151
|
Babenko VA, Silachev DN, Zorova LD, Pevzner IB, Khutornenko AA, Plotnikov EY, Sukhikh GT, Zorov DB. Improving the Post-Stroke Therapeutic Potency of Mesenchymal Multipotent Stromal Cells by Cocultivation With Cortical Neurons: The Role of Crosstalk Between Cells. Stem Cells Transl Med 2015; 4:1011-20. [PMID: 26160961 DOI: 10.5966/sctm.2015-0010] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 05/26/2015] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED The goal of the present study was to maximally alleviate the negative impact of stroke by increasing the therapeutic potency of injected mesenchymal multipotent stromal cells (MMSCs). To pursue this goal, the intercellular communications of MMSCs and neuronal cells were studied in vitro. As a result of cocultivation of MMSCs and rat cortical neurons, we proved the existence of intercellular contacts providing transfer of cellular contents from one cell to another. We present evidence of intercellular exchange with fluorescent probes specifically occupied by cytosol with preferential transfer from neurons toward MMSCs. In contrast, we observed a reversed transfer of mitochondria (from MMSCs to neural cells). Intravenous injection of MMSCs in a postischemic period alleviated the pathological indexes of a stroke, expressed as a lower infarct volume in the brain and partial restoration of neurological status. Also, MMSCs after cocultivation with neurons demonstrated more profound neuroprotective effects than did unprimed MMSCs. The production of the brain-derived neurotrophic factor was slightly increased in MMSCs, and the factor itself was redistributed in these cells after cocultivation. The level of Miro1 responsible for intercellular traffic of mitochondria was increased in MMSCs after cocultivation. We conclude that the exchange by cellular compartments between neural and stem cells improves MMSCs' protective abilities for better rehabilitation after stroke. This could be used as an approach to enhance the therapeutic benefits of stem cell therapy to the damaged brain. SIGNIFICANCE The idea of priming stem cells before practical use for clinical purposes was applied. Thus, cells were preconditioned by coculturing them with the targeted cells (i.e., neurons for the treatment of brain pathological features) before the transfusion of stem cells to the organism. Such priming improved the capacity of stem cells to treat stroke. Some additional minimal study will be required to develop a detailed protocol for coculturing followed by cell separation.
Collapse
Affiliation(s)
- Valentina A Babenko
- Faculty of Bioengineering and Bioinformatics, A.N. Belozersky Institute of Physico-Chemical Biology, and International Laser Center, Lomonosov Moscow State University, Moscow, Russian Federation; Gynecology and Perinatology, Research Center of Obstetrics, Moscow, Russian Federation
| | - Denis N Silachev
- Faculty of Bioengineering and Bioinformatics, A.N. Belozersky Institute of Physico-Chemical Biology, and International Laser Center, Lomonosov Moscow State University, Moscow, Russian Federation; Gynecology and Perinatology, Research Center of Obstetrics, Moscow, Russian Federation
| | - Ljubava D Zorova
- Faculty of Bioengineering and Bioinformatics, A.N. Belozersky Institute of Physico-Chemical Biology, and International Laser Center, Lomonosov Moscow State University, Moscow, Russian Federation; Gynecology and Perinatology, Research Center of Obstetrics, Moscow, Russian Federation
| | - Irina B Pevzner
- Faculty of Bioengineering and Bioinformatics, A.N. Belozersky Institute of Physico-Chemical Biology, and International Laser Center, Lomonosov Moscow State University, Moscow, Russian Federation; Gynecology and Perinatology, Research Center of Obstetrics, Moscow, Russian Federation
| | - Anastasia A Khutornenko
- Faculty of Bioengineering and Bioinformatics, A.N. Belozersky Institute of Physico-Chemical Biology, and International Laser Center, Lomonosov Moscow State University, Moscow, Russian Federation; Gynecology and Perinatology, Research Center of Obstetrics, Moscow, Russian Federation
| | - Egor Y Plotnikov
- Faculty of Bioengineering and Bioinformatics, A.N. Belozersky Institute of Physico-Chemical Biology, and International Laser Center, Lomonosov Moscow State University, Moscow, Russian Federation; Gynecology and Perinatology, Research Center of Obstetrics, Moscow, Russian Federation
| | - Gennady T Sukhikh
- Faculty of Bioengineering and Bioinformatics, A.N. Belozersky Institute of Physico-Chemical Biology, and International Laser Center, Lomonosov Moscow State University, Moscow, Russian Federation; Gynecology and Perinatology, Research Center of Obstetrics, Moscow, Russian Federation
| | - Dmitry B Zorov
- Faculty of Bioengineering and Bioinformatics, A.N. Belozersky Institute of Physico-Chemical Biology, and International Laser Center, Lomonosov Moscow State University, Moscow, Russian Federation; Gynecology and Perinatology, Research Center of Obstetrics, Moscow, Russian Federation
| |
Collapse
|
152
|
Physical Activity Increases the Total Number of Bone-Marrow-Derived Mesenchymal Stem Cells, Enhances Their Osteogenic Potential, and Inhibits Their Adipogenic Properties. Stem Cells Int 2015; 2015:379093. [PMID: 26167185 PMCID: PMC4488015 DOI: 10.1155/2015/379093] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 05/12/2015] [Accepted: 05/18/2015] [Indexed: 01/09/2023] Open
Abstract
Aging and sedentary lifestyle are common nowadays and are associated with the increasing number of chronic diseases. Thus, physical activity is recommended as one of three healthy behavior factors that play a crucial role in health prophylaxis. In the present study, we were interested whether physical activity influences the number and potential of bone-marrow-derived mesenchymal stem cells BMMSCs. In this study, four-week-old male C57Bl/6 mice were trained on a treadmill at progressive speeds over a 5-week period. Comparisons made between exercised (EX) and sedentary animal groups revealed (i) significantly higher number of MSCs in EX animals, (ii) elevated alkaline phosphatase (ALP) activity, (iii) increased level of osteopontin (OPN) and osteocalcin (OCL), and (iv) reduced marrow cavity fat. The results obtained support the thesis that EX may play a substantial role in the regeneration of mesenchymal tissues. Therefore, EX may represent a novel, nonpharmacological strategy of slowing down age-related decline of the musculoskeletal functions.
Collapse
|
153
|
Gray A, Maguire T, Schloss R, Yarmush ML. Identification of IL-1β and LPS as optimal activators of monolayer and alginate-encapsulated mesenchymal stromal cell immunomodulation using design of experiments and statistical methods. Biotechnol Prog 2015; 31:1058-70. [PMID: 25958832 DOI: 10.1002/btpr.2103] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 04/23/2015] [Indexed: 12/13/2022]
Abstract
Induction of therapeutic mesenchymal stromal cell (MSC) function is dependent upon activating factors present in diseased or injured tissue microenvironments. These functions include modulation of macrophage phenotype via secreted molecules including prostaglandin E2 (PGE2). Many approaches aim to optimize MSC-based therapies, including preconditioning using soluble factors and cell immobilization in biomaterials. However, optimization of MSC function is usually inefficient as only a few factors are manipulated in parallel. We utilized fractional factorial design of experiments to screen a panel of 6 molecules (lipopolysaccharide [LPS], polyinosinic-polycytidylic acid [poly(I:C)], interleukin [IL]-6, IL-1β, interferon [IFN]-β, and IFN-γ), individually and in combinations, for the upregulation of MSC PGE2 secretion and attenuation of macrophage secretion of tumor necrosis factor (TNF)-α, a pro-inflammatory molecule, by activated-MSC conditioned medium (CM). We used multivariable linear regression (MLR) and analysis of covariance to determine differences in functions of optimal factors on monolayer MSCs and alginate-encapsulated MSCs (eMSCs). The screen revealed that LPS and IL-1β potently activated monolayer MSCs to enhance PGE2 production and attenuate macrophage TNF-α. Activation by LPS and IL-1β together synergistically increased MSC PGE2, but did not synergistically reduce macrophage TNF-α. MLR and covariate analysis revealed that macrophage TNF-α was strongly dependent on the MSC activation factor, PGE2 level, and macrophage donor but not MSC culture format (monolayer versus encapsulated). The results demonstrate the feasibility and utility of using statistical approaches for higher throughput cell analysis. This approach can be extended to develop activation schemes to maximize MSC and MSC-biomaterial functions prior to transplantation to improve MSC therapies.
Collapse
Affiliation(s)
- Andrea Gray
- Dept. of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854
| | - Timothy Maguire
- Dept. of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854
| | - Rene Schloss
- Dept. of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854
| | - Martin L Yarmush
- Dept. of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854
| |
Collapse
|
154
|
Agadi S, Shetty AK. Concise Review: Prospects of Bone Marrow Mononuclear Cells and Mesenchymal Stem Cells for Treating Status Epilepticus and Chronic Epilepsy. Stem Cells 2015; 33:2093-103. [PMID: 25851047 DOI: 10.1002/stem.2029] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 03/16/2015] [Indexed: 12/22/2022]
Abstract
Mononuclear cells (MNCs) and mesenchymal stem cells (MSCs) derived from the bone marrow and other sources have received significant attention as donor cells for treating various neurological disorders due to their robust neuroprotective and anti-inflammatory effects. Moreover, it is relatively easy to procure these cells from both autogenic and allogenic sources. Currently, there is considerable interest in examining the usefulness of these cells for conditions such as status epilepticus (SE) and chronic epilepsy. A prolonged seizure activity in SE triggers neurodegeneration in the limbic brain areas, which elicits epileptogenesis and evolves into a chronic epileptic state. Because of their potential for providing neuroprotection, diminishing inflammation and curbing epileptogenesis, early intervention with MNCs or MSCs appears attractive for treating SE as such effects may restrain the development of chronic epilepsy typified by spontaneous seizures and learning and memory impairments. Delayed administration of these cells after SE may also be useful for easing spontaneous seizures and cognitive dysfunction in chronic epilepsy. This concise review evaluates the current knowledge and outlook pertaining to MNC and MSC therapies for SE and chronic epilepsy. In the first section, the behavior of these cells in animal models of SE and their efficacy to restrain neurodegeneration, inflammation, and epileptogenesis are discussed. The competence of these cells for suppressing seizures and improving cognitive function in chronic epilepsy are conferred in the next section. The final segment ponders issues that need to be addressed to pave the way for clinical application of these cells for SE and chronic epilepsy.
Collapse
Affiliation(s)
- Satish Agadi
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, Temple, Texas, USA.,Department of Pediatrics, McLane's Children's Hospital, Baylor Scott & White Health, Temple, Texas, USA
| | - Ashok K Shetty
- Institute for Regenerative Medicine, Texas A&M Health Science Center College of Medicine at Scott & White, Temple, Texas, USA.,Research Service, Olin E. Teague Veterans Affairs Medical Center, Central Texas Veterans Health Care System, Temple, Texas, USA.,Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, Texas, USA
| |
Collapse
|
155
|
Vertès AA. The potential of cytotherapeutics in hematologic reconstitution and in the treatment and prophylaxis of graft-versus-host disease. Chapter II: emerging transformational cytotherapies. Regen Med 2015; 10:345-73. [DOI: 10.2217/rme.15.13] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is a life-saving treatment for inherited anemias, immunodeficiencies or hematologic malignancies. A major complication of allo-HSCT associated with high transplant-related mortality rates is graft-versus-host disease (GvHD). Current and future clinical benefits in HSCT enabled by advances in hematopoietic stem cells, mesenchymal stem cells, Tregs and natural killer cells technologies are reviewed here and discussed. Among these evolutions, based on the need for mesenchymal stem cells to be recruited by an inflammatory environment, the development and use of novel GvHD biomarkers could be explored further to deliver the right pharmaceutical to the right patient at the right time. The successful commercialization of cytotherapeutics to efficiently manage GvHD will create a virtuous ‘halo’ effect for regenerative medicine.
Collapse
Affiliation(s)
- Alain A Vertès
- Sloan Fellow, London Business School, London, UK
- NxR Biotechnologies GmbH, Basel, Switzerland
| |
Collapse
|
156
|
Oh KW, Moon C, Kim HY, Oh SI, Park J, Lee JH, Chang IY, Kim KS, Kim SH. Phase I trial of repeated intrathecal autologous bone marrow-derived mesenchymal stromal cells in amyotrophic lateral sclerosis. Stem Cells Transl Med 2015; 4:590-7. [PMID: 25934946 DOI: 10.5966/sctm.2014-0212] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 02/16/2015] [Indexed: 02/07/2023] Open
Abstract
UNLABELLED Stem cell therapy is an emerging alternative therapeutic or disease-modifying strategy for amyotrophic lateral sclerosis (ALS). The aim of this open-label phase I clinical trial was to evaluate the safety of two repeated intrathecal injections of autologous bone marrow (BM)-derived mesenchymal stromal cells (MSCs) in ALS patients. Eight patients with definite or probable ALS were enrolled. After a 3-month lead-in period, autologous MSCs were isolated two times from the BM at an interval of 26 days and were then expanded in vitro for 28 days and suspended in autologous cerebrospinal fluid. Of the 8 patients, 7 received 2 intrathecal injections of autologous MSCs (1 × 10(6) cells per kg) 26 days apart. Clinical or laboratory measurements were recorded to evaluate the safety 12 months after the first MSC injection. The ALS Functional Rating Scale-Revised (ALSFRS-R), the Appel ALS score, and forced vital capacity were used to evaluate the patients' disease status. One patient died before treatment and was withdrawn from the study. With the exception of that patient, no serious adverse events were observed during the 12-month follow-up period. Most of the adverse events were self-limited or subsided after supportive treatment within 4 days. Decline in the ALSFRS-R score was not accelerated during the 6-month follow-up period. Two repeated intrathecal injections of autologous MSCs were safe and feasible throughout the duration of the 12-month follow-up period. SIGNIFICANCE Stem cell therapy is an emerging alternative therapeutic or disease-modifying strategy for amyotrophic lateral sclerosis (ALS). To the authors' best knowledge, there are no clinical trials to evaluate the safety of repeated intrathecal injections of autologous bone marrow mesenchymal stromal cells in ALS. After the clinical trial (phase I/II) was conducted, the stem cell (HYNR-CS, NEURONATA-R) was included in the revision of the regulations on orphan drug designation (number 160; December 31, 2013) and approved as a New Drug Application (Department of Cell and Gene Therapy 233; July 30, 2014) by the Korean Food and Drug Administration. The phase II trial is expected to be reported later.
Collapse
Affiliation(s)
- Ki-Wook Oh
- Department of Neurology, College of Medicine and Cell Therapy Center for Neurologic Disorders, Hanyang University Hospital, Seoul, Republic of Korea; Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea; Bioengineering Institute, Corestem Inc., Seoul, Republic of Korea
| | - Chanil Moon
- Department of Neurology, College of Medicine and Cell Therapy Center for Neurologic Disorders, Hanyang University Hospital, Seoul, Republic of Korea; Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea; Bioengineering Institute, Corestem Inc., Seoul, Republic of Korea
| | - Hyun Young Kim
- Department of Neurology, College of Medicine and Cell Therapy Center for Neurologic Disorders, Hanyang University Hospital, Seoul, Republic of Korea; Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea; Bioengineering Institute, Corestem Inc., Seoul, Republic of Korea
| | - Sung-Il Oh
- Department of Neurology, College of Medicine and Cell Therapy Center for Neurologic Disorders, Hanyang University Hospital, Seoul, Republic of Korea; Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea; Bioengineering Institute, Corestem Inc., Seoul, Republic of Korea
| | - Jinseok Park
- Department of Neurology, College of Medicine and Cell Therapy Center for Neurologic Disorders, Hanyang University Hospital, Seoul, Republic of Korea; Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea; Bioengineering Institute, Corestem Inc., Seoul, Republic of Korea
| | - Jun Ho Lee
- Department of Neurology, College of Medicine and Cell Therapy Center for Neurologic Disorders, Hanyang University Hospital, Seoul, Republic of Korea; Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea; Bioengineering Institute, Corestem Inc., Seoul, Republic of Korea
| | - In Young Chang
- Department of Neurology, College of Medicine and Cell Therapy Center for Neurologic Disorders, Hanyang University Hospital, Seoul, Republic of Korea; Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea; Bioengineering Institute, Corestem Inc., Seoul, Republic of Korea
| | - Kyung Suk Kim
- Department of Neurology, College of Medicine and Cell Therapy Center for Neurologic Disorders, Hanyang University Hospital, Seoul, Republic of Korea; Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea; Bioengineering Institute, Corestem Inc., Seoul, Republic of Korea
| | - Seung Hyun Kim
- Department of Neurology, College of Medicine and Cell Therapy Center for Neurologic Disorders, Hanyang University Hospital, Seoul, Republic of Korea; Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea; Bioengineering Institute, Corestem Inc., Seoul, Republic of Korea
| |
Collapse
|
157
|
Using magnetic nanoparticles for gene transfer to neural stem cells: stem cell propagation method influences outcomes. J Funct Biomater 2015; 6:259-76. [PMID: 25918990 PMCID: PMC4493511 DOI: 10.3390/jfb6020259] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 04/11/2015] [Accepted: 04/16/2015] [Indexed: 12/17/2022] Open
Abstract
Genetically engineered neural stem cell (NSC) transplants offer a key strategy to augment neural repair by releasing therapeutic biomolecules into injury sites. Genetic modification of NSCs is heavily reliant on viral vectors but cytotoxic effects have prompted development of non-viral alternatives, such as magnetic nanoparticle (MNPs). NSCs are propagated in laboratories as either 3-D suspension “neurospheres” or 2-D adherent “monolayers”. MNPs deployed with oscillating magnetic fields (“magnetofection technology”) mediate effective gene transfer to neurospheres but the efficacy of this approach for monolayers is unknown. It is important to address this issue as oscillating magnetic fields dramatically enhance MNP-based transfection in transplant cells (e.g., astrocytes and oligodendrocyte precursors) propagated as monolayers. We report for the first time that oscillating magnetic fields enhanced MNP-based transfection with reporter and functional (basic fibroblast growth factor; FGF2) genes in monolayer cultures yielding high transfection versus neurospheres. Transfected NSCs showed high viability and could re-form neurospheres, which is important as neurospheres yield higher post-transplantation viability versus monolayer cells. Our results demonstrate that the combination of oscillating magnetic fields and a monolayer format yields the highest efficacy for MNP-mediated gene transfer to NSCs, offering a viable non-viral alternative for genetic modification of this important neural cell transplant population.
Collapse
|
158
|
Abstract
Stem cells have great potential in basic research and are being slowly integrated into toxicological research. This symposium provided an overview of the state of the field, stem cell models, described allogenic stem cell treatments and issues of immunogenicity associated with protein therapeutics, and tehn concentrated on stem cell uses in regenerative medicine focusing on lung and testing strategies on engineered tissues from a pathologist's perspective.
Collapse
Affiliation(s)
- Alan Trounson
- Monash University, Hudson Institute for Medical Research, Clayton, Victoria, Australia
| | | | - Thomas Petersen
- United Therapeutics Corporation, Regenerative Medicine, Research Triangle Park, NC, USA
| | | | - Maralee McVean
- Pre-Clinical Research Services, Inc, Ft Collins, CO, USA
| | | |
Collapse
|
159
|
Pierantozzi E, Badin M, Vezzani B, Curina C, Randazzo D, Petraglia F, Rossi D, Sorrentino V. Human pericytes isolated from adipose tissue have better differentiation abilities than their mesenchymal stem cell counterparts. Cell Tissue Res 2015; 361:769-78. [DOI: 10.1007/s00441-015-2166-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 03/02/2015] [Indexed: 01/07/2023]
|
160
|
Son S, Liang MS, Lei P, Xue X, Furlani EP, Andreadis ST. Magnetofection Mediated Transient NANOG Overexpression Enhances Proliferation and Myogenic Differentiation of Human Hair Follicle Derived Mesenchymal Stem Cells. Bioconjug Chem 2015; 26:1314-27. [PMID: 25685943 DOI: 10.1021/bc5005203] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
We used magnetofection (MF) to achieve high transfection efficiency into human mesenchymal stem cells (MSCs). A custom-made magnet array, matching well-to-well to a 24-well plate, was generated and characterized. Theoretical predictions of magnetic force distribution within each well demonstrated that there was no magnetic field interference among magnets in adjacent wells. An optimized protocol for efficient gene delivery to human hair follicle derived MSCs (hHF-MSCs) was established using an egfp-encoding plasmid, reaching approximately ∼50% transfection efficiency without significant cytotoxicity. Then we applied the optimized MF protocol to express the pluripotency-associated transcription factor NANOG, which was previously shown to reverse the effects of organismal aging on MSC proliferation and myogenic differentiation capacity. Indeed, MF-mediated NANOG delivery increased proliferation and enhanced the differentiation of hHF-MSCs into smooth muscle cells (SMCs). Collectively, our results show that MF can achieve high levels of gene delivery to MSCs and, therefore, may be employed to moderate or reverse the effects of cellular senescence or reprogram cells to the pluripotent state without permanent genetic modification.
Collapse
Affiliation(s)
| | | | | | | | | | - Stelios T Andreadis
- ∥Center of Excellence in Bioinformatics and Life Sciences, Buffalo, New York 14203, United States
| |
Collapse
|
161
|
Gu WG. Genome editing-based HIV therapies. Trends Biotechnol 2015; 33:172-9. [DOI: 10.1016/j.tibtech.2014.12.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 10/17/2014] [Accepted: 12/17/2014] [Indexed: 12/26/2022]
|
162
|
Desai N, Rambhia P, Gishto A. Human embryonic stem cell cultivation: historical perspective and evolution of xeno-free culture systems. Reprod Biol Endocrinol 2015; 13:9. [PMID: 25890180 PMCID: PMC4351689 DOI: 10.1186/s12958-015-0005-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 02/09/2015] [Indexed: 01/23/2023] Open
Abstract
Human embryonic stem cells (hESC) have emerged as attractive candidates for cell-based therapies that are capable of restoring lost cell and tissue function. These unique cells are able to self-renew indefinitely and have the capacity to differentiate in to all three germ layers (ectoderm, endoderm and mesoderm). Harnessing the power of these pluripotent stem cells could potentially offer new therapeutic treatment options for a variety of medical conditions. Since the initial derivation of hESC lines in 1998, tremendous headway has been made in better understanding stem cell biology and culture requirements for maintenance of pluripotency. The approval of the first clinical trials of hESC cells for treatment of spinal cord injury and macular degeneration in 2010 marked the beginning of a new era in regenerative medicine. Yet it was clearly recognized that the clinical utility of hESC transplantation was still limited by several challenges. One of the most immediate issues has been the exposure of stem cells to animal pathogens, during hESC derivation and during in vitro propagation. Initial culture protocols used co-culture with inactivated mouse fibroblast feeder (MEF) or human feeder layers with fetal bovine serum or alternatively serum replacement proteins to support stem cell proliferation. Most hESC lines currently in use have been exposed to animal products, thus carrying the risk of xeno-transmitted infections and immune reaction. This mini review provides a historic perspective on human embryonic stem cell culture and the evolution of new culture models. We highlight the challenges and advances being made towards the development of xeno-free culture systems suitable for therapeutic applications.
Collapse
Affiliation(s)
- Nina Desai
- Department of Obstetrics and Gynecology, Cleveland Clinic, Beachwood, OH, USA.
| | - Pooja Rambhia
- Department of Obstetrics and Gynecology, Cleveland Clinic, Beachwood, OH, USA.
| | - Arsela Gishto
- Department of Obstetrics and Gynecology, Cleveland Clinic, Beachwood, OH, USA.
| |
Collapse
|
163
|
Kumashiro Y, Ishihara J, Umemoto T, Itoga K, Kobayashi J, Shimizu T, Yamato M, Okano T. Stripe-patterned thermo-responsive cell culture dish for cell separation without cell labeling. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2015; 11:681-687. [PMID: 25238273 DOI: 10.1002/smll.201400787] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 08/11/2014] [Indexed: 06/03/2023]
Abstract
A stripe-patterned thermo-responsive surface is prepared to enable cell separation without labeling. The thermo-responsive surface containing a 3 μm striped pattern exhibits various cell adhesion and detachment properties. A mixture of three cell types is separated on the patterned surface based on their distinct cell-adhesion properties, and the composition of the cells is analyzed by flow cytometry.
Collapse
Affiliation(s)
- Yoshikazu Kumashiro
- Institute of Advanced Biomedical Engineering and Science (TWIns), Tokyo Women's Medical University, 8-1 Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | | | | | | | | | | | | | | |
Collapse
|
164
|
Lee S, Choi E, Cha MJ, Hwang KC. Cell adhesion and long-term survival of transplanted mesenchymal stem cells: a prerequisite for cell therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:632902. [PMID: 25722795 PMCID: PMC4333334 DOI: 10.1155/2015/632902] [Citation(s) in RCA: 185] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 01/19/2015] [Indexed: 12/13/2022]
Abstract
The literature provides abundant evidence that mesenchymal stem cells (MSCs) are an attractive resource for therapeutics and have beneficial effects in regenerating injured tissues due to their self-renewal ability and broad differentiation potential. Although the therapeutic potential of MSCs has been proven in both preclinical and clinical studies, several questions have not yet been addressed. A major limitation to the use of MSCs in clinical applications is their poor viability at the site of injury due to the harsh microenvironment and to anoikis driven by the loss of cell adhesion. To improve the survival of the transplanted MSCs, strategies to regulate apoptotic signaling and enhance cell adhesion have been developed, such as pretreatment with cytokines, growth factors, and antiapoptotic molecules, genetic modifications, and hypoxic preconditioning. More appropriate animal models and a greater understanding of the therapeutic mechanisms of MSCs will be required for their successful clinical application. Nevertheless, the development of stem cell therapies using MSCs has the potential to treat degenerative diseases. This review discusses various approaches to improving MSC survival by inhibiting anoikis.
Collapse
Affiliation(s)
- Seahyoung Lee
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do 210-701, Republic of Korea
- Catholic Kwandong University International St. Mary's Hospital, Incheon Metropolitan City 404-834, Republic of Korea
| | - Eunhyun Choi
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do 210-701, Republic of Korea
- Catholic Kwandong University International St. Mary's Hospital, Incheon Metropolitan City 404-834, Republic of Korea
| | - Min-Ji Cha
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do 210-701, Republic of Korea
- Catholic Kwandong University International St. Mary's Hospital, Incheon Metropolitan City 404-834, Republic of Korea
| | - Ki-Chul Hwang
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si, Gangwon-do 210-701, Republic of Korea
- Catholic Kwandong University International St. Mary's Hospital, Incheon Metropolitan City 404-834, Republic of Korea
| |
Collapse
|
165
|
Schwerk A, Altschüler J, Roch M, Gossen M, Winter C, Berg J, Kurtz A, Steiner B. Human adipose-derived mesenchymal stromal cells increase endogenous neurogenesis in the rat subventricular zone acutely after 6-hydroxydopamine lesioning. Cytotherapy 2015; 17:199-214. [DOI: 10.1016/j.jcyt.2014.09.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 09/05/2014] [Accepted: 09/20/2014] [Indexed: 01/07/2023]
|
166
|
Lahiani A, Zahavi E, Netzer N, Ofir R, Pinzur L, Raveh S, Arien-Zakay H, Yavin E, Lazarovici P. Human PLacental eXpanded (PLX) mesenchymal-like adherent stromal cells confer neuroprotection to nerve growth factor (NGF)-differentiated PC12 cells exposed to ischemia by secretion of IL-6 and VEGF. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:422-30. [DOI: 10.1016/j.bbamcr.2014.11.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Revised: 11/03/2014] [Accepted: 11/10/2014] [Indexed: 12/21/2022]
|
167
|
Choi Y, Cox C, Lally K, Li Y. The strategy and method in modulating finger regeneration. Regen Med 2015; 9:231-42. [PMID: 24750063 DOI: 10.2217/rme.13.98] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The tip of the human finger can regenerate if the amputation is distal to the nail bed, usually in young children. Studies in regeneration of rodent digits have shown that regeneration occurs if the amputation is distal to the mid-third phalanx for certain ages. The digit contains many different components, such as muscle, tendon, bone, skin, nerves and blood vessels, which must all be regrown in the proper location in order to restore functionality. The mechanism behind the complex healing/regeneration processes is still under investigation; however, improvements in injured finger regeneration have been gradually developing in animal models over the past few years. This review discusses a few strategies and methods to possibly enhance digit regeneration beyond current natural limits, focusing on aspects including scarless wound healing, cell-based treatments, tissue engineering and electrical stimulation.
Collapse
Affiliation(s)
- Yohan Choi
- Children's Regenerative Medicine, Department of Pediatric Surgery, University of Texas Medical School at Houston, TX 77030, USA
| | | | | | | |
Collapse
|
168
|
Cambier T, Honegger T, Vanneaux V, Berthier J, Peyrade D, Blanchoin L, Larghero J, Théry M. Design of a 2D no-flow chamber to monitor hematopoietic stem cells. LAB ON A CHIP 2015; 15:77-85. [PMID: 25338534 DOI: 10.1039/c4lc00807c] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Hematopoietic stem cells (HSCs) are the most commonly used cell type in cell-based therapy. However, the investigation of their behavior in vitro has been limited by the difficulty of monitoring these non-adherent cells under classical culture conditions. Indeed, fluid flow moves cells away from the video-recording position and prevents single cell tracking over long periods of time. Here we describe a large array of 2D no-flow chambers allowing the monitoring of single HSCs for several days. The chamber design has been optimized to facilitate manufacturing and routine use. The chip contains a single inlet and 800 chambers. The chamber medium can be renewed by diffusion within a few minutes. This allowed us to stain live human HSCs with fluorescent primary antibodies in order to reveal their stage in the hematopoiesis differentiation pathway. Thus we were able to correlate human HSCs' growth rate, polarization and migration to their differentiation stage.
Collapse
Affiliation(s)
- Théo Cambier
- Laboratoire de Physiologie Cellulaire et Végétale, Institut de Recherche en Technologie et Science pour le Vivant, UMR5168, CEA, INRA, CNRS, Université Grenoble-Alpes, Grenoble, France.
| | | | | | | | | | | | | | | |
Collapse
|
169
|
Ooi YY, Dheen ST, Tay SSW. Paracrine effects of mesenchymal stem cells-conditioned medium on microglial cytokines expression and nitric oxide production. Neuroimmunomodulation 2015; 22:233-42. [PMID: 25341618 DOI: 10.1159/000365483] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 06/19/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIM Microglia, the resident macrophages in the central nervous system, secrete various proinflammatory cytokines and undergo proliferation upon activation in various neurodegenerative diseases. Activation of microglia has been implicated in exacerbation of various neurodegenerative diseases. Recently, it has been proposed that mesenchymal stem cells (MSC) have immunosuppressive properties and the potential to moderate inflammation. This study aimed to elucidate the effects of MSC-conditioned medium (MSC-CM) in modulating microglial activation by analyzing microglial proinflammatory and anti-inflammatory factors [interleukin (IL)-6, tumor necrosis factor (TNF)-α, inducible nitric oxide synthase (iNOS) and IL-10], signaling pathway molecules [NFκB, c-Jun N-terminal kinase (JNK) and MKP-1) and NO production. METHODS Immortalized murine microglia cell line, BV2 microglia and primary microglia isolated from C57BL/6 mouse pup brains were used in this study. Mouse MSC were isolated from the male C57BL/6 mouse tibia and fibula. The effects of MSC-CM on the expression of inflammatory cytokines and signaling molecules in microglia were elucidated using RT-PCR, immunofluorescence analysis and Western blot analysis. NO production in microglia was assessed using a Griess kit. RESULTS MSC-CM significantly reduced the mRNA and protein expression levels of proinflammatory cytokines (IL-6 and TNF-α) in microglia activated by lipopolysaccharide (LPS). In addition, MSC-CM significantly reduced the protein expression of NFκB, JNK and c-Jun, but increased the expression levels of IL-10 and MKP-1 in activated BV2 microglia. NO production and iNOS expression by BV2 microglia in MSC-CM were increased. CONCLUSIONS Overall, our findings suggest that MSC immunomodulate microglial activities through paracrine effects.
Collapse
Affiliation(s)
- Yin Yin Ooi
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | | |
Collapse
|
170
|
Kaiser SC, Kraume M, Eibl D, Eibl R. Single-Use Bioreactors for Animal and Human Cells. CELL ENGINEERING 2015. [DOI: 10.1007/978-3-319-10320-4_14] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
171
|
Park K, Nam Y, Choi Y. An agarose gel-based neurosphere culture system leads to enrichment of neuronal lineage cells in vitro. In Vitro Cell Dev Biol Anim 2014; 51:455-62. [PMID: 25539864 DOI: 10.1007/s11626-014-9855-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 12/01/2014] [Indexed: 11/26/2022]
Abstract
Stem cell-based therapy holds great potential especially for neurological disorders. However, clinical applications await further understanding of many aspects of stem cell differentiation and development of technology enabling manipulation of stem cells into desired cell types in the central nervous system. Here, we developed a new method that leads to enrichment of neuronal lineage cells in neural stem cell cultures. The protocol involves cultivation of primary cells derived from the forebrains of rat E18 embryos above a layer of nonadhesive hard agarose gel in the form of neurospheres. In contrast to the neurospheres that were cultured above an anti-adhesive hydrogel layer, the primary cells that were cultured above a layer of agarose gel preferentially differentiated into β-III tubulin-positive neurons when allowed to undergo differentiation in vitro.In an effort to investigate the mechanism behind this observation, we found that the gene expression of a vertebrate neuronal determination gene (neurogenin1) was enhanced in the neurospheres that proliferated above a layer of agarose gel as compared with the control, and the gene expression level of neurogenin1 was quite well correlated with the rigidity of agarose gel. These results indicate that agarose gel can contribute, at least in part, to enrich neuronal progenitors and immature postmitotic neurons during neurosphere formation and may provide additional information to establish efficient protocols for the neural stem cell-based study.
Collapse
Affiliation(s)
- Kyuhee Park
- Gyeonggi Bio-Center, Gyeonggi Institute of Science and Technology Promotion, Suwon, Gyeonggi-do, 443-270, South Korea
| | | | | |
Collapse
|
172
|
Disease-in-a-dish: the contribution of patient-specific induced pluripotent stem cell technology to regenerative rehabilitation. Am J Phys Med Rehabil 2014; 93:S155-68. [PMID: 25122102 DOI: 10.1097/phm.0000000000000141] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Advances in regenerative medicine technologies will lead to dramatic changes in how patients in rehabilitation medicine clinics are treated in the upcoming decades. The multidisciplinary field of regenerative medicine is developing new tools for disease modeling and drug discovery based on induced pluripotent stem cells. This approach capitalizes on the idea of personalized medicine by using the patient's own cells to discover new drugs, increasing the likelihood of a favorable outcome. The search for compounds that can correct disease defects in the culture dish is a conceptual departure from how drug screens were done in the past. This system proposes a closed loop from sample collection from the diseased patient, to in vitro disease model, to drug discovery and Food and Drug Administration approval, to delivering that drug back to the same patient. Here, recent progress in patient-specific induced pluripotent stem cell derivation, directed differentiation toward diseased cell types, and how those cells can be used for high-throughput drug screens are reviewed. Given that restoration of normal function is a driving force in rehabilitation medicine, the authors believe that this drug discovery platform focusing on phenotypic rescue will become a key contributor to therapeutic compounds in regenerative rehabilitation.
Collapse
|
173
|
Scott CT. The case for stem cell counselors. Stem Cell Reports 2014; 4:1-6. [PMID: 25483110 PMCID: PMC4297874 DOI: 10.1016/j.stemcr.2014.10.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 10/30/2014] [Accepted: 10/31/2014] [Indexed: 12/14/2022] Open
Abstract
In this article, Scott describes how a new counseling profession could support clinical sites and patients enrolling in stem cell clinical trials. A possible model is proposed, along with a curriculum that would provide counselors with the tools to address challenges facing the clinical stem cell field. Finally, a candidate recruitment and clinical site interface scheme is offered.
Collapse
|
174
|
Popov B, Petrov N. pRb-E2F signaling in life of mesenchymal stem cells: Cell cycle, cell fate, and cell differentiation. Genes Dis 2014; 1:174-187. [PMID: 30258863 PMCID: PMC6150080 DOI: 10.1016/j.gendis.2014.09.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 09/14/2014] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that can differentiate into various mesodermal lines forming fat, muscle, bone, and other lineages of connective tissue. MSCs possess plasticity and under special metabolic conditions may transform into cells of unusual phenotypes originating from ecto- and endoderm. After transplantation, MSCs release the humoral factors promoting regeneration of the damaged tissue. During last five years, the numbers of registered clinical trials of MSCs have increased about 10 folds. This gives evidence that MSCs present a new promising resource for cell therapy of the most dangerous diseases. The efficacy of the MSCs therapy is limited by low possibilities to regulate their conversion into cells of damaged tissues that is implemented by the pRb-E2F signaling. The widely accepted viewpoint addresses pRb as ubiquitous regulator of cell cycle and tumor suppressor. However, current publications suggest that basic function of the pRb-E2F signaling in development is to regulate cell fate and differentiation. Through facultative and constitutive chromatin modifications, pRb-E2F signaling promotes transient and stable cells quiescence, cell fate choice to differentiate, to senesce, or to die. Loss of pRb is associated with cancer cell fate. pRb regulates cell fate by retaining quiescence of one cell population in favor of commitment of another or by suppression of genes of different cell phenotype. pRb is the founder member of the "pocket protein" family possessing functional redundancy. Critical increase in the efficacy of the MSCs based cell therapy will depend on precise understanding of various aspects of the pRb-E2F signaling.
Collapse
Affiliation(s)
- Boris Popov
- Institute of Cytology, Russian Academy of Sciences, St.Petersburg, 4, Tikhoretsky Av., 194064, Russia
| | | |
Collapse
|
175
|
Jin G, Li K. The electrically conductive scaffold as the skeleton of stem cell niche in regenerative medicine. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2014; 45:671-81. [DOI: 10.1016/j.msec.2014.06.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 04/18/2014] [Accepted: 06/09/2014] [Indexed: 12/13/2022]
|
176
|
Chen BK, Staff NP, Knight AM, Nesbitt JJ, Butler GW, Padley DJ, Parisi JE, Dietz AB, Windebank AJ. A safety study on intrathecal delivery of autologous mesenchymal stromal cells in rabbits directly supporting Phase I human trials. Transfusion 2014; 55:1013-20. [PMID: 25413276 DOI: 10.1111/trf.12938] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 09/25/2014] [Accepted: 09/29/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND There are no effective treatments that slow the progression of neurodegenerative diseases. A major challenge of treatment in neurodegenerative diseases is appropriate delivery of pharmaceuticals into the cerebrospinal fluid (CSF) of affected individuals. Mesenchymal stromal cells (MSCs-either naïve or modified) are a promising therapy in neurodegenerative diseases and may be delivered directly into the CSF where they can reside for months. In this preclinical study, we evaluated the safety of intrathecal autologous MSCs in a rabbit model. STUDY DESIGN AND METHODS Autologous adipose-derived MSCs (or artificial CSF) were delivered intrathecally, either with single or with repeated injections into the foramen magnum of healthy rabbits and monitored for 4 and 12 weeks, respectively. RESULTS Rabbits tolerated injections well and no definitive MSC-related side effects were observed apart from three rabbits that had delayed death secondary to traumatic foramen magnum puncture. Functional assessments and body weights were equivalent between groups. Gross pathology and histology did not reveal any abnormalities or tumor growth. Complete blood count data were normal and there were no differences in CSF interleukin-6 levels in all groups tested. CONCLUSION Our data suggest that intrathecal delivery of autologous MSCs is safe in a rabbit model. Data from this study have supported two successful investigational new drug applications to the Food and Drug Administration, resulting in the initiation of two clinical trials using autologous MSCs in amyotrophic lateral sclerosis and multiple system atrophy.
Collapse
Affiliation(s)
- Bingkun K Chen
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | - Nathan P Staff
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Greg W Butler
- Human Cell Therapy Laboratory, Mayo Clinic, Rochester, Minnesota
| | - Douglas J Padley
- Human Cell Therapy Laboratory, Mayo Clinic, Rochester, Minnesota
| | - Joseph E Parisi
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Allan B Dietz
- Human Cell Therapy Laboratory, Mayo Clinic, Rochester, Minnesota
| | | |
Collapse
|
177
|
Kanherkar RR, Bhatia-Dey N, Makarev E, Csoka AB. Cellular reprogramming for understanding and treating human disease. Front Cell Dev Biol 2014; 2:67. [PMID: 25429365 PMCID: PMC4228919 DOI: 10.3389/fcell.2014.00067] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 10/27/2014] [Indexed: 12/15/2022] Open
Abstract
In the last two decades we have witnessed a paradigm shift in our understanding of cells so radical that it has rewritten the rules of biology. The study of cellular reprogramming has gone from little more than a hypothesis, to applied bioengineering, with the creation of a variety of important cell types. By way of metaphor, we can compare the discovery of reprogramming with the archeological discovery of the Rosetta stone. This stone slab made possible the initial decipherment of Egyptian hieroglyphics because it allowed us to see this language in a way that was previously impossible. We propose that cellular reprogramming will have an equally profound impact on understanding and curing human disease, because it allows us to perceive and study molecular biological processes such as differentiation, epigenetics, and chromatin in ways that were likewise previously impossible. Stem cells could be called “cellular Rosetta stones” because they allow also us to perceive the connections between development, disease, cancer, aging, and regeneration in novel ways. Here we present a comprehensive historical review of stem cells and cellular reprogramming, and illustrate the developing synergy between many previously unconnected fields. We show how stem cells can be used to create in vitro models of human disease and provide examples of how reprogramming is being used to study and treat such diverse diseases as cancer, aging, and accelerated aging syndromes, infectious diseases such as AIDS, and epigenetic diseases such as polycystic ovary syndrome. While the technology of reprogramming is being developed and refined there have also been significant ongoing developments in other complementary technologies such as gene editing, progenitor cell production, and tissue engineering. These technologies are the foundations of what is becoming a fully-functional field of regenerative medicine and are converging to a point that will allow us to treat almost any disease.
Collapse
Affiliation(s)
- Riya R Kanherkar
- Epigenetics Laboratory, Department of Anatomy, Howard University Washington, DC, USA
| | - Naina Bhatia-Dey
- Epigenetics Laboratory, Department of Anatomy, Howard University Washington, DC, USA
| | - Evgeny Makarev
- InSilico Medicine, Emerging Technology Center, Johns Hopkins University Eastern Baltimore, MD, USA
| | - Antonei B Csoka
- Epigenetics Laboratory, Department of Anatomy, Howard University Washington, DC, USA
| |
Collapse
|
178
|
Duffy BA, Weitz AJ, Lee JH. In vivo imaging of transplanted stem cells in the central nervous system. Curr Opin Genet Dev 2014; 28:83-8. [PMID: 25461455 DOI: 10.1016/j.gde.2014.09.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 09/14/2014] [Indexed: 12/11/2022]
Abstract
In vivo imaging is increasingly being utilized in studies investigating stem cell-based treatments for neurological disorders. Direct labeling is used in preclinical and clinical studies to track the fate of transplanted cells. To further determine cell viability, experimental studies are able to take advantage of reporter gene technologies. Structural and functional brain imaging can also be used alongside cell imaging as biomarkers of treatment efficacy. Furthermore, it is possible that new imaging techniques could be used to monitor functional integration of stem cell-derived cells with the host nervous system. In this review, we examine recent developments in these areas and identify promising directions for future research at the interface of stem cell therapies and neuroimaging.
Collapse
Affiliation(s)
- Ben A Duffy
- Department of Neurology & Neurological Sciences, Stanford University, CA 94305, USA
| | - Andrew J Weitz
- Department of Bioengineering, Stanford University, CA 94305, USA
| | - Jin Hyung Lee
- Department of Neurology & Neurological Sciences, Stanford University, CA 94305, USA; Department of Bioengineering, Stanford University, CA 94305, USA; Department of Neurosurgery, Stanford University, CA 94305, USA; Department of Electrical Engineering, Stanford University, CA 94305, USA.
| |
Collapse
|
179
|
Sadhukha T, O'Brien TD, Prabha S. Nano-engineered mesenchymal stem cells as targeted therapeutic carriers. J Control Release 2014; 196:243-51. [PMID: 25456830 DOI: 10.1016/j.jconrel.2014.10.015] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 10/07/2014] [Accepted: 10/15/2014] [Indexed: 12/18/2022]
Abstract
Poor availability in deep-seated solid tumors is a significant challenge that limits the effectiveness of currently used anticancer drugs. Approaches that can specifically enhance drug delivery to the tumor tissue can potentially improve therapeutic efficacy. In our current studies, we used nano-engineered mesenchymal stem cells (nano-engineered MSCs) as tumor-targeted therapeutic carriers. In addition to their exquisite tumor homing capabilities, MSCs overexpress efflux transporters such as P-glycoprotein and are highly drug resistant. The inherent tumor-tropic and drug-resistant properties make MSCs ideal carriers for toxic payload. Nano-engineered MSCs were prepared by treating human MSCs with drug-loaded polymeric nanoparticles. Incorporating nanoparticles in MSCs did not affect their viability, differentiation or migration potential. Nano-engineered MSCs induced dose-dependent cytotoxicity in A549 lung adenocarcinoma cells and MA148 ovarian cancer cells in vitro. An orthotopic A549 lung tumor model was used to monitor the in vivo distribution of nanoengineered MSCs. Intravenous injection of nanoparticles resulted in non-specific biodistribution, with significant accumulation in the liver and spleen while nano-engineered MSCs demonstrated selective accumulation and retention in lung tumors. These studies demonstrate the feasibility of developing nano-engineered MSCs loaded with high concentration of anticancer agents without affecting their tumor-targeting or drug resistance properties.
Collapse
Affiliation(s)
- Tanmoy Sadhukha
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, 308 Harvard Street SE, Minneapolis, MN 55455, USA
| | - Timothy D O'Brien
- Stem Cell Institute and Veterinary Population Medicine Department, University of Minnesota, 1365 Gortner Avenue, Saint Paul, MN 55108, USA
| | - Swayam Prabha
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, 308 Harvard Street SE, Minneapolis, MN 55455, USA; Center for Translational Drug Delivery, University of Minnesota, 308 Harvard Street SE, Minneapolis, MN 55455, USA.
| |
Collapse
|
180
|
Skowron K, Tomsia M, Czekaj P. An experimental approach to the generation of human embryonic stem cells equivalents. Mol Biotechnol 2014; 56:12-37. [PMID: 24146427 DOI: 10.1007/s12033-013-9702-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Recently, particular attention has been paid to the human embryonic stem cells (hESC) in the context of their potential application in regenerative medicine; however, ethical concerns prevent their clinical application. Induction of pluripotency in somatic cells seems to be a good alternative for hESC recruitment regarding its potential use in tissue regeneration, disease modeling, and drug screening. Since Yamanaka's team in 2006 restored pluripotent state of somatic cells for the first time, a significant progress has been made in the area of induced pluripotent stem cells (iPSC) generation. Here, we review the current state of knowledge in the issue of techniques applied to establish iPSC. Somatic cell nuclear transfer, cell fusion, cell extracts reprogramming, and techniques of direct reprogramming are described. Retroviral and lentiviral transduction are depicted as ways of cell reprogramming with the use of integrating vectors. Contrary to them, adenoviruses, plasmids, single multiprotein expression vectors, and PiggyBac transposition systems are examples of non-integrative vectors used in iPSC generation protocols. Furthermore, reprogramming with the delivery of specific proteins, miRNA or small chemical compounds are presented. Finally, the changes occurring during the reprogramming process are described. It is concluded that subject to some limitations iPSC could become equivalents for hESC in regenerative medicine.
Collapse
Affiliation(s)
- Katarzyna Skowron
- Students Scientific Society, Medical University of Silesia, Katowice, Poland
| | | | | |
Collapse
|
181
|
Tirotta I, Dichiarante V, Pigliacelli C, Cavallo G, Terraneo G, Bombelli FB, Metrangolo P, Resnati G. (19)F magnetic resonance imaging (MRI): from design of materials to clinical applications. Chem Rev 2014; 115:1106-29. [PMID: 25329814 DOI: 10.1021/cr500286d] [Citation(s) in RCA: 340] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Ilaria Tirotta
- Laboratory of Nanostructured Fluorinated Materials (NFMLab), Department of Chemistry, Materials, and Chemical Engineering "Giulio Natta" and ‡Fondazione Centro Europeo Nanomedicina, Politecnico di Milano , Milan 20131, Italy
| | | | | | | | | | | | | | | |
Collapse
|
182
|
Niemansburg SL, Teraa M, Hesam H, van Delden JJM, Verhaar MC, Bredenoord AL. Stem cell trials for cardiovascular medicine: ethical rationale. Tissue Eng Part A 2014; 20:2567-74. [PMID: 24164351 PMCID: PMC4195508 DOI: 10.1089/ten.tea.2013.0332] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 10/24/2013] [Indexed: 12/29/2022] Open
Abstract
Stem cell-based interventions provide new treatment prospects for many disease conditions, including cardiovascular disorders. Clinical trials are necessary to collect adequate evidence on (long-term) safety and efficacy of novel interventions such as stem cells, but the design and launch of clinical trials, from first-in-human studies to larger randomized controlled trials (RCTs), is scientifically and ethically challenging. Stem cells are different from traditional pharmaceuticals, surgical procedures, and medical devices in the following ways: the novelty and complexity of stem cells, the invasiveness of the procedures, and the novel aim of regeneration. These specifics, combined with the characteristics of the study population, will have an impact on the design and ethics of RCTs. The recently closed JUVENTAS trial will serve as an example to identify the (interwoven) scientific and ethical challenges in the design and launch of stem cell RCTs. The JUVENTAS trial has investigated the efficacy of autologous bone marrow cells in end-stage vascular patients, in a double-blind sham-controlled design. We first describe the choices, considerations, and experiences of the JUVENTAS team. Subsequently, we identify the main ethical and scientific challenges and discuss what is important to consider in the design of future stem cell RCTs: assessment of risks and benefits, the choice for outcome measures, the choice for the comparator, the appropriate selection of participants, and adequate informed consent. Additionally, the stem cell field is highly in the spotlight due to the (commercial) interests and expectations. This warrants a cautious pace of translation and scrupulous set up of clinical trials, as failures could put the field in a negative light. At the same time, knowledge from clinical trials is necessary for the field to progress. We conclude that in the scientifically and ethically challenging field of stem cell RCTs, researchers and clinicians have to maneuver between the Skylla of hyper accelerated translation without rigorously conducted RCTs and the Charybdis of the missed opportunity of valuable knowledge.
Collapse
Affiliation(s)
- Sophie L Niemansburg
- 1 Department of Medical Humanities, Julius Center, University Medical Center Utercht , The Netherlands
| | | | | | | | | | | |
Collapse
|
183
|
Bellavia M, Altomare R, Cacciabaudo F, Santoro A, Allegra A, Concetta Gioviale M, Lo Monte AI. Towards an ideal source of mesenchymal stem cell isolation for possible therapeutic application in regenerative medicine. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2014; 158:356-60. [DOI: 10.5507/bp.2013.051] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 06/27/2013] [Indexed: 12/16/2022] Open
|
184
|
Ishii T, Eto K. Fetal stem cell transplantation: Past, present, and future. World J Stem Cells 2014; 6:404-420. [PMID: 25258662 PMCID: PMC4172669 DOI: 10.4252/wjsc.v6.i4.404] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 08/26/2014] [Accepted: 09/01/2014] [Indexed: 02/06/2023] Open
Abstract
Since 1928, human fetal tissues and stem cells have been used worldwide to treat various conditions. Although the transplantation of the fetal midbrain substantia nigra and dopaminergic neurons in patients suffering from Parkinson’s disease is particularly noteworthy, the history of other types of grafts, such as those of the fetal liver, thymus, and pancreas, should be addressed as there are many lessons to be learnt for future stem cell transplantation. This report describes previous practices and complications that led to current clinical trials of isolated fetal stem cells and embryonic stem (ES) cells. Moreover, strategies for transplantation are considered, with a particular focus on donor cells, cell processing, and the therapeutic cell niche, in addition to ethical issues associated with fetal origin. With the advent of autologous induced pluripotent stem cells and ES cells, clinical dependence on fetal transplantation is expected to gradually decline due to lasting ethical controversies, despite landmark achievements.
Collapse
|
185
|
Yu H, Cheng L, Cho KS. The potential of stem cell-based therapy for retinal repair. Neural Regen Res 2014; 9:1100-3. [PMID: 25206766 PMCID: PMC4146102 DOI: 10.4103/1673-5374.135311] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2014] [Indexed: 12/20/2022] Open
Affiliation(s)
- Honghua Yu
- Department of Ophthalmology, General Hospital of Guangzhou Military Command of PLA, Guangzhou, Guangdong Province, China ; Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, 20 Staniford St., Boston, MA, USA
| | - Lin Cheng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China ; Institute of Clinical Pharmacology, Central South University; Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan Province, China ; Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, 20 Staniford St., Boston, MA, USA
| | - Kin-Sang Cho
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, 20 Staniford St., Boston, MA, USA
| |
Collapse
|
186
|
Srijaya TC, Ramasamy TS, Kasim NHA. Advancing stem cell therapy from bench to bedside: lessons from drug therapies. J Transl Med 2014; 12:243. [PMID: 25182194 PMCID: PMC4163166 DOI: 10.1186/s12967-014-0243-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Accepted: 08/26/2014] [Indexed: 12/20/2022] Open
Abstract
The inadequacy of existing therapeutic tools together with the paucity of organ donors have always led medical researchers to innovate the current treatment methods or to discover new ways to cure disease. Emergence of cell-based therapies has provided a new framework through which it has given the human world a new hope. Though relatively a new concept, the pace of advancement clearly reveals the significant role that stem cells will ultimately play in the near future. However, there are numerous uncertainties that are prevailing against the present setting of clinical trials related to stem cells: like the best route of cell administration, appropriate dosage, duration and several other applications. A better knowledge of these factors can substantially improve the effectiveness of disease cure or organ repair using this latest therapeutic tool. From a certain perspective, it could be argued that by considering certain proven clinical concepts and experience from synthetic drug system, we could improve the overall efficacy of cell-based therapies. In the past, studies on synthetic drug therapies and their clinical trials have shown that all the aforementioned factors have critical ascendancy over its therapeutic outcomes. Therefore, based on the knowledge gained from synthetic drug delivery systems, we hypothesize that by employing many of the clinical approaches from synthetic drug therapies to this new regenerative therapeutic tool, the efficacy of stem cell-based therapies can also be improved.
Collapse
Affiliation(s)
| | - Thamil Selvee Ramasamy
- />Department of Molecular Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Noor Hayaty Abu Kasim
- />Department of Restorative Dentistry, Faculty of Dentistry, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
187
|
Bubela T, McCabe C. Value-engineered translation for regenerative medicine: meeting the needs of health systems. Stem Cells Dev 2014; 22 Suppl 1:89-93. [PMID: 24304083 DOI: 10.1089/scd.2013.0398] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Despite high expectations of economic returns, large investments in regenerative medicine technology have yet to materialize, partly due to a lack of proven business and investment models, regulatory hurdles, and a greater focus on cost-effectiveness for reimbursement decisions by payors. Adoption of new economic modeling methods will better link investment decisions to value-based criteria of health systems.
Collapse
Affiliation(s)
- Tania Bubela
- 1 School of Public Health, University of Alberta , Edmonton, Canada
| | | |
Collapse
|
188
|
Li PS, Lee IL, Yu WL, Sun JS, Jane WN, Shen HH. A novel albumin-based tissue scaffold for autogenic tissue engineering applications. Sci Rep 2014; 4:5600. [PMID: 25034369 PMCID: PMC4102902 DOI: 10.1038/srep05600] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 06/19/2014] [Indexed: 01/12/2023] Open
Abstract
Tissue scaffolds provide a framework for living tissue regeneration. However, traditional tissue scaffolds are exogenous, composed of metals, ceramics, polymers, and animal tissues, and have a defined biocompatibility and application. This study presents a new method for obtaining a tissue scaffold from blood albumin, the major protein in mammalian blood. Human, bovine, and porcine albumin was polymerised into albumin polymers by microbial transglutaminase and was then cast by freeze-drying-based moulding to form albumin tissue scaffolds. Scanning electron microscopy and material testing analyses revealed that the albumin tissue scaffold possesses an extremely porous structure, moderate mechanical strength, and resilience. Using a culture of human mesenchymal stem cells (MSCs) as a model, we showed that MSCs can be seeded and grown in the albumin tissue scaffold. Furthermore, the albumin tissue scaffold can support the long-term osteogenic differentiation of MSCs. These results show that the albumin tissue scaffold exhibits favourable material properties and good compatibility with cells. We propose that this novel tissue scaffold can satisfy essential needs in tissue engineering as a general-purpose substrate. The use of this scaffold could lead to the development of new methods of artificial fabrication of autogenic tissue substitutes.
Collapse
Affiliation(s)
- Pei-Shan Li
- 1] Department of Materials Science and Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan [2] Tissue Regeneration Product Technology Division, Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu County 310, Taiwan [3]
| | - I-Liang Lee
- 1] c/o Rm. 624, Bldg. 53, No. 195, Sec. 4, Chung Hsing Rd., Chutung Township, Hsinchu County 310, Taiwan [2]
| | - Wei-Lin Yu
- Tissue Regeneration Product Technology Division, Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu County 310, Taiwan
| | - Jui-Sheng Sun
- Department of Orthopedic Surgery, School of Medicine, College of Medicine, National Taiwan University, Taipei 10051 and Department of Orthopaedic Surgery, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu 30059, Taiwan
| | - Wann-Neng Jane
- Institute of Plant and Microbial Biology, Academia Sinica, Nankang, Taipei 11529, Taiwan
| | - Hsin-Hsin Shen
- Tissue Regeneration Product Technology Division, Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu County 310, Taiwan
| |
Collapse
|
189
|
Arien-Zakay H, Gincberg G, Nagler A, Cohen G, Liraz-Zaltsman S, Trembovler V, Alexandrovich AG, Matok I, Galski H, Elchalal U, Lelkes PI, Lazarovici P, Shohami E. Neurotherapeutic effect of cord blood derived CD45+ hematopoietic cells in mice after traumatic brain injury. J Neurotrauma 2014; 31:1405-16. [PMID: 24640955 DOI: 10.1089/neu.2013.3270] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Treatment of traumatic brain injury (TBI) is still an unmet need. Cell therapy by human umbilical cord blood (HUCB) has shown promising results in animal models of TBI and is under evaluation in clinical trials. HUCB contains different cell populations but to date, only mesenchymal stem cells have been evaluated for therapy of TBI. Here we present the neurotherapeutic effect, as evaluated by neurological score, using a single dose of HUCB-derived mononuclear cells (MNCs) upon intravenous (IV) administration one day post-trauma in a mouse model of closed head injury (CHI). Delayed (eight days post-trauma) intracerebroventricular administration of MNCs showed improved neurobehavioral deficits thereby extending the therapeutic window for treating TBI. Further, we demonstrated for the first time that HUCB-derived pan-hematopoietic CD45 positive (CD45(+)) cells, isolated by magnetic sorting and characterized by expression of CD45 and CD11b markers (96-99%), improved the neurobehavioral deficits upon IV administration, which persisted for 35 days. The therapeutic effect was in a direct correlation to a reduction in the lesion volume and decreased by pre-treatment of the cells with anti-human-CD45 antibody. At the site of brain injury, 1.5-2 h after transplantation, HUCB-derived cells were identified by near infrared scanning and immunohistochemistry using anti-human-CD45 and anti-human-nuclei antibodies. Nerve growth factor and vascular endothelial growth factor levels were differentially expressed in both ipsilateral and contralateral brain hemispheres, thirty-five days after CHI, measured by enzyme-linked immunosorbent assay. These findings indicate the neurotherapeutic potential of HUCB-derived CD45(+) cell population in a mouse model of TBI and propose their use in the clinical setting of human TBI.
Collapse
Affiliation(s)
- Hadar Arien-Zakay
- 1 School of Pharmacy Institute for Drug Research, The Hebrew University of Jerusalem , Jerusalem, Israel
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
190
|
Chen SY, Mahabole M, Horesh E, Wester S, Goldberg JL, Tseng SCG. Isolation and characterization of mesenchymal progenitor cells from human orbital adipose tissue. Invest Ophthalmol Vis Sci 2014; 55:4842-52. [PMID: 24994870 DOI: 10.1167/iovs.14-14441] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Adipose-derived stem cells (ASCs) have gained importance due to their myriad potential clinical applications. We hypothesize that progenitor cells also exist besides those conventionally isolated from the stromal vascular fraction (SVF). METHOD Central and medial orbital adipose tissues obtained from patients during eyelid surgery were digested with collagenase for 3 or 16 hours at 37°C with or without shaking. After centrifugation, the remaining cell pellet was resuspended and filtered to yield flow through in SVF and retained cells (RC) on the filter. Single cells from RC and SVF were cultured on 5% coated Matrigel in serum-free modified embryonic stem cells medium (MESCM) for 10 passages. The progenitor status was evaluated by the expression of a number of markers by qPCR and immunofluorescence staining as well as their plasticity for endothelial and tri-lineage differentiation. RESULTS Type I collagenase digestion for 3 hours under shaking was significantly less effective in releasing progenitor cells than collagenase A digestion for 16 hours without shaking. Following filtration, cells in SVF and RC, of which the latter were tangled in collagen IV-containing matrix, expressed different markers of progenitor cells. Cells from SVF and RC could be expanded for 10 passages on coated Matrigel in MESCM and exhibited similar or better potential to differentiate into vascular endothelial cells, chondrocytes, osteocytes, and adipocytes than SVF cells expanded on plastic in Dulbecco's modified Eagle's medium (DMEM) with 10% fetal bovine serum (FBS). CONCLUSIONS Different progenitor cells can be isolated and expanded from orbital adipose tissues. Further characterization of their mesodermal or neuroectodermal origin might enhance clinical outcome when used as a source of autologous stem cells for ocular surface regeneration.
Collapse
Affiliation(s)
- Szu-Yu Chen
- TissueTech, Inc., Miami, Florida, United States
| | | | - Elan Horesh
- University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Sara Wester
- University of Miami Miller School of Medicine, Miami, Florida, United States Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Jeffrey L Goldberg
- University of Miami Miller School of Medicine, Miami, Florida, United States
| | - Scheffer C G Tseng
- TissueTech, Inc., Miami, Florida, United States Ocular Surface Center, and Ocular Surface Research Education Foundation, Miami, Florida, United States
| |
Collapse
|
191
|
Abstract
Human heart failure (HF) is one of the leading causes of morbidity and mortality worldwide. Currently, heart transplantation and implantation of mechanical devices represent the only available treatments for advanced HF. Two alternative strategies have emerged to treat patients with HF. One approach relies on transplantation of exogenous stem cells (SCs) of non-cardiac or cardiac origin to induce cardiac regeneration and improve ventricular function. Another complementary strategy relies on stimulation of the endogenous regenerative capacity of uninjured cardiac progenitor cells to rebuild cardiac muscle and restore ventricular function. Various SC types and delivery strategies have been examined in the experimental and clinical settings; however, neither the ideal cell type nor the cell delivery method for cardiac cell therapy has yet emerged. Although the use of bone marrow (BM)-derived cells, most frequently exploited in clinical trials, appears to be safe, the results are controversial. Two recent randomized trials have failed to document any beneficial effects of intracardiac delivery of autologous BM mononuclear cells on cardiac function of patients with HF. The remarkable discovery that various populations of cardiac progenitor cells (CPCs) are present in the adult human heart and that it possesses limited regeneration capacity has opened a new era in cardiac repair. Importantly, unlike BM-derived SCs, autologous CPCs from myocardial biopsies cultured and subsequently delivered by coronary injection to patients have given positive results. Although these data are promising, a better understanding of how to control proliferation and differentiation of CPCs, to enhance their recruitment and survival, is required before CPCs become clinically applicable therapeutics.
Collapse
Affiliation(s)
- Alexander T Akhmedov
- The Molecular Cardiology and Neuromuscular Institute, 75 Raritan Ave., Highland Park, NJ, 08904, USA
| | | |
Collapse
|
192
|
Gálvez P, Martín MJ, Calpena AC, Tamayo JA, Ruiz MA, Clares B. Enhancing effect of glucose microspheres in the viability of human mesenchymal stem cell suspensions for clinical administration. Pharm Res 2014; 31:3515-28. [PMID: 24962511 DOI: 10.1007/s11095-014-1438-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 06/10/2014] [Indexed: 02/07/2023]
Abstract
PURPOSE A critical limiting factor of cell therapy is the short life of the stem cells. In this study, glucose containing alginate microspheres were developed and characterized to provide a sustained release system prolonging the viability of human mesenchymal stem cells (hMSCs) in a suspension for clinical application. METHODS The glucose microspheres were satisfactorily elaborated with alginate by emulsification/internal gelation method. Particle size was evaluated by light diffraction and optical microscopy. Shape and surface texture by scanning electron microscopy (SEM). Zeta potential, infrared spectra and release studies were also conducted. Also, rheological properties and stability of hMSCs suspensions with microspheres were tested. The viability of hMSCs was determined by trypan blue dye exclusion staining. RESULTS Microspheres of 86.62 μm, spherical shaped and -32.54 mV zeta potential with excellent stability, good encapsulation efficiency and providing an exponential release of glucose were obtained. hMSCs had better survival rate when they were packed with glucose microspheres. Microspheres maintained the aseptic conditions of the cell suspension without rheological, morphological or immunophenotypic disturbances on hMSCs. CONCLUSIONS Developed microspheres were able to enhance the functionality of hMSC suspension. This strategy could be broadly applied to various therapeutic approaches in which prolonged viability of cells is necessary.
Collapse
Affiliation(s)
- Patricia Gálvez
- Andalusian Centre for Molecular Biology and Regenerative Medicine (CABIMER), Seville, Spain
| | | | | | | | | | | |
Collapse
|
193
|
Carstairs A, Genever P. Stem cell treatment for musculoskeletal disease. Curr Opin Pharmacol 2014; 16:1-6. [DOI: 10.1016/j.coph.2014.01.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 01/27/2014] [Accepted: 01/27/2014] [Indexed: 01/22/2023]
|
194
|
Mariñas‐Pardo L, Mirones I, Amor‐Carro Ó, Fraga‐Iriso R, Lema‐Costa B, Cubillo I, Rodríguez Milla MÁ, García‐Castro J, Ramos‐Barbón D. Mesenchymal stem cells regulate airway contractile tissue remodeling in murine experimental asthma. Allergy 2014; 69:730-40. [PMID: 24750069 PMCID: PMC4114550 DOI: 10.1111/all.12392] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2014] [Indexed: 01/10/2023]
Abstract
Background Mesenchymal stem cells may offer therapeutic potential for asthma due to their immunomodulatory properties and host tolerability, yet prior evidence suggests that bloodborne progenitor cells may participate in airway remodeling. Here, we tested whether mesenchymal stem cells administered as anti‐inflammatory therapy may favor airway remodeling and therefore be detrimental. Methods Adipose tissue‐derived mesenchymal stem cells were retrovirally transduced to express green fluorescent protein and intravenously injected into mice with established experimental asthma induced by repeat intranasal house dust mite extract. Controls were house dust mite‐instilled animals receiving intravenous vehicle or phosphate‐buffered saline‐instilled animals receiving mesenchymal stem cells. Data on lung function, airway inflammation, and remodeling were collected at 72 h after injection or after 2 weeks of additional intranasal challenge. Results The mesenchymal stem cells homed to the lungs and rapidly downregulated airway inflammation in association with raised T‐helper‐1 lung cytokines, but such effect declined under sustained allergen challenge despite a persistent presence of mesenchymal stem cells. Conversely, airway hyperresponsiveness and contractile tissue underwent a late reduction regardless of continuous pathogenic stimuli and inflammatory rebound. Tracking of green fluorescent protein did not show mesenchymal stem cell integration or differentiation in airway wall tissues. Conclusions Therapeutic mesenchymal stem cell infusion in murine experimental asthma is free of unwanted pro‐remodeling effects and ameliorates airway hyper‐responsiveness and contractile tissue remodeling. These outcomes support furthering the development of mesenchymal stem cell‐based asthma therapies, although caution and solid preclinical data building are warranted.
Collapse
Affiliation(s)
- L. Mariñas‐Pardo
- Respiratory Research Unit Instituto de Investigación Biomédica de A Coruña (INIBIC)/Complexo Hospitalario Universitario A Coruña Spain
- Cellular Biotechnology Unit Instituto de Salud Carlos III Madrid Spain
| | - I. Mirones
- Cellular Biotechnology Unit Instituto de Salud Carlos III Madrid Spain
| | - Ó. Amor‐Carro
- Respiratory Research Unit Instituto de Investigación Biomédica de A Coruña (INIBIC)/Complexo Hospitalario Universitario A Coruña Spain
- Respiratory Department Hospital de la Santa Creu i Sant Pau Universitat Autònoma de Barcelona Barcelona Spain
| | - R. Fraga‐Iriso
- Respiratory Research Unit Instituto de Investigación Biomédica de A Coruña (INIBIC)/Complexo Hospitalario Universitario A Coruña Spain
- Respiratory Department Hospital de la Santa Creu i Sant Pau Universitat Autònoma de Barcelona Barcelona Spain
| | - B. Lema‐Costa
- Respiratory Research Unit Instituto de Investigación Biomédica de A Coruña (INIBIC)/Complexo Hospitalario Universitario A Coruña Spain
| | - I. Cubillo
- Cellular Biotechnology Unit Instituto de Salud Carlos III Madrid Spain
| | | | - J. García‐Castro
- Cellular Biotechnology Unit Instituto de Salud Carlos III Madrid Spain
| | - D. Ramos‐Barbón
- Respiratory Research Unit Instituto de Investigación Biomédica de A Coruña (INIBIC)/Complexo Hospitalario Universitario A Coruña Spain
- Respiratory Department Hospital de la Santa Creu i Sant Pau Universitat Autònoma de Barcelona Barcelona Spain
- Meakins‐Christie Laboratories Department of Medicine McGill University Montreal QCCanada
| |
Collapse
|
195
|
Bach C, Sherman W, Pallis J, Patra P, Bajwa H. Evaluation of novel design strategies for developing zinc finger nucleases tools for treating human diseases. BIOTECHNOLOGY RESEARCH INTERNATIONAL 2014; 2014:970595. [PMID: 24808958 PMCID: PMC3997970 DOI: 10.1155/2014/970595] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 01/02/2014] [Accepted: 01/02/2014] [Indexed: 11/24/2022]
Abstract
Zinc finger nucleases (ZFNs) are associated with cell death and apoptosis by binding at countless undesired locations. This cytotoxicity is associated with the binding ability of engineered zinc finger domains to bind dissimilar DNA sequences with high affinity. In general, binding preferences of transcription factors are associated with significant degenerated diversity and complexity which convolutes the design and engineering of precise DNA binding domains. Evolutionary success of natural zinc finger proteins, however, evinces that nature created specific evolutionary traits and strategies, such as modularity and rank-specific recognition to cope with binding complexity that are critical for creating clinical viable tools to precisely modify the human genome. Our findings indicate preservation of general modularity and significant alteration of the rank-specific binding preferences of the three-finger binding domain of transcription factor SP1 when exchanging amino acids in the 2nd finger.
Collapse
Affiliation(s)
- Christian Bach
- University of Bridgeport, Biomedical Engineering, 221 University Avenue, Bridgeport, CT 06604, USA
| | - William Sherman
- Physics Faculty, BHSEC Queens, 30-20 Thomson Avenue, Long Island City, NY 11101, USA
| | - Jani Pallis
- University of Bridgeport, Mechanical Engineering, 221 University Avenue, Bridgeport, CT 06604, USA
| | - Prabir Patra
- University of Bridgeport, Biomedical Engineering, 221 University Avenue, Bridgeport, CT 06604, USA
| | - Hassan Bajwa
- University of Bridgeport, Electrical Engineering, 221 University Avenue, Bridgeport, CT 06604, USA
| |
Collapse
|
196
|
Establishment and biological characterization of a dermal mesenchymal stem cells line from bovine. Biosci Rep 2014; 34:BSR20130095. [PMID: 27919035 PMCID: PMC3971452 DOI: 10.1042/bsr20130095] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 11/04/2013] [Accepted: 11/25/2013] [Indexed: 11/17/2022] Open
Abstract
The DMSCs (dermal mesenchymal stem cells) are multipotent stem cells, which can differentiate in vitro into many cell types. Much work has been done on DMSCs from humans, mice, rabbits and other mammals, but the related literature has not been published about these cells in cattle. In this study, we isolated and established the DMSC lines from cattle, thereby initiating further research on these cells, such as growth kinetics, detection of special surface antigen and RT-PCR (reverse transcription-PCR) assays to identify the biological characterization of the cell line. Furthermore, the DMSCs are induced to differentiate into adipocytes, osteoblasts and neural cells in vitro Our results suggest that DMSCs isolated from cattle possess similar biological characteristics with those from other species. Their multi-lineage differentiation capabilities herald a probable application model in tissue engineering and induced pluripotent stem cells.
Collapse
|
197
|
Pan XH, Song QQ, Dai JJ, Yao X, Wang JX, Pang RQ, He J, Li ZA, Sun XM, Ruan GP. Transplantation of bone marrow mesenchymal stem cells for the treatment of type 2 diabetes in a macaque model. Cells Tissues Organs 2014; 198:414-27. [PMID: 24686078 DOI: 10.1159/000358383] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2014] [Indexed: 12/22/2022] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs) are self-renewing, multipotent cells that can migrate to pathological sites and thereby provide a new treatment in diabetic animals. Superparamagnetic iron oxide/4',6-diamidino-2-phenylindole (DAPI) double-labeled BMSCs were transplanted into the pancreatic artery of macaques to treat type 2 diabetes mellitus (T2DM). The treatment efficiency of BMSCs was also evaluated. After successful induction of the T2DM model, the treatment group received double-labeled BMSCs via the pancreatic artery. Six weeks after BMSC transplantation, the fasting blood glucose and blood lipid levels measured in the treatment group were significantly lower (p < 0.05) than in the model group, although they were not reduced to normal levels (p < 0.05). Additionally, the serum C-peptide levels were significantly increased (p < 0.05). An intravenous glucose tolerance test and C-peptide release test had significant changes to the area under the curve. Within 14 days of the transplantation of labeled cells, the pancreatic and kidney tissue of the treatment group emitted a negative signal that was visible on magnetic resonance imaging (MRI). Six weeks after transplantation, DAPI signals appeared in the pancreatic and kidney tissue, which indicates that the BMSCs were mainly distributed in damaged tissue. Labeled stem cells can be used to track migration and distribution in vivo by MRI. In conclusion, the transplantation of BMSCs for the treatment of T2DM is safe and effective.
Collapse
Affiliation(s)
- Xing-hua Pan
- Stem Cell Engineering Laboratory of Yunnan Province, Kunming General Hospital of Chengdu Military Command, Kunming, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
198
|
Josh F, Tobita M, Tanaka R, Orbay H, Ogata K, Suzuki K, Hyakusoku H, Mizuno H. Concentration of PDGF-AB, BB and TGF-β1 as valuable human serum parameters in adipose-derived stem cell proliferation. J NIPPON MED SCH 2014; 80:140-7. [PMID: 23657067 DOI: 10.1272/jnms.80.140] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Human serum (HS) has attributes similar to fetal bovine serum (FBS) in the proliferation and differentiation of human adipose-derived stem cells (hASCs) when compared in vitro. The purpose of this study was to determine what types of HS, with respect to the concentrations of endogenous growth factors, could be made available for hASC proliferation. METHODS HS was collected from 2 groups of healthy donor (freshly isolated HS [n=9], and HS preserved for 4 years [n=7]). All sera were isolated with a Cellaid HS isolation device (JMS Co., Ltd, Hiroshima, Japan) and then classified into 3 groups based on the concentrations (high, middle, and low) of platelet-derived growth factor (PDGF)-AB, PDGF-BB, and transforming growth factor-beta 1 (TGF-β1) by means of enzyme-linked immunoassay screening. The hASCs were isolated from subcutaneous fat using a collagenase enzymatic digestion process and were cultured in control media, each supplemented with HS from a different group. Cell numbers were counted on days 2, 4, 7, and 14, and the relationship between cell proliferation and the level of each growth factor was investigated. RESULTS The proliferation of hASCs correlated with the concentration of each growth factor. The cut-off points for PDGF-AB, PDGF-BB, and TGF-β1 in HS [necessary for hASC proliferation when compared with FBS] were 10 ng/mL, 1.5 ng/mL, and 15 ng/mL, respectively. There was no correlation between the storage period of HS and the proliferation potential of hASCs. CONCLUSIONS These results suggest that the effectiveness of HS on hASC proliferation depends on the concentrations of endogenous PDGFs. In addition, the Cellaid device used in this study allows the simultaneous release of several growth factors from platelets, and our results have shown that it can be used to collect HS for future hASC-based therapies.
Collapse
Affiliation(s)
- Fonny Josh
- Department of Plastic and Reconstructive Surgery, Juntendo University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
199
|
Budniatzky I, Gepstein L. Concise review: reprogramming strategies for cardiovascular regenerative medicine: from induced pluripotent stem cells to direct reprogramming. Stem Cells Transl Med 2014; 3:448-57. [PMID: 24591731 DOI: 10.5966/sctm.2013-0163] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Myocardial cell-replacement therapies are emerging as novel therapeutic paradigms for myocardial repair but are hampered by the lack of sources of autologous human cardiomyocytes. The recent advances in stem cell biology and in transcription factor-based reprogramming strategies may provide exciting solutions to this problem. In the current review, we describe the different reprogramming strategies that can give rise to cardiomyocytes for regenerative medicine purposes. Initially, we describe induced pluripotent stem cell technology, a method by which adult somatic cells can be reprogrammed to yield pluripotent stem cells that could later be coaxed ex vivo to differentiate into cardiomyocytes. The generated induced pluripotent stem cell-derived cardiomyocytes could then be used for myocardial cell transplantation and tissue engineering strategies. We also describe the more recent direct reprogramming approaches that aim to directly convert the phenotype of one mature cell type (fibroblast) to another (cardiomyocyte) without going through a pluripotent intermediate cell type. The advantages and shortcomings of each strategy for cardiac regeneration are discussed, along with the hurdles that need to be overcome on the road to clinical translation.
Collapse
Affiliation(s)
- Inbar Budniatzky
- Sohnis Research Laboratory for Cardiac Electrophysiology and Regenerative Medicine and Cardiology Department, Rambam Medical Center, Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa, Israel
| | | |
Collapse
|
200
|
Murray IR, Corselli M, Petrigliano FA, Soo C, Péault B. Recent insights into the identity of mesenchymal stem cells. Bone Joint J 2014; 96-B:291-8. [DOI: 10.1302/0301-620x.96b3.32789] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The ability of mesenchymal stem cells (MSCs) to differentiate in vitro into chondrocytes, osteocytes and myocytes holds great promise for tissue engineering. Skeletal defects are emerging as key targets for treatment using MSCs due to the high responsiveness of bone to interventions in animal models. Interest in MSCs has further expanded in recognition of their ability to release growth factors and to adjust immune responses. Despite their increasing application in clinical trials, the origin and role of MSCs in the development, repair and regeneration of organs have remained unclear. Until recently, MSCs could only be isolated in a process that requires culture in a laboratory; these cells were being used for tissue engineering without understanding their native location and function. MSCs isolated in this indirect way have been used in clinical trials and remain the reference standard cellular substrate for musculoskeletal engineering. The therapeutic use of autologous MSCs is currently limited by the need for ex vivo expansion and by heterogeneity within MSC preparations. The recent discovery that the walls of blood vessels harbour native precursors of MSCs has led to their prospective identification and isolation. MSCs may therefore now be purified from dispensable tissues such as lipo-aspirate and returned for clinical use in sufficient quantity, negating the requirement for ex vivo expansion and a second surgical procedure. In this annotation we provide an update on the recent developments in the understanding of the identity of MSCs within tissues and outline how this may affect their use in orthopaedic surgery in the future. Cite this article: Bone Joint J 2014;96-B:291–8.
Collapse
Affiliation(s)
- I. R. Murray
- Scottish Centre for Regenerative Medicine, The
University of Edinburgh, 5 Little France Drive, Edinburgh, EH16
4UU, UK
| | - M. Corselli
- Orthopaedic Hospital Research Center, David
Geffen School of Medicine, University of California, Los
Angeles, California 90095, USA
| | - F. A. Petrigliano
- UCLA Orthopaedic Hospital, Department
of Orthopaedic Surgery, University of California, Los
Angeles, California 90095, USA
| | - C. Soo
- Division of Plastic and Reconstructive
Surgery, David Geffen School of Medicine, University
of California, Los Angeles, California
90095, USA
| | - B. Péault
- Orthopaedic Hospital Research Center, David
Geffen School of Medicine, University of California, Los
Angeles, California 90095, USA
| |
Collapse
|