1
|
Saksena J, Hamilton AE, Gilbert RJ, Zuidema JM. Nanomaterial payload delivery to central nervous system glia for neural protection and repair. Front Cell Neurosci 2023; 17:1266019. [PMID: 37941607 PMCID: PMC10628439 DOI: 10.3389/fncel.2023.1266019] [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: 07/24/2023] [Accepted: 10/06/2023] [Indexed: 11/10/2023] Open
Abstract
Central nervous system (CNS) glia, including astrocytes, microglia, and oligodendrocytes, play prominent roles in traumatic injury and degenerative disorders. Due to their importance, active pharmaceutical ingredients (APIs) are being developed to modulate CNS glia in order to improve outcomes in traumatic injury and disease. While many of these APIs show promise in vitro, the majority of APIs that are systemically delivered show little penetration through the blood-brain barrier (BBB) or blood-spinal cord barrier (BSCB) and into the CNS, rendering them ineffective. Novel nanomaterials are being developed to deliver APIs into the CNS to modulate glial responses and improve outcomes in injury and disease. Nanomaterials are attractive options as therapies for central nervous system protection and repair in degenerative disorders and traumatic injury due to their intrinsic capabilities in API delivery. Nanomaterials can improve API accumulation in the CNS by increasing permeation through the BBB of systemically delivered APIs, extending the timeline of API release, and interacting biophysically with CNS cell populations due to their mechanical properties and nanoscale architectures. In this review, we present the recent advances in the fields of both locally implanted nanomaterials and systemically administered nanoparticles developed for the delivery of APIs to the CNS that modulate glial activity as a strategy to improve outcomes in traumatic injury and disease. We identify current research gaps and discuss potential developments in the field that will continue to translate the use of glia-targeting nanomaterials to the clinic.
Collapse
Affiliation(s)
- Jayant Saksena
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
- Department of Materials Science and Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
| | - Adelle E. Hamilton
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
| | - Ryan J. Gilbert
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
- Albany Stratton Veterans Affairs Medical Center, Albany, NY, United States
| | - Jonathan M. Zuidema
- Department of Biochemistry and Molecular Pharmacology, Mario Negri Institute for Pharmacological Research IRCCS, Milan, Italy
| |
Collapse
|
2
|
Huang D, Bao H, Wu J, Zhuge Q, Yang J, Ye S. Overexpression of NT3 P75-2 gene modified bone marrow mesenchymal stem cells supernatant promotes neurological function recovery in ICH rats. Neurosci Lett 2023; 796:137067. [PMID: 36641043 DOI: 10.1016/j.neulet.2023.137067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/04/2023] [Accepted: 01/10/2023] [Indexed: 01/13/2023]
Abstract
Intracerebral hemorrhage (ICH) is an acute cerebrovascular disease with high mortality and long-term disability rates. Stem cell transplantation and neurotrophic factor therapy have shown great potential in ICH. It has been established that mutated NT3 (NT3P75 - 2) can enhance the positive biological functions of NT3 by decreasing its affinity to the P75-2 receptor. The present study aimed to explore whether NT3P75-2 could further improve neurological recovery after ICH. First, we constructed three stable BMSC cell lines (GFP, GFP-NT3 overexpressed and GFP-NT3P75 - 2 overexpressed) by lentivirus infection. Next, rats were injected with fresh supernatants of these three cell lines on days 1 (24 h) and 3 (72 h) post-ICH induction. Behavioral evaluations were conducted to assess the neurological recovery of ICH rats. We further evaluated changes in microglia activation, neuron survival and proliferation of neural stem cells. Compared with the GFP group and the GFP-NT3 group, animals in the GFP-NT3P75 - 2 group exhibited better motor function improvements and milder neuroinflammation response. Meanwhile, overexpression of NT3P75 - 2 significantly decreased neuronal apoptosis and increased number of SOX2 - positive cells. Taken together, our study demonstrated that early administration of NT3P75 - 2 enriched BMMSC supernatants significantly enhanced neuro-functional recovery after ICH by regulating neuroinflammation response, neuronal survival and increasing neural stem cell number, providing a new therapeutic strategy and direction for early treatment of ICH.
Collapse
Affiliation(s)
- Dongdong Huang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Han Bao
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Jian Wu
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Qichuan Zhuge
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Jianjing Yang
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Sheng Ye
- Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
3
|
Hall A, Fortino T, Spruance V, Niceforo A, Harrop JS, Phelps PE, Priest CA, Zholudeva LV, Lane MA. Cell transplantation to repair the injured spinal cord. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 166:79-158. [PMID: 36424097 PMCID: PMC10008620 DOI: 10.1016/bs.irn.2022.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Adam Hall
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States
| | - Tara Fortino
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States
| | - Victoria Spruance
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States; Division of Kidney, Urologic, & Hematologic Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Alessia Niceforo
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States
| | - James S Harrop
- Department of Neurological and Orthopedic Surgery, Thomas Jefferson University, Philadelphia, PA, United States
| | - Patricia E Phelps
- Department of Integrative Biology & Physiology, UCLA, Los Angeles, CA, United States
| | | | - Lyandysha V Zholudeva
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States; Gladstone Institutes, San Francisco, CA, United States
| | - Michael A Lane
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States.
| |
Collapse
|
4
|
Zastosowanie fibryny w inżynierii tkankowej. Osiągnięcia i perspektywy. POSTEP HIG MED DOSW 2021. [DOI: 10.2478/ahem-2021-0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstrakt
W ostatnich latach istotnym obszarem zastosowania fibryny stała się inżynieria tkankowa, w której wykorzystuje się naturalne właściwości biostatyczne i bioaktywne fibryny, a także możliwość pułapkowania i wiązania w jej strukturze czynników wzrostu. Fibryna jest najczęściej stosowana w postaci żeli i dysków. Jednak każda postać wskutek pochłaniania wody docelowo przyjmuje postać żelu. Białko to w warunkach in vivo spełnia rolę rusztowania dla komórek, a także może być aplikowane w miejsca trudno dostępne – może wypełniać ubytki tkanek i podtrzymywać tkanki okalające, zapobiegając ich zapadaniu się. Ponadto fibryna hamuje krwawienie i inicjuje proces odnowy, jak również pełni rolę stymulatora wzrostu komórek. Przez modyfikacje struktury fibryny cząsteczkami adhezyjnymi, można przyspieszyć odbudowę prawidłowej struktury tkanek. Jej właściwości strukturalne mogą być także wykorzystywane jako rezerwuar czynników wzrostu i system ich przedłużonego uwalniania. Fibryna jest materiałem biodegradowalnym, umożliwiając skorelowanie ubytku matrycy fibrynowej z odbudową tkanek własnych pacjenta. Wprowadzenie metod druku 3D i elektroprzędzenia umożliwia formulację dopasowanych do uszkodzeń kształtek oraz włóknin bez utraty bioaktywnych funkcji fibryny. Metody te umożliwiają także poprawę właściwości mechanicznych przez otrzymywanie m.in. włóknin fibryny z innymi polimerami, co jest szczególnie uzasadnione w przypadku materiałów stosowanych w odbudowie takich struktur jak ścięgna czy kości. Biotechnologiczna synteza fibrynogenu może w przyszłości uniezależnić pozyskiwanie go z krwi i zwiększyć popularność wyrobów medycznych otrzymywanych z fibryny.
Collapse
|
5
|
Moysidou CM, Barberio C, Owens RM. Advances in Engineering Human Tissue Models. Front Bioeng Biotechnol 2021; 8:620962. [PMID: 33585419 PMCID: PMC7877542 DOI: 10.3389/fbioe.2020.620962] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 12/22/2020] [Indexed: 12/11/2022] Open
Abstract
Research in cell biology greatly relies on cell-based in vitro assays and models that facilitate the investigation and understanding of specific biological events and processes under different conditions. The quality of such experimental models and particularly the level at which they represent cell behavior in the native tissue, is of critical importance for our understanding of cell interactions within tissues and organs. Conventionally, in vitro models are based on experimental manipulation of mammalian cells, grown as monolayers on flat, two-dimensional (2D) substrates. Despite the amazing progress and discoveries achieved with flat biology models, our ability to translate biological insights has been limited, since the 2D environment does not reflect the physiological behavior of cells in real tissues. Advances in 3D cell biology and engineering have led to the development of a new generation of cell culture formats that can better recapitulate the in vivo microenvironment, allowing us to examine cells and their interactions in a more biomimetic context. Modern biomedical research has at its disposal novel technological approaches that promote development of more sophisticated and robust tissue engineering in vitro models, including scaffold- or hydrogel-based formats, organotypic cultures, and organs-on-chips. Even though such systems are necessarily simplified to capture a particular range of physiology, their ability to model specific processes of human biology is greatly valued for their potential to close the gap between conventional animal studies and human (patho-) physiology. Here, we review recent advances in 3D biomimetic cultures, focusing on the technological bricks available to develop more physiologically relevant in vitro models of human tissues. By highlighting applications and examples of several physiological and disease models, we identify the limitations and challenges which the field needs to address in order to more effectively incorporate synthetic biomimetic culture platforms into biomedical research.
Collapse
Affiliation(s)
| | | | - Róisín Meabh Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
6
|
Abstract
Spinal cord injury results in significant loss of motor, sensory, and autonomic functions. Although a wide range of therapeutic agents have been shown to attenuate secondary injury or promote regeneration/repair in animal models of spinal cord injury, clinical translation of these strategies has been limited, in part due to difficulty in safely and effectively achieving therapeutic concentrations in the injured spinal cord tissue. Hydrogel-based drug delivery systems offer unique opportunities to locally deliver drugs to the injured spinal cord with sufficient dose and duration, while avoiding deleterious side effects associated with systemic drug administration. Such local drug delivery systems can be readily fabricated from biocompatible and biodegradable materials. In this review, hydrogel-based strategies for local drug delivery to the injured spinal cord are extensively reviewed, and recommendations are made for implementation.
Collapse
Affiliation(s)
- Robert B Shultz
- School of Biomedical Engineering, Science and Health Systems, Drexel University; Department of Neurosurgery; Department of Bioengineering, University of Pennsylvania; New Jersey Center for Biomaterials, Rutgers - The State University of New Jersey, Piscataway, NJ; Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, PA, USA
| | - Yinghui Zhong
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| |
Collapse
|
7
|
Kastania G, Campbell J, Mitford J, Volodkin D. Polyelectrolyte Multilayer Capsule (PEMC)-Based Scaffolds for Tissue Engineering. MICROMACHINES 2020; 11:E797. [PMID: 32842692 PMCID: PMC7570195 DOI: 10.3390/mi11090797] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/20/2020] [Accepted: 08/21/2020] [Indexed: 12/22/2022]
Abstract
Tissue engineering (TE) is a highly multidisciplinary field that focuses on novel regenerative treatments and seeks to tackle problems relating to tissue growth both in vitro and in vivo. These issues currently involve the replacement and regeneration of defective tissues, as well as drug testing and other related bioapplications. The key approach in TE is to employ artificial structures (scaffolds) to support tissue development; these constructs should be capable of hosting, protecting and releasing bioactives that guide cellular behaviour. A straightforward approach to integrating bioactives into the scaffolds is discussed utilising polyelectrolyte multilayer capsules (PEMCs). Herein, this review illustrates the recent progress in the use of CaCO3 vaterite-templated PEMCs for the fabrication of functional scaffolds for TE applications, including bone TE as one of the main targets of PEMCs. Approaches for PEMC integration into scaffolds is addressed, taking into account the formulation, advantages, and disadvantages of such PEMCs, together with future perspectives of such architectures.
Collapse
Affiliation(s)
| | | | | | - Dmitry Volodkin
- School of Science and Technology, Department of Chemistry and Forensics, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK; (G.K.); (J.C.); (J.M.)
| |
Collapse
|
8
|
Biancotti JC, Walker KA, Jiang G, Di Bernardo J, Shea LD, Kunisaki SM. Hydrogel and neural progenitor cell delivery supports organotypic fetal spinal cord development in an ex vivo model of prenatal spina bifida repair. J Tissue Eng 2020; 11:2041731420943833. [PMID: 32782773 PMCID: PMC7383650 DOI: 10.1177/2041731420943833] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 06/29/2020] [Indexed: 12/13/2022] Open
Abstract
Studying how the fetal spinal cord regenerates in an ex vivo model of spina bifida repair may provide insights into the development of new tissue engineering treatment strategies to better optimize neurologic function in affected patients. Here, we developed hydrogel surgical patches designed for prenatal repair of myelomeningocele defects and demonstrated viability of both human and rat neural progenitor donor cells within this three-dimensional scaffold microenvironment. We then established an organotypic slice culture model using transverse lumbar spinal cord slices harvested from retinoic acid–exposed fetal rats to study the effect of fibrin hydrogel patches ex vivo. Based on histology, immunohistochemistry, gene expression, and enzyme-linked immunoabsorbent assays, these experiments demonstrate the biocompatibility of fibrin hydrogel patches on the fetal spinal cord and suggest this organotypic slice culture system as a useful platform for evaluating mechanisms of damage and repair in children with neural tube defects.
Collapse
Affiliation(s)
- Juan C Biancotti
- Division of General Pediatric Surgery, Department of Surgery, Johns Hopkins University, Baltimore, MD, USA
| | - Kendal A Walker
- Section of Pediatric Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Guihua Jiang
- Section of Pediatric Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Julie Di Bernardo
- Section of Pediatric Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Shaun M Kunisaki
- Division of General Pediatric Surgery, Department of Surgery, Johns Hopkins University, Baltimore, MD, USA.,Fetal Program, Johns Hopkins Children's Center, Baltimore, MD, USA
| |
Collapse
|
9
|
Jose G, Shalumon K, Chen JP. Natural Polymers Based Hydrogels for Cell Culture Applications. Curr Med Chem 2020; 27:2734-2776. [DOI: 10.2174/0929867326666190903113004] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 08/15/2019] [Accepted: 08/20/2019] [Indexed: 02/06/2023]
Abstract
It is well known that the extracellular matrix (ECM) plays a vital role in the growth, survival
and differentiation of cells. Though two-dimensional (2D) materials are generally used as substrates for
the standard in vitro experiments, their mechanical, structural, and compositional characteristics can
alter cell functions drastically. Many scientists reported that cells behave more natively when cultured
in three-dimensional (3D) environments than on 2D substrates, due to the more in vivo-like 3D cell
culture environment that can better mimic the biochemical and mechanical properties of the ECM. In
this regard, water-swollen network polymer-based materials called hydrogels are highly attractive for
developing 3D ECM analogs due to their biocompatibility and hydrophilicity. Since hydrogels can be
tuned and altered systematically, these materials can function actively in a defined culture medium to
support long-term self-renewal of various cells. The physico-chemical and biological properties of the
materials used for developing hydrogel should be tunable in accordance with culture needs. Various
types of hydrogels derived either from natural or synthetic origins are currently being used for cell culture
applications. In this review, we present an overview of various hydrogels based on natural polymers
that can be used for cell culture, irrespective of types of applications. We also explain how each
hydrogel is made, its source, pros and cons in biological applications with a special focus on regenerative
engineering.
Collapse
Affiliation(s)
- Gils Jose
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - K.T. Shalumon
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan
| |
Collapse
|
10
|
Lenzini S, Devine D, Shin JW. Leveraging Biomaterial Mechanics to Improve Pluripotent Stem Cell Applications for Tissue Engineering. Front Bioeng Biotechnol 2019; 7:260. [PMID: 31649928 PMCID: PMC6795675 DOI: 10.3389/fbioe.2019.00260] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 09/26/2019] [Indexed: 12/31/2022] Open
Abstract
A primary goal in tissue engineering is to develop functional tissues by recapitulating salient features of complex biological systems that exhibit a diverse range of physical forces. Induced pluripotent stem cells (iPSCs) are promising autologous cell sources to execute these developmental programs and their functions; however, cells require an extracellular environment where they will sense and respond to mechanical forces. Thus, understanding the biophysical relationships between stem cells and their extracellular environments will improve the ability to design complex biological systems through tissue engineering. This article first describes how the mechanical properties of the environment are important determinants of developmental processes, and then further details how biomaterials can be designed to precisely control the mechanics of cell-matrix interactions in order to study and define their reprogramming, self-renewal, differentiation, and morphogenesis. Finally, a perspective is presented on how insights from the mechanics of cell-matrix interactions can be leveraged to control pluripotent stem cells for tissue engineering applications.
Collapse
Affiliation(s)
- Stephen Lenzini
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, United States
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, United States
| | - Daniel Devine
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, United States
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, United States
| | - Jae-Won Shin
- Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, United States
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
11
|
Kratochvil MJ, Seymour AJ, Li TL, Paşca SP, Kuo CJ, Heilshorn SC. Engineered materials for organoid systems. NATURE REVIEWS. MATERIALS 2019; 4:606-622. [PMID: 33552558 PMCID: PMC7864216 DOI: 10.1038/s41578-019-0129-9] [Citation(s) in RCA: 223] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/04/2019] [Indexed: 04/14/2023]
Abstract
Organoids are 3D cell culture systems that mimic some of the structural and functional characteristics of an organ. Organoid cultures provide the opportunity to study organ-level biology in models that mimic human physiology more closely than 2D cell culture systems or non-primate animal models. Many organoid cultures rely on decellularized extracellular matrices as scaffolds, which are often poorly chemically defined and allow only limited tunability and reproducibility. By contrast, the biochemical and biophysical properties of engineered matrices can be tuned and optimized to support the development and maturation of organoid cultures. In this Review, we highlight how key cell-matrix interactions guiding stem-cell decisions can inform the design of biomaterials for the reproducible generation and control of organoid cultures. We survey natural, synthetic and protein-engineered hydrogels for their applicability to different organoid systems and discuss biochemical and mechanical material properties relevant for organoid formation. Finally, dynamic and cell-responsive material systems are investigated for their future use in organoid research.
Collapse
Affiliation(s)
- Michael J. Kratochvil
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Alexis J. Seymour
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Thomas L. Li
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Sergiu P. Paşca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Calvin J. Kuo
- Division of Hematology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Sarah C. Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| |
Collapse
|
12
|
de la Vega L, Lee C, Sharma R, Amereh M, Willerth SM. 3D bioprinting models of neural tissues: The current state of the field and future directions. Brain Res Bull 2019; 150:240-249. [DOI: 10.1016/j.brainresbull.2019.06.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 05/30/2019] [Accepted: 06/06/2019] [Indexed: 01/01/2023]
|
13
|
Xing H, Ren X, Yin H, Sun C, Jiang T. Construction of a NT-3 sustained-release system cross-linked with an acellular spinal cord scaffold and its effects on differentiation of cultured bone marrow mesenchymal stem cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 104:109902. [PMID: 31500033 DOI: 10.1016/j.msec.2019.109902] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 05/28/2019] [Accepted: 06/16/2019] [Indexed: 12/24/2022]
Abstract
OBJECTIVE This study sought to promote the adhesion, proliferation and differentiation of rat bone marrow mesenchymal stem cells by constructing a neurotrophin-3 (NT-3) sustained-release system cross-linked with an acellular spinal cord scaffold. METHODS 1-Ethyl-3-[3-dimethylaminopropyl] carbodiimide hydrochloride (EDC) chemistry combined with chemical extraction was used to construct an acellular spinal cord scaffold. The decellularization completion was validated. An EDC cross-linking method was used to construct the NT-3 cross-linked acellular spinal scaffold. ELISA was used to verify sustained release of NT-3; the dorsal root ganglion method was used to verify the biological activity of the sustained-release NT-3. DAPI staining was used to confirm the adhesion of the cultured rat bone marrow mesenchymal stem cells (P3) to the NT-3 scaffold, and cell counting kit-8 (CCK-8) analysis was used to verify the cellular proliferation after 24 h and 48 h of culture. Immunohistochemistry was used to confirm the differentiation of the bone marrow cells into neuron-like cells. RESULTS An NT-3 sustained-release system cross-linked to an acellular spinal cord scaffold was successfully constructed. Sustained-release NT-3 could persist for 35 days and had biological activity for at least 21 days. It could promote the adhesion, proliferation and differentiation of rat bone marrow mesenchymal stem cells. CONCLUSION As a composite scaffold, an NT-3 sustained-release system cross-linked with an acellular spinal cord scaffold has potential applications for tissue engineering.
Collapse
Affiliation(s)
- Hui Xing
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China
| | - Xianjun Ren
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China
| | - Hong Yin
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China
| | - Chao Sun
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China
| | - Tao Jiang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, The Third Military Medical University, Chongqing 400037, PR China.
| |
Collapse
|
14
|
Willerth SM, Sakiyama-Elbert SE. Combining Stem Cells and Biomaterial Scaffolds for Constructing Tissues and Cell Delivery. ACTA ACUST UNITED AC 2019. [DOI: 10.3233/stj-180001] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Combining stem cells with biomaterial scaffolds serves as a promising strategy for engineering tissues for both in vitro and in vivo applications. This updated review details commonly used biomaterial scaffolds for engineering tissues from stem cells. We first define the different types of stem cells and their relevant properties and commonly used scaffold formulations. Next, we discuss natural and synthetic scaffold materials typically used when engineering tissues, along with their associated advantages and drawbacks and gives examples of target applications. New approaches to engineering tissues, such as 3D bioprinting, are described as they provide exciting opportunities for future work along with current challenges that must be addressed. Thus, this review provides an overview of the available biomaterials for directing stem cell differentiation as a means of producing replacements for diseased or damaged tissues.
Collapse
Affiliation(s)
- Stephanie M. Willerth
- Department of Mechanical Engineering, University of Victoria, VIC, Canada
- Division of Medical Sciences, University of Victoria, VIC, Canada
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada
| | | |
Collapse
|
15
|
Modulation of cell-cell interactions for neural tissue engineering: Potential therapeutic applications of cell adhesion molecules in nerve regeneration. Biomaterials 2019; 197:327-344. [DOI: 10.1016/j.biomaterials.2019.01.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/08/2018] [Accepted: 01/20/2019] [Indexed: 12/21/2022]
|
16
|
Singh A, Yadav CB, Tabassum N, Bajpeyee AK, Verma V. Stem cell niche: Dynamic neighbor of stem cells. Eur J Cell Biol 2018; 98:65-73. [PMID: 30563738 DOI: 10.1016/j.ejcb.2018.12.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 10/09/2018] [Accepted: 12/11/2018] [Indexed: 12/19/2022] Open
Abstract
Stem cell niche is a specialized and dynamic microenvironment around the stem cells which plays a critical role in maintaining the stemness properties of stem cells. Over the years, advancement in the research activity has revealed the various important aspects of stem cell niche including cell-cell interaction, cell-extracellular matrix interaction, a large number of soluble signaling factors and various biochemical and biophysical cues (such as oxygen tension, flow, and shear and pore size). Stem cells have the potential to be a powerful tool in regenerative medicine due to their self-renewal property and immense differentiation potential. Recent progresses in in vitro culture conditions of embryonic stem cells, adult stem cells and induced pluripotent stem cells have enabled the researchers to investigate and understand the role of the microenvironment in stem cell properties. The engineered artificial stem cell niche has led to a better execution of stem cells in regenerative medicine. Here we elucidate the key components of stem cell niche and their role in niche engineering and stem cell therapeutics.
Collapse
Affiliation(s)
- Anshuman Singh
- Centre of Biotechnology, Nehru Science Complex, University of Allahabad, Allahabad, India
| | - C B Yadav
- Centre of Biotechnology, Nehru Science Complex, University of Allahabad, Allahabad, India
| | - N Tabassum
- Centre of Biotechnology, Nehru Science Complex, University of Allahabad, Allahabad, India
| | - A K Bajpeyee
- Centre of Biotechnology, Nehru Science Complex, University of Allahabad, Allahabad, India
| | - V Verma
- Centre of Biotechnology, Nehru Science Complex, University of Allahabad, Allahabad, India.
| |
Collapse
|
17
|
Heher P, Mühleder S, Mittermayr R, Redl H, Slezak P. Fibrin-based delivery strategies for acute and chronic wound healing. Adv Drug Deliv Rev 2018; 129:134-147. [PMID: 29247766 DOI: 10.1016/j.addr.2017.12.007] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/24/2017] [Accepted: 12/09/2017] [Indexed: 12/17/2022]
Abstract
Fibrin, a natural hydrogel, is the end product of the physiological blood coagulation cascade and naturally involved in wound healing. Beyond its role in hemostasis, it acts as a local reservoir for growth factors and as a provisional matrix for invading cells that drive the regenerative process. Its unique intrinsic features do not only promote wound healing directly via modulation of cell behavior but it can also be fine-tuned to evolve into a delivery system for sustained release of therapeutic biomolecules, cells and gene vectors. To further augment tissue regeneration potential, current strategies exploit and modify the chemical and physical characteristics of fibrin to employ combined incorporation of several factors and their timed release. In this work we show advanced therapeutic approaches employing fibrin matrices in wound healing and cover the many possibilities fibrin offers to the field of regenerative medicine.
Collapse
|
18
|
Endesfelder S, Weichelt U, Schiller C, Winter K, von Haefen C, Bührer C. Caffeine Protects Against Anticonvulsant-Induced Impaired Neurogenesis in the Developing Rat Brain. Neurotox Res 2018; 34:173-187. [PMID: 29417440 DOI: 10.1007/s12640-018-9872-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 01/08/2018] [Accepted: 01/23/2018] [Indexed: 02/06/2023]
Abstract
In preterm infants, phenobarbital is the first-line antiepileptic drug for neonatal seizures while caffeine is used for the treatment of apnea. Data from experimental animals suggest that phenobarbital and other anticonvulsants are toxic for the developing brain, while neuroprotective effects have been reported for caffeine both in newborn rodents and preterm human infants. To characterize the interaction of phenobarbital and caffeine in the hippocampus of the developing rodent brain, we examined the effects of both drugs given separately or together on postnatal neurogenesis after administration to neonatal rats throughout postnatal day (P) 4 to P6. Phenobarbital treatment (50 mg/kg) resulted in a significant decrease of proliferative capacity in the dentate gyrus. Phenobarbital also reduced expression of neuronal markers (doublecortin (DCX), calretinin, NeuN), neuronal transcription factors (Pax6, Sox2, Tbr1/2, Prox1), and neurotrophins (NGF, BDNF, NT-3) up to 24 h after the last administration. The phenobarbital-mediated impairment of neurogenesis was largely ameliorated by preconditioning with caffeine (10 mg/kg). In contrast, caffeine alone reduced proliferative capacity and expression of the neuronal markers DCX and NeuN at 6 h, but increased expression of neurotrophins and neuronal transcription factors at 6 and 12 h. These results indicate that administration of phenobarbital during the vulnerable phase of brain development negatively interferes with neuronal development, which can be prevented in part by co-administration of caffeine.
Collapse
Affiliation(s)
- Stefanie Endesfelder
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Ulrike Weichelt
- Endowed Professorship of Immunotechnology, Institute of Biochemistry and Biology, University of Potsdam, Campus Golm, Karl-Liebknechtstraße 24-25, 14476, Potsdam - Golm, Germany
| | - Cornelia Schiller
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Katja Winter
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Clarissa von Haefen
- Department of Anesthesiology and Operative Intensive Care Medicine, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Christoph Bührer
- Department of Neonatology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| |
Collapse
|
19
|
Hong MH, Hong HJ, Pang H, Lee HJ, Yi S, Koh WG. Controlled Release of Growth Factors from Multilayered Fibrous Scaffold for Functional Recoveries in Crushed Sciatic Nerve. ACS Biomater Sci Eng 2018; 4:576-586. [PMID: 33418747 DOI: 10.1021/acsbiomaterials.7b00801] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
In this study, we designed and fabricated a multilayered fibrous scaffold capable of the controlled release of multiple growth factors for sciatic nerve regeneration in rats. The scaffold consists of three layers prepared by sequential electrospinning, where the first layer is fabricated using polycaprolactone (PCL)-aligned electrospun nanofibers for the attachment and differentiation of cells toward the direction of the sciatic nerve. The second and third layers are fabricated using poly(lactic-co-glycolic acid) 6535 (PLGA 6535) and 8515 (PLGA 8515), respectively. The resultant three nanofiber layers were stacked and fixed by incorporating hydrogel micropatterns at both ends of nanofiber scaffold, which also facilitated the surgical handling of the multilayered scaffolds. The PLGA layers acted as reservoirs to release growth factors neurotrophin (NT-3), brain-derived neurotrophic factor (BDNF), and platelet-derived growth factor (PDGF). The different biodegradation rate of each PLGA layer enabled the controlled release of multiple growth factors such as NT-3, BDNF, and PDGF with different patterns. In a rat model, the injured nerve was rolled up with the multilayered scaffold loading growth factors, and behavior tests were performed five weeks after surgery. Sciatic functional index (SFI) and mechanical allodynia analysis revealed that the fast release of NT-3 and BDNF from PLGA 6535 and subsequent slow release of PDGF from PLGA 8515 proved to be the greatest aid to neural tissue regeneration. In addition to the biochemical cues from growth factors, the aligned PCL layer that directly contacts the injured nerve could provide topographical stimulation, offering practical assistance to new tissue and cells for directional growth parallel to the sciatic nerve. This study demonstrated that our multilayered scaffold performs a function that can be used to promote locomotor activity and enhance nerve regeneration in combination with align-patterned topography and the controlled release of growth factors.
Collapse
Affiliation(s)
- Min-Ho Hong
- Department of Neurosurgery, Spine and Spinal Cord Institute, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
| | - Hye Jin Hong
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Haejeong Pang
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Hyo-Jung Lee
- Department of Neurosurgery, Spine and Spinal Cord Institute, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
| | - Seong Yi
- Department of Neurosurgery, Spine and Spinal Cord Institute, College of Medicine, Yonsei University, Seoul 03722, Republic of Korea
| | - Won-Gun Koh
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
20
|
Thomas M, Willerth SM. 3-D Bioprinting of Neural Tissue for Applications in Cell Therapy and Drug Screening. Front Bioeng Biotechnol 2017; 5:69. [PMID: 29204424 PMCID: PMC5698280 DOI: 10.3389/fbioe.2017.00069] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Accepted: 10/19/2017] [Indexed: 01/18/2023] Open
Abstract
Neurodegenerative diseases affect millions of individuals in North America and cost the health-care industry billions of dollars for treatment. Current treatment options for degenerative diseases focus on physical rehabilitation or drug therapies, which temporarily mask the effects of cell damage, but quickly lose their efficacy. Cell therapies for the central nervous system remain an untapped market due to the complexity involved in growing neural tissues, controlling their differentiation, and protecting them from the hostile environment they meet upon implantation. Designing tissue constructs for the discovery of better drug treatments are also limited due to the resolution needed for an accurate cellular representation of the brain, in addition to being expensive and difficult to translate to biocompatible materials. 3-D printing offers a streamlined solution for engineering brain tissue for drug discovery or, in the future, for implantation. New microfluidic and bioplotting devices offer increased resolution, little impact on cell viability and have been tested with several bioink materials including fibrin, collagen, hyaluronic acid, poly(caprolactone), and poly(ethylene glycol). This review details current efforts at bioprinting neural tissue and highlights promising avenues for future work.
Collapse
Affiliation(s)
- Michaela Thomas
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
| | - Stephanie M. Willerth
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Biomedical Research, University of Victoria, Victoria, BC, Canada
- International Collaboration on Repair Discoveries (ICORD), Vancouver, BC, Canada
| |
Collapse
|
21
|
Robinson M, Chapani P, Styan T, Vaidyanathan R, Willerth SM. Functionalizing Ascl1 with Novel Intracellular Protein Delivery Technology for Promoting Neuronal Differentiation of Human Induced Pluripotent Stem Cells. Stem Cell Rev Rep 2017; 12:476-83. [PMID: 27138845 DOI: 10.1007/s12015-016-9655-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pluripotent stem cells can become any cell type found in the body. Accordingly, one of the major challenges when working with pluripotent stem cells is producing a highly homogenous population of differentiated cells, which can then be used for downstream applications such as cell therapies or drug screening. The transcription factor Ascl1 plays a key role in neural development and previous work has shown that Ascl1 overexpression using viral vectors can reprogram fibroblasts directly into neurons. Here we report on how a recombinant version of the Ascl1 protein functionalized with intracellular protein delivery technology (Ascl1-IPTD) can be used to rapidly differentiate human induced pluripotent stem cells (hiPSCs) into neurons. We first evaluated a range of Ascl1-IPTD concentrations to determine the most effective amount for generating neurons from hiPSCs cultured in serum free media. Next, we looked at the frequency of Ascl1-IPTD supplementation in the media on differentiation and found that one time supplementation is sufficient enough to trigger the neural differentiation process. Ascl1-IPTD was efficiently taken up by the hiPSCs and enabled rapid differentiation into TUJ1-positive and NeuN-positive populations with neuronal morphology after 8 days. After 12 days of culture, hiPSC-derived neurons produced by Ascl1-IPTD treatment exhibited greater neurite length and higher numbers of branch points compared to neurons derived using a standard neural progenitor differentiation protocol. This work validates Ascl1-IPTD as a powerful tool for engineering neural tissue from pluripotent stem cells.
Collapse
Affiliation(s)
- Meghan Robinson
- Biomedical Engineering Program, University of Victoria, Victoria, BC, Canada
| | - Parv Chapani
- Department of Biochemistry, University of Victoria, Victoria, BC, Canada
| | - Tara Styan
- Biomedical Engineering Program, University of Victoria, Victoria, BC, Canada
| | | | - Stephanie Michelle Willerth
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada.
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- International Collaboration on Repair Discoveries (ICORD), Vancouver, BC, Canada.
| |
Collapse
|
22
|
Russell LN, Lampe KJ. Oligodendrocyte Precursor Cell Viability, Proliferation, and Morphology is Dependent on Mesh Size and Storage Modulus in 3D Poly(ethylene glycol)-Based Hydrogels. ACS Biomater Sci Eng 2017; 3:3459-3468. [PMID: 33445383 DOI: 10.1021/acsbiomaterials.7b00374] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Oligodendrocytes in the central nervous system (CNS) are responsible for generating myelin, an electrically insulating layer around neuronal axons. When myelin is damaged, neurons are incapable of sustaining normal communications, which can manifest in patients as pain and loss of mobility and vision. A plethora of research has used biomaterials to promote neuronal regeneration, but despite the wide implications of a disrupted myelin sheath, very little is known about how biomaterial environments impact proliferation of oligodendrocyte precursor cells (OPCs) or their differentiation into myelinating oligodendrocytes. This work investigates how the storage modulus and mesh size of a polyethylene glycol (PEG)-based hydrogel, varied via two different mechanisms, directly affect the proliferation of two OPC lines encapsulated and cultured in 3D. Viability and proliferation of both OPC lines was dependent on hydrogel swelling and stiffness, where the concentration of ATP increased more in the more compliant gels. OPCs multiplied in the 3D hydrogels, creating significantly larger spheroids in the less cross-linked conditions. Stiffer, more highly cross-linked materials lead to greater expression of PDGFRα, an OPC receptor, indicating that fewer cells were committed to the oligodendrocyte lineage or had dedifferentiated in compliant materials. Laminin incorporation in the 3D matrix was found to have little effect on viability or proliferation. These findings provide valuable information on how mesh size and stiffness affect OPCs where more compliant materials favor proliferation of OPCs with less commitment to a mature oligodendrocyte lineage. Such information will be useful in the development of translational biomaterials to stimulate oligodendrocyte maturation for neural regeneration.
Collapse
Affiliation(s)
- Lauren N Russell
- Department of Chemical Engineering, University of Virginia, 102 Engineers' Way, Charlottesville, Virginia 22904, United States
| | - Kyle J Lampe
- Department of Chemical Engineering, University of Virginia, 102 Engineers' Way, Charlottesville, Virginia 22904, United States
| |
Collapse
|
23
|
Yu C, Griffiths LR, Haupt LM. Exploiting Heparan Sulfate Proteoglycans in Human Neurogenesis-Controlling Lineage Specification and Fate. Front Integr Neurosci 2017; 11:28. [PMID: 29089873 PMCID: PMC5650988 DOI: 10.3389/fnint.2017.00028] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 09/25/2017] [Indexed: 12/26/2022] Open
Abstract
Unspecialized, self-renewing stem cells have extraordinary application to regenerative medicine due to their multilineage differentiation potential. Stem cell therapies through replenishing damaged or lost cells in the injured area is an attractive treatment of brain trauma and neurodegenerative neurological disorders. Several stem cell types have neurogenic potential including neural stem cells (NSCs), embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), and mesenchymal stem cells (MSCs). Currently, effective use of these cells is limited by our lack of understanding and ability to direct lineage commitment and differentiation of neural lineages. Heparan sulfate proteoglycans (HSPGs) are ubiquitous proteins within the stem cell microenvironment or niche and are found localized on the cell surface and in the extracellular matrix (ECM), where they interact with numerous signaling molecules. The glycosaminoglycan (GAG) chains carried by HSPGs are heterogeneous carbohydrates comprised of repeating disaccharides with specific sulfation patterns that govern ligand interactions to numerous factors including the fibroblast growth factors (FGFs) and wingless-type MMTV integration site family (Wnts). As such, HSPGs are plausible targets for guiding and controlling neural stem cell lineage fate. In this review, we provide an overview of HSPG family members syndecans and glypicans, and perlecan and their role in neurogenesis. We summarize the structural changes and subsequent functional implications of heparan sulfate as cells undergo neural lineage differentiation as well as outline the role of HSPG core protein expression throughout mammalian neural development and their function as cell receptors and co-receptors. Finally, we highlight suitable biomimetic approaches for exploiting the role of HSPGs in mammalian neurogenesis to control and tailor cell differentiation into specific lineages. An improved ability to control stem cell specific neural lineage fate and produce abundant cells of lineage specificity will further advance stem cell therapy for the development of improved repair of neurological disorders. We propose a deeper understanding of HSPG-mediated neurogenesis can potentially provide novel therapeutic targets of neurogenesis.
Collapse
Affiliation(s)
- Chieh Yu
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Lyn R Griffiths
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Larisa M Haupt
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
24
|
Iyer NR, Wilems TS, Sakiyama-Elbert SE. Stem cells for spinal cord injury: Strategies to inform differentiation and transplantation. Biotechnol Bioeng 2017; 114:245-259. [PMID: 27531038 PMCID: PMC5642909 DOI: 10.1002/bit.26074] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 06/20/2016] [Accepted: 08/07/2016] [Indexed: 12/13/2022]
Abstract
The complex pathology of spinal cord injury (SCI), involving a cascade of secondary events and the formation of inhibitory barriers, hampers regeneration across the lesion site and often results in irreversible loss of motor function. The limited regenerative capacity of endogenous cells after SCI has led to a focus on the development of cell therapies that can confer both neuroprotective and neuroregenerative benefits. Stem cells have emerged as a candidate cell source because of their ability to self-renew and differentiate into a multitude of specialized cell types. While ethical and safety concerns impeded the use of stem cells in the past, advances in isolation and differentiation methods have largely mitigated these issues. A confluence of work in stem cell biology, genetics, and developmental neurobiology has informed the directed differentiation of specific spinal cell types. After transplantation, these stem cell-derived populations can replace lost cells, provide trophic support, remyelinate surviving axons, and form relay circuits that contribute to functional recovery. Further refinement of stem cell differentiation and transplantation methods, including combinatorial strategies that involve biomaterial scaffolds and drug delivery, is critical as stem cell-based treatments enter clinical trials. Biotechnol. Bioeng. 2017;114: 245-259. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Nisha R Iyer
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton St., Stop C0800 BME 3.314, Austin, Texas 78712
| | - Thomas S Wilems
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton St., Stop C0800 BME 3.314, Austin, Texas 78712
| | - Shelly E Sakiyama-Elbert
- Department of Biomedical Engineering, The University of Texas at Austin, 107 W Dean Keeton St., Stop C0800 BME 3.314, Austin, Texas 78712
| |
Collapse
|
25
|
|
26
|
Agbay A, Edgar JM, Robinson M, Styan T, Wilson K, Schroll J, Ko J, Khadem Mohtaram N, Jun MBG, Willerth SM. Biomaterial Strategies for Delivering Stem Cells as a Treatment for Spinal Cord Injury. Cells Tissues Organs 2016; 202:42-51. [DOI: 10.1159/000446474] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2016] [Indexed: 11/19/2022] Open
Abstract
Ongoing clinical trials are evaluating the use of stem cells as a way to treat traumatic spinal cord injury (SCI). However, the inhibitory environment present in the injured spinal cord makes it challenging to achieve the survival of these cells along with desired differentiation into the appropriate phenotypes necessary to regain function. Transplanting stem cells along with an instructive biomaterial scaffold can increase cell survival and improve differentiation efficiency. This study reviews the literature discussing different types of instructive biomaterial scaffolds developed for transplanting stem cells into the injured spinal cord. We have chosen to focus specifically on biomaterial scaffolds that direct the differentiation of neural stem cells and pluripotent stem cells since they offer the most promise for producing the cell phenotypes that could restore function after SCI. In terms of biomaterial scaffolds, this article reviews the literature associated with using hydrogels made from natural biomaterials and electrospun scaffolds for differentiating stem cells into neural phenotypes. It then presents new data showing how these different types of scaffolds can be combined for neural tissue engineering applications and provides directions for future studies.
Collapse
|
27
|
Russell LN, Lampe KJ. Engineering Biomaterials to Influence Oligodendroglial Growth, Maturation, and Myelin Production. Cells Tissues Organs 2016; 202:85-101. [PMID: 27701172 DOI: 10.1159/000446645] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2016] [Indexed: 11/19/2022] Open
Abstract
Millions of people suffer from damage or disease to the nervous system that results in a loss of myelin, such as through a spinal cord injury or multiple sclerosis. Diminished myelin levels lead to further cell death in which unmyelinated neurons die. In the central nervous system, a loss of myelin is especially detrimental because of its poor ability to regenerate. Cell therapies such as stem or precursor cell injection have been investigated as stem cells are able to grow and differentiate into the damaged cells; however, stem cell injection alone has been unsuccessful in many areas of neural regeneration. Therefore, researchers have begun exploring combined therapies with biomaterials that promote cell growth and differentiation while localizing cells in the injured area. The regrowth of myelinating oligodendrocytes from neural stem cells through a biomaterials approach may prove to be a beneficial strategy following the onset of demyelination. This article reviews recent advancements in biomaterial strategies for the differentiation of neural stem cells into oligodendrocytes, and presents new data indicating appropriate properties for oligodendrocyte precursor cell growth. In some cases, an increase in oligodendrocyte differentiation alongside neurons is further highlighted for functional improvements where the biomaterial was then tested for increased myelination both in vitro and in vivo.
Collapse
|
28
|
Hamilton C, Callanan A. Secreted Endothelial Cell Factors Immobilized on Collagen Scaffolds Enhance the Recipient Endothelial Cell Environment. Biores Open Access 2016; 5:61-71. [PMID: 27057474 PMCID: PMC4817599 DOI: 10.1089/biores.2016.0003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Strategies to design novel vascular scaffolds are a continuing aim in tissue engineering and often such designs encompass the use of recombinant factors to enhance the performance of the scaffold. The established use of cell secretion utilized in feeder systems and conditioned media offer a source of paracrine factors, which has potential to be used in tissue-engineered (TE) scaffolds. Here we utilize this principle from endothelial cells (ECs), to create a novel TE scaffold by harnessing secreted factors and immobilizing these to collagen scaffolds. This research revealed increased cellular attachment and positive angiogenic gene upregulation responses in recipient ECs grown on these conditioned scaffolds. Also, the conditioning method did not affect the mechanical structural integrity of the scaffolds. These results may advocate the potential use of this system to improve vascular scaffolds' in vivo performance. In addition, this process may be a future method utilized to improve other tissue engineering scaffold therapies.
Collapse
Affiliation(s)
- Charlotte Hamilton
- Institute of Bioengineering, The University of Edinburgh , The King's Buildings, Edinburgh, United Kingdom
| | - Anthony Callanan
- Institute of Bioengineering, The University of Edinburgh , The King's Buildings, Edinburgh, United Kingdom
| |
Collapse
|
29
|
Regalado-Santiago C, Juárez-Aguilar E, Olivares-Hernández JD, Tamariz E. Mimicking Neural Stem Cell Niche by Biocompatible Substrates. Stem Cells Int 2016; 2016:1513285. [PMID: 26880934 PMCID: PMC4736764 DOI: 10.1155/2016/1513285] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 10/19/2015] [Accepted: 11/23/2015] [Indexed: 01/17/2023] Open
Abstract
Neural stem cells (NSCs) participate in the maintenance, repair, and regeneration of the central nervous system. During development, the primary NSCs are distributed along the ventricular zone of the neural tube, while, in adults, NSCs are mainly restricted to the subependymal layer of the subventricular zone of the lateral ventricles and the subgranular zone of the dentate gyrus in the hippocampus. The circumscribed areas where the NSCs are located contain the secreted proteins and extracellular matrix components that conform their niche. The interplay among the niche elements and NSCs determines the balance between stemness and differentiation, quiescence, and proliferation. The understanding of niche characteristics and how they regulate NSCs activity is critical to building in vitro models that include the relevant components of the in vivo niche and to developing neuroregenerative approaches that consider the extracellular environment of NSCs. This review aims to examine both the current knowledge on neurogenic niche and how it is being used to develop biocompatible substrates for the in vitro and in vivo mimicking of extracellular NSCs conditions.
Collapse
Affiliation(s)
- Citlalli Regalado-Santiago
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Avenida Luis Castelazo Ayala, s/n, 91190 Xalapa, VER, Mexico
| | - Enrique Juárez-Aguilar
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Avenida Luis Castelazo Ayala, s/n, 91190 Xalapa, VER, Mexico
| | - Juan David Olivares-Hernández
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Avenida Luis Castelazo Ayala, s/n, 91190 Xalapa, VER, Mexico
| | - Elisa Tamariz
- Instituto de Ciencias de la Salud, Universidad Veracruzana, Avenida Luis Castelazo Ayala, s/n, 91190 Xalapa, VER, Mexico
| |
Collapse
|
30
|
Elliott Donaghue I, Shoichet MS. Controlled release of bioactive PDGF-AA from a hydrogel/nanoparticle composite. Acta Biomater 2015; 25:35-42. [PMID: 26257128 DOI: 10.1016/j.actbio.2015.08.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 07/28/2015] [Accepted: 08/05/2015] [Indexed: 12/23/2022]
Abstract
Polymer excipients, such as low molar mass poly(ethylene glycol) (PEG), have shown contradictory effects on protein stability when co-encapsulated in polymeric nanoparticles. To gain further insight into these effects, platelet-derived growth factor (PDGF-AA) was encapsulated in polymeric nanoparticles with vs. without PEG. PDGF-AA is a particularly compelling protein, as it has been demonstrated to promote cell survival and induce the oligodendrocyte differentiation of neural stem/progenitor cells (NSPCs) both in vitro and in vivo. Here we show, for the first time, the controlled release of bioactive PDGF-AA from an injectable nanoparticle/hydrogel drug delivery system (DDS). PDGF-AA was encapsulated, with high efficiency, in poly(lactide-co-glycolide) nanoparticles, and its release from the drug delivery system was followed over 21 d. Interestingly, the co-encapsulation of low molecular weight poly(ethylene glycol) increased the PDGF-AA loading but, unexpectedly, accelerated the aggregation of PDGF-AA, resulting in reduced activity and detection by enzyme-linked immunosorbent assay (ELISA). In the absence of PEG, released PDGF-AA remained bioactive as demonstrated with NSPC oligodendrocyte differentiation, similar to positive controls, and significantly different from untreated controls. This work presents a novel delivery method for differentiation factors, such as PDGF-AA, and provides insights into the contradictory effects reported in the literature of excipients, such as PEG, on the loading and release of proteins from polymeric nanoparticles. STATEMENT OF SIGNIFICANCE Previously, the polymer poly(ethylene glycol) (PEG) has been used in many biomaterials applications, from surface coatings to the encapsulation of proteins. In this work, we demonstrate that, unexpectedly, low molecular weight PEG has a deleterious effect on the release of the encapsulated protein platelet-derived growth factor AA (PDGF-AA). We also demonstrate release of bioactive PDGF-AA (in the absence of PEG). Specifically, we demonstrate the differentiation of neural stem and progenitor cells to oligodendrocytes, similar to what is observed with the addition of fresh PDGFAA. A differentiated oligodendrocyte population is a key strategy in central nervous system regeneration. This work is the first demonstration of controlled PDGF-AA release, and also brings new insights to the broader field of protein encapsulation.
Collapse
Affiliation(s)
- Irja Elliott Donaghue
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Canada
| | - Molly S Shoichet
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Canada; Department of Chemistry, University of Toronto, Canada.
| |
Collapse
|
31
|
Ahmad E, Fatima MT, Hoque M, Owais M, Saleemuddin M. Fibrin matrices: The versatile therapeutic delivery systems. Int J Biol Macromol 2015; 81:121-36. [PMID: 26231328 DOI: 10.1016/j.ijbiomac.2015.07.054] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Revised: 07/24/2015] [Accepted: 07/26/2015] [Indexed: 12/12/2022]
Abstract
Fibrin sealants, that have been employed for over a century by surgeons to stop post surgery bleeding, are finding novel applications in the controlled delivery of antibiotics and several other therapeutics. Fibrinogen can be easily purified from blood plasma and converted by thrombolysis to fibrin that undergoes spontaneous aggregation to form insoluble clot. During the gelling, fibrin can be formulated into films, clots, threads, microbeads, nanoconstructs and nanoparticles. Whole plasma clots in the form of beads and microparticles can also be prepared by activating endogenous thrombin, for possible drug delivery. Fibrin formulations offer remarkable scope for controlling the porosity as well as in vivo degradability and hence the release of the associated therapeutics. Binding/covalent-linking of therapeutics to the fibrin matrix, crosslinking of the matrix with bifunctional reagents and coentrapment of protease inhibitors have been successful in regulating both in vitro and in vivo release of the therapeutics. The release rates can also be remarkably lowered by preentrapment of therapeutics in insoluble particles like liposomes or by anchoring them to the matrix via molecules that bind them as well as fibrin.
Collapse
Affiliation(s)
- Ejaj Ahmad
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| | | | - Mehboob Hoque
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| | - Mohammad Owais
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| | - Mohammed Saleemuddin
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
32
|
Weaver CL, Cui XT. Directed Neural Stem Cell Differentiation with a Functionalized Graphene Oxide Nanocomposite. Adv Healthc Mater 2015; 4:1408-16. [PMID: 25943251 DOI: 10.1002/adhm.201500056] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 03/26/2015] [Indexed: 12/15/2022]
Abstract
Neural stem cell (NSC) transplantation has the potential to restore function to diseased or damaged nervous tissue, but poor control over cell survival, differentiation, and maturation limits therapeutic prospects. Engineered scaffolds that have the ability to drive neural stem cell behavior can address these limitations facing cell transplantation. Conducting polymers, which have the ability to electrically interface with cells, are attractive scaffolding candidates, but they lack the capacity for simple covalent modification, which would enable surface patterning of biomolecules. In this work, the NSC scaffolding performance of a nanocomposite composed of conducting polymer poly(3,4-ethylenedioxythiophene) (PEDOT) and graphene oxide (GO) nanosheets (GO/PEDOT) is investigated. The GO/PEDOT material is nontoxic and improves NSC differentiation toward the neuronal lineage. Biomolecules interferon-γ (IFNγ) and platelet-derived growth factor (PDGF) that selectively promote neuronal or oligodendrocyte lineage differentiation, respectively, are covalently cross-linked to the surface of the GO/PEDOT nanocomposite via carboxylic acid functional groups provided by GO using carbodiimide chemistry. The surfaces support a larger population of neurons when modified with IFNγ and a larger population of oligodendrocytes when modified by PDGF. This work demonstrates the customizability of GO/PEDOT for cell scaffolding applications and underlines its potential for controlling NSC behavior to improve therapeutic potential.
Collapse
Affiliation(s)
- Cassandra L. Weaver
- Department of Bioengineering; University of Pittsburgh; Pittsburgh PA 15260 USA
- Center for the Neural Basis of Cognition; University of Pittsburgh; Pittsburgh PA 15260 USA
- McGowan Institute for Regenerative Medicine; University of Pittsburgh; Pittsburgh PA 15260 USA
| | - Xinyan Tracy Cui
- Department of Bioengineering; University of Pittsburgh; Pittsburgh PA 15260 USA
- Center for the Neural Basis of Cognition; University of Pittsburgh; Pittsburgh PA 15260 USA
- McGowan Institute for Regenerative Medicine; University of Pittsburgh; Pittsburgh PA 15260 USA
| |
Collapse
|
33
|
Kuffler DP. Platelet-Rich Plasma Promotes Axon Regeneration, Wound Healing, and Pain Reduction: Fact or Fiction. Mol Neurobiol 2015; 52:990-1014. [PMID: 26048672 DOI: 10.1007/s12035-015-9251-x] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Indexed: 11/25/2022]
Abstract
Platelet-rich plasma (PRP) has been tested in vitro, in animal models, and clinically for its efficacy in enhancing the rate of wound healing, reducing pain associated with injuries, and promoting axon regeneration. Although extensive data indicate that PRP-released factors induce these effects, the claims are often weakened because many studies were not rigorous or controlled, the data were limited, and other studies yielded contrary results. Critical to assessing whether PRP is effective are the large number of variables in these studies, including the method of PRP preparation, which influences the composition of PRP; type of application; type of wounds; target tissues; and diverse animal models and clinical studies. All these variables raise the question of whether one can anticipate consistent influences and raise the possibility that most of the results are correct under the circumstances where PRP was tested. This review examines evidence on the potential influences of PRP and whether PRP-released factors could induce the reported influences and concludes that the preponderance of evidence suggests that PRP has the capacity to induce all the claimed influences, although this position cannot be definitively argued. Well-defined and rigorously controlled studies of the potential influences of PRP are required in which PRP is isolated and applied using consistent techniques, protocols, and models. Finally, it is concluded that, because of the purported benefits of PRP administration and the lack of adverse events, further animal and clinical studies should be performed to explore the potential influences of PRP.
Collapse
Affiliation(s)
- Damien P Kuffler
- Institute of Neurobiology, University of Puerto Rico, Medical Sciences Campus, 201 Blvd. Del Valle, San Juan, 00901, Puerto Rico,
| |
Collapse
|
34
|
Robinson M, Yau SY, Sun L, Gabers N, Bibault E, Christie BR, Willerth SM. Optimizing Differentiation Protocols for Producing Dopaminergic Neurons from Human Induced Pluripotent Stem Cells for Tissue Engineering Applications: Supplementary Issue: Stem Cell Biology. Biomark Insights 2015; 10 Suppl 1:61-70. [PMID: 36876191 PMCID: PMC9980910 DOI: 10.4137/bmi.s20064] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 03/02/2015] [Accepted: 03/05/2015] [Indexed: 11/05/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder that results when the dopaminergic neurons (DNs) present in the substantia nigra necessary for voluntary motor control are depleted, making patients with this disorder ideal candidates for cell replacement therapy. Human induced pluripotent stem cells (hiPSCs), obtained by reprogramming adult cells, possess the properties of pluripotency and immortality while enabling the possibility of patient-specific therapies. An effective cell therapy for PD requires an efficient, defined method of DN generation, as well as protection from the neuroinflammatory environment upon engraftment. Although similar in pluripotency to human embryonic stem cells (hESCs), hiPSCs differentiate less efficiently into neuronal subtypes. Previous work has shown that treatment with guggulsterone can efficiently differentiate hESCs into DNs. Our work shows that guggulsterone is able to derive DNs from hiPSCs with comparable efficiency, and furthermore, this differentiation can be achieved inside three-dimensional fibrin scaffolds that could enhance cell survival upon engraftment.
Collapse
Affiliation(s)
| | - Suk-Yu Yau
- Department of Neuroscience, Division of Medical Sciences
| | - Lin Sun
- Department of Neuroscience, Division of Medical Sciences
| | - Nicole Gabers
- Department of Biology, University of Victoria, Victoria, BC, Canada
| | | | | | - Stephanie M Willerth
- Department of Biomedical Engineering.,Department of Neuroscience, Division of Medical Sciences.,Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
35
|
Montgomery A, Wong A, Gabers N, Willerth SM. Engineering personalized neural tissue by combining induced pluripotent stem cells with fibrin scaffolds. Biomater Sci 2014. [PMID: 26218131 DOI: 10.1039/c4bm00299g] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Induced pluripotent stem cells (iPSCs) are generated from adult somatic cells through the induction of key transcription factors that restore the ability to become any cell type found in the body. These cells are of interest for tissue engineering due to their potential for developing patient-specific therapies. As the technology for generating iPSCs advances, it is important to concurrently investigate protocols for the efficient differentiation of these cells to desired downstream phenotypes in combination with biomaterial scaffolds as a way of engineering neural tissue. For such applications, the generation of neurons within three dimensional fibrin scaffolds has been well characterized as a cell-delivery platform for murine embryonic stem cells (ESCs) but has not yet been applied to murine iPSCs. Given that iPSCs have been reported to differentiate less effectively than ESCs, a key objective of this investigation is to maximize the proportion of iPSC-derived neurons in fibrin through the choice of differentiation protocol. To this end, this study compares two EB-mediated protocols for generating neurons from murine iPSCs and ESCs: an 8 day 4-/4+ protocol using soluble retinoic acid in the last 4 days and a 6 day 2-/4+ protocol using soluble retinoic acid and the small molecule sonic hedgehog agonist purmorphamine in the last 4 days. EBs were then seeded in fibrin scaffolds for 14 days to allow further differentiation into neurons. EBs generated by the 2-/4+ protocol yielded a higher percentage of neurons compared to those from the 4-/4+ protocol for both iPSCs and ESCs. The results demonstrate the successful translation of the fibrin-based cell-delivery platform for use with murine iPSCs and furthermore that the proportion of neurons generated from murine iPSC-derived EBs seeded in fibrin can be maximized using the 2-/4+ differentiation protocol. Together, these findings validate the further exploration of 3D fibrin-based scaffolds as a method of delivering neuronal cells derived from iPSCs - an important step toward the development of iPSC-based tissue engineering strategies for spinal cord injury repair.
Collapse
Affiliation(s)
- Amy Montgomery
- Department of Mechanical Engineering, University of Victoria, Canada.
| | | | | | | |
Collapse
|
36
|
Vulic K, Shoichet MS. Affinity-Based Drug Delivery Systems for Tissue Repair and Regeneration. Biomacromolecules 2014; 15:3867-80. [DOI: 10.1021/bm501084u] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Katarina Vulic
- Department of Chemistry, ‡Department of Chemical
Engineering and Applied Chemistry, §Institute of Biomaterials
and Biomedical Engineering, Donnelly Centre, University of Toronto, 160 College Street, Toronto, Ontario M5S3E1, Canada
| | - Molly S. Shoichet
- Department of Chemistry, ‡Department of Chemical
Engineering and Applied Chemistry, §Institute of Biomaterials
and Biomedical Engineering, Donnelly Centre, University of Toronto, 160 College Street, Toronto, Ontario M5S3E1, Canada
| |
Collapse
|
37
|
Tang S, Liao X, Shi B, Qu Y, Huang Z, Lin Q, Guo X, Pei F. The effects of controlled release of neurotrophin-3 from PCLA scaffolds on the survival and neuronal differentiation of transplanted neural stem cells in a rat spinal cord injury model. PLoS One 2014; 9:e107517. [PMID: 25215612 PMCID: PMC4162607 DOI: 10.1371/journal.pone.0107517] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 08/18/2014] [Indexed: 02/05/2023] Open
Abstract
Neural stem cells (NSCs) have emerged as a potential source for cell replacement therapy following spinal cord injury (SCI). However, poor survival and low neuronal differentiation remain major obstacles to the use of NSCs. Biomaterials with neurotrophic factors are promising strategies for promoting the proliferation and differentiation of NSCs. Silk fibroin (SF) matrices were demonstrated to successfully deliver growth factors and preserve their potency. In this study, by incorporating NT-3 into a SF coating, we successfully developed NT-3-immobilized scaffolds (membranes and conduits). Sustained release of bioactive NT-3 from the conduits for up to 8 weeks was achieved. Cell viability was confirmed using live/dead staining after 14 days in culture. The efficacy of the immobilized NT-3 was confirmed by assessing NSC neuronal differentiation in vitro. NSC neuronal differentiation was 55.2 ± 4.1% on the NT-3-immobilized membranes, which was significantly higher than that on the NT-3 free membrane. Furthermore, 8 weeks after the NSCs were seeded into conduits and implanted in rats with a transected SCI, the conduit+NT-3+NSCs group achieved higher NSC survival (75.8 ± 15.1%) and neuronal differentiation (21.5 ± 5.2%) compared with the conduit+NSCs group. The animals that received the conduit+NT-3+NSCs treatment also showed improved functional outcomes, as well as increased axonal regeneration. These results indicate the feasibility of fabricating NT-3-immobilized scaffolds using the adsorption of NT-3/SF coating method, as well as the potential of these scaffolds to induce SCI repair by promoting survival and neuronal differentiation of transplanted NSCs.
Collapse
Affiliation(s)
- Shuo Tang
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiang Liao
- Department of Pain Medicine, Shenzhen Nanshan Hospital, Shenzhen, China
| | - Bo Shi
- Department of Orthopaedics, Mianyang Center Hospital, Mianyang, China
| | - Yanzhen Qu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zeyu Huang
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, China
| | - Qiang Lin
- Department of Orthopaedics, Guangdong hospital of traditional Chinese medicine, Guangzhou, China
- * E-mail: (QL); (XDG); (FXP)
| | - Xiaodong Guo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- * E-mail: (QL); (XDG); (FXP)
| | - Fuxing Pei
- Department of Orthopaedics, West China Hospital, Sichuan University, Chengdu, China
- * E-mail: (QL); (XDG); (FXP)
| |
Collapse
|
38
|
Adult-Derived Pluripotent Stem Cells. World Neurosurg 2014; 82:500-8. [DOI: 10.1016/j.wneu.2013.08.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 08/07/2013] [Accepted: 08/09/2013] [Indexed: 01/27/2023]
|
39
|
Skop NB, Calderon F, Cho CH, Gandhi CD, Levison SW. Improvements in biomaterial matrices for neural precursor cell transplantation. MOLECULAR AND CELLULAR THERAPIES 2014; 2:19. [PMID: 26056586 PMCID: PMC4452047 DOI: 10.1186/2052-8426-2-19] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 06/05/2014] [Indexed: 12/24/2022]
Abstract
Progress is being made in developing neuroprotective strategies for traumatic brain injuries; however, there will never be a therapy that will fully preserve neurons that are injured from moderate to severe head injuries. Therefore, to restore neurological function, regenerative strategies will be required. Given the limited regenerative capacity of the resident neural precursors of the CNS, many investigators have evaluated the regenerative potential of transplanted precursors. Unfortunately, these precursors do not thrive when engrafted without a biomaterial scaffold. In this article we review the types of natural and synthetic materials that are being used in brain tissue engineering applications for traumatic brain injury and stroke. We also analyze modifications of the scaffolds including immobilizing drugs, growth factors and extracellular matrix molecules to improve CNS regeneration and functional recovery. We conclude with a discussion of some of the challenges that remain to be solved towards repairing and regenerating the brain.
Collapse
Affiliation(s)
- Nolan B Skop
- Department of Neurology & Neurosciences, Rutgers University-New Jersey Medical School, NJMS-Cancer Center, H-1226, 205 South Orange Ave., Newark, NJ 07103 USA ; Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102 USA
| | - Frances Calderon
- Department of Neurology & Neurosciences, Rutgers University-New Jersey Medical School, NJMS-Cancer Center, H-1226, 205 South Orange Ave., Newark, NJ 07103 USA
| | - Cheul H Cho
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102 USA
| | - Chirag D Gandhi
- Department of Neurology & Neurosciences, Rutgers University-New Jersey Medical School, NJMS-Cancer Center, H-1226, 205 South Orange Ave., Newark, NJ 07103 USA ; Department of Neurological Surgery, Rutgers University-New Jersey Medical School, New Jersey Medical School, Newark, NJ 07103 USA
| | - Steven W Levison
- Department of Neurology & Neurosciences, Rutgers University-New Jersey Medical School, NJMS-Cancer Center, H-1226, 205 South Orange Ave., Newark, NJ 07103 USA
| |
Collapse
|
40
|
Rodda AE, Meagher L, Nisbet DR, Forsythe JS. Specific control of cell–material interactions: Targeting cell receptors using ligand-functionalized polymer substrates. Prog Polym Sci 2014. [DOI: 10.1016/j.progpolymsci.2013.11.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
41
|
Skop NB, Calderon F, Cho CH, Gandhi CD, Levison SW. Improvements in biomaterial matrices for neural precursor cell transplantation. MOLECULAR AND CELLULAR THERAPIES 2014; 2:19. [PMID: 26056586 PMCID: PMC4452047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 06/05/2014] [Indexed: 11/21/2023]
Abstract
Progress is being made in developing neuroprotective strategies for traumatic brain injuries; however, there will never be a therapy that will fully preserve neurons that are injured from moderate to severe head injuries. Therefore, to restore neurological function, regenerative strategies will be required. Given the limited regenerative capacity of the resident neural precursors of the CNS, many investigators have evaluated the regenerative potential of transplanted precursors. Unfortunately, these precursors do not thrive when engrafted without a biomaterial scaffold. In this article we review the types of natural and synthetic materials that are being used in brain tissue engineering applications for traumatic brain injury and stroke. We also analyze modifications of the scaffolds including immobilizing drugs, growth factors and extracellular matrix molecules to improve CNS regeneration and functional recovery. We conclude with a discussion of some of the challenges that remain to be solved towards repairing and regenerating the brain.
Collapse
Affiliation(s)
- Nolan B Skop
- />Department of Neurology & Neurosciences, Rutgers University-New Jersey Medical School, NJMS-Cancer Center, H-1226, 205 South Orange Ave., Newark, NJ 07103 USA
- />Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102 USA
| | - Frances Calderon
- />Department of Neurology & Neurosciences, Rutgers University-New Jersey Medical School, NJMS-Cancer Center, H-1226, 205 South Orange Ave., Newark, NJ 07103 USA
| | - Cheul H Cho
- />Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102 USA
| | - Chirag D Gandhi
- />Department of Neurology & Neurosciences, Rutgers University-New Jersey Medical School, NJMS-Cancer Center, H-1226, 205 South Orange Ave., Newark, NJ 07103 USA
- />Department of Neurological Surgery, Rutgers University-New Jersey Medical School, New Jersey Medical School, Newark, NJ 07103 USA
| | - Steven W Levison
- />Department of Neurology & Neurosciences, Rutgers University-New Jersey Medical School, NJMS-Cancer Center, H-1226, 205 South Orange Ave., Newark, NJ 07103 USA
| |
Collapse
|
42
|
Jang HK, Kim BS. Modulation of stem cell differentiation with biomaterials. Int J Stem Cells 2014; 3:80-4. [PMID: 24855545 DOI: 10.15283/ijsc.2010.3.2.80] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2010] [Indexed: 11/09/2022] Open
Abstract
Differentiation of stem cells can be controlled with interactions with microenvironments of the stem cells. The interactions contain various signals including soluble growth factor signal, cell adhesion signal, and mechanical signal, which can modulate differentiation of stem cells. Biomaterials can provide these types of signals to induce desirable cellular differentiation. Biomaterials can deliver soluble growth factors locally to stem cells at a controlled rate for a long period. Stem cell adhesion to specific adhesion molecules presented by biomaterials can induce specific differentiation. Mechanical signals can be delivered to stem cells seeded onto biomaterial scaffolds. These approaches would be invaluable for direction of stem cell differentiation and in vivo tissue regeneration using stem cells.
Collapse
Affiliation(s)
- Hyeon-Ki Jang
- Interdisciplinary Program of Bioengineering, Seoul National University, Seoul, Korea
| | - Byung-Soo Kim
- Interdisciplinary Program of Bioengineering, Seoul National University, Seoul, Korea ; School of Chemical and Biological Engineering, Seoul National University, Seoul, Korea
| |
Collapse
|
43
|
Sakiyama-Elbert SE. Incorporation of heparin into biomaterials. Acta Biomater 2014; 10:1581-7. [PMID: 24021232 DOI: 10.1016/j.actbio.2013.08.045] [Citation(s) in RCA: 133] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 08/27/2013] [Accepted: 08/29/2013] [Indexed: 11/30/2022]
Abstract
This review provides an overview of the incorporation of heparin into biomaterials with a focus on drug delivery and the use of heparin-based biomaterials for self-assembly of polymer networks. Heparin conjugation to biomaterials was originally explored to reduce the thrombogenicity of materials in contact with blood. Many of the conjugation strategies that were developed for these applications are still popular today for other applications. More recently heparin has been conjugated to biomaterials for drug delivery applications. Many of the delivery approaches have taken advantage of the ability of heparin to bind to a wide variety of growth factors, protecting them from degradation and potentiating interactions with cell surface receptors. More recently, the use of heparin as a base polymer for scaffold fabrication has also been explored, often utilizing non-covalent binding of heparin with peptides or proteins to promote self-assembly of hydrogel networks. This review will highlight recent advances in each of these areas.
Collapse
Affiliation(s)
- Shelly E Sakiyama-Elbert
- Department of Biomedical Engineering, Washington University, 1 Brookings Drive, Campus Box 1097, St Louis, MO 63130, USA.
| |
Collapse
|
44
|
de la Puente P, Ludeña D. Cell culture in autologous fibrin scaffolds for applications in tissue engineering. Exp Cell Res 2014; 322:1-11. [DOI: 10.1016/j.yexcr.2013.12.017] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 12/11/2013] [Accepted: 12/18/2013] [Indexed: 12/31/2022]
|
45
|
McCreedy DA, Wilems TS, Xu H, Butts JC, Brown CR, Smith AW, Sakiyama-Elbert SE. Survival, Differentiation, and Migration of High-Purity Mouse Embryonic Stem Cell-derived Progenitor Motor Neurons in Fibrin Scaffolds after Sub-Acute Spinal Cord Injury. Biomater Sci 2014; 2:1672-1682. [PMID: 25346848 DOI: 10.1039/c4bm00106k] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Embryonic stem (ES) cells can be differentiated into many neural cell types that hold great potential as cell replacement therapies following spinal cord injury (SCI). Coupling stem cell transplantation with biomaterial scaffolds can produce a unified combination therapy with several potential advantages including enhanced cell survival, greater transplant retention, reduced scarring, and improved integration at the transplant/host interface. Undesired cell types, however, are commonly present in ES-cell derived cultures due to the limited efficiency of most ES cell induction protocols. Heterogeneous cell populations can confound the interaction between the biomaterial and specific neural populations leading to undesired outcomes. In particular, biomaterials scaffolds may enhance tumor formation by promoting survival and proliferation of undifferentiated ES cells that can persist after induction. Methods for purification of specific ES cell-derived neural populations are necessary to recognize the full potential of combination therapies involving biomaterials and ES cell-derived neural populations. We previously developed a method for enriching ES cell-derived progenitor motor neurons (pMNs) induced from mouse ES cells via antibiotic selection and showed that the enriched cell populations are depleted of pluripotent stem cells. In this study, we demonstrate the survival and differentiation of enriched pMNs within three dimensional (3D) fibrin scaffolds in vitro and when transplanted into a sub-acute dorsal hemisection model of SCI into neurons, oligodendrocytes and astrocytes.
Collapse
Affiliation(s)
- D A McCreedy
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63112, USA
| | - T S Wilems
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63112, USA
| | - H Xu
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63112, USA
| | - J C Butts
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63112, USA
| | - C R Brown
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63112, USA
| | - A W Smith
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63112, USA
| | - S E Sakiyama-Elbert
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63112, USA
| |
Collapse
|
46
|
Abstract
Within the adult organism, stem cells reside in defined anatomical microenvironments called niches. These architecturally diverse microenvironments serve to balance stem cell self-renewal and differentiation. Proper regulation of this balance is instrumental to tissue repair and homeostasis, and any imbalance can potentially lead to diseases such as cancer. Within each of these microenvironments, a myriad of chemical and physical stimuli interact in a complex (synergistic or antagonistic) manner to tightly regulate stem cell fate. The in vitro replication of these in vivo microenvironments will be necessary for the application of stem cells for disease modeling, drug discovery, and regenerative medicine purposes. However, traditional reductionist approaches have only led to the generation of cell culture methods that poorly recapitulate the in vivo microenvironment. To that end, novel engineering and systems biology approaches have allowed for the investigation of the biological and mechanical stimuli that govern stem cell fate. In this review, the application of these technologies for the dissection of stem cell microenvironments will be analyzed. Moreover, the use of these engineering approaches to construct in vitro stem cell microenvironments that precisely control stem cell fate and function will be reviewed. Finally, the emerging trend of using high-throughput, combinatorial methods for the stepwise engineering of stem cell microenvironments will be explored.
Collapse
Affiliation(s)
- David A Brafman
- Department of Cellular and Molecular Medicine, Stem Cell Program, University of California at San Diego, La Jolla, California
| |
Collapse
|
47
|
Prabhakaran MP, Mobarakeh LG, Kai D, Karbalaie K, Nasr-Esfahani MH, Ramakrishna S. Differentiation of embryonic stem cells to cardiomyocytes on electrospun nanofibrous substrates. J Biomed Mater Res B Appl Biomater 2013; 102:447-54. [PMID: 24039141 DOI: 10.1002/jbm.b.33022] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Revised: 06/24/2013] [Accepted: 08/01/2013] [Indexed: 12/31/2022]
Abstract
The potential of pluripotent embryonic stem cells (ESCs) isolated from the inner mass of blastocysts are investigated for its ability to differentiate on biocompatible electrospun nanofibers, for regeneration of the myocardially infracted heart. Nanostructured poly(d,l-lactide-co-glycolide)/collagen (PLGA/Col) scaffolds with fiber diameters in the range of 300 ± 65 nm, was fabricated by electrospinning to mimic the extracellular matrix of the native tissue. During the culture of embryoid bodies outgrowth on the scaffolds, and further differentiation of ESCs to cardiomyocytes, the PLGA/Col nanofibers was found better than that of the electrospun PLGA nanofibers, where a better interaction and growth of ESC differentiated cardiomyocytes was observed on the composite scaffolds. The phenotypical characteristics of ESC-derived cardiomyocytes and molecular protein expression were carried out by scanning electron microscopy and immunocytochemistry, respectively. Our studies highlight the significance of a suitable material, its architecture, and cell-biomaterial interactions that is essential at a nanoscale level signifying the application of a bioengineered cardiac graft for stem cell differentiation and transplantation, which could be an intriguing strategy for cardiac regeneration.
Collapse
Affiliation(s)
- Molamma P Prabhakaran
- Center for Nanofibers and Nanotechnology, E3-05-14, Nanoscience and Nanotechnology Initiative, Faculty of Engineering, Singapore, 117576
| | | | | | | | | | | |
Collapse
|
48
|
Converting stem cells to dendritic cells by agonist antibodies from unbiased morphogenic selections. Proc Natl Acad Sci U S A 2013; 110:14966-71. [PMID: 23980154 DOI: 10.1073/pnas.1313671110] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
When combinatorial antibody libraries are rendered infectious for eukaryotic cells, the integrated antibody genotype and cellular phenotype become permanently linked and each cell becomes a selection system unto itself. These systems should be ideal for the identification of proteins and pathways that regulate differentiation so long as selection systems can be devised. Here we use a selection system based on the ability of secreted antibodies to alter the morphology of colonies expressing them when grown in soft agar. Importantly, this approach is different from all previous studies in that it used a pure discovery format where unbiased libraries that were not preselected against any known protein were used as probes. As such, the strategy is analogous to classical forward genetic approaches except that it operates directly at the protein level. This approach led to the identification of integrin-binding agonist antibodies that efficiently converted human stem cells to dendritic cells.
Collapse
|
49
|
Christ GJ, Saul JM, Furth ME, Andersson KE. The pharmacology of regenerative medicine. Pharmacol Rev 2013; 65:1091-133. [PMID: 23818131 DOI: 10.1124/pr.112.007393] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Regenerative medicine is a rapidly evolving multidisciplinary, translational research enterprise whose explicit purpose is to advance technologies for the repair and replacement of damaged cells, tissues, and organs. Scientific progress in the field has been steady and expectations for its robust clinical application continue to rise. The major thesis of this review is that the pharmacological sciences will contribute critically to the accelerated translational progress and clinical utility of regenerative medicine technologies. In 2007, we coined the phrase "regenerative pharmacology" to describe the enormous possibilities that could occur at the interface between pharmacology, regenerative medicine, and tissue engineering. The operational definition of regenerative pharmacology is "the application of pharmacological sciences to accelerate, optimize, and characterize (either in vitro or in vivo) the development, maturation, and function of bioengineered and regenerating tissues." As such, regenerative pharmacology seeks to cure disease through restoration of tissue/organ function. This strategy is distinct from standard pharmacotherapy, which is often limited to the amelioration of symptoms. Our goal here is to get pharmacologists more involved in this field of research by exposing them to the tools, opportunities, challenges, and interdisciplinary expertise that will be required to ensure awareness and galvanize involvement. To this end, we illustrate ways in which the pharmacological sciences can drive future innovations in regenerative medicine and tissue engineering and thus help to revolutionize the discovery of curative therapeutics. Hopefully, the broad foundational knowledge provided herein will spark sustained conversations among experts in diverse fields of scientific research to the benefit of all.
Collapse
Affiliation(s)
- George J Christ
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA.
| | | | | | | |
Collapse
|
50
|
Reactive oxygen species-responsive miR-210 regulates proliferation and migration of adipose-derived stem cells via PTPN2. Cell Death Dis 2013; 4:e588. [PMID: 23579275 PMCID: PMC3641340 DOI: 10.1038/cddis.2013.117] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hypoxia enhances the proliferation and migration of adipose-derived stem cells (ASCs) via the generation of reactive oxygen species (ROS). Therefore, this study primarily investigated whether or not ROS generation could regulate microRNA-210 (miR-210) expression, and increase proliferation/migration of ASCs. In addition, we tried to identify the signaling pathways involved in miR-210 upregulation and the direct target genes of miR-210 that mediate these functions. Various sources of ROS generation such as hypoxia, antimycin, rotenone, and platelet-derived growth factor (PDGF)-BB upregulated miR-210 expression, and increased the proliferation/migration of ASCs. There is a positive feed-forward loop between ROS generation and miR-210, and miR-210 itself increases ROS generation by downregulation of iron–sulfur cluster scaffold homolog 2 (ISCU2). Although hypoxia-inducible factor-1α was not involved in miR-210 expression, pharmacological or small interfering RNA (siRNA)-driven inhibition of Akt and ERK1/2 molecules reduced miR-210 expression. Transfection of siRNAs of NF-κB and Elk1 also reduced miR-210 expression, indicating that these signaling pathways mediate miR-210 upregulation. Protein tyrosine phosphatase, non-receptor type 2 (PTPN2) was selected for miR-210 target gene, and it was downregulated by ROS generators or miR-210 mimic treatment. PTPN2 was first proven to be a direct miR-210 target in luciferase activity assay, and pharmacological inhibition or overexpression of PTPN2 regulated the proliferation and migration of ASC. In conclusion, ROS generation from diverse sources induces miR-210 expression in ASCs via PDGFR-β, Akt and ERK pathways. Transcription of miR-210 expression is regulated by NF-κB and Elk1, and miR-210 increases the proliferation and migration of ASCs via ISCU2 and PTPN2 downregulation.
Collapse
|