Elsevier

Advanced Drug Delivery Reviews

Volume 62, Issue 12, 30 September 2010, Pages 1156-1166
Advanced Drug Delivery Reviews

Mesenchymal stem cells as therapeutics and vehicles for gene and drug delivery

https://doi.org/10.1016/j.addr.2010.08.010Get rights and content

Abstract

Mesenchymal stem cells (MSCs) possess a set of several fairly unique properties which make them ideally suited both for cellular therapies/regenerative medicine, and as vehicles for gene and drug delivery. These include: 1) relative ease of isolation; 2) the ability to differentiate into a wide variety of seemingly functional cell types of both mesenchymal and non-mesenchymal origin; 3) the ability to be extensively expanded in culture without a loss of differentiative capacity; 4) they are not only hypoimmunogenic, but they produce immunosuppression upon transplantation; 5) their pronounced anti-inflammatory properties; and 6) their ability to home to damaged tissues, tumors, and metastases following in vivo administration. In this review, we summarize the latest research in the use of mesenchymal stem cells in regenerative medicine, as immunomodulatory/anti-inflammatory agents, and as vehicles for transferring both therapeutic genes in genetic disease and genes designed to destroy malignant cells.

Section snippets

Isolation and characterization of MSC

In pioneering studies [1], [2] performed over 30 years ago, Friedenstein demonstrated that fibroblastoid cells obtained from the bone marrow were capable of transferring the hematopoietic microenvironment to ectopic sites, thus establishing the concept that the marrow microenvironment resided within the so-called stromal cells of the marrow. Years later, scientists began to explore the full potential of these microenvironmental cells, and results of these studies led to the realization that this

Sources of MSC

Although much of the work to date has focused on MSC isolated from adult bone marrow, it is important to realize that cells that appear phenotypically and functionally to be MSC have now been isolated by our group and others from numerous tissues, including brain, liver, lung, fetal blood, umbilical cord blood, kidney, and even liposuction material [19], [20], [21], [22], [23], [24], [25], [26]. The broad distribution of MSC throughout the body leads one to postulate that MSC are likely to play

MSC in regenerative medicine

The in vitro and in vivo differentiation of MSC into the various mesenchymal cell types found within the bone marrow, i.e. bone, cartilage, and fat, has now been described by numerous laboratories, and the conditions to bring about each of these differentiative pathways have been delineated in detail [7]. Recent studies employing microarrays [30], [41], [42], [43] have now begun to shed light on the molecular mechanisms responsible for commitment to and progression along each of these lineages,

MSC as trophic “factories”

One issue which has complicated interpretation of the data generated from the aforementioned studies in liver as well as those conducted looking at the potential of MSC to mediate repair in the heart and other organ systems, is the fact that a therapeutic benefit is often observed in the absence of any evidence of sustained engraftment of the transplanted MSC within the damaged organ. Instead, it appears that the transplantation of MSC somehow stimulates the damaged host organ to repair itself

MSC as vehicles for delivering therapeutic genes

While MSC possess tremendous therapeutic potential by virtue of their ability to lodge/engraft within multiple tissues in the body and both give rise to tissue-specific cells and release trophic factors that trigger the tissue's own endogenous repair pathways, gene therapists have realized that these properties are just the beginning of the therapeutic applications for MSC [24], [174], [175]. By using gene therapy to engineer MSC to either augment their own natural production of specific

Use of MSC as immunomodulatory/anti-inflammatory agents

In addition to their broad differentiative capabilities and their potential utility as gene delivery vehicles, MSC represent a rather unique cell from an immunological standpoint, since MSC are known to be relatively hypoimmunogenic. They do not normally express MHC class II or the co-stimulatory molecules CD80 and CD82, unless they are stimulated with IFN-γ. As such, they do not seem to serve as very good targets for lysis by cytotoxic T cells or NK cells, and do not really stimulate the

MSC as anti-cancer agents

Cancer represents another condition in which there is a selective need for new cells created by the forming tumor. Studies over the last several years have now revealed that MSC have the ability to “sense” this need, migrate to the forming tumor following intravenous administration, and contribute to the newly forming tumor “stroma”. While this may not seem ideal, since the MSC could, in fact, provide support to the growing tumor, this property has now been realized to present a very powerful

Conclusions

With numerous investigators around the globe having established and verified that MSC harbor the ability to cross embryonic germ layers and give rise to a wide range of what developmental biology had taught were tissue-specific cells, it is now clear that the differentiative capacity of MSC is far broader than anyone would have foreseen at the time Friedenstein originally described his bone marrow-derived CFU-F. In addition to this tremendous differentiative potential, the relative ease with

References (252)

  • W. Ando et al.

    Cartilage repair using an in vitro generated scaffold-free tissue-engineered construct derived from porcine synovial mesenchymal stem cells

    Biomaterials

    (2007)
  • E.M. Horwitz et al.

    Clinical responses to bone marrow transplantation in children with severe osteogenesis imperfecta

    Blood

    (2001)
  • T.E. Ichim et al.

    Mesenchymal stem cells as anti-inflammatories: implications for treatment of Duchenne muscular dystrophy

    Cell Immunol.

    (2010)
  • P. Bossolasco et al.

    Neuro-glial differentiation of human bone marrow stem cells in vitro

    Exp. Neurol.

    (2005)
  • B. Neuhuber et al.

    Axon growth and recovery of function supported by human bone marrow stromal cells in the injured spinal cord exhibit donor variations

    Brain Res.

    (2005)
  • T. Honma et al.

    Intravenous infusion of immortalized human mesenchymal stem cells protects against injury in a cerebral ischemia model in adult rat

    Exp. Neurol.

    (2006)
  • S.K. Kang et al.

    Improvement of neurological deficits by intracerebral transplantation of human adipose tissue-derived stromal cells after cerebral ischemia in rats

    Exp. Neurol.

    (2003)
  • T. Nomura et al.

    I.V. infusion of brain-derived neurotrophic factor gene-modified human mesenchymal stem cells protects against injury in a cerebral ischemia model in adult rat

    Neuroscience

    (2005)
  • J. Chen et al.

    Therapeutic benefit of intracerebral transplantation of bone marrow stromal cells after cerebral ischemia in rats

    J. Neurol. Sci.

    (2001)
  • S. Iihoshi et al.

    A therapeutic window for intravenous administration of autologous bone marrow after cerebral ischemia in adult rats

    Brain Res.

    (2004)
  • K. Kurozumi et al.

    Mesenchymal stem cells that produce neurotrophic factors reduce ischemic damage in the rat middle cerebral artery occlusion model

    Mol. Ther.

    (2005)
  • K. Kurozumi et al.

    BDNF gene-modified mesenchymal stem cells promote functional recovery and reduce infarct size in the rat middle cerebral artery occlusion model

    Mol. Ther.

    (2004)
  • W. Zheng et al.

    Therapeutic benefits of human mesenchymal stem cells derived from bone marrow after global cerebral ischemia

    Brain Res.

    (2010)
  • E. Zappia et al.

    Mesenchymal stem cells ameliorate experimental autoimmune encephalomyelitis inducing T-cell anergy

    Blood

    (2005)
  • L. Cova et al.

    Multiple neurogenic and neurorescue effects of human mesenchymal stem cell after transplantation in an experimental model of Parkinson's disease

    Brain Res.

    (2010)
  • Y. Li et al.

    Intracerebral transplantation of bone marrow stromal cells in a 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine mouse model of Parkinson's disease

    Neurosci. Lett.

    (2001)
  • D.J. Prockop et al.

    Potential use of marrow stromal cells as therapeutic vectors for diseases of the central nervous system

    Prog. Brain Res.

    (2000)
  • T. Tondreau et al.

    Bone marrow-derived mesenchymal stem cells already express specific neural proteins before any differentiation

    Differentiation

    (2004)
  • K. Fukuda

    Reprogramming of bone marrow mesenchymal stem cells into cardiomyocytes

    C R Biol.

    (2002)
  • W.S. Shim et al.

    Ex vivo differentiation of human adult bone marrow stem cells into cardiomyocyte-like cells

    Biochem. Biophys. Res. Commun.

    (2004)
  • Y. Cao et al.

    Human adipose tissue-derived stem cells differentiate into endothelial cells in vitro and improve postnatal neovascularization in vivo

    Biochem. Biophys. Res. Commun.

    (2005)
  • S. Tomita et al.

    Improved heart function with myogenesis and angiogenesis after autologous porcine bone marrow stromal cell transplantation

    J. Thorac. Cardiovasc. Surg.

    (2002)
  • Q. Li et al.

    Intra-myocardial delivery of mesenchymal stem cells ameliorates left ventricular and cardiomyocyte contractile dysfunction following myocardial infarction

    Toxicol. Lett.

    (2010)
  • S. Ohnishi et al.

    Transplantation of mesenchymal stem cells attenuates myocardial injury and dysfunction in a rat model of acute myocarditis

    J. Mol. Cell. Cardiol.

    (2007)
  • A.J. Friedenstein

    Osteogenic stem cells in the bone marrow

    Bone Miner.

    (1991)
  • A.J. Friedenstein et al.

    Stromal cells responsible for transferring the microenvironment of the hemopoietic tissues. Cloning in vitro and retransplantation in vivo

    Transplantation

    (1974)
  • A.I. Caplan

    Mesenchymal stem cells

    J. Orthop. Res.

    (1991)
  • M. Kassem

    Mesenchymal stem cells: biological characteristics and potential clinical applications

    Cloning Stem Cells

    (2004)
  • E.A. Luria et al.

    Fibroblast colony formation from monolayer cultures of blood cells

    Transfusion

    (1971)
  • M.F. Pittenger et al.

    Multilineage potential of adult human mesenchymal stem cells

    Science

    (1999)
  • J.A. Airey et al.

    Human mesenchymal stem cells form Purkinje fibers in fetal sheep heart

    Circulation

    (2004)
  • M.G. Almeida-Porada et al.

    Human marrow stromal cells (MSC) represent a latent pool of stem cells capable of generating long-term hematopoietic cells

    Blood

    (2001)
  • J. Chamberlain et al.

    Efficient generation of human hepatocytes by the intrahepatic delivery of clonal human mesenchymal stem cells in fetal sheep

    Hepatology

    (2007)
  • E. Colletti et al.

    Tales from the crypt: mesenchymal stem cells for replenishing the intestinal stem cell pool

    Blood

    (2008)
  • E.J. Colletti et al.

    Human mesenchymal stem cells differentiate promptly into tissue-specific cell types without cell fusion, mitochondrial or membrane vesicular transfer in fetal sheep

    Blood

    (2007)
  • S.P. Bruder et al.

    Mesenchymal stem cell surface antigen SB-10 corresponds to activated leukocyte cell adhesion molecule and is involved in osteogenic differentiation

    J. Bone Miner. Res.

    (1998)
  • J.B. Mitchell et al.

    Immunophenotype of human adipose-derived cells: temporal changes in stromal-associated and stem cell-associated markers

    Stem Cells

    (2006)
  • C.G. Fan et al.

    Characterization and neural differentiation of fetal lung mesenchymal stem cells

    Cell Transplant.

    (2005)
  • C. Gotherstrom et al.

    Difference in gene expression between human fetal liver and adult bone marrow mesenchymal stem cells

    Haematologica

    (2005)
  • P.S. in 't Anker et al.

    Mesenchymal stem cells in human second-trimester bone marrow, liver, lung, and spleen exhibit a similar immunophenotype but a heterogeneous multilineage differentiation potential

    Haematologica

    (2003)
  • Cited by (0)

    This review is part of the Advanced Drug Delivery Reviews theme issue on “Stem Cell Gene Manipulation and Delivery as Systemic Therapeutics”.

    View full text