1
|
Umar AK. Stem Cell's Secretome Delivery Systems. Adv Pharm Bull 2023; 13:244-258. [PMID: 37342369 PMCID: PMC10278206 DOI: 10.34172/apb.2023.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 10/05/2021] [Accepted: 12/31/2021] [Indexed: 09/01/2023] Open
Abstract
Stem cells' secretome contains biomolecules that are ready to give therapeutic activities. However, the biomolecules should not be administered directly because of their in vivo instability. They can be degraded by enzymes or seep into other tissues. There have been some advancements in localized and stabilized secretome delivery systems, which have increased their effectiveness. Fibrous, in situ, or viscoelastic hydrogel, sponge-scaffold, bead powder/ suspension, and bio-mimetic coating can maintain secretome retention in the target tissue and prolong the therapy by sustained release. Porosity, young's modulus, surface charge, interfacial interaction, particle size, adhesiveness, water absorption ability, in situ gel/film, and viscoelasticity of the preparation significantly affect the quality, quantity, and efficacy of the secretome. Therefore, the dosage forms, base materials, and characteristics of each system need to be examined to develop a more optimal secretome delivery system. This article discusses the clinical obstacles and potential solutions for secretome delivery, characterization of delivery systems, and devices used or potentially used in secretome delivery for therapeutic applications. This article concludes that secretome delivery for various organ therapies necessitates the use of different delivery systems and bases. Coating, muco-, and cell-adhesive systems are required for systemic delivery and to prevent metabolism. The lyophilized form is required for inhalational delivery, and the lipophilic system can deliver secretomes across the blood-brain barrier. Nano-sized encapsulation and surface-modified systems can deliver secretome to the liver and kidney. These dosage forms can be administered using devices such as a sprayer, eye drop, inhaler, syringe, and implant to improve their efficacy through dosing, direct delivery to target tissues, preserving stability and sterility, and reducing the immune response.
Collapse
Affiliation(s)
- Abd. Kakhar Umar
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor 45363, Indonesia
| |
Collapse
|
2
|
Sahan AZ, Baday M, Patel CB. Biomimetic Hydrogels in the Study of Cancer Mechanobiology: Overview, Biomedical Applications, and Future Perspectives. Gels 2022; 8:gels8080496. [PMID: 36005097 PMCID: PMC9407355 DOI: 10.3390/gels8080496] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/26/2022] [Accepted: 07/02/2022] [Indexed: 11/18/2022] Open
Abstract
Hydrogels are biocompatible polymers that are tunable to the system under study, allowing them to be widely used in medicine, bioprinting, tissue engineering, and biomechanics. Hydrogels are used to mimic the three-dimensional microenvironment of tissues, which is essential to understanding cell–cell interactions and intracellular signaling pathways (e.g., proliferation, apoptosis, growth, and survival). Emerging evidence suggests that the malignant properties of cancer cells depend on mechanical cues that arise from changes in their microenvironment. These mechanobiological cues include stiffness, shear stress, and pressure, and have an impact on cancer proliferation and invasion. The hydrogels can be tuned to simulate these mechanobiological tissue properties. Although interest in and research on the biomedical applications of hydrogels has increased in the past 25 years, there is still much to learn about the development of biomimetic hydrogels and their potential applications in biomedical and clinical settings. This review highlights the application of hydrogels in developing pre-clinical cancer models and their potential for translation to human disease with a focus on reviewing the utility of such models in studying glioblastoma progression.
Collapse
Affiliation(s)
- Ayse Z. Sahan
- Biomedical Sciences Graduate Program, Department of Pharmacology, School of Medicine, University California at San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Murat Baday
- Department of Neurology and Neurological Sciences, School of Medicine, Stanford University, Stanford, CA 94305, USA
- Precision Health and Integrated Diagnostics Center, School of Medicine, Stanford University, Stanford, CA 94305, USA
- Correspondence: (M.B.); (C.B.P.)
| | - Chirag B. Patel
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (GSBS), Houston, TX 77030, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (GSBS), Houston, TX 77030, USA
- Correspondence: (M.B.); (C.B.P.)
| |
Collapse
|
3
|
Gangadaran P, Rajendran RL, Oh JM, Oh EJ, Hong CM, Chung HY, Lee J, Ahn BC. Identification of Angiogenic Cargo in Extracellular Vesicles Secreted from Human Adipose Tissue-Derived Stem Cells and Induction of Angiogenesis In Vitro and In Vivo. Pharmaceutics 2021; 13:495. [PMID: 33916460 PMCID: PMC8066163 DOI: 10.3390/pharmaceutics13040495] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 03/29/2021] [Accepted: 03/29/2021] [Indexed: 12/15/2022] Open
Abstract
Angiogenesis is defined as the generation of new blood vessels or the sprouting of endothelial cells from a pre-existing vascular network. Angiogenesis occurs during the growth and development of an organism, the response of organs or tissues to injury, and during cancer development and progression. The majority of studies on stem-cell-derived extracellular vesicles (EVs) have used cell lines, and have primarily focused on well-known solitary proteins. Here, we isolated stem cells from human adipose tissue (ADSCs), and we isolated EVs from them (ADSC-EVs). The ADSC-EVs were characterised and 20 angiogenic proteins were analysed using an angiogenic antibody array. Furthermore, we analysed the ability of ADSC-EVs to induce angiogenesis in vitro and in vivo. ADSC-EVs were positive for CD81 and negative for GM130, calnexin, and cytochrome-C. ADSC-EVs showed typical EV spherical morphology and were ~200 nm in size. ADSC-EVs were found to contain angiogenic proteins as cargo, among which interleukin 8 (IL-8) was the most abundant, followed by chemokine (C-C motif) ligand 2 (CCL2), a tissue inhibitor of metalloproteinases 1 (TIMP-1), TIMP-2, and vascular endothelial growth factor-D (VEGF-D). ADSC-EVs treatment increased the proliferation, migration, total vessel length, total number of junctions, and junction density of endothelial cells in vitro. The results of an in vivo Matrigel plug assay revealed that ADSC-EVs induced more blood vessels in the Matrigel compared with the control. These results demonstrate that ADSC-EVs contain angiogenic proteins as cargo and promote angiogenesis in vitro and in vivo. Therefore, ADSC-EVs have potential for therapeutic use in ischaemia.
Collapse
Affiliation(s)
- Prakash Gangadaran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (P.G.); (H.Y.C.)
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (R.L.R.); (J.M.O.); (J.L.)
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (R.L.R.); (J.M.O.); (J.L.)
| | - Ji Min Oh
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (R.L.R.); (J.M.O.); (J.L.)
| | - Eun Jung Oh
- Department of Plastic and Reconstructive Surgery, CMRI, School of Medicine, Kyungpook National University Hospital, Kyungpook National University, Daegu 41944, Korea;
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University Hospital, Kyungpook National University, 680 Gukchaebosangro, Junggu, Daegu 41944, Korea;
| | - Ho Yun Chung
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (P.G.); (H.Y.C.)
- Department of Plastic and Reconstructive Surgery, CMRI, School of Medicine, Kyungpook National University Hospital, Kyungpook National University, Daegu 41944, Korea;
| | - Jaetae Lee
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (R.L.R.); (J.M.O.); (J.L.)
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University Hospital, Kyungpook National University, 680 Gukchaebosangro, Junggu, Daegu 41944, Korea;
| | - Byeong-Cheol Ahn
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (P.G.); (H.Y.C.)
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea; (R.L.R.); (J.M.O.); (J.L.)
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University Hospital, Kyungpook National University, 680 Gukchaebosangro, Junggu, Daegu 41944, Korea;
| |
Collapse
|
4
|
Taghiyar L, Jahangir S, Khozaei Ravari M, Shamekhi MA, Eslaminejad MB. Cartilage Repair by Mesenchymal Stem Cell-Derived Exosomes: Preclinical and Clinical Trial Update and Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1326:73-93. [PMID: 33629260 DOI: 10.1007/5584_2021_625] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Osteoarthritis (OA) and other degenerative joint diseases are characterized by articular cartilage destruction, synovial inflammation, sclerosis of subchondral bone, and loss of extracellular matrix (ECM). Worldwide, these diseases are major causes of disability. Cell therapies have been considered to be the best therapeutic strategies for long-term treatment of articular cartilage diseases. It has been suggested that the mechanism of stem cell-based therapy is related to paracrine secretion of extracellular vesicles (EVs), which are recognized as the main secretion factors of stem cells. EVs, and in particular the subclass exosomes (Exos), are novel therapeutic approaches for treatment of cartilage lesions and OA. The results of recent studies have shown that EVs isolated from mesenchymal stem cells (MSCs) could inhibit OA progression. EVs isolated from various stem cell sources, such as MSCs, may contribute to tissue regeneration of the limbs, skin, heart, and other tissues. Here, we summarize recent findings of preclinical and clinical studies on different MSC-derived EVs and their effectiveness as a treatment for damaged cartilage. The Exos isolation techniques in OA treatment are also highlighted.
Collapse
Affiliation(s)
- Leila Taghiyar
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Shahrbano Jahangir
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mojtaba Khozaei Ravari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
5
|
Kshitiz, Ellison DD, Suhail Y, Afzal J, Woo L, Kilic O, Spees J, Levchenko A. Dynamic secretome of bone marrow-derived stromal cells reveals a cardioprotective biochemical cocktail. Proc Natl Acad Sci U S A 2019; 116:14374-14383. [PMID: 31239339 PMCID: PMC6628676 DOI: 10.1073/pnas.1902598116] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Transplanted stromal cells have demonstrated considerable promise as therapeutic agents in diverse disease settings. Paracrine signaling can be an important mediator of these therapeutic effects at the sites of acute or persistent injury and inflammation. As many stromal cell types, including bone marrow-derived stromal cells (BMSCs), display tissue-specific responses, there is a need to explore their secretory dynamics in the context of tissue and injury type. Paracrine signals are not static, and could encode contextual dynamics in the kinetic changes of the concentrations of the secreted ligands. However, precise measurement of dynamic and context-specific cellular secretory signatures, particularly in adherent cells, remains challenging. Here, by creating an experimental and computational analysis platform, we reconstructed dynamic secretory signatures of cells based on a very limited number of time points. By using this approach, we demonstrate that the secretory signatures of CD133-positive BMSCs are uniquely defined by distinct biological contexts, including signals from injured cardiac cells undergoing oxidative stress, characteristic of cardiac infarction. Furthermore, we show that the mixture of recombinant factors reproducing the dynamics of BMSC-generated secretion can mediate a highly effective rescue of cells injured by oxidative stress and an improved cardiac output. These results support the importance of the dynamic multifactorial paracrine signals in mediating remedial effects of stromal stem cells, and pave the way for stem cell-inspired cell-free treatments of cardiac and other injuries.
Collapse
Affiliation(s)
- Kshitiz
- Yale Institute of Systems Biology, Yale University, West Haven, CT 06516;
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030
| | - David D Ellison
- Department of Medicine, The Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Yasir Suhail
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030
| | - Junaid Afzal
- Department of Cardiology, University of California, San Francisco, CA 94115
| | - Laura Woo
- Department of Medicine, The Johns Hopkins School of Medicine, Baltimore, MD 21205
| | - Onur Kilic
- Yale Institute of Systems Biology, Yale University, West Haven, CT 06516
| | - Jeffrey Spees
- Department of Cellular Molecular and Biomedical Sciences, University of Vermont, Burlington, VT 05405
| | - Andre Levchenko
- Yale Institute of Systems Biology, Yale University, West Haven, CT 06516;
| |
Collapse
|
6
|
Ahmed HH, Aglan HA, Mabrouk M, Abd-Rabou AA, Beherei HH. Enhanced mesenchymal stem cell proliferation through complexation of selenium/titanium nanocomposites. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2019; 30:24. [PMID: 30747346 DOI: 10.1007/s10856-019-6224-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 01/19/2019] [Indexed: 06/09/2023]
Abstract
The main target of this work was to explore the proliferative impact of selenium dioxide nanoparticles (SeO2) and selenium dioxide/titanium dioxide nanocomposites (Se/Ti (I), (II) and (III)) on mesenchymal stem cells (MSCs). For this purpose, SeO2 and Se/Ti (I), (II) and (III) were prepared by facile one step method and characterized by transmission electron microscopy (TEM), Zetasizer, X-ray diffraction (XRD), Fourier transform infrared spectroscopy (FT-IR) and scanning electron microscope (SEM) along with energy-dispersive X-ray spectrometry (EDX) with reference to SeO2 nanoparticles. Also, MSCs were isolated from rat bone marrow (BM-MSCs) and adipose tissue (ADSCs), propagated and characterized by flow cytometry. Thereafter, the proliferative effect of the fabricated nanomaterials was investigated by MTT assay. The TEM and DLS results, revealed that the average particle size of the suggested nanomaterials was in nanoscale. XRD pattern showed well crystalline structure for SeO2 nanoparticles and Se/Ti (I), (II) and (III) nanocomposites; the decreasing of the crystalline phase was observed by increasing the wt% of TiO2. The designed nanomaterials showed proliferative effects on MSCs with the most prominent effect exerted by 2 µg/ml of Se/Ti (III) and 5 µg/ml of Se/Ti (II) for ADSCs and 20 µg/ml of Se/Ti (II) and 10 µg/ml of Se/Ti (III) for BM-MSCs. Therefore, these newly designed nanomaterials have a promising influence on MSCs proliferation and they are recommended to be utilized in the filed of tissue engineering.
Collapse
Affiliation(s)
- Hanaa H Ahmed
- Hormones Department, National Research Centre, Giza, Egypt.
- Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, Giza, Egypt.
| | - Hadeer A Aglan
- Hormones Department, National Research Centre, Giza, Egypt
- Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, Giza, Egypt
| | - Mostafa Mabrouk
- Refractories, Ceramics and Building Materials Department, National Research Centre, Giza, Egypt
| | - Ahmed A Abd-Rabou
- Hormones Department, National Research Centre, Giza, Egypt
- Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, Giza, Egypt
| | - Hanan H Beherei
- Refractories, Ceramics and Building Materials Department, National Research Centre, Giza, Egypt
| |
Collapse
|
7
|
Crivelli B, Chlapanidas T, Perteghella S, Lucarelli E, Pascucci L, Brini AT, Ferrero I, Marazzi M, Pessina A, Torre ML. Mesenchymal stem/stromal cell extracellular vesicles: From active principle to next generation drug delivery system. J Control Release 2017; 262:104-117. [PMID: 28736264 DOI: 10.1016/j.jconrel.2017.07.023] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 07/12/2017] [Accepted: 07/15/2017] [Indexed: 02/06/2023]
Abstract
It has been demonstrated that the biological effector of mesenchymal stem/stromal cells (MSCs) is their secretome, which is composed of a heterogeneous pool of bioactive molecules, partially enclosed in extracellular vesicles (EVs). Therefore, the MSC secretome (including EVs) has been recently proposed as possible alternative to MSC therapy. The secretome can be considered as a protein-based biotechnological product, it is probably safer compared with living/cycling cells, it presents virtually lower tumorigenic risk, and it can be handled, stored and sterilized as an Active Pharmaceutical/Principle Ingredient (API). EVs retain some structural and technological analogies with synthetic drug delivery systems (DDS), even if their potential clinical application is also limited by the absence of reproducible/scalable isolation methods and Good Manufacturing Practice (GMP)-compliant procedures. Notably, EVs secreted by MSCs preserve some of their parental cell features such as homing, immunomodulatory and regenerative potential. This review focuses on MSCs and their EVs as APIs, as well as DDS, considering their ability to reach inflamed and damaged tissues and to prolong the release of encapsulated drugs. Special attention is devoted to the illustration of innovative therapeutic approaches in which nanomedicine is successfully combined with stem cell therapy, thus creating a novel class of "next generation drug delivery systems."
Collapse
Affiliation(s)
- Barbara Crivelli
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
| | - Theodora Chlapanidas
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
| | - Sara Perteghella
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
| | - Enrico Lucarelli
- Osteoarticular Regeneration Laboratory, 3rd Orthopaedic and Traumatologic Clinic, Rizzoli Orthopedic Institute, Via di Barbiano 1/10, 40136 Bologna, Italy.
| | - Luisa Pascucci
- Veterinary Medicine Department, University of Perugia, Via San Costanzo 4, 06126 Perugia, Italy.
| | - Anna Teresa Brini
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via Pascal 36, 20100 Milan, Italy; I.R.C.C.S. Galeazzi Orthopedic Institute, Via Riccardo Galeazzi 4, 20161 Milan, Italy.
| | - Ivana Ferrero
- Paediatric Onco-Haematology, Stem Cell Transplantation and Cellular Therapy Division, City of Science and Health of Turin, Regina Margherita Children's Hospital, Piazza Polonia 94, 10126 Turin, Italy; Department of Public Health and Paediatrics, University of Turin, Piazza Polonia 94, 10126 Turin, Italy.
| | - Mario Marazzi
- Tissue Therapy Unit, ASST Niguarda Hospital, Piazza Ospedale Maggiore 3, 20162 Milan, Italy.
| | - Augusto Pessina
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via Pascal 36, 20100 Milan, Italy.
| | - Maria Luisa Torre
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy.
| | | |
Collapse
|
8
|
Chueng STD, Yang L, Zhang Y, Lee KB. Multidimensional nanomaterials for the control of stem cell fate. NANO CONVERGENCE 2016; 3:23. [PMID: 28191433 PMCID: PMC5271342 DOI: 10.1186/s40580-016-0083-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 09/07/2016] [Indexed: 05/17/2023]
Abstract
Current stem cell therapy suffers low efficiency in giving rise to differentiated cell lineages, which can replace the original damaged cells. Nanomaterials, on the other hand, provide unique physical size, surface chemistry, conductivity, and topographical microenvironment to regulate stem cell differentiation through multidimensional approaches to facilitate gene delivery, cell-cell, and cell-ECM interactions. In this review, nanomaterials are demonstrated to work both alone and synergistically to guide selective stem cell differentiation. From three different nanotechnology families, three approaches are shown: (1) soluble microenvironmental factors; (2) insoluble physical microenvironment; and (3) nano-topographical features. As regenerative medicine is heavily invested in effective stem cell therapy, this review is inspired to generate discussions in the potential clinical applications of multi-dimensional nanomaterials.
Collapse
Affiliation(s)
- Sy-Tsong Dean Chueng
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 USA
| | - Letao Yang
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 USA
| | - Yixiao Zhang
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 USA
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 USA
| |
Collapse
|
9
|
Nair A, Shen J, Lotfi P, Ko CY, Zhang CC, Tang L. Biomaterial implants mediate autologous stem cell recruitment in mice. Acta Biomater 2011; 7:3887-95. [PMID: 21784181 DOI: 10.1016/j.actbio.2011.06.050] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 05/26/2011] [Accepted: 06/28/2011] [Indexed: 12/29/2022]
Abstract
Autologous stem cells, recognized as the best cells for stem cell therapy, are associated with difficult extraction procedures which often lead to more traumas for the patients and time-consuming laboratory work, which delays their subsequent application. To combat such challenges, it was recently uncovered that, shortly after biomaterial implantation, following the recruitment of inflammatory cells, substantial numbers of mesenchymal stem cells (MSC) and hematopoietic stem cells (HSC) were recruited to the implantation sites. These multipotent MSC could be differentiated into various lineages in vitro. Inflammatory signals may be responsible for the gathering of stem cells, since there is a good relationship between biomaterial-mediated inflammatory responses and stem cell accumulation in vivo. In addition, the treatment with the anti-inflammatory drug dexamethasone substantially reduced the recruitment of both MSC and HSC. The results from this work support that such strategies could be further developed towards localized recruitment and differentiation of progenitor cells. This may permit the future development of autologous stem cell therapies without the need for tedious cell isolation, culture and transplantation.
Collapse
Affiliation(s)
- A Nair
- Bioengineering Department, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | | | | | | | | | | |
Collapse
|