Cellular and molecular basis of wound healing in diabetes

Similar documents
Defining Outcomes for Clinical Wound Research in Older Adults February 21, 2014

Biomarker Discovery: Prognosis and Management of Chronic Diabetic Foot Ulcers

Tissue repair. (3&4 of 4)

Effectiveness of topical insulin in management of chronic diabetic foot ulcers

Wound Healing Stages

Adjunctive Therapies: The Use of Skin Substitutes and Growth Factors in Venous Leg Ulceration (VLU)

Wound Healing & Limb Salvage. Dr. Harold Brem The Wound Healing Program Columbia University Medical Center November 8, 2006

The Georgetown Team Approach to Diabetic Limb Salvage: 2013

ulcer healing role 118 Bicarbonate, prostaglandins in duodenal cytoprotection 235, 236

YUJI YAMAGUCHI Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences

JMSCR Vol 06 Issue 03 Page March 2018

WOUND CARE UPDATE. -Commonly Used Skin Substitute Products For Wound. -Total Contact Casting. Jack W. Hutter DPM, FACFAS, C. ped.

Hyperbaric Oxygen Utilization in Wound Care

A New Approach To Diabetic Foot Ulcers Using Keratin Gel Technology

Vascular Endothelial Growth Factor Delivery via Gene Therapy for Diabetic Wounds: First Steps

Diabetic Foot Ulcer Treatment and Prevention

Original Article Tissue engineered skin for diabetic foot ulcers: a meta-analysis

Journal Club Semmler Lorenz

Vol. 209,. 2, August 2009 Abbreviations and Acronyms DFU PU VU WEMR diabetic foot ulcer pressure ulcer venous ulcer Wound Electronic Medical Record De

PREPARED FOR: U.S. Army Medical Research and Materiel Command Fort Detrick, Maryland

Calcium alginate/bone marrow mesenchymal stem cells combined with degradation membrane for repair of skin defects

Regenerative Tissue Matrix in Treatment of Wounds

Topically Applicable Stromal Cell Growth Factors - Encapsulated Cosmeceuticals

Scott E. Palmer, V.M.D. Diplomate, A.B.V.P., Eq. Practice New Jersey Equine Clinic

Interleukin-20 is associated with delayed healing in diabetic wounds

Increased Matrix Metalloproteinase-9 Predicts Poor Wound Healing in Diabetic Foot Ulcers

Spherical Nucleic Acids For Advanced Wound Healing Applications Chad A. Mirkin

Muscular Dystrophy. Biol 405 Molecular Medicine

GENE THERAPY IN CARDIOVASCULAR DISEASES ASAN MEDICAL CENTER KI HOON HAN MD

Endothelial PGC 1 - α 1 mediates vascular dysfunction in diabetes

Introduction. Cancer Biology. Tumor-suppressor genes. Proto-oncogenes. DNA stability genes. Mechanisms of carcinogenesis.

Research Article Cytological Evaluation of Hyaluronic Acid on Wound Healing Following Extraction

Clinical Review Criteria

Which molecules of the initial phase of wound healing may be used as markers for early detection of skin damage?

PROLIFERATION AND ABILITY FOR EPIDERMAL AUTOREGENERATION IN PATIENTS WITH CHRONIC LOWER LEG VENOUS ULCERATIONS*

Healing & Repair. Tissue Regeneration

Targeting X-Linked Inhibitor of Apoptosis Protein for Melanoma Therapy: The Need for More Homogeneous Samples and the Importance of Cell Lines

Frequency of Debridements and Time to Heal A Retrospective Cohort Study of Wounds

Cancer and Oncogenes Bioscience in the 21 st Century. Linda Lowe-Krentz

Inflammation is Not the Enemy

Abhd2 regulates alveolar type Ⅱ apoptosis and airway smooth muscle remodeling: a key target of COPD research

Emerging Use of Topical Biologics in Limb Salvage Role of Activated Collagen in Multimodality Treatment

Tissue renewal and Repair. Nisamanee Charoenchon, PhD Department of Pathobiology, Faculty of Science

Cell Death & Renewal (part 2)

CD34 + VEGFR-3 + progenitor cells have a potential to differentiate towards lymphatic endothelial cells

Hyperbaric Oxygen: Putting Principles into Practice Session 245 Sunday, September 11 th 2011.

STUDY. complication of diabetes, affecting 12% to 15% of patients. during their lifetime. 1,2 Diabetic foot ulcers (DFUs) impair quality

Healing and Repair. Dr. Nabila Hamdi MD, PhD

A comprehensive study on effect of recombinant human epidermal growth factor gel in diabetic foot ulcer

The Role of Hyperbaric Therapy in Wound Management

Physiology and Advancements in Wound Healing

Dressings do not heal wounds properly selected dressings enhance the body s ability to heal the wound. Progression Towards Healing

T cell development October 28, Dan Stetson

EB 2009 The DEBRA International Epidermolysis Bullosa Research Conference Vienna, 6-8 September 2009

Follow this and additional works at:

DIABETES AND THE AT-RISK LOWER LIMB:

Hyperbarics in Diabetic Wound Care. Aurel Mihai, MD & Brian Kline, MD

PhD THESIS Epigenetic mechanisms involved in stem cell differentiation

Growth Factors. BIT 230 Walsh Chapter 7

Mesenchymal Stem Cells

Citation for published version (APA): Gostynski, A. (2014). Revertant cell therapy for epidermolysis bullosa [S.l.]: [S.n.]

Abnormally differentiating keratinocytes in the epidermis of systemic sclerosis patients show enhanced secretion of CCN2 and S100A9

THE BIOLOGY OF PLATELET-GEL THERAPY

Small Molecule Inhibitor of the Wnt Pathway (SM04755) as a Potential Topical Scleroderma Treatment

Oncolytic Virotherapy: Targeting Cancer Stem Cells

Potential Role of Sphingosine 1-Phosphate in the. Pathogenesis of Rheumatoid Arthritis

A Post-hoc Analysis of Reduction in Diabetic Foot Ulcer Size at 4 Weeks as a Predictor of Healing by 12 Weeks

Four Types of Vertebrate Tissue

Immunological Lung Diseases

The role of oxygen insufficiency in the onset and development of the vascular complications of diabetes

Current Status of Pressure Ulcer Management in the US Geriatric Population Gregory A Bohn, MD MAPWCA, ABPM/UHMS

Authors' objectives To assess the value of treatments for foot ulcers in patients with Type 2 diabetes mellitus.

Generation of post-germinal centre myeloma plasma B cell.

Research Article Topical Application of Sadat-Habdan Mesenchymal Stimulating Peptide (SHMSP) Accelerates Wound Healing in Diabetic Rabbits

HHS Public Access Author manuscript J Invest Dermatol. Author manuscript; available in PMC 2015 January 01.

Adjunctive Hyperbaric Oxygen Therapy in the Lower Extremity. Helen Gelly, MD, FUHM, FACCWS, FACHM, UHM/ABPM

ESPEN Congress Prague 2007

Correlation between expression and significance of δ-catenin, CD31, and VEGF of non-small cell lung cancer

Clinical Policy Title: Growth factor treatment for wound healing/musculoskeletal uses

The Angiopoietin Axis in Cancer

A review on treatment and management of diabetic foot ulcer

Use of an Acellular Regenerative Tissue Matrix Over Chronic Wounds

In the treatment of partial and full-thickness chronic wounds TRANSFORM YOUR APPROACH TO HEALING: SIGNAL THE BODY, NOT THE WOUND DERMA

Journal Club WS 2012/13 Stefanie Nickl

Expert Recommendations for Optimizing Outcomes Utilizing Apligraf for Diabetic Foot Ulcers

Index. derm.theclinics.com. Note: Page numbers of article titles are in boldface type.

Genetics contributes to size. Bundschu et al., 2005, JBC Shingleton et al., 2005, PLOS

Role of Inflammatory and Progenitor Cells in Pulmonary Vascular Remodeling: Potential Role for Targeted Therapies. Traditional Hypothesis Stress

Hot Topics in Wound Care 2011

4/18/2011. Physiology 67 Lecture on Neural Development

Accelerate Your Research with Conversant Bio

vi Preface Table 2 Association of Fibrosis With Types of Injury: Representative Examples

Department of Dermatology Faculty of Medicine Siriraj Hospital, Mahidol University Bangkok, THAILAND

SMA IS A SEVERE NEUROLOGICAL DISORDER [1]

Adaptive immune responses: T cell-mediated immunity

ULCERS 1/12/ million diabetics in the US (2012) Reamputation Rate 26.7% at 1 year 48.3% at 3 years 60.7% at 5 years

Contents 1 Introduction Structure and function of the skin Wound definition and implications 2 Phases and processes during healing Inflammatory phase

Clinical Policy Title: Growth factor treatment for wound healing/musculoskeletal uses

Evidence-Based Rationale

Transcription:

Cellular and molecular basis of wound healing in diabetes Harold Brem, Marjana Tomic-Canic J Clin Invest. 2007;117(5):1219-1222. https://doi.org/10.1172/jci32169. Commentary Diabetic foot ulcers (DFUs), a leading cause of amputations, affect 15% of people with diabetes. A series of multiple mechanisms, including decreased cell and growth factor response, lead to diminished peripheral blood flow and decreased local angiogenesis, all of which can contribute to lack of healing in persons with DFUs. In this issue of the JCI, Gallagher and colleagues demonstrate that in diabetic mice, hyperoxia enhances the mobilization of circulating endothelial progenitor cells (EPCs) from the bone marrow to the peripheral circulation (see the related article beginning on page 1249). Local injection of the chemokine stromal cell derived factor 1a then recruits these EPCs to the cutaneous wound site, resulting in accelerated wound healing. Thus, Gallagher et al. have identified novel potential targets for therapeutic intervention in diabetic wound healing. Find the latest version: http://jci.me/32169-pdf

16. Meyer, M., and Wagner, E. 2006. Recent developments in the application of plasmid DNA-based vectors and small interfering RNA therapeutics for cancer. Hum. Gene Ther. 17:1062 1076. 17. Xia, X., Zhou, H., Huang, Y., and Xu, Z. 2006. Allele-specific RNAi selectively silences mutant SOD1 and achieves significant therapeutic benefit in vivo. Neurobiol. Dis. 23:578 586. 18. Yang, T.P., Stout, J.T., Patel, P.I., Alsford, R.L., and Caskey, C.T. 1988. Spontaneous reversion of novel Lesch- Nyhan mutation by HPRT gene rearrangement. Somat. Cell Mol. Genet. 14:293 303. 19. Jonkman, M.F. 1999. Revertant mosaicism in human genetic disorders. Am. J. Med. Genet. 85:361 364. 20. Bittar, M., and Happle, R. 2005. Revertant mosaicism and retrotransposons: another explanation of natural gene therapy. Am. J. Med. Genet. A. 137:222. 21. Pizzuti, A., Pieretti, M., Fenwick, R.G., Gibba, R.A., and Caskey, C.T. 1992. A transposon-like element in the deletion-prone region of the dystrophin gene. Genomics. 13:594 600. 22. Thanh, L.T., Nguyen, T.M., Helliwell, T.R., and Morris, G.E. 1995. Characterization of revertant muscle fibers in Duchenne muscular dystrophy, using exon-specific monoclonal antibodies against dystrophin. Am. J. Hum. Genet. 56:725 731. 23. Jonkman, M.F., et al. 1997. Revertant mosaicism in epidermolysis bullosa caused by mitotic gene conversion. Cell. 88:543 551. 24. Bliksrud, Y.T., Brodtkorb, E., Andresen, P.A., van den Berg, I.E.T., and Kvittingen, E.A. 2005. Tyrosinaemia type I de novo mutation in liver tissue suppressing an inborn splicing defect. J. Mol. Med. 83:406 410. 25. Wada, T., et al. 2005. Oligoclonal expansion of T lymphocytes with multiple second-site mutations leads to Omenn syndrome in a patient with RAG1- deficient severe combined immunodeficiency. Blood. 106:2099 2101. 26. Mavilio, F., et al. 2007. Correction of junctional epidermolysis bullosa by transplantation of genetically modified epidermal stem cells. Nat. Med. 12:1397 1402. Cellular and molecular basis of wound healing in diabetes Harold Brem 1 and Marjana Tomic-Canic 2 1 Wound Healing and Vascular Biology Laboratory, Division of Plastic Surgery, Columbia University College of Physicians and Surgeons, New York, New York, USA. 2 Tissue Repair Laboratory, Tissue Engineering, Regeneration, and Repair Program, and Department of Dermatology, Hospital for Special Surgery, Weill Medical College of Cornell University, New York, New York, USA. Diabetic foot ulcers (DFUs), a leading cause of amputations, affect 15% of people with diabetes. A series of multiple mechanisms, including decreased cell and growth factor response, lead to diminished peripheral blood flow and decreased local angiogenesis, all of which can contribute to lack of healing in persons with DFUs. In this issue of the JCI, Gallagher and colleagues demonstrate that in diabetic mice, hyperoxia enhances the mobilization of circulating endothelial progenitor cells (EPCs) from the bone marrow to the peripheral circulation (see the related article beginning on page 1249). Local injection of the chemokine stromal cell derived factor 1α then recruits these EPCs to the cutaneous wound site, resulting in accelerated wound healing. Thus, Gallagher et al. have identified novel potential targets for therapeutic intervention in diabetic wound healing. Diabetes affects approximately 170 million people worldwide, including 20.8 million in the USA (1), and by 2030 these numbers are projected to double (2). The foot ulcer is a leading cause of hospital admissions for people with diabetes in the developed world (3) and is a major morbidity associated with diabetes, often leading to pain, suffering, and a poor quality of life for patients. Diabetic foot ulcers (DFUs) are estimated to occur in 15% of all patients with diabetes (3) and precede 84% of all diabetes-related lowerleg amputations (4). Nonstandard abbreviations used: DFU, diabetic foot ulcer; EPC, endothelial progenitor cell; HBO, hyperbaric oxygen therapy; SDF-1α, stromal cell derived factor 1α. Conflict of interest: The authors have declared that no conflict of interest exists. Citation for this article: J. Clin. Invest. 117:1219 1222 (2007). doi:10.1172/jci32169. Despite the existence of protocols to standardize care, the physiological impairments that can result in a DFU complicate the healing process. Currently, the only FDAapproved growth factor and cell therapies for DFUs are not routinely used during treatment, preventing professionals from implementing evidence-based protocols (5). Molecular pathogenesis of diabetic wound healing The moment a person with diabetes suffers a break in the skin of their foot, they become at danger for amputation. Most commonly, patients have neuropathy, which could be causative. When coupled with an impaired ability to fight infection, these patients become largely unable to mount an adequate inflammatory response. Thus, the DFU that may look like a healing wound becomes a portal for infection that can lead to sepsis and require limb amputation. Over 100 known physiologic factors contribute to wound healing deficiencies in individuals with diabetes (Figure 1). These include decreased or impaired growth factor production (6 8), angiogenic response (8, 9), macrophage function (10), collagen accumulation, epidermal barrier function, quantity of granulation tissue (8), keratinocyte and fibroblast migration and proliferation, number of epidermal nerves (11), bone healing, and balance between the accumulation of ECM components and their remodeling by MMPs (12). Wound healing occurs as a cellular response to injury and involves activation of keratinocytes, fibroblasts, endothelial cells, macrophages, and platelets. Many growth factors and cytokines released by these cell types are needed to coordinate and maintain healing. Molecular analyses of biopsies from the epidermis of patients have identified pathogenic markers that correlate with delayed wound healing. These include overexpression of c-myc and nuclear localization of β-catenin (13). Coupled with a reduction in and abnormal localization of EGFR and activation of the glucocorticoid pathway, keratinocyte migration is inhibited (13, 14). At the nonhealing edge (callus) of DFUs, keratinocytes show an absence of migration, hyperproliferation, and incomplete differentiation (13, 14). Fibroblasts demonstrate a phenotypic change as well as decreased migration and proliferation. The Journal of Clinical Investigation http://www.jci.org Volume 117 Number 5 May 2007 1219

Figure 1 Mechanisms of wound healing in healthy people versus people with diabetes. In healthy individuals (left), the acute wound healing process is guided and maintained through integration of multiple signals (in the form of cytokines and chemokines) released by keratinocytes, fibroblasts, endothelial cells, macrophages, and platelets. During wound-induced hypoxia, VEGF released by macrophages, fibroblasts, and epithelial cells induces the phosphorylation and activation of enos in the bone marrow, resulting in an increase in NO levels, which triggers the mobilization of bone marrow EPCs to the circulation. The chemokine SDF-1a promotes the homing of these EPCs to the site of injury, where they participate in neovasculogenesis. In this issue of the JCI, Gallagher et al. (18) show that, in a murine model of diabetes (right), enos phosphorylation in the bone marrow is impaired, which directly limits EPC mobilization from the bone marrow into the circulation. They also show that SDF-1a expression is decreased in epithelial cells and myofibroblasts in the diabetic wound, which prevents EPC homing to wounds and therefore limits wound healing. The authors further show that establishing hyperoxia in wound tissue (via HBO therapy) activated many NOS isoforms, increased NO levels, and enhanced EPC mobilization to the circulation. However, local administration of SDF-1a was required to trigger homing of these cells to the wound site. These results suggest that HBO therapy combined with SDF-1a administration may be a potential therapeutic option to accelerate diabetic wound healing alone or in combination with existing clinical protocols. In contrast, cells from an adjacent nonulcerated area display the appearance of a normal phenotype but are still physiologically impaired. However, they are able to respond to administration of additional growth factors or cellular therapy. Microarray analyses of patient biopsies have confirmed these clinical findings by showing that the transcription profiles of epithelial cells from the two locations (callus and adjacent nonulcerated skin) are distinct and recognizable (14). Molecular surgery: integrative approach to healing Proper debridement, defined as the removal of hyperkeratotic, infected, and nonviable tissue from a wound, is essential as it accelerates wound healing (15, 16) via a multitude of mechanisms (15, 17). It is essential that pathological diagnosis is used to assure elimination of the hyperkeratotic epithelium and evaluate the extent of debridement (Figure 2). Molecular markers (13) may be utilized in the future development of rapid pathology tests for the guidance of effective debridement procedure. DFU debridement should be done in a sequential fashion until no callus or hyperkeratotic tissue is seen in the periphery of the wound and no scar or infection is present in the base of the wound. Bone marrow progenitors in diabetic wound healing Bone marrow derived endothelial progenitor cells (EPCs) play a significant role in the process of neovascularization in response to 1220 The Journal of Clinical Investigation http://www.jci.org Volume 117 Number 5 May 2007

Figure 2 Molecular basis of debridement. A typical foot ulcer in a person with diabetes is shown at top. (i) The nonhealing edge (callus) containing ulcerogenic cells with molecular markers indicative of healing impairment. (ii) Phenotypically normal but physiologically impaired cells, which can be stimulated to heal. Despite a wound s appearance after debridement, it may not be healing and may need to be evaluated for the presence of molecular markers of inhibition and/or hyperkeratotic tissue (e.g., c-myc and β-catenin). We expect more such molecular markers will be identified in the future. Once a wound is debrided, pathology analyses along with immunohistochemistry should determine whether the extent of debridement was sufficient. If the extent of debridement was not sufficient (lower left diagram), cells positive for c-myc (green) and nuclear β-catenin (purple) will be found, indicating the presence of ulcerogenic cells, which will prevent the wound from healing and indicate that additional debridement is necessary. Lack of healing is also demarcated by a thicker epidermis, thicker cornified layer, and presence of nuclei in the cornified layer. If the debridement was successful (lower right lower diagram), no staining for c-myc or β-catenin will be found, indicating an absence of ulcerogenic cells and successful debridement. These markers of inhibition are useful, but the most important goal is actual healing as defined by the appearance of new epithelium, decreased area of the wound, and no drainage. This information should be stored electronically in the Wound Electronic Medical Record (WEMR), which provides an objective analysis coupled with pathology and microbiology reports. ischemic conditions, as may be the case in diabetic wounds complicated by decreased peripheral blood flow. In this issue of the JCI, Gallagher and colleagues (18) show that EPCs in the bone marrow respond to ischemia by following chemokine gradients, which results in the homing of these cells to sites of hypoxia, where they then participate in the formation of new blood vessels. Bone marrow derived EPCs are mobilized by enos activation in the bone marrow, a process that the authors hypothesized is impaired in diabetics, thus preventing these cells from reaching the wound site in significant numbers. Hyperoxia has been shown to stimulate enos activation in some tissues (19). Therefore, to test the effect of hyperoxia on enos activation and EPC recruitment, the authors wounded and later exposed diabetic and nondiabetic mice to hyperbaric oxygen therapy (HBO). Results showed that although the total numbers of active EPCs were much lower in diabetic mice than in controls, hyperoxia does indeed spur the mobilization of EPCs from the bone marrow to the bloodstream (Figure 1). EPC mobilization into the bloodstream occurs through an increase in bone marrow enos activation as a result of hyperoxia. The increased enos stimulates NO production, which in turn helps to produce EPCs from the bone marrow. Though hyperoxia can increase the levels of circulating EPCs in the bloodstream, the cells are not effectively mobilized to the wound site, creating another roadblock in the path to healing. The term homing relates to the signals that attract and stimulate the cells involved in healing to migrate to sites of injury and aid in repair. EPC recruitment to the wound site depends on ischemia-induced upregulation of stromal cell derived factor 1α (SDF-1α). Gallagher et al. (18) also report a decrease in SDF-1α expression particularly by epithelial cells and myofibroblasts derived from wounds of streptozocin-induced diabetic mice. The decrease in SDF-1α was found to be responsible for decreased EPC homing. This defect was largely corrected by the simultaneous administration of HBO and SDF-1α at the wound site. These results imply The Journal of Clinical Investigation http://www.jci.org Volume 117 Number 5 May 2007 1221

that the decreased expression of SDF-1α by epithelial cells and myofibroblasts may be responsible for the lack of EPC homing to the periphery of diabetic wounds. This work is extremely important because it underscores the complexity of regulatory responses in diabetic wounds and explains the inconsistent response to currently approved hyperoxia protocols (i.e., HBO) in patients with diabetes (20). From bench to bedside A combination of therapeutic approaches is likely to lead to a successful treatment outcome for diabetic wounds. Although EPCs seem to be ideal candidates for in vivo cellbased therapies for ischemia, Gallagher and colleagues show here that singularly targeting one aspect of EPC function is not completely efficient in correcting diabetic vascular complications due to the deficits affecting both the cellular and cytokine-mediated responses to ischemia (18). Therefore, future therapies for ischemic complications will have to be based on correcting multiple deficits simultaneously. Therapeutic interventions, including correcting both EPC activation via HBO and EPC homing via administration of SDF-1α, may significantly accelerate diabetic wound healing by correcting the EPC deficit inherent to diabetic wounds. Clinically, approved evidence-based protocols based on adequate off-loading (21) coupled with the use of FDA-approved biological therapies that have undergone rigorous controlled randomized trials (5, 22) must be utilized. These FDA-approved treatments are clinically efficacious and include PDGF-BB (23), fibroblasts delivered in an absorbable mesh (24), and fibroblasts and keratinocytes delivered in type 1 collagen (25). If simultaneously combined with current therapies, potential treatments targeting enos activation and EPC recruitment might further stimulate healing. Wound progression should be monitored with a Wound Electronic Medical Record. This allows nonhealing wounds (those that have not healed after 2 weeks of treatment) to be objectively measured and treatment to be tailored accordingly. From the laboratory perspective, there is a compelling need to develop technologies to rapidly convert current discoveries into effective therapy for people with diabetes as well as to provide understanding of the molecular and cellular etiologies of patients with foot ulcers. Identification of the DFU genome and correlation of transcriptional profiles with clinical outcomes will determine specific genes that prevent a wound from healing and should further be utilized to develop therapies to prevent wound progression and promote rapid healing. The promise of the future Technologies for various molecular analyses (such as genomics, proteomics, transgenic mice), systems for sustained topical delivery (such as polymers and adenovirus vectors), major advances in tissue engineering (such as human skin engineering, cellular matrices, and bone marrow derived cell therapy), novel discoveries of disease molecular pathogenesis from studies of patient biopsies and animal models, and developments in molecular targeting (in areas such as antisense oligonucleotides, sirna, antibodies, and small molecules), coupled with breakthroughs in stem cell research, hold the promise of a bright future. All of these powerful technologies could potentially be applied to people with diabetic wounds in the near future. One of the major remaining steps is the integration of these resources into a coordinated effort to make the technology developed at the bench available to patients at the bedside. It is these synergistic therapies, starting at the molecular and cellular level, that will help to eliminate amputations in patients with diabetes. Acknowledgments We are very grateful to members of the Brem and Tomic laboratories for their assistance and helpful comments, especially Lisa Martínez and Irena Pastar. We apologize to our colleagues whose contributions were not cited due to space limitations. Our research is supported by the NIH grants R01NR08029, R01LM008443, and DK0859424. Address correspondence to: Harold Brem, Department of Surgery, Columbia University, 5141 Broadway, Rm. 3020, New York, New York 10034, USA. Phone: (212) 932-4325; Fax: (212) 658-9806; E-mail: hb2133@ columbia.edu. 1. National Diabetes Information Clearinghouse. National Diabetes Statistics fact sheet. http://diabetes. niddk.nih.gov/dm/pubs/statistics/index.htm. 2. Wild, S., Roglic, G., Green, A., Sicree, R., and King, H. 2004. Global prevalence of diabetes: estimates for the year 2000 and projections for 2030. Diabetes Care. 27:1047 1053. 3. Reiber, G.E., et al. 1999. Causal pathways for incident lower-extremity ulcers in patients with diabetes from two settings. Diabetes Care. 22:157 162. 4. Reiber, G.E., Boyko, E.J., and Smith, D.G. 1995. Lower extremity foot ulcers and amputations in diabetes. In Diabetes in America. M.I. Harris and M.P. Stern, editors. U.S. Government Printing Office. Bethesda, Maryland, USA. 409 428. 5. Brem, H., Sheehan, P., Rosenberg, H.J., Schneider, J.S., and Boulton, A.J. 2006. Evidence-based protocol for diabetic foot ulcers. Plast. Reconstr. Surg. 117:193S 209S; discussion 210S 211S. 6. Galkowska, H., Wojewodzka, U., and Olszewski, W.L. 2006. Chemokines, cytokines, and growth factors in keratinocytes and dermal endothelial cells in the margin of chronic diabetic foot ulcers. Wound Repair Regen. 14:558 565. 7. Goren, I., Muller, E., Pfeilschifter, J., and Frank, S. 2006. Severely impaired insulin signaling in chronic wounds of diabetic ob/ob mice: a potential role of tumor necrosis factor-alpha. Am. J. Pathol. 168:765 777. 8. Falanga, V. 2005. Wound healing and its impairment in the diabetic foot. Lancet. 366:1736 1743. 9. Galiano, R.D., et al. 2004. Topical vascular endothelial growth factor accelerates diabetic wound healing through increased angiogenesis and by mobilizing and recruiting bone marrow derived cells. Am. J. Pathol. 164:1935 1947. 10. Maruyama, K., et al. 2007. Decreased macrophage number and activation lead to reduced lymphatic vessel formation and contribute to impaired diabetic would healing. Am. J. Pathol. 170:1178 1191. 11. Gibran, N.S., et al. 2002. Diminished neuropeptide levels contribute to the impaired cutaneous healing response associated with diabetes mellitus. J. Surg. Res. 108:122 128. 12. Lobmann, R., et al. 2002. Expression of matrixmetalloproteinases and their inhibitors in the wounds of diabetic and non-diabetic patients. Diabetologia. 45:1011 1016. 13. Stojadinovic, O., et al. 2005. Molecular pathogenesis of chronic wounds: the role of beta-catenin and c-myc in the inhibition of epithelialization and wound healing. Am. J. Pathol. 167:59 69. 14. Brem, H., et al. 2007. Molecular markers in patients with chronic wounds to guide surgical debridement. Mol. Med. In press. 15. Saap, L.J., and Falanga, V. 2002. Debridement performance index and its correlation with complete closure of diabetic foot ulcers. Wound Repair Regen. 10:354 359. 16. Steed, D.L., Donohoe, D., Webster, M.W., and Lindsley, L. 1996. Effect of extensive debridement and treatment on the healing of diabetic foot ulcers. Diabetic Ulcer Study Group. J. Am. Coll. Surg. 183:61 64. 17. Steed, D.L. 2004. Debridement. Am. J. Surg. 187:71S 74S. 18. Gallagher, K.A., et al. 2007. Diabetic impairments in NO-mediated endothelial progenitor cell mobilization and homing are reversed by hyperoxia and SDF-1α. J. Clin. Invest. 117:1249 1259. doi:10.1172/ JCI29710. 19. Potter, C.F., et al. 1999. Effects of hyperoxia on nitric oxide synthase expression, nitric oxide activity, and lung injury in rat pups. Pediatr. Res. 45:8 13. 20. Kranke, P., Bennett, M., Roeckl-Wiedmann, I., and Debus, S. 2004. Hyperbaric oxygen therapy for chronic wounds. The Cochrane Library. http://www.thecochranelibrary.com. 21. Boulton, A.J., Vileikyte, L., Ragnarson-Tennvall, G., and Apelqvist, J. 2005. The global burden of diabetic foot disease. Lancet. 366:1719 1724. 22. Boulton, A.J., Kirsner, R.S., and Vileikyte, L. 2004. Clinical practice. Neuropathic diabetic foot ulcers. N. Engl. J. Med. 351:48 55. 23. Smiell, J.M. 1998. Clinical safety of becaplermin (rhpdgf-bb) gel. Becaplermin Studies Group. Am. J. Surg. 176:68S 73S. 24. Marston, W.A., Hanft, J., Norwood, P., and Pollak, R. 2003. The efficacy and safety of Dermagraft in improving the healing of chronic diabetic foot ulcers: results of a prospective randomized trial. Diabetes Care. 26:1701 1705. 25. Brem, H., Balledux, J., Bloom, T., Kerstein, M.D., and Hollier, L. 2000. Healing of diabetic foot ulcers and pressure ulcers with human skin equivalent: a new paradigm in wound healing. Arch. Surg. 135:627 634. 1222 The Journal of Clinical Investigation http://www.jci.org Volume 117 Number 5 May 2007