1
|
Liu S, Zhang R, Hallajzadeh J. Role of exercise on ncRNAs and exosomal ncRNAs in preventing neurodegenerative diseases: a narrative review. Mol Med 2025; 31:51. [PMID: 39920595 PMCID: PMC11803956 DOI: 10.1186/s10020-025-01091-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 01/17/2025] [Indexed: 02/09/2025] Open
Abstract
Engaging in activity has proven to have beneficial effects on different facets of well-being, such as conditions related to the deterioration of the nervous system. Non-coding RNAs (ncRNAs) and exosomal ncRNAs associated with vesicles have been recognized as influencers of gene expression and cell signaling, potentially contributing to the positive impact of physical activity on neurodegenerative conditions. It is hypothesized that exercise-induced changes in ncRNA expression may regulate key processes involved in neuroprotection, including neuroinflammation, oxidative stress, protein aggregation, and synaptic function. Exercise has shown promise in preventing neurodegenerative diseases (NDs), and ncRNAs and exosomal ncRNAs are emerging as potential mediators of these benefits. In review, we explored how ncRNAs and exosomal ncRNAs play a role in enhancing the impacts of activity on neurodegenerative disorders for future treatments. Research studies, both preclinical and clinical, that have documented the use of various exercises and their effects on ncRNAs and exosomal ncRNAs for the treatment of NDs have been compiled and enlisted from the PubMed database, spanning the time period from the year 2000 up to the current time. Studies show that manipulating specific ncRNAs or harnessing exercise-induced changes in ncRNA expression and exosomal cargo could potentially be utilized as therapeutic strategies for preventing or treating NDs. In conclusion, studies suggest that various exercise modalities, including aerobic, resistance, and high-intensity interval training, can modulate the expression of ncRNAs and exosomal ncRNAs in the context of NDs. The altered ncRNA profiles may contribute to the neuroprotective and therapeutic effects observed with exercise interventions. However, more research is needed to fully understand the underlying mechanisms and to further explore the potential of exercise-induced ncRNA signatures as biomarkers and therapeutic targets for neurodegenerative disorders.
Collapse
Affiliation(s)
- Shangwu Liu
- Department of Physical Education, Lyuliang University, Lishi, 033000, Shanxi, China
| | - Runhong Zhang
- Department of Physical Education, Lyuliang University, Lishi, 033000, Shanxi, China.
| | - Jamal Hallajzadeh
- Research Center for Evidence-Based Health Management, Maragheh University of Medical Sciences, Maragheh, Iran.
| |
Collapse
|
2
|
Bradshaw KJ, Leipzig ND. Applications of Regenerative Tissue-Engineered Scaffolds for Treatment of Spinal Cord Injury. Tissue Eng Part A 2025; 31:108-125. [PMID: 39556330 DOI: 10.1089/ten.tea.2024.0194] [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] [Indexed: 11/19/2024] Open
Abstract
Tissue engineering provides a path forward for emerging personalized medicine therapies as well as the ability to bring about cures for diseases or chronic injuries. Traumatic spinal cord injuries (SCIs) are an example of a chronic injury in which no cure or complete functional recovery treatment has been developed. In part, this has been due to the complex and interconnected nature of the central nervous system (CNS), the cellular makeup, its extracellular matrix (ECM), and the injury site pathophysiology. One way to combat the complex nature of an SCI has been to create functional tissue-engineered scaffolds that replace or replenish the aspects of the CNS and tissue/ECM that are damaged following the immediate injury and subsequent immune response. This can be achieved by employing the tissue-engineering triad consisting of cells, biomaterial(s), and environmental factors. Stem cells, with their innate ability to proliferate and differentiate, are a common choice for cellular therapies. Natural or synthetic biomaterials that have tunable characteristics are normally used as the scaffold base. Environmental factors can range from drugs to growth factors (GFs) or proteins, depending on if the idea would be to stimulate exogeneous or endogenous cell populations or just simply retain cells on the scaffold for effective transplantation. For functional regeneration and integration for SCI, the scaffold must promote neuroprotection and neuroplasticity. Tissue-engineering strategies have shown benefits including neuronal differentiation, axonal regeneration, axonal outgrowth, integration into the native spinal cord, and partial functional recovery. Overall, this review focuses on the background that causes SCI to be so difficult to treat, the individual components of the tissue-engineering triad, and how combinatorial scaffolds can be beneficial toward the prospects of future SCI recovery.
Collapse
Affiliation(s)
- Katherine J Bradshaw
- Department of Biomedical Engineering, Auburn Science and Engineering Center #275, The University of Akron, Akron, Ohio, USA
| | - Nic D Leipzig
- Department of Biomedical Engineering, Auburn Science and Engineering Center #275, The University of Akron, Akron, Ohio, USA
- Department of Chemical, Biomolecular, and Corrosion Engineering, The University of Akron, Akron, Ohio, USA
| |
Collapse
|
3
|
Ross BC, Kent RN, Saunders MN, Schwartz SR, Smiley BM, Hocevar SE, Chen SC, Xiao C, Williams LA, Anderson AJ, Cummings BJ, Baker BM, Shea LD. Building-Block Size Mediates Microporous Annealed Particle Hydrogel Tube Microenvironment Following Spinal Cord Injury. Adv Healthc Mater 2024; 13:e2302498. [PMID: 37768019 PMCID: PMC10972780 DOI: 10.1002/adhm.202302498] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/10/2023] [Indexed: 09/29/2023]
Abstract
Spinal cord injury (SCI) is a life-altering event, which often results in loss of sensory and motor function below the level of trauma. Biomaterial therapies have been widely investigated in SCI to promote directional regeneration but are often limited by their pre-constructed size and shape. Herein, the design parameters of microporous annealed particles (MAPs) are investigated with tubular geometries that conform to the injury and direct axons across the defect to support functional recovery. MAP tubes prepared from 20-, 40-, and 60-micron polyethylene glycol (PEG) beads are generated and implanted in a T9-10 murine hemisection model of SCI. Tubes attenuate glial and fibrotic scarring, increase innate immune cell density, and reduce inflammatory phenotypes in a bead size-dependent manner. Tubes composed of 60-micron beads increase the cell density of the chronic macrophage response, while neutrophil infiltration and phenotypes do not deviate from those seen in controls. At 8 weeks postinjury, implantation of tubes composed of 60-micron beads results in enhanced locomotor function, robust axonal ingrowth, and remyelination through both lumens and the inter-tube space. Collectively, these studies demonstrate the importance of bead size in MAP construction and highlight PEG tubes as a biomaterial therapy to promote regeneration and functional recovery in SCI.
Collapse
Affiliation(s)
- Brian C Ross
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Blvd, Ann Arbor, MI, 48109, USA
| | - Robert N Kent
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Blvd, Ann Arbor, MI, 48109, USA
| | - Michael N Saunders
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Blvd, Ann Arbor, MI, 48109, USA
| | - Samantha R Schwartz
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Blvd, Ann Arbor, MI, 48109, USA
| | - Brooke M Smiley
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Blvd, Ann Arbor, MI, 48109, USA
| | - Sarah E Hocevar
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Blvd, Ann Arbor, MI, 48109, USA
- Neuroscience Graduate Program, University of Michigan Medical School, 204 Washtenaw Ave, Ann Arbor, MI, 48109, USA
| | - Shao-Chi Chen
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Blvd, Ann Arbor, MI, 48109, USA
| | - Chengchuan Xiao
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, 1105 North University Ave, Ann Arbor, MI, 48109, USA
| | - Laura A Williams
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Blvd, Ann Arbor, MI, 48109, USA
| | - Aileen J Anderson
- Institute for Memory Impairments and Neurological Disorders, University of California, Biological Sciences III, 2642, Irvine, CA, 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, 845 Health Sciences Rd, Irvine, CA, 92697, USA
- Physical Medicine and Rehabilitation, University of California, 18124 Culver Dr # F, Irvine, CA, 92612, USA
| | - Brian J Cummings
- Institute for Memory Impairments and Neurological Disorders, University of California, Biological Sciences III, 2642, Irvine, CA, 92697, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, 845 Health Sciences Rd, Irvine, CA, 92697, USA
- Physical Medicine and Rehabilitation, University of California, 18124 Culver Dr # F, Irvine, CA, 92612, USA
| | - Brendon M Baker
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Blvd, Ann Arbor, MI, 48109, USA
- Department of Chemical Engineering, University of Michigan, 2300 Hayward St, Ann Arbor, MI, 48109, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Blvd, Ann Arbor, MI, 48109, USA
- Neuroscience Graduate Program, University of Michigan Medical School, 204 Washtenaw Ave, Ann Arbor, MI, 48109, USA
- Department of Chemical Engineering, University of Michigan, 2300 Hayward St, Ann Arbor, MI, 48109, USA
| |
Collapse
|
4
|
Kim J, Jia X. Flexible multimaterial fibers in modern biomedical applications. Natl Sci Rev 2024; 11:nwae333. [PMID: 39411353 PMCID: PMC11476783 DOI: 10.1093/nsr/nwae333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/12/2024] [Accepted: 07/26/2024] [Indexed: 10/19/2024] Open
Abstract
Biomedical devices are indispensable in modern healthcare, significantly enhancing patients' quality of life. Recently, there has been a drastic increase in innovations for the fabrication of biomedical devices. Amongst these fabrication methods, the thermal drawing process has emerged as a versatile and scalable process for the development of advanced biomedical devices. By thermally drawing a macroscopic preform, which is meticulously designed and integrated with functional materials, hundreds of meters of multifunctional fibers are produced. These scalable flexible multifunctional fibers are embedded with functionalities such as electrochemical sensing, drug delivery, light delivery, temperature sensing, chemical sensing, pressure sensing, etc. In this review, we summarize the fabrication method of thermally drawn multifunctional fibers and highlight recent developments in thermally drawn fibers for modern biomedical application, including neural interfacing, chemical sensing, tissue engineering, cancer treatment, soft robotics and smart wearables. Finally, we discuss the existing challenges and future directions of this rapidly growing field.
Collapse
Affiliation(s)
- Jongwoon Kim
- The Bradley Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, VA 24060, USA
| | - Xiaoting Jia
- The Bradley Department of Electrical and Computer Engineering, Virginia Tech, Blacksburg, VA 24060, USA
- School of Neuroscience, Virginia Tech, Blacksburg, VA 24060, USA
- Department of Materials Science and Engineering, Virginia Tech, Blacksburg, VA 24060, USA
| |
Collapse
|
5
|
Li Q, Li C, Zhang X. Research Progress on the Effects of Different Exercise Modes on the Secretion of Exerkines After Spinal Cord Injury. Cell Mol Neurobiol 2024; 44:62. [PMID: 39352588 PMCID: PMC11445308 DOI: 10.1007/s10571-024-01497-y] [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: 06/13/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024]
Abstract
Exercise training is a conventional treatment strategy throughout the entire treatment process for patients with spinal cord injury (SCI). Currently, exercise modalities for SCI patients primarily include aerobic exercise, endurance training, strength training, high-intensity interval training, and mind-body exercises. These exercises play a positive role in enhancing skeletal muscle function, inducing neuroprotection and regeneration, thereby influencing neural plasticity, reducing limb spasticity, and improving motor function and daily living abilities in SCI patients. However, the mechanism by which exercise training promotes functional recovery after SCI is still unclear, and there is no consensus on a unified and standardized exercise treatment plan. Different exercise methods may bring different benefits. After SCI, patients' physical activity levels decrease significantly due to factors such as motor dysfunction, which may be a key factor affecting changes in exerkines. The changes in exerkines of SCI patients caused by exercise training are an important and highly relevant and visual evaluation index, which may provide a new research direction for revealing the intrinsic mechanism by which exercise promotes functional recovery after SCI. Therefore, this article summarizes the changes in the expression of common exerkines (neurotrophic factors, inflammatory factors, myokines, bioactive peptides) after SCI, and intends to analyze the impact and role of different exercise methods on functional recovery after SCI from the perspective of exerkines mechanism. We hope to provide theoretical basis and data support for scientific exercise treatment programs after SCI.
Collapse
Affiliation(s)
- Qianxi Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China
| | - Chenyu Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China
| | - Xin Zhang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| |
Collapse
|
6
|
Nekanti U, Sakthivel PS, Zahedi A, Creasman DA, Nishi RA, Dumont CM, Piltti KM, Guardamondo GL, Hernandez N, Chen X, Song H, Lin X, Martinez J, On L, Lakatos A, Pawar K, David BT, Guo Z, Seidlits SK, Xu X, Shea LD, Cummings BJ, Anderson AJ. Multichannel bridges and NSC synergize to enhance axon regeneration, myelination, synaptic reconnection, and recovery after SCI. NPJ Regen Med 2024; 9:12. [PMID: 38499577 PMCID: PMC10948859 DOI: 10.1038/s41536-024-00356-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 02/15/2024] [Indexed: 03/20/2024] Open
Abstract
Regeneration in the injured spinal cord is limited by physical and chemical barriers. Acute implantation of a multichannel poly(lactide-co-glycolide) (PLG) bridge mechanically stabilizes the injury, modulates inflammation, and provides a permissive environment for rapid cellularization and robust axonal regrowth through this otherwise inhibitory milieu. However, without additional intervention, regenerated axons remain largely unmyelinated (<10%), limiting functional repair. While transplanted human neural stem cells (hNSC) myelinate axons after spinal cord injury (SCI), hNSC fate is highly influenced by the SCI inflammatory microenvironment, also limiting functional repair. Accordingly, we investigated the combination of PLG scaffold bridges with hNSC to improve histological and functional outcome after SCI. In vitro, hNSC culture on a PLG scaffold increased oligodendroglial lineage selection after inflammatory challenge. In vivo, acute PLG bridge implantation followed by chronic hNSC transplantation demonstrated a robust capacity of donor human cells to migrate into PLG bridge channels along regenerating axons and integrate into the host spinal cord as myelinating oligodendrocytes and synaptically integrated neurons. Axons that regenerated through the PLG bridge formed synaptic circuits that connected the ipsilateral forelimb muscle to contralateral motor cortex. hNSC transplantation significantly enhanced the total number of regenerating and myelinated axons identified within the PLG bridge. Finally, the combination of acute bridge implantation and hNSC transplantation exhibited robust improvement in locomotor recovery. These data identify a successful strategy to enhance neurorepair through a temporally layered approach using acute bridge implantation and chronic cell transplantation to spare tissue, promote regeneration, and maximize the function of new axonal connections.
Collapse
Affiliation(s)
- Usha Nekanti
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA.
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA.
| | - Pooja S Sakthivel
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
| | - Atena Zahedi
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Dana A Creasman
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
| | - Rebecca A Nishi
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Courtney M Dumont
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Katja M Piltti
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Glenn L Guardamondo
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Norbert Hernandez
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Xingyuan Chen
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Hui Song
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Xiaoxiao Lin
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
| | - Joshua Martinez
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Lillian On
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Anita Lakatos
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Kiran Pawar
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Brian T David
- Department of Neurosurgery, Rush University Medical Center, Chicago, IL, USA
| | - Zhiling Guo
- Department of Medicine & Susan Samueli Integrative Health Institute, University of California, Irvine, CA, USA
| | - Stephanie K Seidlits
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
- Center for Neural Circuit Mapping, University of California Irvine, Irvine, CA, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Brian J Cummings
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
- Department of Physical Medicine and Rehabilitation, University of California, Irvine, CA, USA
- Institute for Memory Impairments & Neurological Disorder, University of California Irvine, Irvine, CA, USA
| | - Aileen J Anderson
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA.
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA.
- Department of Physical Medicine and Rehabilitation, University of California, Irvine, CA, USA.
- Institute for Memory Impairments & Neurological Disorder, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
7
|
Saunders MN, Griffin KV, Kalashnikova I, Kolpek D, Smith DR, Saito E, Cummings BJ, Anderson AJ, Shea LD, Park J. Biodegradable nanoparticles targeting circulating immune cells reduce central and peripheral sensitization to alleviate neuropathic pain following spinal cord injury. Pain 2024; 165:92-101. [PMID: 37463227 PMCID: PMC10787809 DOI: 10.1097/j.pain.0000000000002989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 05/26/2023] [Indexed: 07/20/2023]
Abstract
ABSTRACT Neuropathic pain is a critical source of comorbidity following spinal cord injury (SCI) that can be exacerbated by immune-mediated pathologies in the central and peripheral nervous systems. In this article, we investigate whether drug-free, biodegradable, poly(lactide- co -glycolide) (PLG) nanoparticle treatment mitigates the development of post-SCI neuropathic pain in female mice. Our results show that acute treatment with PLG nanoparticles following thoracic SCI significantly reduces tactile and cold hypersensitivity scores in a durable fashion. Nanoparticles primarily reduce peripheral immune-mediated mechanisms of neuropathic pain, including neuropathic pain-associated gene transcript frequency, transient receptor potential ankyrin 1 nociceptor expression, and MCP-1 (CCL2) chemokine production in the subacute period after injury. Altered central neuropathic pain mechanisms during this period are limited to reduced innate immune cell cytokine expression. However, in the chronic phase of SCI, nanoparticle treatment induces changes in both central and peripheral neuropathic pain signaling, driving reductions in cytokine production and other immune-relevant markers. This research suggests that drug-free PLG nanoparticles reprogram peripheral proalgesic pathways subacutely after SCI to reduce neuropathic pain outcomes and improve chronic central pain signaling.
Collapse
Affiliation(s)
- Michael N Saunders
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI USA
| | - Kate V Griffin
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI USA
| | - Irina Kalashnikova
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY USA
| | - Daniel Kolpek
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY USA
| | - Dominique R Smith
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI USA
| | - Eiji Saito
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI USA
| | - Brian J Cummings
- Department of Anatomy and Neurobiology, University of California, Irvine, CA USA
- Department of Physical Medicine and Rehabilitation, University of California, Irvine, CA USA
| | - Aileen J Anderson
- Department of Anatomy and Neurobiology, University of California, Irvine, CA USA
- Department of Physical Medicine and Rehabilitation, University of California, Irvine, CA USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI USA
| | - Jonghyuck Park
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY USA
| |
Collapse
|
8
|
Cheng C, Li Q, Lin G, Opara EC, Zhang Y. Neurobiological insights into lower urinary tract dysfunction: evaluating the role of brain-derived neurotrophic factor. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL UROLOGY 2023; 11:559-577. [PMID: 38148930 PMCID: PMC10749380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/17/2023] [Indexed: 12/28/2023]
Abstract
Lower urinary tract dysfunction (LUTD) encompasses a range of debilitating conditions that affect both sexes and different age groups. Understanding the underlying neurobiological mechanisms contributing to LUTD has emerged as a critical avenue for the development of targeted therapeutic strategies. Brain-derived neurotrophic factor (BDNF), a prominent member of the neurotrophin family, has attracted attention due to its multiple roles in neural development, plasticity, and maintenance. This review examines the intricate interplay between neurobiological factors and LUTD, focusing on the central involvement of BDNF. The review emphasizes the bidirectional relationship between LUTD and BDNF and explores how LUTD-induced neural changes may affect BDNF dynamics and vice versa. Growth factor therapy and the combined administration of controlled release growth factors and stem cells are minimally invasive treatment strategies for neuromuscular injury. Among the many growth factors and cytokines, brain-derived neurotrophic factor (BDNF) plays a prominent role in neuromuscular repair. As an essential neurotrophin, BDNF is involved in the modulation of neuromuscular regeneration through tropomyosin receptor kinase B (TrkB). Increasing BDNF levels facilitates the regeneration of the external urethral sphincter and contributes to the regulation of bladder contraction. Treatments targeting the BDNF pathway and sustained release of BDNF may become novel treatment options for urinary incontinence and other forms of lower urinary tract dysfunction. This review discusses the applications of BDNF and the theoretical basis for its use in the treatment of lower urinary tract dysfunction, including urinary incontinence (UI), overactive bladder (OAB), and benign prostatic hyperplasia (BPH), and in the clinical diagnosis of bladder dysfunction.
Collapse
Affiliation(s)
- Chen Cheng
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200011, China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of MedicineShanghai 200011, China
| | - Guiting Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of CaliforniaSan Francisco, CA 94143, USA
| | - Emmanuel C Opara
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health SciencesWinston-Salem, NC 27101, USA
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health SciencesWinston-Salem, NC 27101, USA
| |
Collapse
|
9
|
Zhu F, Nie G, Liu C. Engineered biomaterials in stem cell-based regenerative medicine. LIFE MEDICINE 2023; 2:lnad027. [PMID: 39872549 PMCID: PMC11749850 DOI: 10.1093/lifemedi/lnad027] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 07/17/2023] [Indexed: 01/30/2025]
Abstract
Stem cell-based regenerative therapies, which harness the self-renewal and differentiation properties of stem cells, have been in the spotlight due to their widespread applications in treating degenerative, aging, and other, generally intractable diseases. Therapeutically effective hematopoietic stem cells, mesenchymal stem cells, embryonic stem cells, and induced pluripotent stem cells have been used in numerous basic and translational studies with exciting results. However, pre-/post-transplantation issues of poor cell survival and retention, uncontrolled differentiation, and insufficient numbers of cells engrafted into host tissues are the major challenges in stem cell-based regenerative therapies. Engineered biomaterials have adjustable biochemical and biophysical properties that significantly affect cell behaviors, such as cell engraftment, survival, migration, and differentiation outcomes, thereby enhancing the engraftment of implanted stem cells and guiding tissue regeneration. Therefore, the combination of stem cell biology with bioengineered materials is a promising strategy to improve the therapeutic outcomes of stem cell-based regenerative therapy. In this review, we summarize the advances in the modulation of behaviors of stem cells via engineered biomaterials. We then present different approaches to harnessing bioengineered materials to enhance the transplantation of stem cells. Finally, we will provide future directions in regenerative therapy using stem cells.
Collapse
Affiliation(s)
- Fei Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Centre for Excellence in Nanoscience, National Centre for Nanoscience and Technology, Beijing 100190, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Centre for Excellence in Nanoscience, National Centre for Nanoscience and Technology, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
10
|
Park J, Farmer M, Casson C, Kalashnikova I, Kolpek D. Therapeutic Potential of Combinative shRNA-Encoded Lentivirus-Mediated Gene Silencing to Accelerate Somatosensory Recovery After Spinal Cord Trauma. Neurotherapeutics 2023; 20:564-577. [PMID: 36401079 PMCID: PMC10121969 DOI: 10.1007/s13311-022-01331-7] [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] [Accepted: 11/03/2022] [Indexed: 11/19/2022] Open
Abstract
Neuropathic pain following spinal cord injury (SCI) remains a difficult problem that affects more than 80% of SCI patients. Growing evidence indicates that neuroinflammatory responses play a key role in neuropathic pain after SCI. Short hairpin RNA (shRNA) interference is an efficient tool for the knockdown of disease-related specific gene expression after SCI, yet insufficient data is available to establish guidelines. In this study, we have constructed the transient receptor potential ankyrin 1 (TRPA1) shRNA encoded-lentiviral vector (LV-shTRPA1) and P38 MAPK shRNA encoded-lentiviral vector (LV-shP38) to investigate the silencing effects of shRNAs and their ability to reprogram the neuroinflammatory responses, thereby enhancing somatosensory recovery after SCI. Our in vitro data employing HEK293-FT and activated macrophages demonstrated that delivered LV-shRNAs showed high transduction efficacy with no cytotoxicity. Furthermore, a combination of LV-shP38 and LV-shTRPA1 was found to be most effective at suppressing target genes, cutting the expression of pro-inflammatory and pro-nociceptive factors in the dorsal horn of the spinal cord and dorsal root ganglia, thus contributing to the alleviation of neuronal hypersensitivities after SCI. Overall, our data demonstrated that the combination LV-shP38/shTRPA1 produced a synergistic effect for immunomodulation and reduced neuropathic pain with a favorable risk-to-benefit ratio. Collectively, our LV-mediated shRNA delivery will provide an efficient tool for gene silencing therapeutic approaches to treat various incurable disorders.
Collapse
Affiliation(s)
- Jonghyuck Park
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40506, USA.
- Spinal Cord and Brain Injury Research Center, University of Kentucky, 741 S. Limestone, Lexington, KY, 40506, USA.
| | - Matthew Farmer
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40506, USA
| | - Camara Casson
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40506, USA
| | - Irina Kalashnikova
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40506, USA
| | - Daniel Kolpek
- Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone, Lexington, KY, 40506, USA
| |
Collapse
|
11
|
Sousa JPM, Stratakis E, Mano J, Marques PAAP. Anisotropic 3D scaffolds for spinal cord guided repair: Current concepts. BIOMATERIALS ADVANCES 2023; 148:213353. [PMID: 36848743 DOI: 10.1016/j.bioadv.2023.213353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023]
Abstract
A spinal cord injury (SCI) can be caused by unforeseen events such as a fall, a vehicle accident, a gunshot, or a malignant illness, which has a significant impact on the quality of life of the patient. Due to the limited regenerative potential of the central nervous system (CNS), SCI is one of the most daunting medical challenges of modern medicine. Great advances have been made in tissue engineering and regenerative medicine, which include the transition from two-dimensional (2D) to three-dimensional (3D) biomaterials. Combinatory treatments that use 3D scaffolds may significantly enhance the repair and regeneration of functional neural tissue. In an effort to mimic the chemical and physical properties of neural tissue, scientists are researching the development of the ideal scaffold made of synthetic and/or natural polymers. Moreover, in order to restore the architecture and function of neural networks, 3D scaffolds with anisotropic properties that replicate the native longitudinal orientation of spinal cord nerve fibres are being designed. In an effort to determine if scaffold anisotropy is a crucial property for neural tissue regeneration, this review focuses on the most current technological developments relevant to anisotropic scaffolds for SCI. Special consideration is given to the architectural characteristics of scaffolds containing axially oriented fibres, channels, and pores. By analysing neural cell behaviour in vitro and tissue integration and functional recovery in animal models of SCI, the therapeutic efficacy is evaluated for its successes and limitations.
Collapse
Affiliation(s)
- Joana P M Sousa
- TEMA - Centre for Mechanical Technology and Automation, Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal; LASI - Intelligent Systems Associate Laboratory, Portugal; Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas (FORTH-IESL), Heraklion, Greece; CICECO - Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro 3810-193, Portugal
| | - Emmanuel Stratakis
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas (FORTH-IESL), Heraklion, Greece
| | - João Mano
- CICECO - Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, Aveiro 3810-193, Portugal
| | - Paula A A P Marques
- TEMA - Centre for Mechanical Technology and Automation, Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal; LASI - Intelligent Systems Associate Laboratory, Portugal.
| |
Collapse
|
12
|
Neurotrophic Factors as Regenerative Therapy for Neurodegenerative Diseases: Current Status, Challenges and Future Perspectives. Int J Mol Sci 2023; 24:ijms24043866. [PMID: 36835277 PMCID: PMC9968045 DOI: 10.3390/ijms24043866] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/25/2023] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), multiple sclerosis (MS), spinal cord injury (SCI), and amyotrophic lateral sclerosis (ALS), are characterized by acute or chronic progressive loss of one or several neuronal subtypes. However, despite their increasing prevalence, little progress has been made in successfully treating these diseases. Research has recently focused on neurotrophic factors (NTFs) as potential regenerative therapy for neurodegenerative diseases. Here, we discuss the current state of knowledge, challenges, and future perspectives of NTFs with a direct regenerative effect in chronic inflammatory and degenerative disorders. Various systems for delivery of NTFs, such as stem and immune cells, viral vectors, and biomaterials, have been applied to deliver exogenous NTFs to the central nervous system, with promising results. The challenges that currently need to be overcome include the amount of NTFs delivered, the invasiveness of the delivery route, the blood-brain barrier permeability, and the occurrence of side effects. Nevertheless, it is important to continue research and develop standards for clinical applications. In addition to the use of single NTFs, the complexity of chronic inflammatory and degenerative diseases may require combination therapies targeting multiple pathways or other possibilities using smaller molecules, such as NTF mimetics, for effective treatment.
Collapse
|
13
|
Feng C, Deng L, Yong YY, Wu JM, Qin DL, Yu L, Zhou XG, Wu AG. The Application of Biomaterials in Spinal Cord Injury. Int J Mol Sci 2023; 24:816. [PMID: 36614259 PMCID: PMC9821025 DOI: 10.3390/ijms24010816] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/19/2022] [Accepted: 12/30/2022] [Indexed: 01/05/2023] Open
Abstract
The spinal cord and the brain form the central nervous system (CNS), which is the most important part of the body. However, spinal cord injury (SCI) caused by external forces is one of the most difficult types of neurological injury to treat, resulting in reduced or even absent motor, sensory and autonomic functions. It leads to the reduction or even disappearance of motor, sensory and self-organizing nerve functions. Currently, its incidence is increasing each year worldwide. Therefore, the development of treatments for SCI is urgently needed in the clinic. To date, surgery, drug therapy, stem cell transplantation, regenerative medicine, and rehabilitation therapy have been developed for the treatment of SCI. Among them, regenerative biomaterials that use tissue engineering and bioscaffolds to transport cells or drugs to the injured site are considered the most promising option. In this review, we briefly introduce SCI and its molecular mechanism and summarize the application of biomaterials in the repair and regeneration of tissue in various models of SCI. However, there is still limited evidence about the treatment of SCI with biomaterials in the clinic. Finally, this review will provide inspiration and direction for the future study and application of biomaterials in the treatment of SCI.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiao-Gang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Drugability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Key Laboratory of Medical Electrophysiology of Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
14
|
Molecular Mechanisms in the Vascular and Nervous Systems following Traumatic Spinal Cord Injury. LIFE (BASEL, SWITZERLAND) 2022; 13:life13010009. [PMID: 36675958 PMCID: PMC9866624 DOI: 10.3390/life13010009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/26/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
Traumatic spinal cord injury (SCI) induces various complex pathological processes that cause physical impairment and psychological devastation. The two phases of SCI are primary mechanical damage (the immediate result of trauma) and secondary injury (which occurs over a period of minutes to weeks). After the mechanical impact, vascular disruption, inflammation, demyelination, neuronal cell death, and glial scar formation occur during the acute phase. This sequence of events impedes nerve regeneration. In the nervous system, various extracellular secretory factors such as neurotrophic factors, growth factors, and cytokines are involved in these events. In the vascular system, the blood-spinal cord barrier (BSCB) is damaged, allowing immune cells to infiltrate the parenchyma. Later, endogenous angiogenesis is promoted during the subacute phase. In this review, we describe the roles of secretory factors in the nervous and vascular systems following traumatic SCI, and discuss the outcomes of their therapeutic application in traumatic SCI.
Collapse
|
15
|
Ciciriello AJ, Surnar B, Medy GD, Su X, Dhar S, Dumont CM. Biomaterial-targeted precision nanoparticle delivery to the injured spinal cord. Acta Biomater 2022; 152:532-545. [PMID: 36087868 PMCID: PMC10551882 DOI: 10.1016/j.actbio.2022.08.077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/29/2022] [Accepted: 08/31/2022] [Indexed: 11/01/2022]
Abstract
Drug delivery requires precision in timing, location, and dosage to achieve therapeutic benefits. Challenges in addressing all three of these critical criteria result in poor temporal dexterity, widespread accumulation and off-target effects, and high doses with the potential for toxicity. To address these challenges, we have developed the BiomatErial Accumulating Carriers for On-demand Nanotherapy (BEACON) platform that utilizes an implantable biomaterial to serve as a target for systemically delivered nanoparticles (NPs). With the BEACON system, administered NPs are conjugated with a ligand that has high affinity for a receptor in the implanted biomaterial. To test BEACON, an in vivo spinal cord injury (SCI) model was used as it provides an injury model where the three identified criteria can be tested as it is a dynamic and complicated injury model with no currently approved therapies. Through our work, we have demonstrated temporal dexterity in NP administration by injecting 6 days post-SCI, decreased off-target accumulation with a significant drop in liver accumulation, and retention of our NPs in the target biomaterial. The BEACON system can be applied broadly, beyond the nervous system, to improve systemically delivered NP accumulation at an implanted biomaterial target. STATEMENT OF SIGNIFICANCE: Targeted drug delivery approaches have the potential to improve therapeutic regimens for patients on a case-by-case basis. Improved localization of a therapeutic to site of interest can result in increased efficacy and limit the need for repeat dosing. Unfortunately, targeted strategies can fall short when receptors on cells or tissues are too widespread or change over the course of disease or injury progression. The BEACON system developed herein eliminates the need to target a cell or tissue receptor by targeting an implantable biomaterial with location-controllable accumulation and sustained presentation over time. The targeting paradigm presented by BEACON is widely applicable throughout tissue engineering and regenerative medicine without the need to retool for each new application.
Collapse
Affiliation(s)
- Andrew J Ciciriello
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, FL 33146, United States; Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, 1951 NW 7th Avenue, Miami, Florida 33136, United States
| | - Bapurao Surnar
- Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, 1951 NW 7th Avenue, Miami, Florida 33136, United States; Department of Biochemistry & Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United States
| | - Giovanni D Medy
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, FL 33146, United States
| | - Xiaoyu Su
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, FL 33146, United States
| | - Shanta Dhar
- Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, 1951 NW 7th Avenue, Miami, Florida 33136, United States; Department of Biochemistry & Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United States; Sylvester Comprehensive Cancer Center, University of Miami, 1475 NW 12th Avenue, Miami, Florida 33136, United States
| | - Courtney M Dumont
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, FL 33146, United States; Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, 1951 NW 7th Avenue, Miami, Florida 33136, United States; Department of Biochemistry & Molecular Biology, University of Miami, 1011 NW 15th Street, Miami, Florida 33136, United States.
| |
Collapse
|
16
|
Multimodular Bio-Inspired Organized Structures Guiding Long-Distance Axonal Regeneration. Biomedicines 2022; 10:biomedicines10092228. [PMID: 36140328 PMCID: PMC9496454 DOI: 10.3390/biomedicines10092228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022] Open
Abstract
Axonal bundles or axonal tracts have an aligned and unidirectional architecture present in many neural structures with different lengths. When peripheral nerve injury (PNI), spinal cord injury (SCI), traumatic brain injury (TBI), or neurodegenerative disease occur, the intricate architecture undergoes alterations leading to growth inhibition and loss of guidance through large distance. In order to overcome the limitations of long-distance axonal regeneration, here we combine a poly-L-lactide acid (PLA) fiber bundle in the common lumen of a sequence of hyaluronic acid (HA) conduits or modules and pre-cultured Schwann cells (SC) as cells supportive of axon extension. This multimodular preseeded conduit is then used to induce axon growth from a dorsal root ganglion (DRG) explant placed at one of its ends and left for 21 days to follow axon outgrowth. The multimodular conduit proved effective in promoting directed axon growth, and the results may thus be of interest for the regeneration of long tissue defects in the nervous system. Furthermore, the hybrid structure grown within the HA modules consisting in the PLA fibers and the SC can be extracted from the conduit and cultured independently. This “neural cord” proved to be viable outside its scaffold and opens the door to the generation of ex vivo living nerve in vitro for transplantation.
Collapse
|
17
|
Clarke E, Stocki P, Sinclair EH, Gauhar A, Fletcher EJR, Krawczun-Rygmaczewska A, Duty S, Walsh FS, Doherty P, Rutkowski JL. A Single Domain Shark Antibody Targeting the Transferrin Receptor 1 Delivers a TrkB Agonist Antibody to the Brain and Provides Full Neuroprotection in a Mouse Model of Parkinson’s Disease. Pharmaceutics 2022; 14:pharmaceutics14071335. [PMID: 35890231 PMCID: PMC9318160 DOI: 10.3390/pharmaceutics14071335] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 02/04/2023] Open
Abstract
Single domain shark antibodies that bind to the transferrin receptor 1 (TfR1) on brain endothelial cells have been used to shuttle antibodies and other cargos across the blood brain barrier (BBB) to the brain. For these studies the TXB4 brain shuttle was fused to a TrkB neurotrophin receptor agonist antibody. The TXB4-TrkB fusion retained potent agonist activity at its cognate receptor and after systemic administration showed a 12-fold increase in brain levels over the unmodified antibody. Only the TXB4-TrkB antibody fusion was detected within the brain and localized to TrkB positive cells in the cortex and tyrosine hydroxylase (TH) positive dopaminergic neurons in the substantia nigra pars compacta (SNc), where it was associated with activated ERK1/2 signaling. When tested in the 6-hydroxydopamine (6-OHDA) mouse model of Parkinson’s disease (PD), TXB4-TrkB, but not the unmodified antibody, completely prevented the 6-OHDA induced death of TH positive neurons in the SNc. In conclusion, the fusion of the TXB4 brain shuttle allows a TrkB agonist antibody to reach neuroprotective concentrations in the brain parenchyma following systemic administration.
Collapse
Affiliation(s)
- Emily Clarke
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Wolfson Centre for Age-Related Disease, Guy’s Campus, London SE1 1UL, UK; (E.C.); (E.J.R.F.); (A.K.-R.); (S.D.); (P.D.)
| | - Pawel Stocki
- Ossianix, Inc., Gunnels Wood Rd., Stevenage SG1 2FX, UK; (P.S.); (E.H.S.); (A.G.); (F.S.W.)
| | - Elizabeth H. Sinclair
- Ossianix, Inc., Gunnels Wood Rd., Stevenage SG1 2FX, UK; (P.S.); (E.H.S.); (A.G.); (F.S.W.)
| | - Aziz Gauhar
- Ossianix, Inc., Gunnels Wood Rd., Stevenage SG1 2FX, UK; (P.S.); (E.H.S.); (A.G.); (F.S.W.)
| | - Edward J. R. Fletcher
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Wolfson Centre for Age-Related Disease, Guy’s Campus, London SE1 1UL, UK; (E.C.); (E.J.R.F.); (A.K.-R.); (S.D.); (P.D.)
| | - Alicja Krawczun-Rygmaczewska
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Wolfson Centre for Age-Related Disease, Guy’s Campus, London SE1 1UL, UK; (E.C.); (E.J.R.F.); (A.K.-R.); (S.D.); (P.D.)
| | - Susan Duty
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Wolfson Centre for Age-Related Disease, Guy’s Campus, London SE1 1UL, UK; (E.C.); (E.J.R.F.); (A.K.-R.); (S.D.); (P.D.)
| | - Frank S. Walsh
- Ossianix, Inc., Gunnels Wood Rd., Stevenage SG1 2FX, UK; (P.S.); (E.H.S.); (A.G.); (F.S.W.)
| | - Patrick Doherty
- King’s College London, Institute of Psychiatry, Psychology and Neuroscience, Wolfson Centre for Age-Related Disease, Guy’s Campus, London SE1 1UL, UK; (E.C.); (E.J.R.F.); (A.K.-R.); (S.D.); (P.D.)
| | - Julia Lynn Rutkowski
- Ossianix, Inc., Gunnels Wood Rd., Stevenage SG1 2FX, UK; (P.S.); (E.H.S.); (A.G.); (F.S.W.)
- Correspondence: ; Tel.: +1-(610)-291-1724
| |
Collapse
|
18
|
Abstract
The successful transplantation of stem cells has the potential to transform regenerative medicine approaches and open promising avenues to repair, replace, and regenerate diseased, damaged, or aged tissues. However, pre-/post-transplantation issues of poor cell survival, retention, cell fate regulation, and insufficient integration with host tissues constitute significant challenges. The success of stem cell transplantation depends upon the coordinated sequence of stem cell renewal, specific lineage differentiation, assembly, and maintenance of long-term function. Advances in biomaterials can improve pre-/post-transplantation outcomes by integrating biophysiochemical cues and emulating tissue microenvironments. This review highlights leading biomaterials-based approaches for enhancing stem cell transplantation.
Collapse
Affiliation(s)
- Bhushan N Kharbikar
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Priya Mohindra
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA 94158, USA
| | - Tejal A Desai
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA; UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA 94158, USA; Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA; School of Engineering, Brown University, Providence, RI, 02912, USA.
| |
Collapse
|
19
|
Lai BQ, Bai YR, Han WT, Zhang B, Liu S, Sun JH, Liu JL, Li G, Zeng X, Ding Y, Ma YH, Zhang L, Chen ZH, Wang J, Xiong Y, Wu JH, Quan Q, Xing LY, Zhang HB, Zeng YS. Construction of a niche-specific spinal white matter-like tissue to promote directional axon regeneration and myelination for rat spinal cord injury repair. Bioact Mater 2021; 11:15-31. [PMID: 34938909 DOI: 10.1016/j.bioactmat.2021.10.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/29/2021] [Accepted: 10/03/2021] [Indexed: 12/16/2022] Open
Abstract
Directional axon regeneration and remyelination are crucial for repair of spinal cord injury (SCI), but existing treatments do not effectively promote those processes. Here, we propose a strategy for construction of niche-specific spinal white matter-like tissue (WMLT) using decellularized optic nerve (DON) loaded with neurotrophin-3 (NT-3)-overexpressing oligodendrocyte precursor cells. A rat model with a white matter defect in the dorsal spinal cord of the T10 segment was used. The WMLT transplantation group showed significant improvement in coordinated motor functions compared with the control groups. WMLT transplants integrated well with host spinal cord white matter, effectively addressing several barriers to directional axonal regeneration and myelination during SCI repair. In WMLT, laminin was found to promote development of oligodendroglial lineage (OL) cells by binding to laminin receptors. Interestingly, laminin could also guide linear axon regeneration via interactions with specific integrins on the axon surface. The WMLT developed here utilizes the unique microstructure and bioactive matrix of DON to create a niche rich in laminin, NT-3 and OL cells to achieve significant structural repair of SCI. Our protocol can help to promote research on repair of nerve injury and construction of neural tissues and organoids that form specific cell niches.
Collapse
Affiliation(s)
- Bi-Qin Lai
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China.,Department of Histology and Embryology, Zhongshan School of Medicine, SunYat-sen University, Guangzhou, 510080, China.,Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, 510120, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.,Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yu-Rong Bai
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Wei-Tao Han
- Department of Histology and Embryology, Zhongshan School of Medicine, SunYat-sen University, Guangzhou, 510080, China
| | - Bao Zhang
- Department of Histology and Embryology, Zhongshan School of Medicine, SunYat-sen University, Guangzhou, 510080, China
| | - Shu Liu
- Department of Anatomy, Anhui Medical University, Hefei, 230032, China
| | - Jia-Hui Sun
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Jia-Lin Liu
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Ge Li
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Xiang Zeng
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China.,Department of Histology and Embryology, Zhongshan School of Medicine, SunYat-sen University, Guangzhou, 510080, China.,Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, 510120, China.,Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ying Ding
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China.,Department of Histology and Embryology, Zhongshan School of Medicine, SunYat-sen University, Guangzhou, 510080, China.,Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yuan-Huan Ma
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Ling Zhang
- Department of Geriatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Zheng-Hong Chen
- Department of Geriatrics, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jun Wang
- Physiotherapy Department, Guangdong Work Injury Rehabilitation Hospital, Guangzhou, China
| | - Yuan Xiong
- Physiotherapy Department, Guangdong Work Injury Rehabilitation Hospital, Guangzhou, China
| | - Jin-Hua Wu
- Physiotherapy Department, Guangdong Work Injury Rehabilitation Hospital, Guangzhou, China
| | - Qi Quan
- Department of Orthopedic Surgery, Key Laboratory of Musculoskeletal Trauma &War Injuries PLA, Beijing Key Lab of Regenerative Medicine in Orthopedics, The 4th Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Ling-Yan Xing
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hong-Bo Zhang
- Department of Histology and Embryology, Zhongshan School of Medicine, SunYat-sen University, Guangzhou, 510080, China
| | - Yuan-Shan Zeng
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China.,Department of Histology and Embryology, Zhongshan School of Medicine, SunYat-sen University, Guangzhou, 510080, China.,Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, 510120, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.,Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| |
Collapse
|
20
|
Bahlakeh G, Rahbarghazi R, Mohammadnejad D, Abedelahi A, Karimipour M. Current knowledge and challenges associated with targeted delivery of neurotrophic factors into the central nervous system: focus on available approaches. Cell Biosci 2021; 11:181. [PMID: 34641969 PMCID: PMC8507154 DOI: 10.1186/s13578-021-00694-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 09/28/2021] [Indexed: 12/23/2022] Open
Abstract
During the last decades, numerous basic and clinical studies have been conducted to assess the delivery efficiency of therapeutic agents into the brain and spinal cord parenchyma using several administration routes. Among conventional and in-progress administrative routes, the eligibility of stem cells, viral vectors, and biomaterial systems have been shown in the delivery of NTFs. Despite these manifold advances, the close association between the delivery system and regeneration outcome remains unclear. Herein, we aimed to discuss recent progress in the delivery of these factors and the pros and cons related to each modality.
Collapse
Affiliation(s)
- Gozal Bahlakeh
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Daruosh Mohammadnejad
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Abedelahi
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Karimipour
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran. .,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
21
|
Lentiviral Vectors Delivered with Biomaterials as Therapeutics for Spinal Cord Injury. Cells 2021; 10:cells10082102. [PMID: 34440872 PMCID: PMC8394044 DOI: 10.3390/cells10082102] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/10/2021] [Accepted: 08/12/2021] [Indexed: 12/15/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating trauma that can cause permanent disability, life-long chronic issues for sufferers and is a big socioeconomic burden. Regenerative medicine aims to overcome injury caused deficits and restore function after SCI through gene therapy and tissue engineering approaches. SCI has a multifaceted pathophysiology. Due to this, producing therapies that target multiple different cellular and molecular mechanisms might prove to be a superior approach in attempts at regeneration. Both biomaterials and nucleic acid delivery via lentiviral vectors (LVs) have proven to promote repair and restoration of function post SCI in animal models. Studies indicate that a combination of biomaterials and LVs is more effective than either approach alone. This review presents studies supporting the use of LVs and LVs delivered with biomaterials in therapies for SCI and summarises methods to combine LVs with biomaterials for SCI treatment. By summarising this knowledge this review aims to demonstrate how LV delivery with biomaterials can augment/compliment both LV and biomaterial therapeutic effects in SCI.
Collapse
|
22
|
Otsuka T, Maeda Y, Kurose T, Nakagawa K, Mitsuhara T, Kawahara Y, Yuge L. Comparisons of Neurotrophic Effects of Mesenchymal Stem Cells Derived from Different Tissues on Chronic Spinal Cord Injury Rats. Stem Cells Dev 2021; 30:865-875. [PMID: 34148410 DOI: 10.1089/scd.2021.0070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cell-based therapies with mesenchymal stem cells (MSCs) are considered as promising strategies for spinal cord injury (SCI). MSCs have unique characteristics due to differences in the derived tissues. However, relatively few studies have focused on differences in the therapeutic effects of MSCs derived from different tissues. In this study, the therapeutic effects of adipose tissue-derived MSCs, bone marrow-derived MSCs, and cranial bone-derived MSCs (cMSCs) on chronic SCI model rats were compared. MSCs were established from the collected adipose tissue, bone marrow, and cranial bone. Neurotrophic factor expression of each MSC type was analyzed by real-time PCR. SCI rats were established using the weight-drop method and transplanted intravenously with MSCs at 4 weeks after SCI. Hindlimb motor function was evaluated from before injury to 4 weeks after transplantation. Endogenous neurotrophic factor and neural repair factor expression in spinal cord (SC) tissue were examined by real-time PCR and western blot analyses. Although there were no differences in the expression levels of cell surface markers and multipotency, expression of Bdnf, Ngf, and Sort1 (Nt-3) was relatively higher in cMSCs. Transplantation of cMSCs improved motor function of chronic SCI model rats. Although there was no difference in the degree of engraftment of transplanted cells in the injured SC tissue, transplantation of cMSCs enhanced Bdnf, TrkB, and Gap-43 messenger RNA expression and synaptophysin protein expression in injured SC tissue. As compared with MSCs derived other tissues, cMSCs highly express many neurotrophic factors, which improved motor function in chronic SCI model rats by promoting endogenous neurotrophic and neural plasticity factors. These results demonstrate the efficacy of cMSCs in cell-based therapy for chronic SCI.
Collapse
Affiliation(s)
- Takashi Otsuka
- Division of Bio-Environmental Adaptation Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuyo Maeda
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tomoyuki Kurose
- Division of Bio-Environmental Adaptation Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kei Nakagawa
- Division of Bio-Environmental Adaptation Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takafumi Mitsuhara
- Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | | | - Louis Yuge
- Division of Bio-Environmental Adaptation Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.,Space Bio-Laboratories Co., Ltd., Hiroshima, Japan
| |
Collapse
|
23
|
Ciciriello AJ, Smith DR, Munsell MK, Boyd SJ, Shea LD, Dumont CM. IL-10 lentivirus-laden hydrogel tubes increase spinal progenitor survival and neuronal differentiation after spinal cord injury. Biotechnol Bioeng 2021; 118:2609-2625. [PMID: 33835500 DOI: 10.1002/bit.27781] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022]
Abstract
A complex cellular cascade characterizes the pathophysiological response following spinal cord injury (SCI) limiting regeneration. Biomaterial and stem cell combination therapies together have shown synergistic effects, compared to the independent benefits of each intervention, and represent a promising approach towards regaining function after injury. In this study, we combine our polyethylene glycol (PEG) cell delivery platform with lentiviral-mediated overexpression of the anti-inflammatory cytokine interleukin (IL)-10 to improve mouse embryonic Day 14 (E14) spinal progenitor transplant survival. Immediately following injury in a mouse SCI hemisection model, five PEG tubes were implanted followed by direct injection into the tubes of lentivirus encoding for IL-10. Two weeks after tube implantation, mouse E14 spinal progenitors were injected directly into the integrated tubes, which served as a soft substrate for cell transplantation. Together, the tubes with the IL-10 encoding lentivirus improved E14 spinal progenitor survival, assessed at 2 weeks posttransplantation (4 weeks postinjury). On average, 8.1% of E14 spinal progenitors survived in mice receiving IL-10 lentivirus-laden tubes compared with 0.7% in mice receiving transplants without tubes, an 11.5-fold difference. Surviving E14 spinal progenitors gave rise to neurons when injected into tubes. Axon elongation and remyelination were observed, in addition to a significant increase in functional recovery in mice receiving IL-10 lentivirus-laden tubes with E14 spinal progenitor delivery compared to the injury only control by 4 weeks postinjury. All other conditions did not exhibit increased stepping until 8 or 12 weeks postinjury. This system affords increased control over the transplantation microenvironment, offering the potential to improve stem cell-mediated tissue regeneration.
Collapse
Affiliation(s)
- Andrew J Ciciriello
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA.,DJTMF Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, Miami, Florida, USA
| | - Dominique R Smith
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Mary K Munsell
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Sydney J Boyd
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA.,Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Courtney M Dumont
- Department of Biomedical Engineering, University of Miami, Coral Gables, Florida, USA.,DJTMF Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, Miami, Florida, USA
| |
Collapse
|
24
|
Ehsanipour A, Sathialingam M, Rad LM, de Rutte J, Bierman RD, Liang J, Xiao W, Di Carlo D, Seidlits SK. Injectable, macroporous scaffolds for delivery of therapeutic genes to the injured spinal cord. APL Bioeng 2021; 5:016104. [PMID: 33728392 PMCID: PMC7946441 DOI: 10.1063/5.0035291] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/03/2021] [Indexed: 02/06/2023] Open
Abstract
Biomaterials are being developed as therapeutics for spinal cord injury (SCI) that can stabilize and bridge acute lesions and mediate the delivery of transgenes, providing a localized and sustained reservoir of regenerative factors. For clinical use, direct injection of biomaterial scaffolds is preferred to enable conformation to unique lesions and minimize tissue damage. While an interconnected network of cell-sized macropores is necessary for rapid host cell infiltration into-and thus integration of host tissue with-implanted scaffolds, injectable biomaterials have generally suffered from a lack of control over the macrostructure. As genetic vectors have short lifetimes in vivo, rapid host cell infiltration into scaffolds is a prerequisite for efficient biomaterial-mediated delivery of transgenes. We present scaffolds that can be injected and assembled in situ from hyaluronic acid (HA)-based, spherical microparticles to form scaffolds with a network of macropores (∼10 μm). The results demonstrate that addition of regularly sized macropores to traditional hydrogel scaffolds, which have nanopores (∼10 nm), significantly increases the expression of locally delivered transgene to the spinal cord after a thoracic injury. Maximal cell and axon infiltration into scaffolds was observed in scaffolds with more regularly sized macropores. The delivery of lentiviral vectors encoding the brain-derived neurotrophic factor (BDNF), but not neurotrophin-3, from these scaffolds further increased total numbers and myelination of infiltrating axons. Modest improvements to the hindlimb function were observed with BDNF delivery. The results demonstrate the utility of macroporous and injectable HA scaffolds as a platform for localized gene therapies after SCI.
Collapse
Affiliation(s)
- Arshia Ehsanipour
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Mayilone Sathialingam
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Laila M Rad
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Joseph de Rutte
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Rebecca D Bierman
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Jesse Liang
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | - Weikun Xiao
- Department of Bioengineering, University of California, Los Angeles, California 90095, USA
| | | | | |
Collapse
|
25
|
Huang RY, Liu ZH, Weng WH, Chang CW. Magnetic nanocomplexes for gene delivery applications. J Mater Chem B 2021; 9:4267-4286. [PMID: 33942822 DOI: 10.1039/d0tb02713h] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Gene delivery is an indispensable technique for various biomedical applications such as gene therapy, stem cell engineering and gene editing. Recently, magnetic nanoparticles (MNPs) have received increasing attention for their use in promoting gene delivery efficiency. Under magnetic attraction, gene delivery efficiency using viral or nonviral gene carriers could be universally enhanced. Besides, magnetic nanoparticles could be utilized in magnetic resonance imaging or magnetic hyperthermia therapy, providing extra theranostic opportunities. In this review, recent research integrating MNPs with a viral or nonviral gene vector is summarized from both technical and application perspectives. Applications of MNPs in cutting-edge research technologies, such as biomimetic cell membrane nano-gene carriers, exosome-based gene delivery, cell-based drug delivery systems or CRISPR/Cas9 gene editing, are also discussed.
Collapse
Affiliation(s)
- Rih-Yang Huang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| | - Zhuo-Hao Liu
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Chang Gung Medical College and University, Taiwan.
| | - Wei-Han Weng
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| | - Chien-Wen Chang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
26
|
Youngblood R, Flesher CG, Delproposto J, Baker NA, Neeley CK, Li F, Lumeng CN, Shea LD, O'Rourke RW. Regulation of adipose tissue inflammation and systemic metabolism in murine obesity by polymer implants loaded with lentiviral vectors encoding human interleukin-4. Biotechnol Bioeng 2020; 117:3891-3901. [PMID: 32729936 PMCID: PMC8358590 DOI: 10.1002/bit.27523] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 07/26/2020] [Accepted: 07/28/2020] [Indexed: 11/08/2022]
Abstract
Dysfunctional adipose tissue plays a central role in the pathogenesis of the obesity-related metabolic disease, including type 2 diabetes. Targeting adipose tissue using biopolymer implants is a novel therapeutic approach for metabolic disease. We transplanted porous poly(lactide-co-glycolide) (PLG) implants coated with human interleukin-4 (hIL-4)-expressing lentivirus into epididymal white adipose tissue (eWAT) of mice fed a high-fat diet. Tissue and systemic inflammation and metabolism were studied with flow cytometry, immunohistochemistry, quantitative real-time polymerase chain reaction, adipose tissue histology, and in vivo glucose tolerance testing at 2 and 10 weeks of a high-fat diet. PLG implants carrying hIL-4-expressing lentivirus implanted into epididymal white adipose tissue of mice-regulated adipose tissue inflammation, including increased CD3+ CD4+ T-cell frequency, increased eWAT adipocyte hypertrophy, and decreased FASN and ATGL expression, along with reduced fasting blood glucose levels. These effects were observed in early obesity but were not maintained in established obesity. Local delivery of bioimplants loaded with cytokine-expressing lentivirus vectors to adipose tissue influences tissue inflammation and systemic metabolism in early obesity. Further study will be required to show more durable metabolic effects. These data demonstrate that polymer biomaterials implanted into adipose tissue have the potential to modulate local tissue and systemic inflammation and metabolism.
Collapse
Affiliation(s)
- Richard Youngblood
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Carmen G Flesher
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jennifer Delproposto
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
| | - Nicki A Baker
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Christopher K Neeley
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Fanghua Li
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Carey N Lumeng
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, Michigan
- Graduate Program in Immunology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
- Graduate Program in Cellular and Molecular Biology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Robert W O'Rourke
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan
- Department of Surgery, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
| |
Collapse
|
27
|
Sahib S, Sharma A, Menon PK, Muresanu DF, Castellani RJ, Nozari A, Lafuente JV, Bryukhovetskiy I, Tian ZR, Patnaik R, Buzoianu AD, Wiklund L, Sharma HS. Cerebrolysin enhances spinal cord conduction and reduces blood-spinal cord barrier breakdown, edema formation, immediate early gene expression and cord pathology after injury. PROGRESS IN BRAIN RESEARCH 2020; 258:397-438. [PMID: 33223040 DOI: 10.1016/bs.pbr.2020.09.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Spinal cord evoked potentials (SCEP) are good indicators of spinal cord function in health and disease. Disturbances in SCEP amplitudes and latencies during spinal cord monitoring predict spinal cord pathology following trauma. Treatment with neuroprotective agents preserves SCEP and reduces cord pathology after injury. The possibility that cerebrolysin, a balanced composition of neurotrophic factors improves spinal cord conduction, attenuates blood-spinal cord barrier (BSCB) disruption, edema formation, and cord pathology was examined in spinal cord injury (SCI). SCEP is recorded from epidural space over rat spinal cord T9 and T12 segments after peripheral nerves stimulation. SCEP consists of a small positive peak (MPP), followed by a prominent negative peak (MNP) that is stable before SCI. A longitudinal incision (2mm deep and 5mm long) into the right dorsal horn (T10 and T11 segments) resulted in an immediate long-lasting depression of the rostral MNP with an increase in the latencies. Pretreatment with either cerebrolysin (CBL 5mL/kg, i.v. 30min before) alone or TiO2 nanowired delivery of cerebrolysin (NWCBL 2.5mL/kg, i.v.) prevented the loss of MNP amplitude and even enhanced further from the pre-injury level after SCI without affecting latencies. At 5h, SCI induced edema, BSCB breakdown, and cell injuries were significantly reduced by CBL and NWCBL pretreatment. Interestingly this effect on SCEP and cord pathology was still prominent when the NWCBL was delivered 2min after SCI. Moreover, expressions of c-fos and c-jun genes that are prominent at 5h in untreated SCI are also considerably reduced by CBL and NWCBL treatment. These results are the first to show that CBL and NWCBL enhanced SCEP activity and thwarted the development of cord pathology after SCI. Furthermore, NWCBL in low doses has superior neuroprotective effects on SCEP and cord pathology, not reported earlier. The functional significance and future clinical potential of CBL and NWCBL in SCI are discussed.
Collapse
Affiliation(s)
- Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Preeti K Menon
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden; Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Igor Bryukhovetskiy
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
28
|
Miranda-Lourenço C, Ribeiro-Rodrigues L, Fonseca-Gomes J, Tanqueiro SR, Belo RF, Ferreira CB, Rei N, Ferreira-Manso M, de Almeida-Borlido C, Costa-Coelho T, Freitas CF, Zavalko S, Mouro FM, Sebastião AM, Xapelli S, Rodrigues TM, Diógenes MJ. Challenges of BDNF-based therapies: From common to rare diseases. Pharmacol Res 2020; 162:105281. [PMID: 33161136 DOI: 10.1016/j.phrs.2020.105281] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/20/2020] [Accepted: 10/22/2020] [Indexed: 12/11/2022]
Abstract
Neurotrophins are a well-known family of neurotrophic factors that play an important role both in the central and peripheral nervous systems, where they modulate neuronal survival, development, function and plasticity. Brain-derived neurotrophic factor (BDNF) possesses diverse biological functions which are mediated by the activation of two main classes of receptors, the tropomyosin-related kinase (Trk) B and the p75 neurotrophin receptor (p75NTR). The therapeutic potential of BDNF has drawn attention since dysregulation of its signalling cascades has been suggested to underlie the pathogenesis of both common and rare diseases. Multiple strategies targeting this neurotrophin have been tested; most have found obstacles that ultimately hampered their effectiveness. This review focuses on the involvement of BDNF and its receptors in the pathophysiology of Alzheimer's disease (AD), Amyotrophic Lateral Sclerosis (ALS) and Rett Syndrome (RTT). We describe the known mechanisms leading to the impairment of BDNF/TrkB signalling in these disorders. Such mechanistic insight highlights how BDNF signalling compromise can take various shapes, nearly disease-specific. Therefore, BDNF-based therapeutic strategies must be specifically tailored and are more likely to succeed if a combination of resources is employed.
Collapse
Affiliation(s)
- Catarina Miranda-Lourenço
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Leonor Ribeiro-Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - João Fonseca-Gomes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Sara R Tanqueiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Rita F Belo
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Catarina B Ferreira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Nádia Rei
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Mafalda Ferreira-Manso
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Carolina de Almeida-Borlido
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Tiago Costa-Coelho
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Céline Felicidade Freitas
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Svitlana Zavalko
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Francisco M Mouro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Sara Xapelli
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Tiago M Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Institute of Molecular and Clinical Ophthalmology Basel (IOB), Mittlere Strasse 91, 4031 Basel, Switzerland
| | - Maria J Diógenes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, 1649-028 Lisboa, Portugal.
| |
Collapse
|
29
|
Ciciriello AJ, Smith DR, Munsell MK, Boyd SJ, Shea LD, Dumont CM. Acute Implantation of Aligned Hydrogel Tubes Supports Delayed Spinal Progenitor Implantation. ACS Biomater Sci Eng 2020; 6:5771-5784. [DOI: 10.1021/acsbiomaterials.0c00844] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Andrew J. Ciciriello
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, Florida 33156, United States
- Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, 1951 NW Seventh Avenue Suite 475, Miami, Florida 33136, United States
| | - Dominique R. Smith
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Boulevard, Ann Arbor, Michigan 48109, United States
| | - Mary K. Munsell
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Boulevard, Ann Arbor, Michigan 48109, United States
| | - Sydney J. Boyd
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, Florida 33156, United States
| | - Lonnie D. Shea
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Boulevard, Ann Arbor, Michigan 48109, United States
- Department of Chemical Engineering, University of Michigan, 2300 Hayward Street, Ann Arbor, Michigan 48109, United States
| | - Courtney M. Dumont
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, Florida 33156, United States
- Biomedical Nanotechnology Institute at the University of Miami (BioNIUM), University of Miami, 1951 NW Seventh Avenue Suite 475, Miami, Florida 33136, United States
| |
Collapse
|
30
|
Wu P, Zhao Y, Chen F, Xiao A, Du Q, Dong Q, Ke M, Liang X, Zhou Q, Chen Y. Conductive Hydroxyethyl Cellulose/Soy Protein Isolate/Polyaniline Conduits for Enhancing Peripheral Nerve Regeneration via Electrical Stimulation. Front Bioeng Biotechnol 2020; 8:709. [PMID: 32719783 PMCID: PMC7347754 DOI: 10.3389/fbioe.2020.00709] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/08/2020] [Indexed: 12/15/2022] Open
Abstract
Nerve regeneration remains a challenge to the treatment of peripheral nerve injury. Electrical stimulation (ES) is an assistant treatment to enhance recovery from peripheral nerve injury. A conductive nerve guide conduit was prepared from hydroxyethyl cellulose (HEC)/soy protein isolate (SPI)/PANI sponge (HSPS) and then the HSPS conduits were used to repair 10 mm sciatic nerve injury in rat model with or without ES, using HSPS+brain-derived neurotrophic factor (BDNF) and autografts as controls. The nerve repairing capacities were evaluated by animal experiments of behavioristics, electrophysiology, toluidine blue staining, and transmission electron microscopy (TEM) in the regenerated nerves. The results revealed that the nerve regeneration efficiency of HSPS conduits with ES (HSPS+ES) group was the best among the conduit groups but slightly lower than that of autografts group. HSPS+ES group even exhibited notably increased in the BDNF expression of regenerated nerve tissues, which was also confirmed through in vitro experiments that exogenous BDNF could promote Schwann cells proliferation and MBP protein expression. As a result, this work provided a strategy to repair nerve defect using conductive HSPS as nerve guide conduit and using ES as an extrinsic physical cue to promote the expression of endogenous BDNF.
Collapse
Affiliation(s)
- Ping Wu
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immune Related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Yanan Zhao
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immune Related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Feixiang Chen
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immune Related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Ao Xiao
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immune Related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Qiaoyue Du
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immune Related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Qi Dong
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immune Related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Meifang Ke
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immune Related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Xiao Liang
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immune Related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Qing Zhou
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yun Chen
- Department of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Province Key Laboratory of Allergy and Immune Related Diseases, School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Engineering Center of Natural Polymers-Based Medical Materials, Wuhan University, Wuhan, China
| |
Collapse
|
31
|
Adler AF, Björklund A, Parmar M. Transsynaptic tracing and its emerging use to assess graft-reconstructed neural circuits. Stem Cells 2020; 38:716-726. [PMID: 32101353 DOI: 10.1002/stem.3166] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/20/2020] [Accepted: 02/19/2020] [Indexed: 12/16/2022]
Abstract
Fetal neural progenitor grafts have been evaluated in preclinical animal models of spinal cord injury and Parkinson's disease for decades, but the initial reliance on primary tissue as a cell source limited the scale of their clinical translatability. With the development of robust methods to differentiate human pluripotent stem cells to specific neural subtypes, cell replacement therapy holds renewed promise to treat a variety of neurodegenerative diseases and injuries at scale. As these cell sources are evaluated in preclinical models, new transsynaptic tracing methods are making it possible to study the connectivity between host and graft neurons with greater speed and detail than was previously possible. To date, these studies have revealed that widespread, long-lasting, and anatomically appropriate synaptic contacts are established between host and graft neurons, as well as new aspects of host-graft connectivity which may be relevant to clinical cell replacement therapy. It is not yet clear, however, whether the synaptic connectivity between graft and host neurons is as cell-type specific as it is in the endogenous nervous system, or whether that connectivity is responsible for the functional efficacy of cell replacement therapy. Here, we review evidence suggesting that the new contacts established between host and graft neurons may indeed be cell-type specific, and how transsynaptic tracing can be used in the future to further elucidate the mechanisms of graft-mediated functional recovery in spinal cord injury and Parkinson's disease.
Collapse
Affiliation(s)
- Andrew F Adler
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Anders Björklund
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Malin Parmar
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
32
|
Smith DR, Dumont CM, Park J, Ciciriello AJ, Guo A, Tatineni R, Cummings BJ, Anderson AJ, Shea LD. Polycistronic Delivery of IL-10 and NT-3 Promotes Oligodendrocyte Myelination and Functional Recovery in a Mouse Spinal Cord Injury Model. Tissue Eng Part A 2020; 26:672-682. [PMID: 32000627 DOI: 10.1089/ten.tea.2019.0321] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
One million estimated cases of spinal cord injury (SCI) have been reported in the United States and repairing an injury has constituted a difficult clinical challenge. The complex, dynamic, inhibitory microenvironment postinjury, which is characterized by proinflammatory signaling from invading leukocytes and lack of sufficient factors that promote axonal survival and elongation, limits regeneration. Herein, we investigated the delivery of polycistronic vectors, which have the potential to coexpress factors that target distinct barriers to regeneration, from a multiple channel poly(lactide-co-glycolide) (PLG) bridge to enhance spinal cord regeneration. In this study, we investigated polycistronic delivery of IL-10 that targets proinflammatory signaling, and NT-3 that targets axonal survival and elongation. A significant increase was observed in the density of regenerative macrophages for IL-10+NT-3 condition relative to conditions without IL-10. Furthermore, combined delivery of IL-10+NT-3 produced a significant increase of axonal density and notably myelinated axons compared with all other conditions. A significant increase in functional recovery was observed for IL-10+NT-3 delivery at 12 weeks postinjury that was positively correlated to oligodendrocyte myelinated axon density, suggesting oligodendrocyte-mediated myelination as an important target to improve functional recovery. These results further support the use of multiple channel PLG bridges as a growth supportive substrate and platform to deliver bioactive agents to modulate the SCI microenvironment and promote regeneration and functional recovery. Impact statement Spinal cord injury (SCI) results in a complex microenvironment that contains multiple barriers to regeneration and functional recovery. Multiple factors are necessary to address these barriers to regeneration, and polycistronic lentiviral gene therapy represents a strategy to locally express multiple factors simultaneously. A bicistronic vector encoding IL-10 and NT-3 was delivered from a poly(lactide-co-glycolide) bridge, which provides structural support that guides regeneration, resulting in increased axonal growth, myelination, and subsequent functional recovery. These results demonstrate the opportunity of targeting multiple barriers to SCI regeneration for additive effects.
Collapse
Affiliation(s)
- Dominique R Smith
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Courtney M Dumont
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan.,Department of Biomedical Engineering, University of Miami, Coral Gables, Florida.,Biomedical Nanotechnology Institute at University of Miami (BioNIUM), University of Miami, Miami, Florida
| | - Jonghyuck Park
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky.,Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky
| | - Andrew J Ciciriello
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan.,Department of Biomedical Engineering, University of Miami, Coral Gables, Florida
| | - Amina Guo
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Ravindra Tatineni
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Brian J Cummings
- Institute for Memory Impairments and Neurological Disorders (iMIND), University of California, Irvine, California.,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California.,Department of Anatomy and Neurobiology, University of California, Irvine, California.,Department of Physical Medicine and Rehabilitation, University of California, Irvine, California
| | - Aileen J Anderson
- Institute for Memory Impairments and Neurological Disorders (iMIND), University of California, Irvine, California.,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, California.,Department of Anatomy and Neurobiology, University of California, Irvine, California.,Department of Physical Medicine and Rehabilitation, University of California, Irvine, California
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan.,Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
33
|
Chen X, Wu J, Sun R, Zhao Y, Li Y, Pan J, Chen Y, Wang X. Tubular scaffold with microchannels and an H-shaped lumen loaded with bone marrow stromal cells promotes neuroregeneration and inhibits apoptosis after spinal cord injury. J Tissue Eng Regen Med 2020; 14:397-411. [PMID: 31821733 PMCID: PMC7155140 DOI: 10.1002/term.2996] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 11/13/2019] [Accepted: 11/19/2019] [Indexed: 12/11/2022]
Abstract
As a result of its complex histological structure, regeneration patterns of grey and white matter are quite different in the spinal cord. Therefore, tissue engineering scaffolds for repairing spinal cord injury must be able to adapt to varying neural regeneration patterns. The aim of the present study was to improve a previously reported spinal cord‐mimicking partition‐type scaffold by adding microchannels on a single tubular wall along its longitudinal axis, thus integrating the two architectures of a single H‐shaped central tube and many microchannels. Next, the integrated scaffold was loaded with bone marrow stromal cells (BMSCs) and transplanted to bridge the 5‐mm defect of a complete transverse lesion in the thoracic spinal cord of rats. Subsequently, effects on nerve regeneration, locomotion function recovery, and early neuroprotection were observed. After 1 year of repair, the integrated scaffold could guide the regeneration of axons appearing in the debris of degraded microchannels, especially serotonin receptor 1A receptor‐positive axonal tracts, which were relatively orderly arranged. Moreover, a network of nerve fibres was present, and a few BMSCs expressed neuronal markers in tubular lumens. Functionally, electrophysiological and locomotor functions of rats were partially recovered. In addition, we found that BMSCs could protect neurons and oligodendrocytes from apoptosis during the early stage of implantation. Taken together, our results demonstrate the potential of this novel integrated scaffold loaded with BMSCs to promote spinal cord regeneration through mechanical guidance and neuroprotective mechanisms.
Collapse
Affiliation(s)
- Xue Chen
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China.,Wuxi Medical College, Jiangnan University, Wuxi, Jiangsu, China
| | - Jian Wu
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Rongcheng Sun
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Yahong Zhao
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| | - Yi Li
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Jingying Pan
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Ying Chen
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Xiaodong Wang
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China.,Key Laboratory for Neuroregeneration of Ministry of Education and Co-innovation Center for Neuroregeneration of Jiangsu Province, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
34
|
Smith DR, Dumont CM, Ciciriello AJ, Guo A, Tatineni R, Munsell MK, Cummings BJ, Anderson AJ, Shea LD. PLG Bridge Implantation in Chronic SCI Promotes Axonal Elongation and Myelination. ACS Biomater Sci Eng 2019; 5:6679-6690. [PMID: 33423486 PMCID: PMC11283858 DOI: 10.1021/acsbiomaterials.9b01012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Spinal cord injury (SCI) is a devastating condition that may cause permanent functional loss below the level of injury, including paralysis and loss of bladder, bowel, and sexual function. Patients are rarely treated immediately, and this delay is associated with tissue loss and scar formation that can make regeneration at chronic time points more challenging. Herein, we investigated regeneration using a poly(lactide-co-glycolide) multichannel bridge implanted into a chronic SCI following surgical resection of necrotic tissue. We characterized the dynamic injury response and noted that scar formation decreased at 4 and 8 weeks postinjury (wpi), yet macrophage infiltration increased between 4 and 8 wpi. Subsequently, the scar tissue was resected and bridges were implanted at 4 and 8 wpi. We observed robust axon growth into the bridge and remyelination at 6 months after initial injury. Axon densities were increased for 8 week bridge implantation relative to 4 week bridge implantation, whereas greater myelination, particularly by Schwann cells, was observed with 4 week bridge implantation. The process of bridge implantation did not significantly decrease the postinjury function. Collectively, this chronic model follows the pathophysiology of human SCI, and bridge implantation allows for clear demarcation of the regenerated tissue. These data demonstrate that bridge implantation into chronic SCI supports regeneration and provides a platform to investigate strategies to buttress and expand regeneration of neural tissue at chronic time points.
Collapse
Affiliation(s)
- Dominique R. Smith
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Avenue, Ann Arbor, MI, 48109
| | - Courtney M. Dumont
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, FL, 33156
- Biomedical Nanotechnology Institute at University of Miami (BioNIUM), University of Miami, 1951 NW 7 Avenue Ste 475, Miami, FL, 33136
| | - Andrew J. Ciciriello
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Avenue, Ann Arbor, MI, 48109
- Department of Biomedical Engineering, University of Miami, 1251 Memorial Drive, Coral Gables, FL, 33156
- Biomedical Nanotechnology Institute at University of Miami (BioNIUM), University of Miami, 1951 NW 7 Avenue Ste 475, Miami, FL, 33136
| | - Amina Guo
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Avenue, Ann Arbor, MI, 48109
| | - Ravindra Tatineni
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Avenue, Ann Arbor, MI, 48109
| | - Mary K. Munsell
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Avenue, Ann Arbor, MI, 48109
| | - Brian J. Cummings
- Institute for Memory Impairments and Neurological Disorders (iMIND), University of California, 1100 Gottschalk Medical Plaza, Irvine, CA, 92697
- Sue and Bill Gross Stem Cell Research Center, University of California, 845 Health Science Road, Irvine, CA, 92697
- Department of Anatomy and Neurobiology, University of California, 364 Med Surge II, Irvine, CA, 92697
- Department of Physical Medicine and Rehabilitation, University of California, 101 The City Drive South, Building 53, Suite 311A, Orange, CA, 92868
| | - Aileen J. Anderson
- Institute for Memory Impairments and Neurological Disorders (iMIND), University of California, 1100 Gottschalk Medical Plaza, Irvine, CA, 92697
- Sue and Bill Gross Stem Cell Research Center, University of California, 845 Health Science Road, Irvine, CA, 92697
- Department of Anatomy and Neurobiology, University of California, 364 Med Surge II, Irvine, CA, 92697
- Department of Physical Medicine and Rehabilitation, University of California, 101 The City Drive South, Building 53, Suite 311A, Orange, CA, 92868
| | - Lonnie D. Shea
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Avenue, Ann Arbor, MI, 48109
- Department of Chemical Engineering, University of Michigan, 2300 Hayward Street, Ann Arbor, MI, 48109
| |
Collapse
|
35
|
Song YH, Agrawal NK, Griffin JM, Schmidt CE. Recent advances in nanotherapeutic strategies for spinal cord injury repair. Adv Drug Deliv Rev 2019; 148:38-59. [PMID: 30582938 PMCID: PMC6959132 DOI: 10.1016/j.addr.2018.12.011] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 11/12/2018] [Accepted: 12/17/2018] [Indexed: 12/11/2022]
Abstract
Spinal cord injury (SCI) is a devastating and complicated condition with no cure available. The initial mechanical trauma is followed by a secondary injury characterized by inflammatory cell infiltration and inhibitory glial scar formation. Due to the limitations posed by the blood-spinal cord barrier, systemic delivery of therapeutics is challenging. Recent development of various nanoscale strategies provides exciting and promising new means of treating SCI by crossing the blood-spinal cord barrier and delivering therapeutics. As such, we discuss different nanomaterial fabrication methods and provide an overview of recent studies where nanomaterials were developed to modulate inflammatory signals, target inhibitory factors in the lesion, and promote axonal regeneration after SCI. We also review emerging areas of research such as optogenetics, immunotherapy and CRISPR-mediated genome editing where nanomaterials can provide synergistic effects in developing novel SCI therapy regimens, as well as current efforts and barriers to clinical translation of nanomaterials.
Collapse
Affiliation(s)
- Young Hye Song
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Nikunj K Agrawal
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Jonathan M Griffin
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Christine E Schmidt
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
36
|
Shahriari D, Loke G, Tafel I, Park S, Chiang PH, Fink Y, Anikeeva P. Scalable Fabrication of Porous Microchannel Nerve Guidance Scaffolds with Complex Geometries. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1902021. [PMID: 31168865 PMCID: PMC6663568 DOI: 10.1002/adma.201902021] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 05/15/2019] [Indexed: 05/24/2023]
Abstract
Microchannel scaffolds accelerate nerve repair by guiding growing neuronal processes across injury sites. Although geometry, materials chemistry, stiffness, and porosity have been shown to influence nerve growth within nerve guidance scaffolds, independent tuning of these properties in a high-throughput manner remains a challenge. Here, fiber drawing is combined with salt leaching to produce microchannels with tunable cross sections and porosity. This technique is applicable to an array of biochemically inert polymers, and it delivers hundreds of meters of porous microchannel fibers. Employing these fibers as filaments during 3D printing enables the production of microchannel scaffolds with geometries matching those of biological nerves, including branched topographies. Applied to sensory neurons, fiber-based porous microchannels enhance growth as compared to non-porous channels with matching materials and geometries. The combinatorial scaffold fabrication approach may advance the studies of neural regeneration and accelerate the development of nerve repair devices.
Collapse
Affiliation(s)
- Dena Shahriari
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Gabriel Loke
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Ian Tafel
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Seongjun Park
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Po-Han Chiang
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Yoel Fink
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Advanced Functional Fabrics of America, Cambridge, MA, 02139, USA
| | - Polina Anikeeva
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
37
|
Dumont CM, Carlson MA, Munsell MK, Ciciriello AJ, Strnadova K, Park J, Cummings BJ, Anderson AJ, Shea LD. Aligned hydrogel tubes guide regeneration following spinal cord injury. Acta Biomater 2019; 86:312-322. [PMID: 30610918 DOI: 10.1016/j.actbio.2018.12.052] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 12/22/2018] [Accepted: 12/31/2018] [Indexed: 12/21/2022]
Abstract
Directing the organization of cells into a tissue with defined architectures is one use of biomaterials for regenerative medicine. To this end, hydrogels are widely investigated as they have mechanical properties similar to native soft tissues and can be formed in situ to conform to a defect. Herein, we describe the development of porous hydrogel tubes fabricated through a two-step polymerization process with an intermediate microsphere phase that provides macroscale porosity (66.5%) for cell infiltration. These tubes were investigated in a spinal cord injury model, with the tubes assembled to conform to the injury and to provide an orientation that guides axons through the injury. Implanted tubes had good apposition and were integrated with the host tissue due to cell infiltration, with a transient increase in immune cell infiltration at 1 week that resolved by 2 weeks post injury compared to a gelfoam control. The glial scar was significantly reduced relative to control, which enabled robust axon growth along the inner and outer surface of the tubes. Axon density within the hydrogel tubes (1744 axons/mm2) was significantly increased more than 3-fold compared to the control (456 axons/mm2), with approximately 30% of axons within the tube myelinated. Furthermore, implantation of hydrogel tubes enhanced functional recovery relative to control. This modular assembly of porous tubes to fill a defect and directionally orient tissue growth could be extended beyond spinal cord injury to other tissues, such as vascular or musculoskeletal tissue. STATEMENT OF SIGNIFICANCE: Tissue engineering approaches that mimic the native architecture of healthy tissue are needed following injury. Traditionally, pre-molded scaffolds have been implemented but require a priori knowledge of wound geometries. Conversely, hydrogels can conform to any injury, but do not guide bi-directional regeneration. In this work, we investigate the feasibility of a system of modular hydrogel tubes to promote bi-directional regeneration after spinal cord injury. This system allows for tubes to be cut to size during surgery and implanted one-by-one to fill any injury, while providing bi-directional guidance. Moreover, this system of tubes can be broadly applied to tissue engineering approaches that require a modular guidance system, such as repair to vascular or musculoskeletal tissues.
Collapse
|
38
|
Smith DR, Margul DJ, Dumont CM, Carlson MA, Munsell MK, Johnson M, Cummings BJ, Anderson AJ, Shea LD. Combinatorial lentiviral gene delivery of pro-oligodendrogenic factors for improving myelination of regenerating axons after spinal cord injury. Biotechnol Bioeng 2019; 116:155-167. [PMID: 30229864 PMCID: PMC6289889 DOI: 10.1002/bit.26838] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 08/30/2018] [Accepted: 09/12/2018] [Indexed: 12/12/2022]
Abstract
Spinal cord injury (SCI) results in paralysis below the injury and strategies are being developed that support axonal regrowth, yet recovery lags, in part, because many axons are not remyelinated. Herein, we investigated strategies to increase myelination of regenerating axons by overexpression of platelet-derived growth factor (PDGF)-AA and noggin either alone or in combination in a mouse SCI model. Noggin and PDGF-AA have been identified as factors that enhance recruitment and differentiation of endogenous progenitors to promote myelination. Lentivirus encoding for these factors was delivered from a multichannel bridge, which we have previously shown creates a permissive environment and supports robust axonal growth through channels. The combination of noggin+PDGF enhanced total myelination of regenerating axons relative to either factor alone, and importantly, enhanced functional recovery relative to the control condition. The increase in myelination was consistent with an increase in oligodendrocyte-derived myelin, which was also associated with a greater density of cells of an oligodendroglial lineage relative to each factor individually and control conditions. These results suggest enhanced myelination of regenerating axons by noggin+PDGF that act on oligodendrocyte-lineage cells post-SCI, which ultimately led to improved functional outcomes.
Collapse
Affiliation(s)
- Dominique R. Smith
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Daniel J. Margul
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA
| | - Courtney M. Dumont
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Mitchell A. Carlson
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Mary K. Munsell
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Mitchell Johnson
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Brian J. Cummings
- Institute for Memory Impairments and Neurological Disorders (iMIND), University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
- Department of Physical Medicine and Rehabilitation, University of California, Irvine, CA, USA
| | - Aileen J. Anderson
- Institute for Memory Impairments and Neurological Disorders (iMIND), University of California, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
- Department of Physical Medicine and Rehabilitation, University of California, Irvine, CA, USA
| | - Lonnie D. Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
39
|
Zhu W, Zhang H, Chen X, Jin K, Ning L. Numerical characterization of regenerative axons growing along a spherical multifunctional scaffold after spinal cord injury. PLoS One 2018; 13:e0205961. [PMID: 30365562 PMCID: PMC6203361 DOI: 10.1371/journal.pone.0205961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 10/04/2018] [Indexed: 11/18/2022] Open
Abstract
Spinal cord injury (SCI) followed by extensive cell loss, inflammation, and scarring, often permanently damages neurological function. Biomaterial scaffolds are promising but currently have limited applicability in SCI because after entering the scaffold, regenerating axons tend to become trapped and rarelyre-enter the host tissue, the reasons for which remain to be completely explored. Here, we propose a mathematical model and computer simulation for characterizing regenerative axons growing along a scaffold following SCI, and how their growth may be guided. The model assumed a solid, spherical, multifunctional, biomaterial scaffold, that would bridge the rostral and caudal stumps of a completely transected spinal cord in a rat model and would guide the rostral regenerative axons toward the caudal tissue. Other assumptions include the whole scaffold being coated with extracellular matrix components, and the caudal area being additionally seeded with chemoattractants. The chemical factors on and around the scaffold were formulated to several coupled variables, and the parameter values were derived fromexisting experimental data. Special attention was given to the effects of coating strength, seeding location, and seeding density, as well as the ramp slope of the scaffold, on axonal regeneration. In numerical simulations, a slimmer scaffold provided a small slope at the entry "on-ramp" area that improved the success rate of axonal regeneration. If success rates are high, an increased number of regenerative axons traverse through the narrow channels, causing congestion and lowering the growth rate. An increase in the number of severed axons (300-12000) did not significantly affect the growth rate, but it reduced the success rate of axonal regeneration. However, an increase in the seeding densities of the complexes on the whole scaffold, and that in the seeding densities of the chemoattractants on the caudal area, improved both the success and growth rates. However, an increase in the density of thecomplexes on the whole scaffold risks an over-eutrophic surface that harms axonal regeneration.Although theoretical predictions are yet to be validated directly by experiments, this theoretical tool can advance the treatment of SCI, and is also applicable to scaffolds with other architectures.
Collapse
Affiliation(s)
- Weiping Zhu
- Shanghai Institute of Applied Mathematics and Mechanics, Shanghai University, Shanghai, People's Republic of China
- * E-mail:
| | - Han Zhang
- Shanghai Institute of Applied Mathematics and Mechanics, Shanghai University, Shanghai, People's Republic of China
| | - Xuning Chen
- Shanghai Institute of Applied Mathematics and Mechanics, Shanghai University, Shanghai, People's Republic of China
| | - Kan Jin
- Shanghai Institute of Applied Mathematics and Mechanics, Shanghai University, Shanghai, People's Republic of China
| | - Le Ning
- Shanghai Institute of Applied Mathematics and Mechanics, Shanghai University, Shanghai, People's Republic of China
| |
Collapse
|
40
|
Dumont CM, Munsell MK, Carlson MA, Cummings BJ, Anderson AJ, Shea LD. Spinal Progenitor-Laden Bridges Support Earlier Axon Regeneration Following Spinal Cord Injury. Tissue Eng Part A 2018; 24:1588-1602. [PMID: 30215293 DOI: 10.1089/ten.tea.2018.0053] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
IMPACT STATEMENT Spinal cord injury (SCI) results in loss of tissue innervation below the injury. Spinal progenitors have a greater ability to repair the damage and can be injected into the injury, but their regenerative potential is hampered by their poor survival after transplantation. Biomaterials can create a cell delivery platform and generate a more hospitable microenvironment for the progenitors within the injury. In this work, polymeric bridges are used to deliver embryonic spinal progenitors to the injury, resulting in increased progenitor survival and subsequent regeneration and functional recovery, thus demonstrating the importance of combined therapeutic approaches for SCI.
Collapse
Affiliation(s)
- Courtney M Dumont
- 1 Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan
| | - Mary K Munsell
- 1 Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan
| | - Mitchell A Carlson
- 1 Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan
| | - Brian J Cummings
- 2 Institute for Memory Impairments and Neurological Disorders (iMIND), University of California , Irvine, California.,3 Sue and Bill Gross Stem Cell Research Center, University of California , Irvine, California.,4 Department of Anatomy and Neurobiology and University of California , Irvine, California.,5 Department of Physical Medicine and Rehabilitation, University of California , Irvine, California
| | - Aileen J Anderson
- 2 Institute for Memory Impairments and Neurological Disorders (iMIND), University of California , Irvine, California.,3 Sue and Bill Gross Stem Cell Research Center, University of California , Irvine, California.,4 Department of Anatomy and Neurobiology and University of California , Irvine, California.,5 Department of Physical Medicine and Rehabilitation, University of California , Irvine, California
| | - Lonnie D Shea
- 1 Department of Biomedical Engineering, University of Michigan , Ann Arbor, Michigan.,6 Department of Chemical Engineering, University of Michigan , Ann Arbor, Michigan
| |
Collapse
|
41
|
Park J, Decker JT, Smith DR, Cummings BJ, Anderson AJ, Shea LD. Reducing inflammation through delivery of lentivirus encoding for anti-inflammatory cytokines attenuates neuropathic pain after spinal cord injury. J Control Release 2018; 290:88-101. [PMID: 30296461 DOI: 10.1016/j.jconrel.2018.10.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 09/26/2018] [Accepted: 10/03/2018] [Indexed: 01/21/2023]
Abstract
Recently, many clinical trials have challenged the efficacy of current therapeutics for neuropathic pain after spinal cord injury (SCI) due to their life-threatening side-effects including addictions. Growing evidence suggests that persistent inflammatory responses after primary SCI lead to an imbalance between anti-inflammation and pro-inflammation, resulting in pathogenesis and maintenance of neuropathic pain. Conversely, a variety of data suggest that inflammation contributes to regeneration. Herein, we investigated long-term local immunomodulation using anti-inflammatory cytokine IL-10 or IL-4-encoding lentivirus delivered from multichannel bridges. Multichannel bridges provide guidance for axonal outgrowth and act as delivery vehicles. Anti-inflammatory cytokines were hypothesized to modulate the pro-nociceptive inflammatory niche and promote axonal regeneration, leading to neuropathic pain attenuation. Gene expression analyses demonstrated that IL-10 and IL-4 decreased pro-nociceptive genes expression versus control. Moreover, these factors resulted in an increased number of pro-regenerative macrophages and restoration of normal nociceptors expression pattern. Furthermore, the combination of bridges with anti-inflammatory cytokines significantly alleviated both mechanical and thermal hypersensitivity relative to control and promoted axonal regeneration. Collectively, these studies highlight that immunomodulatory strategies target multiple barriers to decrease secondary inflammation and attenuate neuropathic pain after SCI.
Collapse
Affiliation(s)
- Jonghyuck Park
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Joseph T Decker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Dominique R Smith
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Brian J Cummings
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA; Department of Physical Medicine and Rehabilitation, University of California, Irvine, CA, USA
| | - Aileen J Anderson
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA; Department of Physical Medicine and Rehabilitation, University of California, Irvine, CA, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
42
|
Park J, Decker JT, Margul DJ, Smith DR, Cummings BJ, Anderson AJ, Shea LD. Local Immunomodulation with Anti-inflammatory Cytokine-Encoding Lentivirus Enhances Functional Recovery after Spinal Cord Injury. Mol Ther 2018; 26:1756-1770. [PMID: 29778523 PMCID: PMC6037204 DOI: 10.1016/j.ymthe.2018.04.022] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 04/20/2018] [Accepted: 04/23/2018] [Indexed: 12/18/2022] Open
Abstract
Trauma to the spinal cord and associated secondary inflammation can lead to permanent loss of sensory and motor function below the injury level, with the resulting environment serving as a barrier that limits regeneration. In this study, we investigate the localized expression of anti-inflammatory cytokines IL-10 and IL-4 via lentiviral transduction in multichannel bridges. Porous multichannel bridges provide physical guidance for axonal outgrowth with the cytokines hypothesized to modulate the neuroinflammatory microenvironment and enhance axonal regeneration. Gene expression analyses indicated that induced IL-10 and IL-4 expression decreased expression of pro-inflammatory genes and increased pro-regenerative genes relative to control. Moreover, these factors led to increased numbers of axons and myelination, with approximately 45% of axons myelinated and the number of oligodendrocyte myelinated axons significantly increased by 3- to 4-fold. Furthermore, the combination of a bridge with IL-10 and IL-4 expression improved locomotor function after injury to an average score of 6 relative to an average score of 3 for injury alone. Collectively, these studies highlight the potential for localized immunomodulation to decrease secondary inflammation and enhance regeneration that may have numerous applications.
Collapse
Affiliation(s)
- Jonghyuck Park
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| | - Joseph T Decker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| | - Daniel J Margul
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA; Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Dominique R Smith
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| | - Brian J Cummings
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA 92697, USA
| | - Aileen J Anderson
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA 92697, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48105, USA.
| |
Collapse
|
43
|
Anzalone A, Chacko JV, Nishi RA, Dumont C, Smith D, Shea LD, Digman MA, Cummings BJ, Anderson AJ. Feasibility study on mouse live imaging after spinal cord injury and poly(lactide-co-glycolide) bridge implantation. JOURNAL OF BIOMEDICAL OPTICS 2018; 23:1-6. [PMID: 29959835 PMCID: PMC8357334 DOI: 10.1117/1.jbo.23.6.065007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 06/11/2018] [Indexed: 06/08/2023]
Abstract
Spinal cord injury (SCI) causes permanent paralysis below the damaged area. SCI is linked to neuronal death, demyelination, and limited ability of neuronal fibers to regenerate. Regeneration capacity is limited by the presence of many inhibitory factors in the spinal cord environment. The use of poly(lactide-co-glycolide) (PLG) bridges has demonstrated the ability to sustain long-term regeneration after SCI in a cervical hemisection mouse model. Critically, imaging of regenerating fibers and the myelination status of these neuronal filaments is a severe limitation to progress in SCI research. We used a transgenic mouse model that selectively expresses fluorescent reporters (eGFP) in the neuronal fibers of the spinal cord. We implanted a PLG bridge at C5 vertebra after hemisection and evaluated in live animals' neuronal fibers at the bridge interface and within the bridge 8 weeks postimplant. These in vivo observations were correlated with in situ evaluation 12 weeks postimplantation. We sectioned the spinal cords and performed fluorescent bioimaging on the sections to observe neuronal fibers going through the bridge. In parallel, to visualize myelination of regenerated axons, we exploited the characteristics of the third-harmonic generation arising from the myelin structure in these fixed sections.
Collapse
Affiliation(s)
- Andrea Anzalone
- University of California, Institute for Memory Impairments and Neurological Disorders, Irvine, California, United States
| | - Jenu V. Chacko
- University of California Irvine, Department of Biomedical Engineering, Irvine, California, United States
| | - Rebecca A. Nishi
- University of California, Institute for Memory Impairments and Neurological Disorders, Irvine, California, United States
- University of California, Sue and Bill Gross Stem Cell Research Center, Irvine, California, United States
| | - Courtney Dumont
- University of Michigan, Department of Chemical Engineering, Ann Arbor, Michigan, United States
- University of Michigan, Department of Biomedical Engineering, Ann Arbor, Michigan, United States
| | - Dominique Smith
- University of Michigan, Department of Chemical Engineering, Ann Arbor, Michigan, United States
- University of Michigan, Department of Biomedical Engineering, Ann Arbor, Michigan, United States
| | - Lonnie D. Shea
- University of Michigan, Department of Chemical Engineering, Ann Arbor, Michigan, United States
- University of Michigan, Department of Biomedical Engineering, Ann Arbor, Michigan, United States
| | - Michelle A. Digman
- University of California Irvine, Department of Biomedical Engineering, Irvine, California, United States
- University of California Irvine, Laboratory for Fluorescence Dynamics, Irvine, California, United States
| | - Brian J. Cummings
- University of California, Institute for Memory Impairments and Neurological Disorders, Irvine, California, United States
- University of California, Sue and Bill Gross Stem Cell Research Center, Irvine, California, United States
- University of California, Department of Physical Medicine and Rehabilitation, Irvine, California, United States
- University of California, Department of Anatomy and Neurobiology, Irvine, California, United States
| | - Aileen J. Anderson
- University of California, Institute for Memory Impairments and Neurological Disorders, Irvine, California, United States
- University of California, Sue and Bill Gross Stem Cell Research Center, Irvine, California, United States
- University of California, Department of Physical Medicine and Rehabilitation, Irvine, California, United States
- University of California, Department of Anatomy and Neurobiology, Irvine, California, United States
| |
Collapse
|
44
|
Application of the sodium hyaluronate-CNTF scaffolds in repairing adult rat spinal cord injury and facilitating neural network formation. SCIENCE CHINA-LIFE SCIENCES 2017; 61:559-568. [DOI: 10.1007/s11427-017-9217-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 11/05/2017] [Indexed: 12/23/2022]
|
45
|
Recent advances in regenerative medicine approaches for spinal cord injuries. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2017. [DOI: 10.1016/j.cobme.2017.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
46
|
Zhao Y, Zhang Q, Zhao L, Gan L, Yi L, Zhao Y, Xue J, Luo L, Du Q, Geng R, Sun Z, Benkirane-Jessel N, Chen P, Li Y, Chen Y. Enhanced Peripheral Nerve Regeneration by a High Surface Area to Volume Ratio of Nerve Conduits Fabricated from Hydroxyethyl Cellulose/Soy Protein Composite Sponges. ACS OMEGA 2017; 2:7471-7481. [PMID: 30023554 PMCID: PMC6044839 DOI: 10.1021/acsomega.7b01003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Accepted: 10/19/2017] [Indexed: 05/05/2023]
Abstract
Multichannel nerve guide conduits (MCNGCs) have been widely studied and exhibited outstanding nerve repair function. However, the effect of the geometric structure of MCNGCs on the nerve repair function was still not clear. Herein, we postulated that MCNGCs with different inner surface area-to-volume ratios (ISA/V) of the channels inside the nerve guide conduits (NGCs) would show different nerve repair functions. Therefore, in current work, we constructed a series of hydroxyethyl cellulose/soy protein sponge-based nerve conduit (HSSN) with low, medium, and high ISA/V from hydroxyethyl cellulose (HEC)/soy protein isolate (SPI) composite sponges, which were abbreviated as HSSN-L, HSSN-M and HSSN-H, respectively. These NGCs were applied to bridge and repair a 10 mm long sciatic nerve defect in a rat model. Finally, the influence of ISA/V on nerve repair function was evaluated by electrophysiological assessment, histological investigation, and in vivo biodegradability testing. The results of electrophysiological assessment and histological investigation showed that the regenerative nerve tissues bridged with HSSN-H and HSSN-M had higher compound muscle action potential amplitude ratio, higher percentage of positive NF200 and S100 staining, larger axon diameter, lower G-ratio, and greater myelination thickness. Furthermore, the regenerative nerve tissues bridged with HSSN-H also showed higher density of regenerated myelinated nerve fibers and more number of myelin sheath layers. On the whole, the repair efficiency of the peripheral nerve in HSSN-H and HSSN-M groups might be better than that in HSSN-L. These results indicated that higher ISA/V based on HEC/SPI composite sponge may result in greater nerve repair functions. The conclusion provided a probable guiding principle for the structural designs of NGCs in the future.
Collapse
Affiliation(s)
- Yanteng Zhao
- Department
of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
- Department
of Transfusion, The First Affiliated Hospital
of Zhengzhou University, Zhengzhou 450052, China
| | - Qiang Zhang
- Department
of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Lei Zhao
- Department
of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Li Gan
- Department
of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
- Department
of Cell Biology, School of Medicine, Wuhan
University of Science and Technology, Wuhan 430065, China
| | - Li Yi
- Department
of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Yanan Zhao
- Department
of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Jingling Xue
- Department
of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Lihua Luo
- Laboratory
of Stem Cells and Tissue Engineering, School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China
| | - Qiaoyue Du
- Department
of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Rongxin Geng
- Department
of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Zhihong Sun
- Department
of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Nadia Benkirane-Jessel
- INSERM
(French National Institute of Health and Medical Research), Osteoarticular
and Dental Regenerative Nanomedicine Laboratory, UMR 1109, Faculté
de Médecine, Strasbourg F-67000, France
- Université
de Strasbourg, Faculté de Chirurgie Dentaire, 1 place de l’Hôpital, Strasbourg F-67000, France
| | - Pu Chen
- Department
of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Yinping Li
- Department
of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
- E-mail: (Y.L.)
| | - Yun Chen
- Department
of Biomedical Engineering, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
- E-mail: (Y.C.)
| |
Collapse
|
47
|
Mu XP, Wang HB, Cheng X, Yang L, Sun XY, Qu HL, Zhao SS, Zhou ZK, Liu TT, Xiao T, Song B, Jolkkonen J, Zhao CS. Inhibition of Nkcc1 promotes axonal growth and motor recovery in ischemic rats. Neuroscience 2017; 365:83-93. [PMID: 28964752 DOI: 10.1016/j.neuroscience.2017.09.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 09/03/2017] [Accepted: 09/21/2017] [Indexed: 12/15/2022]
Abstract
Bumetanide is a selective inhibitor of the Na+-K+-Cl--co-transporter 1(NKCC1). We studied whether bumetanide could affect axonal growth and behavioral outcome in stroke rats. Adult male Wistar rats were randomly assigned to four groups: sham-operated rats treated with vehicle or bumetanide, and ischemic rats treated with vehicle or bumetanide. Endothelin-1 was used to induce focal cerebral ischemia. Bumetanide administration (i.c.v.) started on postoperative day 7 and continued for 3 weeks. Biotinylated dextran amine (BDA) was injected into the right imotor cortex on postoperative day 14 to trace corticospinal tract (CST) fibers sprouting into the denervated cervical spinal cord. Nogo-A, NKCC1, KCC2 and BDNF in the perilesional cortex and BDA, PSD-95 and vGlut1 in the denervated spinal cord were measured by immunohistochemistry and/or Western blot. Behavioral outcome of rats was assessed by the beam walking and cylinder tests. The total length of CST fibers sprouting into the denervated cervical spinal cord significantly increased after stroke and bumetanide further increased this sprouting. Bumetanide treatment also decreased the expressions of NKCC1 and Nogo-A, increased the expressions of KCC2 and BDNF in the perilesional cortex and enhanced the synaptic plasticity in the denervated cervical spinal cord after cerebral ischemia. The behavioral performance of ischemic rats was significantly improved by bumetanide. In conclusion, bumetanide promoted post-stroke axonal sprouting together accompanied by an improved behavioral outcome possibly through restoring and maintaining neuronal chloride homeostasis and creating a recovery-promoting microenvironment by overcoming the axonal growth inhibition encountered after cerebral ischemia in rats.
Collapse
Affiliation(s)
- X P Mu
- Department of Neurology, The First Affiliated Hospital, China Medical University, Shenyang, China; Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - H B Wang
- Department of Neurology, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - X Cheng
- Department of Neurology, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - L Yang
- Department of Cardiology, The Affiliated Center Hospital, Shenyang Medical College, Shenyang, China
| | - X Y Sun
- Department of Neurology, The People's Hospital of Liaoning Province, Shenyang, China
| | - H L Qu
- Department of Neurology, The People's Hospital of Liaoning Province, Shenyang, China
| | - S S Zhao
- Department of Neurology, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - Z K Zhou
- Department of Geriatrics, The First Affiliated Hospital, China Medical University, Shenyang, China
| | - T T Liu
- Department of Neurology, The People's Hospital of Liaoning Province, Shenyang, China
| | - T Xiao
- Department of Dermatology, The First Affiliated Hospital, China Medical University, Shenyang, China; Key Laboratory of Immunodermatology, Ministry of Health, Ministry of Education, Shenyang, China
| | - B Song
- Regenerative Medicine, Cardiff Institute of Tissue Engineering and Repair, School of Dentistry, Cardiff University, Cardiff, UK
| | - J Jolkkonen
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, P. O. Box 1627, 70211 Kuopio, Finland
| | - C S Zhao
- Department of Neurology, The First Affiliated Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
48
|
Madrigal JL, Stilhano R, Silva EA. Biomaterial-Guided Gene Delivery for Musculoskeletal Tissue Repair. TISSUE ENGINEERING. PART B, REVIEWS 2017; 23:347-361. [PMID: 28166711 PMCID: PMC5749599 DOI: 10.1089/ten.teb.2016.0462] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/11/2017] [Indexed: 02/07/2023]
Abstract
Gene therapy is a promising strategy for musculoskeletal tissue repair and regeneration where local and sustained expression of proteins and/or therapeutic nucleic acids can be achieved. However, the musculoskeletal tissues present unique engineering and biological challenges as recipients of genetic vectors. Targeting specific cell populations, regulating expression in vivo, and overcoming the harsh environment of damaged tissue accompany the general concerns of safety and efficacy common to all applications of gene therapy. In this review, we will first summarize these challenges and then discuss how biomaterial carriers for genetic vectors can address these issues. Second, we will review how limitations specific to given vectors further motivate the utility of biomaterial carriers. Finally, we will discuss how these concepts have been combined with tissue engineering strategies and approaches to improve the delivery of these vectors for musculoskeletal tissue regeneration.
Collapse
Affiliation(s)
- Justin L Madrigal
- Department of Biomedical Engineering, University of California , Davis, Davis, California
| | - Roberta Stilhano
- Department of Biomedical Engineering, University of California , Davis, Davis, California
| | - Eduardo A Silva
- Department of Biomedical Engineering, University of California , Davis, Davis, California
| |
Collapse
|
49
|
Tu WZ, Jiang SH, Zhang L, Li SS, Gu PP, He R, Hu J, Gao LP, Sun QS. Electro-acupuncture at Governor Vessel improves neurological function in rats with spinal cord injury. Chin J Integr Med 2017:10.1007/s11655-017-2968-9. [PMID: 28762132 DOI: 10.1007/s11655-017-2968-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To determine the effects of electro-acupuncture (EA) at Governor Vessel (GV) on the locomotor function in spinal cord injury (SCI) rats and explore the underlying mechanism. METHODS Thirtytwo male Sprague-Dawley rats were randomly divided into four groups namely: the sham group (with sham operation); the untreated group (without treatment after spinal cord impact); the EA-1 group [EA applied at Baihui (GV 20) and Fengfu (GV 16) after spinal cord impact] and the EA-2 group [with EA applied at Dazhui (GV 14) and Mingmen (GV 4) after spinal cord impact]. Real-time quantitative-polymerase chain reaction (RT-PCR) and Western Blotting were used to assess changes in the mRNA and protein expression levels of brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3) at 7 weeks following EA administration. In addition, the Basso-Beattie-Bresnahan (BBB) Locomotor Rating Scale was assessed at 1 day, 1 week, 3 weeks and 7 weeks post-injury. RESULTS The results showed that EA stimulation induced neuroprotective effects after SCI correlated with the up-regulation of BDNF and NT-3 (P<0.05). Furthermore, EA stimulation at GV 14 and GV 4 could significantly promote the recovery of locomotor function and this may be linked to the up-regulation of BDNF and NT-3 (P<0.05). CONCLUSIONS EA treatment applied at GV acupoints either within the injury site or adjacent undamaged regions near the brain can improve functional recovery, which may be correlated with the upregulation of BDNF and NT-3. In addition, it would be more effective to administer EA at GV 14 and GV 4 near the injury site of the SCI rats.
Collapse
Affiliation(s)
| | - Song-He Jiang
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Li Zhang
- Department of Rehabilitation, Dongyang People's Hospital, Dongyang 322100, Zhejiang Province, China
| | - Si-Si Li
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Peng-Peng Gu
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Rong He
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Jie Hu
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Li-Ping Gao
- Department of Physical Medicine and Rehabilitation, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, Zhejiang Province, China
| | - Qiang-San Sun
- Department of Rehabilitation Medicine, The Second Hospital of Shandong University, Jinan, 250033, China.
| |
Collapse
|
50
|
Nejati-Koshki K, Mortazavi Y, Pilehvar-Soltanahmadi Y, Sheoran S, Zarghami N. An update on application of nanotechnology and stem cells in spinal cord injury regeneration. Biomed Pharmacother 2017; 90:85-92. [DOI: 10.1016/j.biopha.2017.03.035] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 03/12/2017] [Accepted: 03/14/2017] [Indexed: 02/08/2023] Open
|