Keratinocyte Induced Differentiation of Mesenchymal Stem Cells into Dermal Myofibroblasts: A Role in Effective Wound Healing
PDF
HTML

Keywords

Mesenchymal stem cells, keratinocyte conditioned medium, dermal myofibroblast, cytokine secretion, wound healing, scarring, animal models.

How to Cite

Mishra, P. J., Mishra, P. J., & Banerjee, D. (2016). Keratinocyte Induced Differentiation of Mesenchymal Stem Cells into Dermal Myofibroblasts: A Role in Effective Wound Healing. International Journal of Translational Science, 2016, 5–32. https://doi.org/10.13052/ijts2246-8765.2016.002

Abstract

We have previously demonstrated that human mesenchymal stem cells
(hMSCs) migrate toward human keratinocytes as well as toward conditioned
medium from cultured human keratinocytes (KCM) indicating that the hMSCs
respond to signals from keratinocytes [1]. Using fluorescently labeled cells
we now show that in vitro hMSCs appear to surround keratinocytes, and
this organization is recapitulated in vivo. Incubation of hMSCs with KCM
induced dermal myofibroblast like differentiation characterized by expression
of cytoskeletal markers and increased expression of cytokines including
SDF-1, IL-8, IL-6 and CXCL5. Interaction of keratinocytes with hMSCs
appears to be important in the wound healing process. Therapeutic efficacy of
hMSCs in wound healing was examined in two animal models representing
normal and chronic wound healing. Accelerated wound healing was observed when hMSCs and KCM exposed hMSCs (KCMSCs) were injected near
wound site in nude and NOD/SCID mice. Long term follow up of wound
healing revealed that in the hMSC treated wounds there was little evidence of
residual scarring. These dermal myofibroblast like hMSCs add to the wound
healing process. Together, the keratinocyte and hMSCs morphed dermal
myofibroblast like cells as well as the factors secreted by these cells support
wound healing with minimal scarring. The ability of hMSCs to support wound
healing process represents another striking example of the importance of
keratinocyte and hMSCs interplay in the wound microenvironment resulting
in effective wound healing with minimal scarring.

https://doi.org/10.13052/ijts2246-8765.2016.002
PDF
HTML

References

Mishra, P.J., Mishra, P.J., and Banerjee, D. (2012) Cell free derivatives

from mesenchymal stem cells are effectives in wound healing. World

J. Stem Cells 4(5): 35–43.

Singer, A.J., and Clark, R.A. (1999). Cutaneous wound healing. N. Engl.

J. Med. 341, 738–46.

Fowler E. (1990) “Chronic wounds: an overview,” in Chronic wound

care: a clinical source book for healthcare professionals, ed. D. Krasner,

–18 King of Prussia, PA: Health Management Publications, Inc.

P. J. Mishra et al.

Rees, R. S., and Hirshberg, J. A. (1999). Wound care centers: costs, care,

and strategies. Adv. Wound Care 12, 4–7.

Callam, M. (1992). Prevalence of chronic leg ulceration and severe

chronic venous disease in Western countries. Phlebology 7, S6–S12.

Goldman, R. (2004). Growth factors and chronic wound healing: past,

present, and future. Adv. Skin Wound Care 17, 24–35.

Baum, C. L., and Arpey, C. J. (2005). Normal cutaneous wound healing:

clinical correlation with cellular and molecular events. Dermatol Surg.

, 674–686.

Sibbald, R. G., and Cameron, J. (2001). “Dermatological aspects of

wound care,” in Chronic wound care: a clinical source book for

healthcare professionals, 3rd Edn. Wayne, PA: Health Management

Publications. 273–285.

Mathus-Vliegen, E. M. (2004). Old age, malnutrition, and pressure sores:

an ill-fated alliance, J. Gerontol A Biol. Sci. Med. Sci. 59, 355–360.

Robles, D. T., and Berg, D. (2004). Abnormal wound healing: keloids.

Clin Dermatol. 25, 26–32.

Falanga, V. (2005). Wound healing and its impairment in the diabetic

foot. Lancet. 366, 1736–1743.

Margolis, D. J., Allen-Taylor, L., Hoffstad, D., and Berlin, J. A. (2005).

Diabetic neuropathic foot ulcers and amputation, Wound Repair Regen.

, 230–236.

Bansal, C., Scott, R., Stewart, D., and Cockerell, C. J. (2005). Decubitus

ulcers: a review of the literature, Int J Dermatol. 44, 805–810.

Ochoa, O., Torres, F. M., Shireman, P. K. (2007). Chemokines and

diabetic wound healing. Vascular 15, 350–355.

Grey, J. E., Harding, K. G., and Enoch, S. (2006). Venous and arterial

leg ulcers, BMJ 332, 347–350.

Wynn, T.A. (2007). Common and unique mechanisms regulate fibrosis

in various fibroproliferative diseases. J. Clin. Invest. 117, 524–529.

Chen, S. L. et al. (2004). Effect on left ventricular function of intracoro-

nary transplantation of autologous bone marrow mesenchymal stem cell

in patients with acute myocardial infarction. Am. J. Cardiol. 94, 92–95.

Giordano, A., Galderisi, U., Marino I. R. (2007). From the labora-

tory bench to the patient’s bedside: an update on clinical trials with

mesenchymal stem cells. J Cell Physiol. 211, 27–35.

Chen, S., et al. (2006). Intracoronary transplantation of autologous bone

marrow mesenchymal stem cells for ischemic cardiomyopathy due to

Mesenchymal Stem Cells and Wound Healing 27

isolated chronic occluded left anterior descending artery. J. Invasive

Cardiol.18, 552–556.

Ball, L. M. et al. (2007). Cotransplantation of ex vivo expanded

mesenchymal stem cells accelerates lymphocyte recovery and may

reduce the risk of graft failure in haploidentical hematopoietic stem-cell

transplantation. Blood 1, 2764–2767.

Le Blanc, K., and Ringdén, O. (2007). Immunomodulation by mesenchy-

mal stem cells and clinical experience. Journal of Internal Medicine.

, 509–525.

Pittenger, M. F., et al. (1999). Multilineage poptential of adult human

mesenchymal stem cells. Science. 284, 143–147.

Laflamme, M. A., and Murry, C. E. (2005). Regenerating the heart. Nat.

Rev. 23, 845–856.

Gregory, C. A., Prockop, D. J., and Spees, J. L. (2005). Non hematopoi-

etic bone marrow stem cells: molecular control of expansion and

differentiation. Exp. Cell Res. 306, 330–335.

Satake, K., Lou, J., Lenke, L. G. (2004). Migration of mesenchymal stem

cells through cerebrospinal fluid into injured spinal cord tissue. Spine

, 1971–1979.

Wynn, R. F. et al. (2004). A small proportion of mesenchymal stem

cells strongly expresses functionally active CXCR4 receptor capable of

promoting migration to bone marrow. Blood 104, 2643–2645.

Ji, J. F., He, B. P., Dheen, S. T., and Tay, S. S. (2004). Interactions of

chemokines and chemokine receptors mediate the migration of mes-

enchymal stem cells to the impaired site in the brain after hypoglossal

nerve injury. Stem Cells 22, 415–427.

Mahmood, A., Lu, D., Lu, M., and Chopp, M. (2003). Treatment of

traumatic brain injury in adult rats with intravenous administration of

human bone marrow stromal cells. Neurosurgery 53, 697–702.

Mishra, P.J. et al. (2008). Carcinoma associated fibroblast like differen-

tiation of Bone marrow derived mesenchymal stem cells. Cancer Res.

, 4331–4339.

Gabbiani, G. (1996). The cellular derivation and the life span of the

myofibroblast. Pathol Res. Pract.192, 708–711.

Helary, C., Ovtracht, L., Coulomb, B., Godeau, G., and Giraud-

Guille, M. M. (2006). Dense fibrillar collagen matrices: a model to

study myofibroblast behaviour during wound healing. Biomaterials 27,

–4452.

P. J. Mishra et al.

Darby, I., Skalli, O., and Gabbiani, G. (1990).Alpha-smooth muscle actin

is transiently expressed by myofibroblasts during experimental wound

healing. Lab Invest. 63, 21–9.

Hinz, B. and Gabbiani, G. (2003). Mechanisms of force generation and

transmission by myofibroblasts. Curr. Opin. Biotechnol.14, 538–546.

Arora, P. D., and McCulloch C. A. (1994). Dependence of collagen

remodelling on alpha-smooth muscle actin expression by fibroblasts.

J. Cell Physiol.159, 161–175.

Hinz, B., and Gabbiani, G. (2003). Cell–matrix and cell–cell contacts of

myofibroblasts: role in connective tissue remodeling. Thromb. Haemost.

, 993–1002.

Germain, L., Jean, A., Auger, F. A., Garrel, D. R. (1994). Human

wound healing fibroblasts have greater contractile properties than dermal

fibroblasts. J. Surg. Res. 57, 268–273.

Hinz, B. (2007). Formation and function of the myofibroblast during

tissue repair. J Invest. Dermatol.127, 526–537.

Sykova, E., and Jendelova, P. (2007). In vivo tracking of stem cells in

brain and spinal cord injury. Prog. Brain Res.161, 367–383.

Nedeau, A.E. et al. (2008). A CXCL5 and bFGF-dependent effect of

PDGF-B-activated fibroblasts in promoting trafficking and differentia-

tion of bone marrow derived mesenchymal stem cells. Exp. Cell Res.

, 2176–2186.

Ozaki, Y. et al. (2007). Comprehensive analysis of chemotactic factors

for bone marrow mesenchymal stem cells. Stem Cells Dev.16, 119–129.

Mansilla, E. et al. (2006). Bloodstream cells phenotypically identical to

human mesenchymal bone marrow stem cells circulate in large amounts

under the influence of acute large skin damage: New evidence for their

use in regenerative medicine. Transplantation Proc. 38, 967–969.

Fukuda, K., and Fujita, J. (2005). Mesenchymal, but not hematopoietic,

stem cells can be mobilized and differentiate into cardiomyocytes after

myocardial infarction in mice. Kidney Int. 68(5), 1940–1943.

Wu, Y., Chen, L., Scott, P. G., and Tredget, E. E. (2007). Mesenchy-

mal stem cells enhance wound healing through differentiation and

angiogenesis. Stem Cells 25, 2648–2659.

Fu, X., Fang, L., Li, X., Cheng, B., and Sheng, Z. (2006). Enhanced

wound healing quality with bone marrow mesenchymal stem cells

autografting after skin injury. Wound Repair Regen.14, 325–335.

Mesenchymal Stem Cells and Wound Healing 29

Li, H., Fu, X., Ouyang, Y., Cai, C. Wang, J. and Sun, T. (2006).Adult bone

marrow derivedmesenchymal stem cells contribute to wound healing of

skin appendages. Cell Tissue Res. 326, 725–736.

Menon, L.G. et al. (2007). Differential gene expression associated with

migration of mesenchymal stem cells to conditioned medium from tumor

cells or bone marrow cells. Stem Cells 25, 520–528.

Vernon, R. B. and Sage, E. H. (1996). Contraction of fibrillar type I

collagen by endothelial cells: a study in vitro. J. Cell. Biochem. 60,

–197.

Vernon, R. B., and Gooden, M.J. (2002). An improved method for

the collagen gel contraction assay. In vitro Cell. Dev. Biol.-Animal 38,

–101.

Wysocki, A. (1996). Wound measurement. Int J Dermatol 35, 82–91.

Sasaki, M. et al. (2008). Mesenchymal stem cells are recruited into

wounded skin and contribute to wound repair by transdifferentiation

into multiple skin types. J. Immunol.180, 2581–2587.

Schneider, R. K. M., et al. (2008). Three dimensional epidermis like

growth of human mesenchymal stem cells on dermal equivalents:

Contribution to tissue organization by adaptation of myofibroblastsic

phenotype and function. Differentiation 76, 156–67.

Wipff, P.J., Rifkin, D.B., Meister, J.J., and Hinz, B. (2007). Myofibrob-

last contraction activates latent TGF-beta1 from the extracellular matrix.

J. Cell Biol. 179, 1311–1323.

Rennekampff, H.O. et al. (2000). Bioactive interleukin-8 is expressed

in wounds and enhances wound healing. J. Surg. Res. 93, 41–54.

Lin, Z.-Q., Kondo, T., Ishida, Y., Takayasu, T., and Mukaida, N. (2003).

Essential involvement of IL-6 in the skin wound-healing process as

evidenced by delayed wound healing in IL-6-deficient mice. J. Leukoc.

Biol. 73, 713–721.

Shephard, P., et al. (2004). Myofibroblast Differentiation is induced in

keratinocyte cocultures and is antagonistically regulated by endogenous

transforming growth factor-b and interleukin-1. Am J Pathol. 164,

–2066.

Werner, S., Krieg, T., and Smola, H. (2007). Keratinocyte–Fibroblast

Interactions in Wound Healing. J. Investig. Dermatol. 127, 998–1008.

Smith, E. et al. (2008). Duffy antigen receptor for chemokines and

CXCL5 are essential for the recruitment of neutrophils in a multi-

cellular model of rheumatoid arthritis synovium. Arthritis Rheum. 58,

–1973.

P. J. Mishra et al.

Lee, J. S. et al. (2003). Duffy antigen facilitates movement of chemokine

across the endothelium in vitro and promotes neutrophil transmigration

in vitro and in vivo. J. Immunol. 170, 5244–5251.

Seeber, R. M., John, A. E., Eidne, K. A., Greaves, D. R. (2008). The

duffy antigen/receptor for chemokines exists in an oligomeric form

in living cells and functionally antagonizes CCR5 signaling through

hetero-oligomerization. Mol. Pharmacol. 73, 1362–1370.

Chen, L., Tredget, E. E., Wu, P. Y. G., Wu, Y. (2008). Paracrine factors of

Mesenchymal Stem Cells recruit macrophages and endothelial lineage

cells and enhance wound healing. PloS ONE. 3, 1–12

Downloads

Download data is not yet available.