentiation of neural stem cells (NSCs) into multi-type and mature functional neurons at a high percentage in two week. When jointly transplanting the bFGF-CRS and NSCs into the CA1 zone of the traumatic brain injured area of adult rats, the bFGF-CRS could provide an optimal microenvironment, which promoted survival, migration and neuronal differentiation of transplanted NSCs and functional synapse formation among iN cells, as well as between iN cells and host brain tissue in TBI rats, consequently leading to the cognitive function recovery of TBI rats.
Collapse
Affiliation(s)
- Hongmei Duan
- Department of Neurobiology, School of Basic Medical Sciences, Captial Medical University, Beijing 100069, China
| | - Xiaoguang Li
- Department of Neurobiology, School of Basic Medical Sciences, Captial Medical University, Beijing 100069, China
| | - Cong Wang
- Department of Neurobiology, School of Basic Medical Sciences, Captial Medical University, Beijing 100069, China
| | - Peng Hao
- Department of Neurobiology, School of Basic Medical Sciences, Captial Medical University, Beijing 100069, China
| | - Wei Song
- School of Rehabilitation Medicine, Captial Medical University, Beijing 100068, China; China Rehabilitation Research Center, Beijing 100068, China
| | - Manli Li
- Department of Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Wen Zhao
- Department of Neurobiology, School of Basic Medical Sciences, Captial Medical University, Beijing 100069, China
| | - Yudan Gao
- Department of Neurobiology, School of Basic Medical Sciences, Captial Medical University, Beijing 100069, China
| | - Zhaoyang Yang
- Department of Neurobiology, School of Basic Medical Sciences, Captial Medical University, Beijing 100069, China.
| |
Collapse
|
43
|
|
44
|
Russo T, Tunesi M, Giordano C, Gloria A, Ambrosio L. Hydrogels for central nervous system therapeutic strategies. Proc Inst Mech Eng H 2016; 229:905-16. [PMID: 26614804 DOI: 10.1177/0954411915611700] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The central nervous system shows a limited regenerative capacity, and injuries or diseases, such as those in the spinal, brain and retina, are a great problem since current therapies seem to be unable to achieve good results in terms of significant functional recovery. Different promising therapies have been suggested, the aim being to restore at least some of the lost functions. The current review deals with the use of hydrogels in developing advanced devices for central nervous system therapeutic strategies. Several approaches, involving cell-based therapy, delivery of bioactive molecules and nanoparticle-based drug delivery, will be first reviewed. Finally, some examples of injectable hydrogels for the delivery of bioactive molecules in central nervous system will be reported, and the key features as well as the basic principles in designing multifunctional devices will be described.
Collapse
Affiliation(s)
- Teresa Russo
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, Naples, Italy
| | - Marta Tunesi
- Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano and Unità di Ricerca Consorzio INSTM, Politecnico di Milano, Milan, Italy
| | - Carmen Giordano
- Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano and Unità di Ricerca Consorzio INSTM, Politecnico di Milano, Milan, Italy
| | - Antonio Gloria
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, Naples, Italy
| | - Luigi Ambrosio
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, Naples, Italy
| |
Collapse
|
45
|
Lim TC, Spector M. Biomaterials for Enhancing CNS Repair. Transl Stroke Res 2016; 8:57-64. [PMID: 27251413 DOI: 10.1007/s12975-016-0470-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 05/07/2016] [Accepted: 05/10/2016] [Indexed: 01/11/2023]
Abstract
The health of the central nervous system (CNS) does not only rely on the state of the neural cells but also on how various extracellular components organize cellular behaviors into proper tissue functions. Biomaterials have been valuable in restoring or augmenting the roles of extracellular components in the CNS in the event of injury and disease. In this review, we highlight how biomaterials have been enabling tools in important therapeutic strategies involving cell transplantation and drug/protein delivery. We further discuss advances in biomaterial design and applications that can potentially be translated into the CNS to provide unprecedented benefits.
Collapse
Affiliation(s)
- Teck Chuan Lim
- Institute of Bioengineering and Nanotechnology, Singapore, Singapore.
| | - Myron Spector
- Tissue Engineering, VA Boston Healthcare System, Boston, MA, USA.,Department of Orthopedics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
46
|
Biomaterial Applications in Cell-Based Therapy in Experimental Stroke. Stem Cells Int 2016; 2016:6810562. [PMID: 27274738 PMCID: PMC4870368 DOI: 10.1155/2016/6810562] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/11/2016] [Accepted: 04/04/2016] [Indexed: 01/08/2023] Open
Abstract
Stroke is an important health issue corresponding to the second cause of mortality and first cause of severe disability with no effective treatments after the first hours of onset. Regenerative approaches such as cell therapy provide an increase in endogenous brain structural plasticity but they are not enough to promote a complete recovery. Tissue engineering has recently aroused a major interesting development of biomaterials for use into the central nervous system. Many biomaterials have been engineered based on natural compounds, synthetic compounds, or a mix of both with the aim of providing polymers with specific properties. The mechanical properties of biomaterials can be exquisitely regulated forming polymers with different stiffness, modifiable physical state that polymerizes in situ, or small particles encapsulating cells or growth factors. The choice of biomaterial compounds should be adapted for the different applications, structure target, and delay of administration. Biocompatibilities with embedded cells and with the host tissue and biodegradation rate must be considerate. In this paper, we review the different applications of biomaterials combined with cell therapy in ischemic stroke and we explore specific features such as choice of biomaterial compounds and physical and mechanical properties concerning the recent studies in experimental stroke.
Collapse
|
47
|
Gao Y, Yang Z, Li X. Regeneration strategies after the adult mammalian central nervous system injury-biomaterials. Regen Biomater 2016; 3:115-22. [PMID: 27047678 PMCID: PMC4817328 DOI: 10.1093/rb/rbw004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 01/04/2016] [Indexed: 01/12/2023] Open
Abstract
The central nervous system (CNS) has very restricted intrinsic regeneration ability under the injury or disease condition. Innovative repair strategies, therefore, are urgently needed to facilitate tissue regeneration and functional recovery. The published tissue repair/regeneration strategies, such as cell and/or drug delivery, has been demonstrated to have some therapeutic effects on experimental animal models, but can hardly find clinical applications due to such methods as the extremely low survival rate of transplanted cells, difficulty in integrating with the host or restriction of blood–brain barriers to administration patterns. Using biomaterials can not only increase the survival rate of grafts and their integration with the host in the injured CNS area, but also sustainably deliver bioproducts to the local injured area, thus improving the microenvironment in that area. This review mainly introduces the advances of various strategies concerning facilitating CNS regeneration.
Collapse
Affiliation(s)
- Yudan Gao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Zhaoyang Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China,; Department of Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Xiaoguang Li
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China,; Department of Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| |
Collapse
|
48
|
Abstract
INTRODUCTION Ischemic stroke is caused by reduced blood supply and leads to loss of brain function. The reduced oxygen and nutrient supply stimulates various physiological responses, including induction of growth factors. Growth factors prevent neuronal cell death, promote neovascularization, and induce cell growth. However, the concentration of growth factors is not sufficient to recover brain function after the ischemic damage, suggesting that delivery of growth factors into the ischemic brain may be a useful treatment for ischemic stroke. AREAS COVERED In this review, various approaches for the delivery of growth factors to ischemic brain tissue are discussed, including local and targeting delivery systems. EXPERT OPINION To develop growth factor therapy for ischemic stroke, important considerations should be taken into account. First, growth factors may have possible side effects. Thus, concentration of growth factors should be restricted to the ischemic tissues by local administration or targeted delivery. Second, the duration of growth factor therapy should be optimized. Growth factor proteins may be degraded too fast to have a high enough therapeutic effect. Therefore, delivery systems for controlled release or gene delivery may be useful. Third, the delivery systems to the brain should be optimized according to the delivery route.
Collapse
Affiliation(s)
- Taiyoun Rhim
- a Department of Bioengineering, College of Engineering , Hanyang University , Seoul , Republic of Korea
| | - Minhyung Lee
- a Department of Bioengineering, College of Engineering , Hanyang University , Seoul , Republic of Korea
| |
Collapse
|
49
|
Maclean FL, Rodriguez AL, Parish CL, Williams RJ, Nisbet DR. Integrating Biomaterials and Stem Cells for Neural Regeneration. Stem Cells Dev 2016; 25:214-26. [DOI: 10.1089/scd.2015.0314] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Francesca L. Maclean
- Research School of Engineering, the Australian National University, Canberra, Australia
| | | | - Clare L. Parish
- Florey Institute of Neuroscience and Mental Health, the University of Melbourne, Parkville, Australia
| | - Richard J. Williams
- School of Aerospace, Mechanical and Manufacturing Engineering and Health Innovations Research Institute, RMIT University, Melbourne, Australia
| | - David R. Nisbet
- Research School of Engineering, the Australian National University, Canberra, Australia
| |
Collapse
|
50
|
Haddad T, Noel S, Liberelle B, El Ayoubi R, Ajji A, De Crescenzo G. Fabrication and surface modification of poly lactic acid (PLA) scaffolds with epidermal growth factor for neural tissue engineering. BIOMATTER 2016; 6:e1231276. [PMID: 27740881 PMCID: PMC5098722 DOI: 10.1080/21592535.2016.1231276] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 03/22/2016] [Accepted: 08/26/2016] [Indexed: 12/11/2022]
Abstract
In an effort to design biomaterials that may promote repair of the central nervous system, 3-dimensional scaffolds made of electrospun poly lactic acid nanofibers with interconnected pores were fabricated. These scaffolds were functionalized with polyallylamine to introduce amine groups by wet chemistry. Experimental conditions of the amination protocol were thoroughly studied and selected to introduce a high amount of amine group while preserving the mechanical and structural properties of the scaffold. Subsequent covalent grafting of epidermal growth factor was then performed to further tailor these aminated structures. The scaffolds were then tested for their ability to support Neural Stem-Like Cells (NSLCs) culture. Of interest, NSLCs were able to proliferate on these EGF-grafted substrates and remained viable up to 14 d even in the absence of soluble growth factors in the medium.
Collapse
Affiliation(s)
- Tanit Haddad
- Department of Chemical Engineering, École Polytechnique de Montréal, Centre-Ville, Montréal (QC), Canada
| | - Samantha Noel
- Department of Chemical Engineering, École Polytechnique de Montréal, Centre-Ville, Montréal (QC), Canada
| | - Benoît Liberelle
- Department of Chemical Engineering, École Polytechnique de Montréal, Centre-Ville, Montréal (QC), Canada
| | | | - Abdellah Ajji
- Department of Chemical Engineering, École Polytechnique de Montréal, Centre-Ville, Montréal (QC), Canada
| | - Gregory De Crescenzo
- Department of Chemical Engineering, École Polytechnique de Montréal, Centre-Ville, Montréal (QC), Canada
| |
Collapse
|