951
|
Carrade DD, Lame MW, Kent MS, Clark KC, Walker NJ, Borjesson DL. Comparative Analysis of the Immunomodulatory Properties of Equine Adult-Derived Mesenchymal Stem Cells(). CELL MEDICINE 2012; 4:1-11. [PMID: 23152950 DOI: 10.3727/215517912x647217] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSCs) derived from bone marrow (BM), adipose tissue (AT), umbilical cord blood (CB), and umbilical cord tissue (CT) are increasingly being used to treat equine inflammatory and degenerative lesions. MSCs modulate the immune system in part through mediator secretion. Animal species and MSC tissue of origin are both important determinants of MSC function. In spite of widespread clinical use, how equine MSCs function to heal tissues is fully unknown. In this study, MSCs derived from BM, AT, CB, and CT were compared for their ability to inhibit lymphocyte proliferation and secrete mediators in response to activation. Five MSC lines from each tissue were isolated. Lymphocyte proliferation was assessed in a mixed leukocyte reaction, and mediator secretion was determined by ELISA. Regardless of tissue of origin, quiescent MSCs did not alter lymphocyte proliferation or secrete mediators, except for transforming growth factor-β (TGF-β1). When stimulated, MSCs of all tissue types decreased lymphocyte proliferation, increased prostaglandin (PGE(2)) and interleukin-6 (IL-6) secretion, and decreased production of tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ). BM-MSCs and CB-MSCs also produced nitric oxide (NO), while AT-MSCs and CT-MSCs did not. Equine MSCs did not produce indoleamine 2,3-dioxygenase (IDO). These data suggest that activated equine MSCs derived from BM, AT, CT, and CB secrete high concentration of mediators and are similar to MSCs from rodents and humans in their immunomodulatory profiles. These findings have implication for the treatment of inflammatory lesions dominated by activated lymphocytes and TNF-α and IFN-γ in vivo.
Collapse
Affiliation(s)
- Danielle D Carrade
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, USA
| | | | | | | | | | | |
Collapse
|
952
|
Koning JJ, Mebius RE. Interdependence of stromal and immune cells for lymph node function. Trends Immunol 2011; 33:264-70. [PMID: 22153930 DOI: 10.1016/j.it.2011.10.006] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 09/15/2011] [Accepted: 10/31/2011] [Indexed: 01/09/2023]
Abstract
Lymph nodes are strategically located throughout the body to allow lymphocytes to efficiently encounter their cognate antigen and become activated. The structure of the lymph nodes is such that B and T lymphocytes each have their own microdomain. This structure is provided by lymph node stromal cells, which also provide the lymphocytes with a scaffold upon which to migrate. Here, we discuss how stromal cells differentiate from mesenchymal precursor cells in response to the interaction with lymphocytes, while these stromal cells in turn provide necessary survival factors for the lymphocytes. We propose that during immune reactions, the interactions of stromal and immune cells are similarly important for controlling the expanding lymphocyte pool.
Collapse
Affiliation(s)
- Jasper J Koning
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | | |
Collapse
|
953
|
Siniscalco D, Giordano C, Galderisi U, Luongo L, de Novellis V, Rossi F, Maione S. Long-lasting effects of human mesenchymal stem cell systemic administration on pain-like behaviors, cellular, and biomolecular modifications in neuropathic mice. Front Integr Neurosci 2011. [PMID: 22164136 DOI: 10.3389/fnint.2011.00079.ecollection] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Neuropathic pain (NP) is an incurable disease caused by a primary lesion in the nervous system. NP is a progressive nervous system disease that results from poorly defined neurophysiological and neurochemical changes. Its treatment is very difficult. Current available therapeutic drugs have a generalized nature, sometime acting only on the temporal pain properties rather than targeting the several mechanisms underlying the generation and propagation of pain. METHODS Using biomolecular and immunohistochemical methods, we investigated the effect of the systemic injection of human mesenchymal stem cells (hMSCs) on NP relief. We used the spared nerve injury (SNI) model of NP in the mouse. hMSCs were injected into the tail vein of the mouse. Stem cell injection was performed 4 days after sciatic nerve surgery. Neuropathic mice were monitored every 10 days starting from day 11 until 90 days after surgery. RESULTS hMSCs were able to reduce pain-like behaviors, such as mechanical allodynia and thermal hyperalgesia, once injected into the tail vein. An anti-nociceptive effect was detectable from day 11 post surgery (7 days post cell injection). hMSCs were mainly able to home in the spinal cord and pre-frontal cortex of neuropathic mice. Injected hMSCs reduced the protein levels of the mouse pro-inflammatory interleukin IL-1β and IL-17 and increased protein levels of the mouse anti-inflammatory interleukin IL-10, and the marker of alternatively activated macrophages CD106 in the spinal cord of SNI mice. CONCLUSION As a potential mechanism of action of hMSCs in reducing pain, we suggest that they could exert their beneficial action through a restorative mechanism involving: (i) a cell-to-cell contact activation mechanism, through which spinal cord homed hMSCs are responsible for switching pro-inflammatory macrophages to anti-inflammatory macrophages; (ii) secretion of a broad spectrum of molecules to communicate with other cell types. This study could provide novel findings in MSC pre-clinical biology and their therapeutic potential in regenerative medicine.
Collapse
Affiliation(s)
- Dario Siniscalco
- Division of Pharmacology "L. Donatelli," Department of Experimental Medicine, Second University of Naples Naples, Italy
| | | | | | | | | | | | | |
Collapse
|
954
|
Siniscalco D, Giordano C, Galderisi U, Luongo L, de Novellis V, Rossi F, Maione S. Long-lasting effects of human mesenchymal stem cell systemic administration on pain-like behaviors, cellular, and biomolecular modifications in neuropathic mice. Front Integr Neurosci 2011; 5:79. [PMID: 22164136 PMCID: PMC3230031 DOI: 10.3389/fnint.2011.00079] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Accepted: 11/16/2011] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Neuropathic pain (NP) is an incurable disease caused by a primary lesion in the nervous system. NP is a progressive nervous system disease that results from poorly defined neurophysiological and neurochemical changes. Its treatment is very difficult. Current available therapeutic drugs have a generalized nature, sometime acting only on the temporal pain properties rather than targeting the several mechanisms underlying the generation and propagation of pain. METHODS Using biomolecular and immunohistochemical methods, we investigated the effect of the systemic injection of human mesenchymal stem cells (hMSCs) on NP relief. We used the spared nerve injury (SNI) model of NP in the mouse. hMSCs were injected into the tail vein of the mouse. Stem cell injection was performed 4 days after sciatic nerve surgery. Neuropathic mice were monitored every 10 days starting from day 11 until 90 days after surgery. RESULTS hMSCs were able to reduce pain-like behaviors, such as mechanical allodynia and thermal hyperalgesia, once injected into the tail vein. An anti-nociceptive effect was detectable from day 11 post surgery (7 days post cell injection). hMSCs were mainly able to home in the spinal cord and pre-frontal cortex of neuropathic mice. Injected hMSCs reduced the protein levels of the mouse pro-inflammatory interleukin IL-1β and IL-17 and increased protein levels of the mouse anti-inflammatory interleukin IL-10, and the marker of alternatively activated macrophages CD106 in the spinal cord of SNI mice. CONCLUSION As a potential mechanism of action of hMSCs in reducing pain, we suggest that they could exert their beneficial action through a restorative mechanism involving: (i) a cell-to-cell contact activation mechanism, through which spinal cord homed hMSCs are responsible for switching pro-inflammatory macrophages to anti-inflammatory macrophages; (ii) secretion of a broad spectrum of molecules to communicate with other cell types. This study could provide novel findings in MSC pre-clinical biology and their therapeutic potential in regenerative medicine.
Collapse
Affiliation(s)
- Dario Siniscalco
- Division of Pharmacology “L. Donatelli,” Department of Experimental Medicine, Second University of NaplesNaples, Italy
| | - Catia Giordano
- Division of Pharmacology “L. Donatelli,” Department of Experimental Medicine, Second University of NaplesNaples, Italy
| | - Umberto Galderisi
- Division of Biotechnology and Molecular Biology “A. Cascino,” Second University of NaplesNaples, Italy
| | - Livio Luongo
- Division of Pharmacology “L. Donatelli,” Department of Experimental Medicine, Second University of NaplesNaples, Italy
| | - Vito de Novellis
- Division of Pharmacology “L. Donatelli,” Department of Experimental Medicine, Second University of NaplesNaples, Italy
| | - Francesco Rossi
- Division of Pharmacology “L. Donatelli,” Department of Experimental Medicine, Second University of NaplesNaples, Italy
| | - Sabatino Maione
- Division of Pharmacology “L. Donatelli,” Department of Experimental Medicine, Second University of NaplesNaples, Italy
| |
Collapse
|
955
|
Gebler A, Zabel O, Seliger B. The immunomodulatory capacity of mesenchymal stem cells. Trends Mol Med 2011; 18:128-34. [PMID: 22118960 DOI: 10.1016/j.molmed.2011.10.004] [Citation(s) in RCA: 267] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 10/17/2011] [Accepted: 10/21/2011] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) are currently being tested in clinical trials for the treatment of various diseases owing to the ease of generating and expanding these cells, the ability to differentiate them into various specialized mesenchymal tissue types and their immunosuppressive properties. However, their immunomodulatory potential remains controversial. This review describes the constitutive and regulated expression of molecules of the major histocompatibility complex (MHC) class I antigen processing machinery (APM), co-stimulatory B7 molecules and HLA-G. Furthermore, this review focuses on the secretion of factors, such as cytokines, in mesenchymal stem cells, their functional role in mounting and controlling immune responses mediated by different immune cell subpopulations, their medical significance, and the obstacles that limit their clinical application.
Collapse
Affiliation(s)
- Antje Gebler
- Institute of Medical Immunology, Martin-Luther-University Halle-Wittenberg, 06112 Halle (Saale), Germany
| | | | | |
Collapse
|
956
|
Balasubramanian S, Venugopal P, Sundarraj S, Zakaria Z, Majumdar AS, Ta M. Comparison of chemokine and receptor gene expression between Wharton's jelly and bone marrow-derived mesenchymal stromal cells. Cytotherapy 2011; 14:26-33. [PMID: 22091833 DOI: 10.3109/14653249.2011.605119] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AIMS Because of their multilineage differentiation capacity, immunomodulatory role and homing ability, mesenchymal stromal cells (MSC) are emerging as a new therapeutic strategy for treating a variety of disorders. Although bone marrow (BM) is the best characterized source of MSC, Wharton's jelly (WJ) of the umbilical cord holds great promise as an alternative. As delivery direct to the site of injury is not always feasible, efficient homing of MSC to the site of injury is critical for inducing tissue repair and regeneration. MSC express a wide variety of growth factors, chemokines and receptors that are important for cell migration, homing and re-establishment of blood supply for recovery of damaged tissues. METHODS Detailed chemokine and receptor gene expression profiles of WJ MSC were established, and subsequently compared with those of BM-derived MSC using a polymerase chain reaction (PCR) array. Secretion of growth factors was analyzed and evaluated using culture supernatant from WJ and BM MSC. RESULTS Our results revealed a differential expression pattern of the chemokines and their receptors between WJ- and BM-derived MSC. Several Glutamic acid-Leucine-Arginine; ELR-positive CXC chemokine genes and secretion of growth factors, which promote angiogenesis, were found to be up-regulated in WJ MSC. CONCLUSIONS To understand better the localization and mechanism of tissue repair by transplanted WJ MSC, we attempted chemokine and their receptor transcription profiling, followed by analysis of growth factors secreted by WJ MSC, and compared them against those of BM MSC. The data suggest that MSC from different sources can be explored for distinct therapeutic roles.
Collapse
|
957
|
Pontikoglou C, Deschaseaux F, Sensebé L, Papadaki HA. Bone marrow mesenchymal stem cells: biological properties and their role in hematopoiesis and hematopoietic stem cell transplantation. Stem Cell Rev Rep 2011; 7:569-89. [PMID: 21249477 DOI: 10.1007/s12015-011-9228-8] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent adult stem cells that are present in practically all tissues as a specialized population of mural cells/pericytes that lie on the abluminal side of blood vessels. Originally identified within the bone marrow (BM) stroma, not only do they provide microenvironmental support for hematopoietic stem cells (HSCs), but can also differentiate into various mesodermal lineages. MSCs can easily be isolated from the BM and subsequently expand in vitro and in addition they exhibit intriguing immunomodulatory properties, thereby emerging as attractive candidates for various therapeutic applications. This review addresses the concept of BM MSCs via a hematologist's point of view. In this context it discusses the stem cell properties that have been attributed to BM MSCs, as compared to those of the prototypic hematopoietic stem cell model and then gives a brief overview of the in vitro and vivo features of the former, emphasizing on their immunoregulatory properties and their hematopoiesis-supporting role. In addition, the qualitative and quantitative characteristics of BM MSCs within the context of a defective microenvironment, such as the one characterizing Myelodysplastic Syndromes are described and the potential involvement of these cells in the pathophysiology of the disease is discussed. Finally, emerging clinical applications of BM MSCs in the field of hematopoietic stem cell transplantation are reviewed and potential hazards from MSC use are outlined.
Collapse
|
958
|
Potential of human embryonic stem cells in cartilage tissue engineering and regenerative medicine. Stem Cell Rev Rep 2011; 7:544-59. [PMID: 21188652 DOI: 10.1007/s12015-010-9222-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The current surgical intervention of using autologous chondrocyte implantation (ACI) for cartilage repair is associated with several problems such as donor site morbidity, de-differentiation upon expansion and fibrocartilage repair following transplantation. This has led to exploration of the use of stem cells as a model for chondrogenic differentiation as well as a potential source of chondrogenic cells for cartilage tissue engineering and repair. Embryonic stem cells (ESCs) are advantageous, due to their unlimited self-renewal and pluripotency, thus representing an immortal cell source that could potentially provide an unlimited supply of chondrogenic cells for both cell and tissue-based therapies and replacements. This review aims to present an overview of emerging trends of using ESCs in cartilage tissue engineering and regenerative medicine. In particular, we will be focusing on ESCs as a promising cell source for cartilage regeneration, the various strategies and approaches employed in chondrogenic differentiation and tissue engineering, the associated outcomes from animal studies, and the challenges that need to be overcome before clinical application is possible.
Collapse
|
959
|
Marigo I, Dazzi F. The immunomodulatory properties of mesenchymal stem cells. Semin Immunopathol 2011; 33:593-602. [PMID: 21499984 DOI: 10.1007/s00281-011-0267-7] [Citation(s) in RCA: 130] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Accepted: 03/28/2011] [Indexed: 12/12/2022]
Abstract
Research on mesenchymal stem cells (MSC) has evolved rapidly during the last decade prompted by their potential use for tissue repair and immunotherapy. Not only can MSC differentiate into cells of the mesodermal lineage, but they also exhibit immunomodulatory functions depending on their interaction with cells of both innate and adaptive immune systems. Most aspects of MSC biology remain to be elucidated. It is emerging even more clearly that these cells are not always a panacea. Only the knowledge of their physiological role and their interactions with other cells will allow us to use them as a therapeutic tool.
Collapse
Affiliation(s)
- Ilaria Marigo
- Stem Cell Biology, Department of Medicine, Division of Experimental Medicine, Hammersmith Hospital, Du Cane Road, W12 ONN, London, UK.
| | | |
Collapse
|
960
|
Peterson M, Butler S, Eriksson M, Svärdsudd K. A randomized controlled trial of exercise versus wait-list in chronic tennis elbow (lateral epicondylosis). Ups J Med Sci 2011; 116:269-79. [PMID: 22066975 PMCID: PMC3207303 DOI: 10.3109/03009734.2011.600476] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2011] [Accepted: 06/22/2011] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Chronic tennis elbow (lateral epicondylosis) is a common disorder. Like other chronic soft-tissue pain conditions it is often difficult to treat successfully. The effects of exercise have been discussed, but no convincing evidence has been put forward so far, and a simple protocol for exercise is lacking. AIMS OF THE STUDY This study is a randomized, controlled, clinical trial of the effect of exercise versus expectation (wait-list) on pain, muscle strength, function, and quality of life in patients with long-standing lateral epicondylosis. METHODS Eighty-one subjects with tennis elbow lasting for more than 3 months were randomly allocated to an exercise group (n = 40) or a reference group (n = 41). The exercise group performed daily exercise, with weekly load increase, for 3 months. The reference group was wait-listed, but otherwise followed in the same way. Outcome measures were pain during maximum voluntary muscle contraction (Cozen's test) and pain during maximum muscle elongation with a load (modified Empty-can-test); muscle strength was measured with a Chatillon MSE 100 hand-held dynamometer, and the Disability of the Arm, Shoulder and Hand (DASH) and the Gothenburg Quality of Life questionnaires. RESULTS The exercise group had greater and faster regression of pain, both during muscle contraction and muscle elongation, than the reference group (p = 0.0005 and p = 0.0016, respectively). There was a non-significant muscle strength difference between the groups, but no differences regarding DASH scores or quality of life measures. CONCLUSIONS Exercise appears to be superior to expectation in reducing pain in chronic lateral epicondylosis.
Collapse
Affiliation(s)
- Magnus Peterson
- Department of Public Health and Caring Sciences, Family Medicine and Clinical Epidemiology Section, Uppsala University, Uppsala, Sweden
| | | | | | | |
Collapse
|
961
|
Nair A, Shen J, Lotfi P, Ko CY, Zhang CC, Tang L. Biomaterial implants mediate autologous stem cell recruitment in mice. Acta Biomater 2011; 7:3887-95. [PMID: 21784181 DOI: 10.1016/j.actbio.2011.06.050] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 05/26/2011] [Accepted: 06/28/2011] [Indexed: 12/29/2022]
Abstract
Autologous stem cells, recognized as the best cells for stem cell therapy, are associated with difficult extraction procedures which often lead to more traumas for the patients and time-consuming laboratory work, which delays their subsequent application. To combat such challenges, it was recently uncovered that, shortly after biomaterial implantation, following the recruitment of inflammatory cells, substantial numbers of mesenchymal stem cells (MSC) and hematopoietic stem cells (HSC) were recruited to the implantation sites. These multipotent MSC could be differentiated into various lineages in vitro. Inflammatory signals may be responsible for the gathering of stem cells, since there is a good relationship between biomaterial-mediated inflammatory responses and stem cell accumulation in vivo. In addition, the treatment with the anti-inflammatory drug dexamethasone substantially reduced the recruitment of both MSC and HSC. The results from this work support that such strategies could be further developed towards localized recruitment and differentiation of progenitor cells. This may permit the future development of autologous stem cell therapies without the need for tedious cell isolation, culture and transplantation.
Collapse
Affiliation(s)
- A Nair
- Bioengineering Department, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | | | | | | | | | | |
Collapse
|
962
|
Guiducci S, Manetti M, Romano E, Mazzanti B, Ceccarelli C, Dal Pozzo S, Milia AF, Bellando-Randone S, Fiori G, Conforti ML, Saccardi R, Ibba-Manneschi L, Matucci-Cerinic M. Bone marrow-derived mesenchymal stem cells from early diffuse systemic sclerosis exhibit a paracrine machinery and stimulate angiogenesis in vitro. Ann Rheum Dis 2011; 70:2011-21. [PMID: 21821866 DOI: 10.1136/ard.2011.150607] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To characterise bone marrow-derived mesenchymal stem cells (MSCs) from patients with systemic sclerosis (SSc) for the expression of factors implicated in MSC recruitment at sites of injury, angiogenesis and fibrosis. The study also analysed whether the production/release of bioactive mediators by MSCs were affected by stimulation with cytokines found upregulated in SSc serum and tissues, and whether MSCs could modulate dermal microvascular endothelial cell (MVEC) angiogenesis. METHODS MSCs obtained from five patients with early severe diffuse SSc (SSc-MSCs) and five healthy donors (H-MSCs) were stimulated with vascular endothelial growth factor (VEGF), transforming growth factor β (TGFβ) or stromal cell-derived factor-1 (SDF-1). Transcript and protein levels of SDF-1 and its receptor CXCR4, VEGF, TGFβ(1) and receptors TβRI and TβRII were evaluated by quantitative real-time PCR, western blotting and confocal microscopy. VEGF, SDF-1 and TGFβ(1) secretion in culture supernatant was measured by ELISA. MVEC capillary morphogenesis was performed on Matrigel with the addition of MSC-conditioned medium. RESULTS In SSc-MSCs the basal expression of proangiogenic SDF-1/CXCR4 and VEGF was significantly increased compared with H-MSCs. SSc-MSCs constitutively released higher levels of SDF-1 and VEGF. SDF-1/CXCR4 were upregulated after VEGF stimulation and CXCR4 redistributed from the cytoplasm to the cell surface. VEGF was increased by SDF-1 challenge. VEGF, TGFβ and SDF-1 stimulation upregulated TGFβ(1), TβRI and TβRII in SSc-MSCs. TβRII redistributed from the cytoplasm to focal adhesion contacts. SSc-MSC-conditioned medium showed a greater proangiogenic effect on MVECs than H-MSCs. Experiments with blocking antibodies showed that MSC-derived cytokines were responsible for this potent proangiogenic effect. CONCLUSION SSc-MSCs constitutively overexpress and release bioactive mediators/proangiogenic factors and potentiate dermal MVEC angiogenesis.
Collapse
Affiliation(s)
- Serena Guiducci
- Department of Biomedicine, Division of Rheumatology, AOUC, Excellence Centre for Research, Transfer and High Education DENOthe, University of Florence, Florence, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
963
|
Ra JC, Kang SK, Shin IS, Park HG, Joo SA, Kim JG, Kang BC, Lee YS, Nakama K, Piao M, Sohl B, Kurtz A. Stem cell treatment for patients with autoimmune disease by systemic infusion of culture-expanded autologous adipose tissue derived mesenchymal stem cells. J Transl Med 2011; 9:181. [PMID: 22017805 PMCID: PMC3222617 DOI: 10.1186/1479-5876-9-181] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 10/21/2011] [Indexed: 12/14/2022] Open
Abstract
Prolonged life expectancy, life style and environmental changes have caused a changing disease pattern in developed countries towards an increase of degenerative and autoimmune diseases. Stem cells have become a promising tool for their treatment by promoting tissue repair and protection from immune-attack associated damage. Patient-derived autologous stem cells present a safe option for this treatment since these will not induce immune rejection and thus multiple treatments are possible without any risk for allogenic sensitization, which may arise from allogenic stem cell transplantations. Here we report the outcome of treatments with culture expanded human adipose-derived mesenchymal stem cells (hAdMSCs) of 10 patients with autoimmune associated tissue damage and exhausted therapeutic options, including autoimmune hearing loss, multiple sclerosis, polymyotitis, atopic dermatitis and rheumatoid arthritis. For treatment, we developed a standardized culture-expansion protocol for hAdMSCs from minimal amounts of fat tissue, providing sufficient number of cells for repetitive injections. High expansion efficiencies were routinely achieved from autoimmune patients and from elderly donors without measurable loss in safety profile, genetic stability, vitality and differentiation potency, migration and homing characteristics. Although the conclusions that can be drawn from the compassionate use treatments in terms of therapeutic efficacy are only preliminary, the data provide convincing evidence for safety and therapeutic properties of systemically administered AdMSC in human patients with no other treatment options. The authors believe that ex-vivo-expanded autologous AdMSCs provide a promising alternative for treating autoimmune diseases. Further clinical studies are needed that take into account the results obtained from case studies as those presented here.
Collapse
Affiliation(s)
- Jeong Chan Ra
- Stem Cell Research Center, RNL BIO, Seoul, 153-768, Republic of Korea.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
964
|
Topical application of autologous adipose-derived mesenchymal stem cells (MSCs) for persistent sterile corneal epithelial defect. Graefes Arch Clin Exp Ophthalmol 2011; 250:455-7. [PMID: 22012407 DOI: 10.1007/s00417-011-1841-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Revised: 09/01/2011] [Accepted: 10/03/2011] [Indexed: 11/27/2022] Open
|
965
|
Pereira MJN, Carvalho IF, Karp JM, Ferreira LS. Sensing the cardiac environment: exploiting cues for regeneration. J Cardiovasc Transl Res 2011; 4:616-30. [PMID: 21735303 PMCID: PMC3334359 DOI: 10.1007/s12265-011-9299-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 06/20/2011] [Indexed: 12/11/2022]
Abstract
Recent pre-clinical and clinical studies indicate that certain exogenous stem cells and biomaterials can preserve cardiac tissue after myocardial infarction. Regarding stem cells, a growing body of data suggests that the short-term positive outcomes are mainly attributed to paracrine signaling mechanisms. The release of such factors is due to the cell's ability to sense cardiac environmentally derived cues, though the exact feedback loops are still poorly understood. However, given the limited engraftment and survival of transplanted cells in the ischemic environment, the long-term clinical benefits of these therapies have not yet been realized. To overcome this, the long-term controlled delivery of bioactive factors using biomaterials is a promising approach. A major challenge has been the ability to develop timely and spatially controlled gradients of different cues, pivotal for the development and regeneration of tissues. In addition, given the complexity of the remodeling process after myocardial infarction, multiple factors may be required at distinct disease stages to maximize therapeutic outcomes. Therefore, novel smart materials that can sense the surrounding environment and generate cues through on demand mechanisms will be of major importance in the translation of these promising advanced therapies. This article reviews how the cardiac environment can mediate the release profiles of bioactive cues from cells and biomaterials and how the controlled delivery impacts heart regeneration.
Collapse
Affiliation(s)
- Maria José Nunes Pereira
- Center of Neurosciences and Cell Biology (CNC), University of Coimbra, 3004-417 Coimbra, Portugal. Center for Regenerative Therapeutics and Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Harvard-MIT Division of Health Sciences and Technology, 65 Landsdowne Street, Cambridge, MA 02139, USA. Harvard Stem Cell Institute, Harvard University, 1350 Massachusetts Avenue, Cambridge, MA 02138, USA
| | | | - Jeffrey M. Karp
- Center for Regenerative Therapeutics and Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Harvard-MIT Division of Health Sciences and Technology, 65 Landsdowne Street, Cambridge, MA 02139, USA. Harvard Stem Cell Institute, Harvard University, 1350 Massachusetts Avenue, Cambridge, MA 02138, USA
| | - Lino S. Ferreira
- Center of Neurosciences and Cell Biology (CNC), University of Coimbra, 3004-417 Coimbra, Portugal. Biocant- Center of Biotechnology Innovation Center, 3060-197 Cantanhede, Portugal
| |
Collapse
|
966
|
Ko IK, Ju YM, Chen T, Atala A, Yoo JJ, Lee SJ. Combined systemic and local delivery of stem cell inducing/recruiting factors for in situ tissue regeneration. FASEB J 2011; 26:158-68. [PMID: 21965595 DOI: 10.1096/fj.11-182998] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Whereas the conventional tissue engineering strategy involves the use of scaffolds combined with appropriate cell types to restore normal functions, the concept of in situ tissue regeneration uses host responses to a target-specific scaffold to mobilize host cells to a site of injury without the need for cell seeding. For this purpose, local delivery of bioactive molecules from scaffolds has been generally used. However, this approach has limited stem cell recruitment into the implants. Thus, we developed a combination of systemic delivery of substance P (SP) and local release of stromal-derived factor-1α (SDF-1α) from an implant. In this study, we examined whether this combined system would significantly enhance recruitment of host stem cells into the implants. Flow cytometry and immunohistochemistry for CD29/CD45, CD146/α-smooth muscle actin, and c-kit demonstrated that this system significantly increased the number of stem cell-like cells within the implants when compared with other systems. In vitro culture of the cells that had infiltrated into the scaffolds from the combined system confirmed that host stem cells were recruited into these implants and indicated that they were capable of differentiation into multiple lineages. These results indicate that this combined system may lead to more efficient tissue regeneration.
Collapse
Affiliation(s)
- In Kap Ko
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | | | | | | | | | | |
Collapse
|
967
|
Sorrell JM, Baber MA, Traktuev DO, March KL, Caplan AI. The creation of an in vitro adipose tissue that contains a vascular-adipocyte complex. Biomaterials 2011; 32:9667-76. [PMID: 21959010 DOI: 10.1016/j.biomaterials.2011.08.090] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Accepted: 08/31/2011] [Indexed: 11/16/2022]
Abstract
An increased demand for soft-tissue substitutes has impelled the development of an in vitro adipose tissue. Ideally, such a tissue should contain a vascular network that can deliver blood throughout the construct following its engraftment. This study describes the in vitro fabrication of a pre-vascularized adipose tissue entirely using a self-assembly approach. Adult human adipose stromal cells (ASCs) provided the foundation for this construct. These cells were cultured at high density in the presence of elevated levels of ascorbate prior to adipocytic induction. Vascular support cells consisting of dermal fibroblasts, mixtures of adipose stromal cells and bone marrow mesenchymal stem cells (MSCs) were introduced to sustain an extensive vascular network formed by human umbilical vein endothelial cells (HUVECs). MSCs were introduced to serve as perivascular cells. The resulting construct contained a vascular-adipose tissue continuum that was held together by basement membrane molecules. This construct contains multiple cell types that are typically found in adipose tissue: adipocytes, pre-adipocytes, stem cells, fibroblasts, vascular cells, and perivascular support cells. As such, these constructs can be employed both for in vitro studies to assay cellular interactions between vasculature and other components of adipose tissue. Further, they can also be engrafted into athymic hosts to study vascular and adipocyte stability.
Collapse
Affiliation(s)
- J Michael Sorrell
- Skeletal Research Center, Department of Biology, Case Western Reserve University, Cleveland, OH 44106, United States.
| | | | | | | | | |
Collapse
|
968
|
The Amazing Power of Cancer Cells to Recapitulate Extraembryonic Functions: The Cuckoo's Tricks. JOURNAL OF ONCOLOGY 2011; 2012:521284. [PMID: 21969829 PMCID: PMC3182376 DOI: 10.1155/2012/521284] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 07/06/2011] [Accepted: 07/07/2011] [Indexed: 12/14/2022]
Abstract
Inflammation is implicated in tumor development, invasion, and metastasis. Hence, it has been suggested that common cellular and molecular mechanisms are activated in wound repair and in cancer development. In addition, it has been previously proposed that the inflammatory response, which is associated with the wound healing process, could recapitulate ontogeny through the reexpression of the extraembryonic, that is, amniotic and vitelline, functions in the interstitial space of the injured tissue. If so, the use of inflammation by the cancer-initiating cell can also be supported in the ability to reacquire extraembryonic functional axes for tumor development, invasion, and metastasis. Thus, the diverse components of the tumor microenvironment could represent the overlapping reexpression of amniotic and vitelline functions. These functions would favor a gastrulation-like process, that is, the creation of a reactive stroma in which fibrogenesis and angiogenesis stand out.
Collapse
|
969
|
Cargnoni A, Ressel L, Rossi D, Poli A, Arienti D, Lombardi G, Parolini O. Conditioned medium from amniotic mesenchymal tissue cells reduces progression of bleomycin-induced lung fibrosis. Cytotherapy 2011; 14:153-61. [PMID: 21954836 PMCID: PMC3279140 DOI: 10.3109/14653249.2011.613930] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background and aims We have demonstrated recently that transplantation of placental membrane-derived cells reduces bleomycin-induced lung fibrosis in mice, despite a limited presence of transplanted cells in host lungs. Because placenta-derived cells are known to release factors with potential immunomodulatory and trophic activities, we hypothesized that transplanted cells may promote lung tissue repair via paracrine-acting molecules. To test this hypothesis, we examined whether administration of conditioned medium (CM) generated from human amniotic mesenchymal tissue cells (AMTC) was able to reduce lung fibrosis in this same animal model. Methods Bleomycin-challenged mice were either treated with AMTC-CM or control medium, or were left untreated (Bleo group). After 9 and 14 days, the distribution and severity of lung fibrosis were assessed histologically with a scoring system. Collagen deposition was also evaluated by quantitative image analysis. Results At day 14, lung fibrosis scores in AMTC-CM-treated mice were significantly lower (P<0.05) compared with mice of the Bleo group, in terms of fibrosis distribution [1.0 (interquartile range, IQR 0.9) versus 3.0 (IQR 1.8)], fibroblast proliferation [0.8 (IQR 0.4) versus 1.6 (IQR 1.0)], collagen deposition [1.4 (IQR 0.5) versus 2.0 (IQR 1.2)] and alveolar obliteration [2.3 (IQR 0.8) versus 3.2 (IQR 0.5)]. No differences were observed between mice of the Bleo group and mice treated with control medium. Quantitative analysis of collagen deposition confirmed these findings. Importantly, AMTC-CM treatment significantly reduced the fibrosis progression between the two observation time-points. Conclusions This pilot study supports the notion that AMTC exert anti-fibrotic effects through release of yet unknown soluble factors.
Collapse
Affiliation(s)
- Anna Cargnoni
- Centro di Ricerca E. Menni, Fondazione Poliambulanza-Istituto Ospedaliero, Brescia, Italy
| | | | | | | | | | | | | |
Collapse
|
970
|
Abstract
Now that mesenchymal stem cells (MSCs) have been shown to be perivascular in vivo, the existing traditional view that focuses on the multipotent differentiation capacity of these cells should be expanded to include their equally interesting role as cellular modulators that brings them into a broader therapeutic scenario. We discuss existing evidence that leads us to propose that during local injury, MSCs are released from their perivascular location, become activated, and establish a regenerative microenvironment by secreting bioactive molecules and regulating the local immune response. These trophic and immunomodulatory activities suggest that MSCs may serve as site-regulated "drugstores" in vivo.
Collapse
Affiliation(s)
- Arnold I Caplan
- Skeletal Research Center, Department of Biology, Case Western Reserve University, Cleveland, OH 44106-7080, USA.
| | | |
Collapse
|
971
|
Abstract
Acute respiratory distress syndrome (ARDS) is a clinical syndrome of acute respiratory failure presenting with hypoxemia and bilateral pulmonary infiltrates, most often in the setting of pneumonia, sepsis, or major trauma. The pathogenesis of ARDS involves lung endothelial injury, alveolar epithelial injury, and the accumulation of protein-rich fluid and cellular debris in the alveolar space. No pharmacologic therapy has so far proved effective. A potential strategy involves cell-based therapies, including mesenchymal stem cells (MSCs). Herein we review basic properties of MSCs, their use in preclinical models of lung injury and ARDS, and potential therapeutic mechanisms.
Collapse
Affiliation(s)
- Jeffrey E Gotts
- Department of Medicine Anesthesiology, The Cardiovascular Research Institute, University of California, 505 Parnassus Avenue, Moffitt Hospital, Room M-917, San Francisco, CA 94143-0624, USA.
| | | |
Collapse
|
972
|
Abstract
There are a range of definitions for nanomaterials and a range of length scales that are considered nano, but one thing is consistent among fields: nanomaterials are small and special. Nanomaterials have the potential to have tremendous impact on medical treatments. In one example, nanomaterials are permitting the tracking of cells via magnetic resonance imaging (MRI) in clinical trials to assess the efficacy and safety of cellular therapies. In a second example, nanomaterials are acting as drug delivery vehicles for the targeted delivery of therapies to increase efficacy and to reduce side effects. However, there are distinct challenges that must be considered in the development and application of these materials, including careful analysis of the distribution and clearance of nanomaterials and their potential off-target effects. By carefully assessing materials early in their development at the bench, one may be able to move successful approaches through to the clinic more rapidly, which is indeed the goal of the field. For far too many conditions and diseases, the tools we have are less than adequate, and nanomaterials have the potential to fill that void. To realize this potential, investigators must be willing to invest time and resources to develop and to translate these technologies to the point where the risk is low enough that they have real commercial possibilities. Working collaboratively and leveraging resources and experience play important roles in moving technologies through preclinical and clinical testing. It requires incredible dedication of teams of researchers, but the result is new treatments and therapies.
Collapse
Affiliation(s)
- Erin Lavik
- Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States.
| | | |
Collapse
|
973
|
Liang H, Hou H, Yi W, Yang G, Gu C, Lau WB, Gao E, Ma X, Lu Z, Wei X, Pei J, Yi D. Increased expression of pigment epithelium-derived factor in aged mesenchymal stem cells impairs their therapeutic efficacy for attenuating myocardial infarction injury. Eur Heart J 2011; 34:1681-90. [PMID: 21606086 PMCID: PMC3675387 DOI: 10.1093/eurheartj/ehr131] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
AIMS Mesenchymal stem cells (MSCs) can ameliorate myocardial infarction (MI) injury. However, older-donor MSCs seem less efficacious than those from younger donors, and the contributing underlying mechanisms remain unknown. Here, we determine how age-related expression of pigment epithelium-derived factor (PEDF) affects MSC therapeutic efficacy for MI. METHODS AND RESULTS Reverse transcriptase-polymerized chain reaction and enzyme-linked immunosorbent assay analyses revealed dramatically increased PEDF expression in MSCs from old mice compared to young mice. Morphological and functional experiments demonstrated significantly impaired old MSC therapeutic efficacy compared with young MSCs in treatment of mice subjected to MI. Immunofluorescent staining demonstrated that administration of old MSCs compared with young MSCs resulted in an infarct region containing fewer endothelial cells, vascular smooth muscle cells, and macrophages, but more fibroblasts. Pigment epithelium-derived factor overexpression in young MSCs impaired the beneficial effects against MI injury, and induced cellular profile changes in the infarct region similar to administration of old MSCs. Knocking down PEDF expression in old MSCs improved MSC therapeutic efficacy, and induced a cellular profile similar to young MSCs administration. Studies in vitro showed that PEDF secreted by MSCs regulated the proliferation and migration of cardiac fibroblasts. CONCLUSIONS This is the first evidence that paracrine factor PEDF plays critical role in the regulatory effects of MSCs against MI injury. Furthermore, the impaired therapeutic ability of aged MSCs is predominantly caused by increased PEDF secretion. These findings indicate PEDF as a promising novel genetic modification target for improving aged MSC therapeutic efficacy.
Collapse
Affiliation(s)
- Hongliang Liang
- Department of Cardiovascular Surgery, Institute of Cardiovascular Disease of Chinese PLA, Xijing Hospital, the Fourth Military Medical University, No.127, West Changle Road, Xi'an, Shaanxi Province 710032, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
974
|
Markert CD, Ambrosio F, Call JA, Grange RW. Exercise and Duchenne muscular dystrophy: toward evidence-based exercise prescription. Muscle Nerve 2011; 43:464-78. [PMID: 21404285 DOI: 10.1002/mus.21987] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
To develop a rational framework for answering questions about the role of exercise in Duchenne muscular dystrophy (DMD), we focused on five pathophysiological mechanisms and offer brief hypotheses regarding how exercise may beneficially modulate pertinent cellular and molecular pathways. We aimed to provide an integrative overview of mechanisms of DMD pathology that may improve or worsen as a result of exercise. We also sought to stimulate discussion of what outcomes/dependent variables most appropriately measure these mechanisms, with the purpose of defining criteria for well-designed, controlled studies of exercise in DMD. The five mechanisms include pathways that are both intrinsic and extrinsic to the diseased muscle cells.
Collapse
Affiliation(s)
- Chad D Markert
- Wake Forest Institute for Regenerative Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157, USA.
| | | | | | | |
Collapse
|
975
|
Strategies for regeneration of the bone using porcine adult adipose-derived mesenchymal stem cells. Theriogenology 2011; 75:1381-99. [DOI: 10.1016/j.theriogenology.2010.11.020] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 11/11/2010] [Accepted: 11/12/2010] [Indexed: 12/17/2022]
|
976
|
Choi YH, Kurtz A, Stamm C. Mesenchymal stem cells for cardiac cell therapy. Hum Gene Ther 2011; 22:3-17. [PMID: 21062128 DOI: 10.1089/hum.2010.211] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Despite refinements of medical and surgical therapies, heart failure remains a fatal disease. Myocardial infarction is the most common cause of heart failure, and only palliative measures are available to relieve symptoms and prolong the patient's life span. Because mammalian cardiomyocytes irreversibly exit the cell cycle at about the time of birth, the heart has traditionally been considered to lack any regenerative capacity. This paradigm, however, is currently shifting, and the cellular composition of the myocardium is being targeted by various regeneration strategies. Adult progenitor and stem cell treatment of diseased human myocardium has been carried out for more than 10 years (Menasche et al., 2001; Stamm et al., 2003), and it has become clear that, in humans, the regenerative capacity of hematopoietic stem cells and endothelial progenitor cells, despite potent proangiogenic effects, is limited (Stamm et al., 2009). More recently, mesenchymal stem cells (MSCs) and related cell types are being evaluated in preclinical models of heart disease as well as in clinical trials (see Published Clinical Trials, below). MSCs have the capacity to self-renew and to differentiate into lineages that normally originate from the embryonic mesenchyme (connective tissues, blood vessels, blood-related organs) (Caplan, 1991; Prockop, 1997; Pittenger et al., 1999). The current definition of MSCs includes plastic adherence in cell culture, specific surface antigen expression (CD105(+)/CD90(+)/CD73(+), CD34(-)/CD45(-)/CD11b(-) or CD14(-)/CD19(-) or CD79α(-)/HLA-DR1(-)), and multilineage in vitro differentiation potential (osteogenic, chondrogenic, and adipogenic) (Dominici et al., 2006 ). If those criteria are not met completely, the term "mesenchymal stromal cells" should be used for marrow-derived adherent cells, or other terms for MSC-like cells of different origin. For the purpose of this review, MSCs and related cells are discussed in general, and cell type-specific properties are indicated when appropriate. We first summarize the preclinical data on MSCs in models of heart disease, and then appraise the clinical experience with MSCs for cardiac cell therapy.
Collapse
|
977
|
Wang F, Yu M, Yan X, Wen Y, Zeng Q, Yue W, Yang P, Pei X. Gingiva-derived mesenchymal stem cell-mediated therapeutic approach for bone tissue regeneration. Stem Cells Dev 2011; 20:2093-102. [PMID: 21361847 DOI: 10.1089/scd.2010.0523] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Up to now, the gingiva-derived mesenchymal stem cells (GMSCs) as a new postnatal stem cells have been isolated and characterized with multipotential differentiation capabilities in vitro. However, the in vivo efficacy of utilizing the GMSCs in bone regeneration remains obscure. First of all, we identified canonical MSCs in human gingival tissue, which possessed homogenous immunophenotype (CD34(-)CD45(-)CD29(+)CD105(+)CD90(+) STRO-1(+)) and had tri-lineage differentiation potential (osteoblasts, adipocytes, and chondrocytes). Next, we examined the efficacy of utilizing these stem cells in bone tissue regeneration; the enhanced green fluorescent protein-labeled GMSCs seeded on type I collagen gel were implanted into the mandibular defects as well as the critical-sized calvarial defects in Sprague Dawley rats. We first demonstrated that GMSCs could repair the mandibular wounds and calvarial defects at 2 months in rats postsurgical reconstruction. Histomorphological analysis and image of fluorescence microscope certified that new bone in the defect areas was derived from the transplanted GMSCs. Immunohistochemical analysis of green fluorescent protein, human collagen I, and osteopontin further confirmed our conclusion. The above results implied that mesenchymal stem cells derived from gingival tissue could be a novel source for stem cell-based therapy in bone reconstruction in clinical applications.
Collapse
Affiliation(s)
- Fang Wang
- Department of Periodontology, School of Stomatology, Shandong University, Jinan, Shandong Province, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
978
|
Pandit SR, Sullivan JM, Egger V, Borecki AA, Oleskevich S. Functional Effects of Adult Human Olfactory Stem Cells on Early-Onset Sensorineural Hearing Loss. Stem Cells 2011; 29:670-7. [DOI: 10.1002/stem.609] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
979
|
Hatlapatka T, Moretti P, Lavrentieva A, Hass R, Marquardt N, Jacobs R, Kasper C. Optimization of culture conditions for the expansion of umbilical cord-derived mesenchymal stem or stromal cell-like cells using xeno-free culture conditions. Tissue Eng Part C Methods 2011; 17:485-493. [PMID: 21166520 DOI: 10.1089/ten.tec.2010.0406] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
First isolated from bone marrow, mesenchymal stem or stromal cells (MSC) were shown to be present in several postnatal and extraembryonic tissues as well as in a large variety of fetal tissues (e.g., fatty tissue, dental pulp, placenta, umbilical cord blood, and tissue). In this study, an optimized protocol for the expansion of MSC-like cells from whole umbilical cord tissue under xeno-free culture conditions is proposed. Different fetal calf sera and human serum (HS) were compared with regard to cell proliferation and MSC marker stability in long-term expansion experiments, and HS was shown to support optimal growth conditions. Additionally, the optimal concentration of HS during the cultivation was determined. With regard to cell proliferative potential, apoptosis, colony-forming unit fibroblast frequency, and cell senescence, our findings suggest that an efficient expansion of the cells is carried out best in media supplemented with 10% HS. Under our given xeno-free culture conditions, MSC-like cells were found to display in vitro immunoprivileged and immunomodulatory properties, which were assessed by co-culture and transwell culture experiments with carboxyfluorescein diacetate succinimidyl ester-labeled peripheral blood mononuclear cells. These findings may be of great value for the establishment of biotechnological protocols for the delivery of sufficient cell numbers of high quality for regenerative medicine purposes.
Collapse
Affiliation(s)
- Tim Hatlapatka
- Institute of Technical Chemistry, Leibniz University Hannover, Hannover, Germany
| | | | | | | | | | | | | |
Collapse
|
980
|
Hilfiker A, Kasper C, Hass R, Haverich A. Mesenchymal stem cells and progenitor cells in connective tissue engineering and regenerative medicine: is there a future for transplantation? Langenbecks Arch Surg 2011; 396:489-497. [PMID: 21373941 DOI: 10.1007/s00423-011-0762-2] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Accepted: 02/21/2011] [Indexed: 12/14/2022]
Abstract
PURPOSE Transplantation surgery suffers from a shortage of donor organs worldwide. Cell injection and tissue engineering (TE), thus emerge as alternative therapy options. The purpose of this article is to review the progress of TE technology, focusing on mesenchymal stem cells (MSC) as a cell source for artificial functional tissue. RESULTS MSC from many different sources can be minimally invasively harvested: peripheral blood, fat tissue, bone marrow, amniotic fluid, cord blood. In comparison to embryonic stem cells (ESC), there are no ethical concerns; MSC can be extracted from autologous or allogenic tissue and cause an immune modulatory effect by suppressing the graft-versus-host reaction (GvHD). Furthermore, MSC do not develop into teratomas when transplanted, a consequence observed with ESC and iPS cells. CONCLUSION MSC as multipotent cells are capable of differentiating into mesodermal and non-mesodermal lineages. However, further studies must be performed to elucidate the differentiation capacity of MSC from different sources, and to understand the involved pathways and processes. Already, MSC have been successfully applied in clinical trials, e.g., to heal large bone defects, cartilage lesions, spinal cord injuries, cardiovascular diseases, hematological pathologies, osteogenesis imperfecta, and GvHD. A detailed understanding of the behavior and homing of MSC is desirable to enlarge the clinical application spectrum of MSC towards the in vitro generation of functional tissue for implantation, for example, resilient cartilage, contractile myocardial replacement tissue, and bioartificial heart valves.
Collapse
Affiliation(s)
- Andres Hilfiker
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany.
| | | | | | | |
Collapse
|
981
|
Deng W, Chen QW, Li XS, Liu H, Niu SQ, Zhou Y, Li GQ, Ke DZ, Mo XG. Bone marrow mesenchymal stromal cells with support of bispecific antibody and ultrasound-mediated microbubbles prevent myocardial fibrosis via the signal transducer and activators of transcription signaling pathway. Cytotherapy 2011; 13:431-40. [DOI: 10.3109/14653249.2010.542458] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
982
|
Majore I, Moretti P, Stahl F, Hass R, Kasper C. Growth and differentiation properties of mesenchymal stromal cell populations derived from whole human umbilical cord. Stem Cell Rev Rep 2011; 7:17-31. [PMID: 20596801 DOI: 10.1007/s12015-010-9165-y] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Up to 2.8 × 10(7) fibroblast-like cells displaying an abundant presence of mesenchymal stem cell (MSC) markers CD73, CD90, CD105 and a low level of HLA-I expression can be isolated from one whole human umbilical cord (UC) using a simple and highly reproducible explant culture approach. Cells derived from whole UC, similar to cells collected from separate compartments of UC, display a distinct chondrogenic and adipogenic potential. Therefore they are potential candidates for cartilage and adipose tissue engineering. Cell differentiation along the osteogenic pathway is, however, less efficient, even after the addition of 1.25-dihydroxyvitamin D3, a potent osteoinductive substance. Isolated cells are highly proliferative, tolerate cryopreservation with an average survival rate of about 75% and after thawing can be propagated further, at least over 20 population doublings before their proliferative activity begins to decline. More importantly, they synthesize numerous trophic factors including neurotrophins and factors which facilitate angiogenesis and hematopoiesis. In conclusion, cells isolated from whole UC satisfies all requirements essential for the generation of stem cell banks containing permanently available cell material for applications in the field of regenerative medicine. Nevertheless, further studies are needed to improve and adjust the methods which are already employed for adult MSC expansion and differentiation to specific properties and requirements of the primitive stem cells collected from UC. So, our data verify that the choice of individual parameters for cell propagation, such as duration of cell expansion and cell seeding density, has a substantial impact on the quality of UC-derived cell populations.
Collapse
Affiliation(s)
- Ingrida Majore
- Institute of Technical Chemistry, Leibniz University of Hannover, Callinstraße 5, Hannover 30167, Germany.
| | | | | | | | | |
Collapse
|
983
|
Albersen M, Fandel TM, Lin G, Wang G, Banie L, Lin CS, Lue TF. Injections of adipose tissue-derived stem cells and stem cell lysate improve recovery of erectile function in a rat model of cavernous nerve injury. J Sex Med 2011; 7:3331-40. [PMID: 20561166 DOI: 10.1111/j.1743-6109.2010.01875.x] [Citation(s) in RCA: 200] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Erectile dysfunction (ED) remains a major complication after radical prostatectomy. The use of adipose tissue-derived stem cells (ADSCs) has shown promising results for the treatment of ED. However, the mechanisms of action for stem cell therapy remain controversial, with increasing evidence pointing to paracrine pathways. AIM To determine the effects and to identify the mechanism of action of ADSC and ADSC-derived lysate in a rat model of cavernous nerve (CN) crush injury. METHODS Thirty-two male Sprague-Dawley rats were randomly divided into four equal groups: one group underwent sham operation, while three groups underwent bilateral CN crush. Crush-injury groups were treated at the time of injury with intracavernous injection of ADSC, lysate, or vehicle only (injured controls). Erectile function was assessed by CN electrostimulation at 4 weeks. Penile tissue was collected for histology. MAIN OUTCOME MEASURES Intracavernous pressure increase upon CN stimulation; neuronal nitric oxide synthase (nNOS) content in the dorsal penile nerve; smooth muscle content, collagen content, and number of apoptotic cells in the corpus cavernosum. RESULTS Both ADSC and lysate treatments resulted in significant recovery of erectile function, as compared with vehicle treatment. nNOS content was preserved in both the ADSC and lysate group, with significantly higher expression compared with vehicle-treated animals. There was significantly less fibrosis and a significant preservation of smooth muscle content in the ADSC and lysate groups compared with injured controls. The observed functional improvement after lysate injection supports the hypothesis that ADSCs act through release of intracellular preformed substances or by active secretion of certain biomolecules. The underlying mechanism of recovery appears to involve neuron preservation and cytoprotection by inhibition of apoptosis. CONCLUSIONS Penile injection of both ADSC and ADSC-derived lysate can improve recovery of erectile function in a rat model of neurogenic ED.
Collapse
Affiliation(s)
- Maarten Albersen
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, CA 94143-0738, USA
| | | | | | | | | | | | | |
Collapse
|
984
|
Shamis Y, Hewitt KJ, Carlson MW, Margvelashvilli M, Dong S, Kuo CK, Daheron L, Egles C, Garlick JA. Fibroblasts derived from human embryonic stem cells direct development and repair of 3D human skin equivalents. Stem Cell Res Ther 2011; 2:10. [PMID: 21338517 PMCID: PMC3092150 DOI: 10.1186/scrt51] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Accepted: 02/21/2011] [Indexed: 12/25/2022] Open
Abstract
Introduction Pluripotent, human stem cells hold tremendous promise as a source of progenitor and terminally differentiated cells for application in future regenerative therapies. However, such therapies will be dependent upon the development of novel approaches that can best assess tissue outcomes of pluripotent stem cell-derived cells and will be essential to better predict their safety and stability following in vivo transplantation. Methods In this study we used engineered, human skin equivalents (HSEs) as a platform to characterize fibroblasts that have been derived from human embryonic stem (hES) cell. We characterized the phenotype and the secretion profile of two distinct hES-derived cell lines with properties of mesenchymal cells (EDK and H9-MSC) and compared their biological potential upon induction of differentiation to bone and fat and following their incorporation into the stromal compartment of engineered, HSEs. Results While both EDK and H9-MSC cell lines exhibited similar morphology and mesenchymal cell marker expression, they demonstrated distinct functional properties when incorporated into the stromal compartment of HSEs. EDK cells displayed characteristics of dermal fibroblasts that could support epithelial tissue development and enable re-epithelialization of wounds generated using a 3D tissue model of cutaneous wound healing, which was linked to elevated production of hepatocyte growth factor (HGF). Lentiviral shRNA-mediated knockdown of HGF resulted in a dramatic decrease of HGF secretion from EDK cells that led to a marked reduction in their ability to promote keratinocyte proliferation and re-epithelialization of cutaneous wounds. In contrast, H9-MSCs demonstrated features of mesenchymal stem cells (MSC) but not those of dermal fibroblasts, as they underwent multilineage differentiation in monolayer culture, but were unable to support epithelial tissue development and repair and produced significantly lower levels of HGF. Conclusions Our findings demonstrate that hES-derived cells could be directed to specified and alternative mesenchymal cell fates whose function could be distinguished in engineered HSEs. Characterization of hES-derived mesenchymal cells in 3D, engineered HSEs demonstrates the utility of this tissue platform to predict the functional properties of hES-derived fibroblasts before their therapeutic transplantation.
Collapse
Affiliation(s)
- Yulia Shamis
- Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, 136 Harrison Avenue, Boston, MA, 02111, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
985
|
Nunes SS, Krishnan L, Gerard CS, Dale JR, Maddie MA, Benton RL, Hoying JB. Angiogenic potential of microvessel fragments is independent of the tissue of origin and can be influenced by the cellular composition of the implants. Microcirculation 2011; 17:557-67. [PMID: 21040121 DOI: 10.1111/j.1549-8719.2010.00052.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
UNLABELLED We have demonstrated that MFs isolated from adipose retain angiogenic potential in vitro and form a mature, perfused network when implanted. However, adipose-derived microvessels are rich in provascularizing cells that could uniquely drive neovascularization in adipose-derived MFs implants. OBJECTIVE Investigate the ability of MFs from a different vascular bed to recapitulate adipose-derived microvessel angiogenesis and network formation and analyze adipose-derived vessel plasticity by assessing whether vessel function could be modulated by astrocyte-like cells. METHODS MFs were isolated by limited collagenase digestion from rodent brain or adipose and assembled into 3D collagen gels in the presence or absence of GRPs. The resulting neovasculatures that formed following implantation were assessed by measuring 3D vascularity and vessel permeability to small and large molecular tracers. RESULTS Similar to adipose-derived MFs, brain-derived MFs can sprout and form a perfused neovascular network when implanted. Furthermore, when co-implanted in the constructs, GRPs caused adipose-derived vessels to express the brain endothelial marker glucose transporter-1 and to significantly reduce microvessel permeability. CONCLUSION Neovascularization involving isolated microvessel elements is independent of the tissue origin and degree of vessel specialization. In addition, adipose-derived vessels have the ability to respond to environmental signals and change vessel characteristics.
Collapse
Affiliation(s)
- Sara S Nunes
- Cardiovascular Innovation Institute, Louisville, Kentucky 40202, USA
| | | | | | | | | | | | | |
Collapse
|
986
|
Nombela-Arrieta C, Ritz J, Silberstein LE. The elusive nature and function of mesenchymal stem cells. Nat Rev Mol Cell Biol 2011; 12:126-31. [PMID: 21253000 PMCID: PMC3346289 DOI: 10.1038/nrm3049] [Citation(s) in RCA: 484] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) are a diverse subset of multipotent precursors present in the stromal fraction of many adult tissues and have drawn intense interest from translational and basic investigators. MSCs have been operationally defined by their ability to differentiate into osteoblasts, adipocytes and chondrocytes after in vitro expansion. Nevertheless, their identity in vivo, heterogeneity, anatomical localization and functional roles in adult tissue homeostasis have remained enigmatic and are only just starting to be uncovered.
Collapse
Affiliation(s)
| | - Jerome Ritz
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, MA
- Center for Human Cell Therapy, Division of Molecular and Cellular Medicine, Children's Hospital Boston, Boston, MA
| | - Leslie E. Silberstein
- Joint program in Transfusion Medicine, Children's Hospital Boston, Boston, MA
- Center for Human Cell Therapy, Division of Molecular and Cellular Medicine, Children's Hospital Boston, Boston, MA
| |
Collapse
|
987
|
Affiliation(s)
- Arnold I Caplan
- Skeletal Research Center, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH 44106-7080, USA.
| |
Collapse
|
988
|
Pan RL, Wang P, Xiang LX, Shao JZ. Delta-like 1 serves as a new target and contributor to liver fibrosis down-regulated by mesenchymal stem cell transplantation. J Biol Chem 2011; 286:12340-8. [PMID: 21239501 DOI: 10.1074/jbc.m110.194498] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Chronic liver injury always progresses to fibrosis and eventually to cirrhosis, a massive health care burden worldwide. Delta-like 1 (Dlk1) is well known as an inhibitor of adipocyte differentiation. However, whether it is involved in liver fibrosis remains unclear. Here, we provide the first evidence that Dlk1 is a critical contributor to liver fibrosis through promoting activation of hepatic stellate cells (HSCs) during chronic liver injury. We found that upon liver injury, Dlk1 was dramatically induced and initially expressed in hepatocytes and then into the HSCs by a paracrine manner. It leads to the activation of HSCs, which is considered to be a pivotal event in liver fibrogenesis. Two forms (∼50 and ∼25 kDa) of the Dlk1 protein were detected by Western blot analysis. In vitro administration of Dlk1 significantly promoted HSC activation, whereas in vivo knockdown of Dlk1 dramatically inhibited HSC activation and the subsequent fibrosis. The large soluble form (∼50 kDa) of Dlk1 was shown to contribute to HSC activation. We were encouraged to find the Dlk1-promoted HSC activation and liver fibrosis can be depressed by transplantation of bone marrow-mesenchymal stem cells (BM-MSCs). Furthermore, we demonstrated that FGF2 was up-regulated in BM-MSCs under injury stimulation, and it probably participated in the inhibition of Dlk1 by BM-MSCs. Our findings provide a novel role of Dlk1 in liver fibrosis leading to a better understanding of the molecular basis in fibrosis and cirrhosis and also give insights into the cellular and molecular mechanisms of MSC biology in liver repair.
Collapse
Affiliation(s)
- Ruo-Lang Pan
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | | | | | | |
Collapse
|
989
|
Tolar J, Le Blanc K, Keating A, Blazar BR. Concise review: hitting the right spot with mesenchymal stromal cells. Stem Cells 2011; 28:1446-55. [PMID: 20597105 DOI: 10.1002/stem.459] [Citation(s) in RCA: 296] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Mesenchymal stromal cells or mesenchymal stem cells (MSCs) have captured considerable scientific and public interest because of their potential to limit physical and immune injury, to produce bioactive molecules and to regenerate tissues. MSCs are phenotypically heterogeneous and distinct subpopulations within MSC cultures are presumed to contribute to tissue repair and the modulation of allogeneic immune responses. As the first example of efficacy, clinical trials for prevention and treatment of graft-versus-host disease after hematopoietic cell transplantation show that MSCs can effectively treat human disease. The view of the mechanisms whereby MSCs function as immunomodulatory and reparative cells has evolved simultaneously. Initially, donor MSCs were thought to replace damaged cells in injured tissues of the recipient. More recently, however, it has become increasingly clear that even transient MSC engraftment may exert favorable effects through the secretion of cytokines and other paracrine factors, which engage and recruit recipient cells in productive tissue repair. Thus, an important reason to investigate MSCs in mechanistic preclinical models and in clinical trials with well-defined end points and controls is to better understand the therapeutic potential of these multifunctional cells. Here, we review the controversies and recent insights into MSC biology, the regulation of alloresponses by MSCs in preclinical models, as well as clinical experience with MSC infusions (Table 1) and the challenges of manufacturing a ready supply of highly defined transplantable MSCs.
Collapse
Affiliation(s)
- Jakub Tolar
- Division of Hematology, Oncology, Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA.
| | | | | | | |
Collapse
|
990
|
Bitencourt CS, Pereira PA, Ramos SG, Sampaio SV, Arantes EC, Aronoff DM, Faccioli LH. Hyaluronidase recruits mesenchymal-like cells to the lung and ameliorates fibrosis. FIBROGENESIS & TISSUE REPAIR 2011; 4:3. [PMID: 21232095 PMCID: PMC3035036 DOI: 10.1186/1755-1536-4-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Accepted: 01/13/2011] [Indexed: 12/12/2022]
Abstract
Hyaluronidases (HYALs) comprise a group of enzymes that degrade hyaluronic acid (HA). In this report, we reveal that a single intranasal inoculation of HYAL induces an increase in mononuclear cells within the bronchoalveolar space demonstrating a mesenchymal-like phenotype, expressing stem cell antigen-1 (SCA-1), CD44 and CD73 but not CD34, CD45, CD3, CD4, CD8 or CD19. This influx of mesenchymal stem cell (MSC)-like cells was dependent on leukotriene production within the lung parenchyma. These findings prompted experiments demonstrating that HYAL treatment potently blocked bleomycin-induced lung injury and fibrosis while decreasing transforming growth factor (TGF)-β production and collagen deposition. These data suggest that HYAL is a novel and promising tool to use autologous MSC-like cells in the treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Claudia S Bitencourt
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, 14040-903, Brazil.
| | | | | | | | | | | | | |
Collapse
|
991
|
|
992
|
Yun SP, Ryu JM, Jang MW, Han HJ. Interaction of profilin-1 and F-actin via a β-arrestin-1/JNK signaling pathway involved in prostaglandin E(2)-induced human mesenchymal stem cells migration and proliferation. J Cell Physiol 2011; 226:559-71. [PMID: 20717968 DOI: 10.1002/jcp.22366] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Although many previous reports have examined the function of prostaglandin E(2) (PGE(2)) in the migration and proliferation of various cell types, the role of the actin cytoskeleton in human mesenchymal stem cells (hMSCs) migration and proliferation has not been reported. The present study examined the involvement of profilin-1 (Pfn-1) and filamentous-actin (F-actin) in PGE(2)-induced hMSC migration and proliferation and its related signal pathways. PGE(2) (10(-6) M) increased both cell migration and proliferation, and also increased E-type prostaglandin receptor 2 (EP2) mRNA expression, β-arrestin-1 phosphorylation, and c-Jun N-terminal kinase (JNK) phosphorylation. Small interfering RNA (siRNA)-mediated knockdown of β-arrestin-1 and JNK (-1, -2, -3) inhibited PGE(2)-induced growth of hMSCs. PGE(2) also activated Pfn-1, which was blocked by JNK siRNA, and induced F-actin level and organization. Downregulation of Pfn-1 by siRNA decreased the level and organization of F-actin. In addition, specific siRNA for TRIO and F-actin-binding protein (TRIOBP) reduced the PGE(2)-induced increase in hMSC migration and proliferation. Together, these experimental data demonstrate that PGE(2) partially stimulates hMSCs migration and proliferation by interaction of Pfn-1 and F-actin via EP2 receptor-dependent β-arrestin-1/JNK signaling pathways.
Collapse
Affiliation(s)
- Seung Pil Yun
- Department of Veterinary Physiology, College of Veterinary Medicine, Biotherapy Human Resources Center (BK21), Chonnam National University, Gwangju, Korea
| | | | | | | |
Collapse
|
993
|
Si YL, Zhao YL, Hao HJ, Fu XB, Han WD. MSCs: Biological characteristics, clinical applications and their outstanding concerns. Ageing Res Rev 2011; 10:93-103. [PMID: 20727988 DOI: 10.1016/j.arr.2010.08.005] [Citation(s) in RCA: 172] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 07/29/2010] [Accepted: 08/04/2010] [Indexed: 12/19/2022]
Abstract
Mesenchymal stem cells (MSCs) are multi-potent adult stem cells harboring multi-lineage differentiation potential and immunosuppressive properties that make MSCs an ideal candidate cell type for immunomodulation and regenerative medicine. Currently, MSC-related researches and clinical trials have evoked exciting promise in a variety of disorders and tissue regeneration. However, it must be recognized that several critical potential problems have also emerged from current clinical trials, for example: (1) the indefinite association between the phenotypic characteristics and the biological functions of MSCs; (2) the lack of clinical data to support the long-term safety of MSCs; (3) the need for further clarification of multiple mechanisms of MSC transplant actions in vivo; and (4) the lack of comparability of MSC transplant efficacy. Therefore, MSC-based therapies could not yet be considered a routine treatment in the clinic. Based on these, we proposed that large-scale and multi-center clinical trials of MSC-based therapies should be initiated under strict supervision. These interventions might help to establish a new clinical paradigm to turn MSC transplantation into a routine therapy for at least some diseases in the near future.
Collapse
Affiliation(s)
- Yi-Ling Si
- Institute of Basic Medicine Science, Chinese PLA General Hospital, 28 Fuxing Road, Beijing 100853, China
| | | | | | | | | |
Collapse
|
994
|
Delaine-Smith RM, Reilly GC. The effects of mechanical loading on mesenchymal stem cell differentiation and matrix production. VITAMINS AND HORMONES 2011; 87:417-80. [PMID: 22127254 DOI: 10.1016/b978-0-12-386015-6.00039-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells or stromal cells (MSCs) have the potential to be used therapeutically in tissue engineering and regenerative medicine to replace or restore the function of damaged tissues. Therefore, considerable effort has been ongoing in the research community to optimize culture conditions for predifferentiation of MSCs. All mesenchymal tissues are subjected to mechanical forces in vivo and all fully differentiated mesenchymal lineage cells respond to mechanical stimulation in vivo and in vitro. Therefore, it is not surprising that MSCs are highly mechanosensitive. We present a summary of current methods of mechanical stimulation of MSCs and an overview of the outcomes of the different mechanical culture techniques tested. Tissue engineers and stem cell researchers should be able to harness this mechanosensitivity to modulate MSC differentiation and matrix production; however, more research needs to be undertaken to understand the complex interactions between the mechanosensitive and biochemically stimulated differentiation pathways.
Collapse
Affiliation(s)
- Robin M Delaine-Smith
- The Kroto Research Institute, Department of Materials Science and Engineering, University of Sheffield, Sheffield, United Kingdom
| | | |
Collapse
|
995
|
Genever PG. The Generation of Three-dimensional Tissue Structures with Mesenchymal Stem Cells. Altern Lab Anim 2010; 38 Suppl 1:31-4. [DOI: 10.1177/026119291003801s08] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Mesenchymal stem cells (MSCs) are multipotent stem cells, found in the bone-marrow and other adult tissues, which give rise to various cell lineages. Although MSCs are biologically important, and may have widespread therapeutic potential, they are not well-characterised, particularly in terms of their cell surface receptors and in vivo phenotype. We aimed to develop a three-dimensional (3-D) MSC in vitro model, in order to understand the factors involved in the regulation of lineage specification routes. A suitable model, which replicates the MSC microenvironment as accurately as possible, will allow more-detailed investigations into the phenotype of the cells. Our MSC spheroids appear to have an enhanced mesenchymal differentiation compared to two-dimensional MSC monolayers. With this in vitro system, it is possible to perform real-time analysis of cellular differentiation status. MSC spheroids may also be amenable for use in high-throughput assays. A more-recent research project aims to generate knockout micro-tissues, based on human 3-D MSCs, as an alternative to animal studies.
Collapse
Affiliation(s)
- Paul G. Genever
- Biomedical Tissue Research, Department of Biology, University of York, UK
| |
Collapse
|
996
|
Yan XL, Lan Y, Wang XY, He WY, Yao HY, Chen DB, Xiong JX, Gao J, Li Z, Yang G, Li XS, Liu YL, Zhang JY, Liu B, Mao N. Migration of dorsal aorta mesenchymal stem cells induced by mouse embryonic circulation. Dev Dyn 2010; 240:65-74. [DOI: 10.1002/dvdy.22490] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
997
|
Uccelli A, Prockop DJ. Why should mesenchymal stem cells (MSCs) cure autoimmune diseases? Curr Opin Immunol 2010; 22:768-74. [PMID: 21093239 DOI: 10.1016/j.coi.2010.10.012] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Revised: 10/19/2010] [Accepted: 10/19/2010] [Indexed: 12/13/2022]
Abstract
The adult stem/progenitor cells from bone marrow and other tissues referred to as mesenchymal stem cells or multipotent mesenchymal stromal cells (MSCs) display a significant therapeutic plasticity as reflected by their ability to enhance tissue repair and influence the immune response both in vitro and in vivo. In this review we will focus on the paradigmatic preclinical experience achieved testing MSCs in experimental autoimmune encephalomyelitis (EAE), a model for multiple sclerosis. We will emphasize how the paradigm changed over time from the original prediction that MSCs would enhance tissue repair through their transdifferentiation into somatic cells to the current paradigm that they can produce therapeutic benefits without engraftment into the injured tissues. The data will be reviewed in terms of the potentials of MSCs for therapy of autoimmune diseases.
Collapse
Affiliation(s)
- Antonio Uccelli
- Department of Neurosciences Ophthalmology and Genetics, University of Genoa, Italy.
| | | |
Collapse
|
998
|
Bunnell BA, Betancourt AM, Sullivan DE. New concepts on the immune modulation mediated by mesenchymal stem cells. Stem Cell Res Ther 2010; 1:34. [PMID: 21092149 PMCID: PMC3025436 DOI: 10.1186/scrt34] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are the nonhematopoietic multipotent progenitor cells found in various adult tissues. They are characterized by their ease of isolation and their rapid growth in vitro while maintaining their differentiation potential, allowing for extensive expansion in culture that yields large quantities suitable for therapeutic use. This article reviews the immunomodulatory activities associated with MSCs. Numerous studies have demonstrated that MSCs are potently immunosuppressive in vitro and in vivo. However, this article presents a new paradigm in MSC biology, in which MSCs, at least in vitro, can undergo polarization into either a pro-inflammatory or an immunosuppressive phenotype.
Collapse
Affiliation(s)
- Bruce A Bunnell
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, 1430 Tulane Ave, SL-99, New Orleans, LA 70112, USA.
| | | | | |
Collapse
|
999
|
Sullivan JM, Cohen MA, Pandit SR, Sahota RS, Borecki AA, Oleskevich S. Effect of epithelial stem cell transplantation on noise-induced hearing loss in adult mice. Neurobiol Dis 2010; 41:552-9. [PMID: 21059389 DOI: 10.1016/j.nbd.2010.11.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2010] [Revised: 10/19/2010] [Accepted: 11/01/2010] [Indexed: 12/20/2022] Open
Abstract
Noise trauma in mammals can result in damage to multiple epithelial cochlear cell types, producing permanent hearing loss. Here we investigate whether epithelial stem cell transplantation can ameliorate noise-induced hearing loss in mice. Epithelial stem/progenitor cells isolated from adult mouse tongue displayed extensive proliferation in vitro as well as positive immunolabelling for the epithelial stem cell marker p63. To examine the functional effects of cochlear transplantation of these cells, mice were exposed to noise trauma and the cells were transplanted via a lateral wall cochleostomy 2 days post-trauma. Changes in auditory function were assessed by determining auditory brainstem response (ABR) threshold shifts 4 weeks after stem cell transplantation or sham surgery. Stem/progenitor cell transplantation resulted in a significantly reduced permanent ABR threshold shift for click stimuli compared to sham-injected mice, as corroborated using two distinct analyses. Cell fate analyses revealed stem/progenitor cell survival and integration into suprastrial regions of the spiral ligament. These results suggest that transplantation of adult epithelial stem/progenitor cells can attenuate the ototoxic effects of noise trauma in a mammalian model of noise-induced hearing loss.
Collapse
Affiliation(s)
- Jeremy M Sullivan
- Hearing Research Group, Neuroscience Program, Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia
| | | | | | | | | | | |
Collapse
|
1000
|
Boeuf S, Richter W. Chondrogenesis of mesenchymal stem cells: role of tissue source and inducing factors. Stem Cell Res Ther 2010; 1:31. [PMID: 20959030 PMCID: PMC2983444 DOI: 10.1186/scrt31] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Multipotent mesenchymal stromal cells (MSCs) are an attractive cell source for cell therapy in cartilage. Although their therapeutic potential is clear, the requirements and conditions for effective induction of chondrogenesis in MSCs and for the production of a stable cartilaginous tissue by these cells are far from being understood. Different sources of MSCs have been considered for cartilage tissue engineering, mainly based on criteria of availability, as for adipose tissue, or of proximity to cartilage and the joint environment in vivo, as for bone marrow and synovial tissues. Focussing on human MSCs, this review will provide an overview of studies featuring comparative analysis of the chondrogenic differentiation of MSCs from different sources. In particular, it will examine the influence of the cells' origin on the requirements for the induction of chondrogenesis and on the phenotype achieved by the cells after differentiation.
Collapse
Affiliation(s)
- Stephane Boeuf
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Schlierbacher Landstrasse 200a, 69118 Heidelberg, Germany.
| | | |
Collapse
|