Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters










Publication year range
1.
Exp Dermatol ; 32(4): 379-391, 2023 04.
Article in English | MEDLINE | ID: mdl-36398464

ABSTRACT

Chemokines are a group of small proteins that induce chemoattraction and inflammation and contribute to the differentiation and homeostasis of various cell types. Here we explored the role of chemokines, extracellular matrix production, and myofibroblast differentiation in self-assembled skin equivalents (SASE), a three-dimensional (3D) skin-equivalent tissue model. We found that the expression of three chemokines, C-C motif chemokine ligand (CCL) 20, C-X-C motif chemokine ligand (CXCL) 5, and CXCL8, increased with differentiation to myofibroblasts. Addition of recombinant CCL20 to human skin fibroblast induced collagen Type I alpha 2 gene expression, but did not affect the expression of alpha smooth muscle actin expression. Conversely, siRNA gene knockdown of CCL20 effectively inhibited the expression of collagen Type I gene and protein. Furthermore, when the CCL20 gene in fibroblasts was knocked down in SASE, collagen Type I synthesis and stromal thickness were decreased. Taken together, these results have indicated the utility of SASE in understanding how cytokines such as CCL20 positively regulate extracellular matrix proteins such as collagen Type I production during myofibroblast differentiation in 3D tissues that mimic human skin.


Subject(s)
Chemokines, CC , Collagen Type I , Humans , Chemokines, CC/metabolism , Collagen Type I/genetics , Collagen Type I/metabolism , Ligands , Skin/metabolism , Fibroblasts/metabolism , Myofibroblasts/metabolism , Cell Differentiation/physiology , Chemokine CCL20/genetics , Chemokine CCL20/metabolism , Cells, Cultured , Actins/metabolism
2.
Rheumatology (Oxford) ; 62(SI): SI114-SI124, 2023 02 06.
Article in English | MEDLINE | ID: mdl-35946522

ABSTRACT

OBJECTIVES: Prior work demonstrates that co-cultured macrophages and fibroblasts from patients with SSc engage in reciprocal activation. However, the mechanism by which these cell types communicate and contribute to fibrosis and inflammation in SSc is unknown. METHODS: Fibroblasts were isolated from skin biopsies obtained from 7 SSc patients or 6 healthy age and gender-matched control subjects following written informed consent. Human donor-derived macrophages were cultured with exosomes isolated from control or SSc fibroblasts for an additional 48 h. Macrophages were immunophenotyped using flow cytometry, qRT-PCR and multiplex. For mutual activation studies, exosome-activated macrophages were co-cultured with SSc or healthy fibroblasts using Transwells. RESULTS: Macrophages activated with dermal fibroblast-derived exosomes from SSc patients upregulated surface expression of CD163, CD206, MHC Class II and CD16 and secreted increased levels of IL-6, IL-10, IL-12p40 and TNF compared with macrophages incubated with healthy control fibroblasts (n = 7, P < 0.05). Exosome-stimulated macrophages and SSc fibroblasts engaged in reciprocal activation, as production of collagen and fibronectin was significantly increased in SSc fibroblasts receiving signals from SSc exosome-stimulated macrophages (n = 7, P < 0.05). CONCLUSION: In this work, we demonstrate for the first time that human SSc dermal fibroblasts mediate macrophage activation through exosomes. Our findings suggest that macrophages and fibroblasts engage in cross-talk in SSc skin, resulting in mutual activation, inflammation, and extracellular matrix (ECM) deposition. Collectively, these studies implicate macrophages and fibroblasts as cooperative mediators of fibrosis in SSc and suggest therapeutic targeting of both cell types may provide maximal benefit in ameliorating disease in SSc patients.


Subject(s)
Exosomes , Scleroderma, Systemic , Humans , Macrophage Activation , Scleroderma, Systemic/pathology , Skin/pathology , Fibrosis , Cells, Cultured , Inflammation/metabolism , Fibroblasts/metabolism
3.
Methods Mol Biol ; 2454: 273-283, 2022.
Article in English | MEDLINE | ID: mdl-33755908

ABSTRACT

Tissue engineering solutions have been widely explored for enhanced healing of skin wounds. Diabetic foot ulcers (DFU) are particularly challenging wounds to heal for a variety of reasons, including aberrant ECM, dysregulation of vascularization, and persistent inflammation. Tissue engineering approaches, such as porous collagen-based scaffolds, have shown promise in replacing the current treatments of surgical debridement and topical treatments. Collagen-glycosaminoglycan scaffolds, which are FDA approved for diabetic foot ulcers, can benefit from further functionalization by incorporation of additional signaling factors or extracellular matrix molecules. One option for this is to incorporate matrix from a rejuvenated cell source, as wounds in younger patients heal more quickly. Induced pluripotent stem cells (iPS) are generated from somatic cells and share many functional similarities with embryonic stem cells (ES), while avoiding the ethical concerns. Fibroblasts differentiated from iPS cells have been shown to enrich their ECM with glycosaminoglycan (GAGs), collagen Type III and fibronectin, to have an increased ECM production, and to be pro-angiogenic. Here we describe a technique to grow matrix from post-iPS fibroblasts, and to develop a scaffold from this matrix, in combination with collagen, with the goal of enhancing wound healing. By activating scaffolds with extracellular matrix (ECM) from fibroblasts derived from an iPS source (post-iPSF), the scaffolds are enriched with beneficial elements like GAGs, collagen type III, fibronectin, and VEGF. We believe these scaffolds can enhance skin regeneration and that the techniques can be modified for other tissue engineering applications.


Subject(s)
Diabetic Foot , Induced Pluripotent Stem Cells , Collagen/metabolism , Collagen Type III/metabolism , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Fibronectins/metabolism , Glycosaminoglycans/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Tissue Engineering/methods , Tissue Scaffolds
4.
Exp Dermatol ; 30(8): 1065-1072, 2021 08.
Article in English | MEDLINE | ID: mdl-34114688

ABSTRACT

Diabetic foot ulcers (DFUs), a prevalent complication of diabetes, constitute a major medical challenge with a critical need for development of cell-based therapies. We previously generated induced pluripotent stem cells (iPSCs) from dermal fibroblasts derived from the DFU patients, location-matched skin of diabetic patients and normal healthy donors and re-differentiated them into fibroblasts. To assess the epigenetic microRNA (miR) regulated changes triggered by cellular reprogramming, we performed miRs expression profiling. We found let-7c, miR-26b-5p, -29c-3p, -148a-3p, -196a-5p, -199b-5p and -374a-5p suppressed in iPSC-derived fibroblasts in vitro and in 3D dermis-like self-assembly tissue, whereas their corresponding targets involved in cellular migration were upregulated. Moreover, targets involved in organization of extracellular matrix were induced after fibroblast reprogramming. PLAT gene, the crucial fibrinolysis factor, was upregulated in iPSC-derived fibroblasts and was confirmed as a direct target of miR-196a-5p. miR-197-3p and miR-331-3p were found upregulated specifically in iPSC-derived diabetic fibroblasts, while their targets CAV1 and CDKN3 were suppressed. CAV1, an important negative regulator of wound healing, was confirmed as a direct miR-197-3p target. Together, our findings demonstrate that iPSC reprogramming is an effective approach for erasing the diabetic non-healing miR-mediated epigenetic signature and promoting a pro-healing cellular phenotype.


Subject(s)
Cellular Reprogramming/genetics , Diabetic Foot/genetics , Epigenesis, Genetic , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/metabolism , MicroRNAs/genetics , Wound Healing/genetics , Cell Movement/genetics , Humans , Up-Regulation
5.
Immunity ; 54(7): 1447-1462.e5, 2021 07 13.
Article in English | MEDLINE | ID: mdl-33979579

ABSTRACT

Two sets of innate immune proteins detect pathogens. Pattern recognition receptors (PRRs) bind microbial products, whereas guard proteins detect virulence factor activities by the surveillance of homeostatic processes within cells. While PRRs are well known for their roles in many types of infections, the role of guard proteins in most infectious contexts remains less understood. Here, we demonstrated that inhibition of protein synthesis during viral infection is sensed as a virulence strategy and initiates pyroptosis in human keratinocytes. We identified the BCL-2 family members MCL-1 and BCL-xL as sensors of translation shutdown. Virus- or chemical-induced translation inhibition resulted in MCL-1 depletion and inactivation of BCL-xL, leading to mitochondrial damage, caspase-3-dependent cleavage of gasdermin E, and release of interleukin-1α (IL-1α). Blocking this pathway enhanced virus replication in an organoid model of human skin. Thus, MCL-1 and BCL-xL can act as guard proteins within barrier epithelia and contribute to antiviral defense.


Subject(s)
Apoptosis/immunology , Epithelial Cells/immunology , Proto-Oncogene Proteins c-bcl-2/immunology , Pyroptosis/immunology , Receptors, Estrogen/immunology , Viruses/immunology , Animals , Apoptosis Regulatory Proteins/immunology , Caspase 3/immunology , Cell Line , Chlorocebus aethiops , HEK293 Cells , Humans , Interleukin-1alpha/immunology , Mice , Mitochondria/immunology , NIH 3T3 Cells , Vero Cells , Virus Replication/immunology , bcl-X Protein/immunology
6.
J Biomed Mater Res A ; 109(10): 1803-1811, 2021 10.
Article in English | MEDLINE | ID: mdl-33755305

ABSTRACT

Extracellular matrix is a key component of all tissues, including skin and it plays a crucial role in the complex events of wound healing. These events are impaired in chronic wounds, with chronic inflammation and infection often present in these non-healing wounds. Many tissue engineering approaches for wound healing provide a scaffold to mimic the native matrix. Fibroblasts derived from iPS cells (iPSF) represent a novel source of matrix rich in pro-regenerative components, which can be used for scaffold fabrication to improve wound healing. However, in vitro production of matrix by cells for scaffold fabrication requires long cell culturing times which increases cost. The aim of this work is to optimize the iPSF matrix production by boosting matrix deposition, without affecting its composition. A good candidate technique to achieve this goal is macromolecular crowding, which is known to promote conversion of procollagen into mature collagen and its accumulation. We tested two molecular crowders, Ficoll and Carrageenan-in combination with ascorbic acid-over a prolonged period of time. Ficoll in combination with ascorbic acid notably increased collagen deposition and matrix dry weight compared to ascorbic acid alone, and did not affect matrix composition as measured by RT-PCR. Interestingly, Carrageenan did not affect collagen quantity, but it significantly increased glycosaminoglycan deposition. Finally, we successfully fabricated scaffolds from harvested matrix and confirmed their ability for cell growth and viability. This work lays the foundation for development of a time and cost effective protocol for novel iPSF ECM production for tissue engineering scaffolds.


Subject(s)
Extracellular Matrix/metabolism , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/cytology , Tissue Scaffolds/chemistry , Wound Healing , Animals , Cattle , Collagen/metabolism , Glycosaminoglycans/metabolism , Induced Pluripotent Stem Cells/metabolism , Macromolecular Substances/metabolism
7.
J Mech Behav Biomed Mater ; 114: 104174, 2021 02.
Article in English | MEDLINE | ID: mdl-33191173

ABSTRACT

Tissue engineering products, like collagen-glycosaminoglycan scaffolds, have been successfully applied to chondrogenic defects. Inducible Pluripotent Stem cell (iPS) technology allows reprograming of somatic cells into an embryonic-like state, allowing for redifferentiation. We postulated that a fibroblast cell line (BJ cells - 'pre-iPSF') cycled through iPS reprogramming and redifferentiated into fibroblasts (post-iPSF) could lubricate collagen-glycosaminoglycan scaffolds; fibroblasts are known to produce lubricating molecules (e.g., lubricin) in the synovium. Herein, we quantified the coefficient of friction (CoF) of collagen-glycosaminoglycan scaffolds seeded with post-iPSF; tested whether cell-free scaffolds made of post-iPSF derived extracellular matrix had reduced friction vs. pre-iPSF; and assessed lubricin quantity as a possible protein responsible for lubrication. Post-iPSF seeded CG had 6- to 10-fold lower CoF versus pre-iPSF. Scaffolds consisting of a collagen and pre-/post-iPSF extracellular matrix blend outperformed these cell-seeded scaffolds (~5-fold lower CoF), yielding excellent CoF values close to synovial fluid. Staining revealed an increased presence of lubricin within post-iPSF scaffolds (confirmed by western blotting) and on the surface of iPSF-seeded collagen-glycosaminoglycan scaffolds. Interestingly, when primary cells from patient biopsy-derived fibroblasts were used, iPS reprogramming did not further reduce the already low CoF of these cells and no lubricin expression was found. We conclude that iPS reprogramming activates lubricating properties in iPS-derived cells in a source cell-specific manner. Additionally, lubricin appears to play a lubricating role, yet other proteins also contribute to lubrication. This work constitutes an important step for understanding post-iPSF lubrication of scaffolds and its potential for cartilage tissue engineering.


Subject(s)
Chondrogenesis , Collagen , Pluripotent Stem Cells , Tissue Scaffolds , Cartilage , Fibroblasts , Humans
8.
J Tissue Eng Regen Med ; 14(8): 1019-1027, 2020 08.
Article in English | MEDLINE | ID: mdl-32483913

ABSTRACT

Three-dimensional (3D) tissue models of human skin are being developed to better understand disease phenotypes and to screen new drugs for potential therapies. Several factors will increase the value of these in vitro 3D skin tissues for these purposes. These include the need for human-derived extracellular matrix (ECM), higher throughput tissue formats, and greater cellular complexity. Here, we present an approach for the fabrication of 3D skin-like tissues as a platform that addresses these three considerations. We demonstrate that human adult and neonatal fibroblasts deposit an endogenous ECM de novo that serves as an effective stroma for full epithelial tissue development and differentiation. We have miniaturized these tissues to a 24-well format to adapt them for eventual higher throughput drug screening. We have shown that monocytes from the peripheral blood can be incorporated into this model as macrophages to increase tissue complexity. This humanized skin-like tissue decreases dependency on animal-derived ECM while increasing cellular complexity that can enable screening inflammatory responses in tissue models of human skin.


Subject(s)
Extracellular Matrix , Fibroblasts/metabolism , Models, Biological , Monocytes/metabolism , Skin , Adult , Cell Culture Techniques , Cells, Cultured , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Humans , Infant, Newborn , Male , Skin/chemistry , Skin/metabolism
9.
Adv Healthc Mater ; 9(16): e2000307, 2020 08.
Article in English | MEDLINE | ID: mdl-32597577

ABSTRACT

Diabetic foot ulcers (DFUs) are chronic wounds, with 20% of cases resulting in amputation, despite intervention. A recently approved tissue engineering product-a cell-free collagen-glycosaminoglycan (GAG) scaffold-demonstrates 50% success, motivating its functionalization with extracellular matrix (ECM). Induced pluripotent stem cell (iPSC) technology reprograms somatic cells into an embryonic-like state. Recent findings describe how iPSCs-derived fibroblasts ("post-iPSF") are proangiogenic, produce more ECM than their somatic precursors ("pre-iPSF"), and their ECM has characteristics of foetal ECM (a wound regeneration advantage, as fetuses heal scar-free). ECM production is 45% higher from post-iPSF and has favorable components (e.g., Collagen I and III, and fibronectin). Herein, a freeze-dried scaffold using ECM grown by post-iPSF cells (Post-iPSF Coll) is developed and tested vs precursors ECM-activated scaffolds (Pre-iPSF Coll). When seeded with healthy or DFU fibroblasts, both ECM-derived scaffolds have more diverse ECM and more robust immune responses to cues. Post-iPSF-Coll had higher GAG, higher cell content, higher Vascular Endothelial Growth Factor (VEGF) in DFUs, and higher Interleukin-1-receptor antagonist (IL-1ra) vs. pre-iPSF Coll. This work constitutes the first step in exploiting ECM from iPSF for tissue engineering scaffolds.


Subject(s)
Diabetes Mellitus , Induced Pluripotent Stem Cells , Extracellular Matrix , Fibroblasts , Humans , Tissue Engineering , Tissue Scaffolds , Vascular Endothelial Growth Factor A , Wound Healing
10.
FASEB J ; 33(1): 1262-1277, 2019 01.
Article in English | MEDLINE | ID: mdl-30088952

ABSTRACT

Diabetic foot ulcers (DFUs) are a major complication of diabetes, and there is a critical need to develop novel cell- and tissue-based therapies to treat these chronic wounds. Induced pluripotent stem cells (iPSCs) offer a replenishing source of allogeneic and autologous cell types that may be beneficial to improve DFU wound-healing outcomes. However, the biologic potential of iPSC-derived cells to treat DFUs has not, to our knowledge, been investigated. Toward that goal, we have performed detailed characterization of iPSC-derived fibroblasts from both diabetic and nondiabetic patients. Significantly, gene array and functional analyses reveal that iPSC-derived fibroblasts from both patients with and those without diabetes are more similar to each other than were the primary cells from which they were derived. iPSC-derived fibroblasts showed improved migratory properties in 2-dimensional culture. iPSC-derived fibroblasts from DFUs displayed a unique biochemical composition and morphology when grown as 3-dimensional (3D), self-assembled extracellular matrix tissues, which were distinct from tissues fabricated using the parental DFU fibroblasts from which they were reprogrammed. In vivo transplantation of 3D tissues with iPSC-derived fibroblasts showed they persisted in the wound and facilitated diabetic wound closure compared with primary DFU fibroblasts. Taken together, our findings support the potential application of these iPSC-derived fibroblasts and 3D tissues to improve wound healing.-Kashpur, O., Smith, A., Gerami-Naini, B., Maione, A. G., Calabrese, R., Tellechea, A., Theocharidis, G., Liang, L., Pastar, I., Tomic-Canic, M., Mooney, D., Veves, A., Garlick, J. A. Differentiation of diabetic foot ulcer-derived induced pluripotent stem cells reveals distinct cellular and tissue phenotypes.


Subject(s)
Cell Differentiation , Diabetic Foot/pathology , Induced Pluripotent Stem Cells/cytology , Animals , Cell Line , Cell Movement , Cell Proliferation , Diabetic Foot/metabolism , Extracellular Matrix Proteins/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Glycosaminoglycans/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, SCID , Phenotype , Wound Healing/genetics
11.
Mol Cell ; 71(5): 825-840.e6, 2018 09 06.
Article in English | MEDLINE | ID: mdl-30100266

ABSTRACT

Virulent pathogens often cause the release of host-derived damage-associated molecular patterns (DAMPs) from infected cells. During encounters with immune-evasive viruses that block inflammatory gene expression, preformed DAMPs provide backup inflammatory signals that ensure protective immunity. Whether DAMPs exhibit additional backup defense activities is unknown. Herein, we report that viral infection of barrier epithelia (keratinocytes) elicits the release of preformed interleukin-1 (IL-1) family cytokines, including the DAMP IL-1α. Mechanistic studies revealed that IL-1 acts on skin fibroblasts to induce an interferon (IFN)-like state that restricts viral replication. We identified a branch in the IL-1 signaling pathway that induces IFN-stimulated gene expression in infected cells and found that IL-1 signaling is necessary to restrict viral replication in human skin explants. These activities are most important to control immune-evasive virus replication in fibroblasts and other barrier cell types. These findings highlight IL-1 as an important backup antiviral system to ensure barrier defense.


Subject(s)
Immune Evasion/immunology , Interleukin-1/immunology , Signal Transduction/immunology , Virus Replication/immunology , Animals , Cell Line , Chlorocebus aethiops , Female , Fibroblasts/immunology , Fibroblasts/virology , Gene Expression/immunology , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred C57BL , Skin/immunology , Skin/virology , Vero Cells
12.
Wound Repair Regen ; 24(6): 943-953, 2016 11.
Article in English | MEDLINE | ID: mdl-27607190

ABSTRACT

Diabetic foot ulcers (DFUs) are one of the major complications of diabetes. Its molecular pathology remains poorly understood, impeding the development of effective treatments. Although it has been established that multiple cell types, including fibroblasts, keratinocytes, macrophages, and endothelial cells, all contribute to inhibition of healing, less is known regarding contributions of individual cell type. Thus, we generated primary fibroblasts from nonhealing DFUs and evaluated their cellular and molecular properties in comparison to nondiabetic foot fibroblasts (NFFs). Specifically, we analyzed both micro-RNA and mRNA expression profiles of primary DFU fibroblasts. Paired genomic analyses identified a total of 331 reciprocal miRNA-mRNA pairs including 21 miRNAs (FC > 2.0) along with 239 predicted target genes (FC > 1.5) that are significantly and differentially expressed. Of these, we focused on three miRNAs (miR-21-5p, miR-34a-5p, miR-145-5p) that were induced in DFU fibroblasts as most differentially regulated. The involvement of these microRNAs in wound healing was investigated by testing the expression of their downstream targets as well as by quantifying cellular behaviors in prospectively collected and generated cell lines from 15 patients (seven DFUF and eight NFF samples). We found large number of downstream targets of miR-21-5p, miR-34a-5p, miR-145-5p to be coordinately regulated in mRNA profiles, which was confirmed by quantitative real-time PCR. Pathway analysis on paired miRNA-mRNA profiles predicted inhibition of cell movement and cell proliferation, as well as activation of cell differentiation and senescence in DFU fibroblasts, which was confirmed by cellular assays. We concluded that induction of miR-21-5p, miR-34a-5p, miR-145-5p in DFU dermal fibroblasts plays an important role in impairing multiple cellular functions, thus contributing to overall inhibition of healing in DFUs.


Subject(s)
Diabetic Foot/genetics , Diabetic Foot/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Profiling , MicroRNAs/metabolism , RNA, Messenger/metabolism , Wound Healing , Blotting, Western , Cell Differentiation , Cellular Senescence , Gene Expression Regulation , Humans , Immunohistochemistry , Microarray Analysis , Signal Transduction
13.
Cell Reprogram ; 18(4): 214-23, 2016 08.
Article in English | MEDLINE | ID: mdl-27328415

ABSTRACT

Diabetic foot ulcers (DFUs) are nonhealing chronic wounds that are a serious complication of diabetes. Since induced pluripotent stem cells (iPSCs) may offer a potent source of autologous cells to heal these wounds, we studied if repair-deficient fibroblasts, derived from DFU patients and age- and site-matched control fibroblasts, could be reprogrammed to iPSCs. To establish this, we used Sendai virus to successfully reprogram six primary fibroblast cell lines derived from ulcerated skin of two DFU patients (DFU8, DFU25), nonulcerated foot skin from two diabetic patients (DFF24, DFF9), and healthy foot skin from two nondiabetic patients (NFF12, NFF14). We confirmed reprogramming to a pluripotent state through three independent criteria: immunofluorescent staining for SSEA-4 and TRA-1-81, formation of embryoid bodies with differentiation potential to all three embryonic germ layers in vitro, and formation of teratomas in vivo. All iPSC lines showed normal karyotypes and typical, nonmethylated CpG sites for OCT4 and NANOG. iPSCs derived from DFUs were similar to those derived from site-matched nonulcerated skin from both diabetic and nondiabetic patients. These results have established for the first time that multiple, DFU-derived fibroblast cell lines can be reprogrammed with efficiencies similar to control fibroblasts, thus demonstrating their utility for future regenerative therapy of DFUs.


Subject(s)
Cell Differentiation , Cellular Reprogramming , Diabetic Foot/pathology , Fibroblasts/cytology , Induced Pluripotent Stem Cells/cytology , Sendai virus/genetics , Teratoma/pathology , Animals , Cells, Cultured , Diabetic Foot/genetics , Fibroblasts/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, SCID , Teratoma/etiology
14.
Wound Repair Regen ; 24(4): 630-43, 2016 07.
Article in English | MEDLINE | ID: mdl-27102877

ABSTRACT

Current chronic wound treatments often fail to promote healing of diabetic foot ulcers (DFU), leading to amputation and increased patient morbidity. A critical mediator of proper wound healing is the production, assembly, and remodeling of the extracellular matrix (ECM) by fibroblasts. However, little is known about how these processes are altered in fibroblasts within the DFU microenvironment. Thus, we investigated the capacity of multiple, primary DFU-derived fibroblast strains to express, produce, and assemble ECM proteins compared to diabetic patient-derived fibroblasts and healthy donor-derived fibroblasts. Gene expression microarray analysis showed differential expression of ECM and ECM-regulatory genes by DFU-derived fibroblasts which translated to functional differences in a 3D in vitro ECM tissue model. DFU-derived fibroblasts produced thin, fibronectin-rich matrices, and responded abnormally when challenged with transforming growth factor-beta, a key regulator of matrix production during healing. These results provide novel evidence that DFU-derived fibroblasts contribute to the defective matrices of DFUs and chronic wound pathogenesis.


Subject(s)
Diabetic Foot/pathology , Diabetic Foot/physiopathology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibroblasts/metabolism , Fibronectins/metabolism , Wound Healing , Collagen Type I/metabolism , Diabetic Foot/metabolism , Extracellular Matrix/chemistry , Extracellular Matrix/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/pathology , Gene Expression Profiling , Humans , Immunohistochemistry , Microarray Analysis , Neovascularization, Physiologic , Transforming Growth Factor beta/pharmacology
15.
PLoS One ; 10(8): e0137133, 2015.
Article in English | MEDLINE | ID: mdl-26318001

ABSTRACT

Diabetes Mellitus (DM) is a chronic, severe disease rapidly increasing in incidence and prevalence and is associated with numerous complications. Patients with DM are at high risk of developing diabetic foot ulcers (DFU) that often lead to lower limb amputations, long term disability, and a shortened lifespan. Despite this, the effects of DM on human foot skin biology are largely unknown. Thus, the focus of this study was to determine whether DM changes foot skin biology predisposing it for healing impairment and development of DFU. Foot skin samples were collected from 20 patients receiving corrective foot surgery and, using a combination of multiple molecular and cellular approaches, we performed comparative analyses of non-ulcerated non-neuropathic diabetic foot skin (DFS) and healthy non-diabetic foot skin (NFS). MicroRNA (miR) profiling of laser captured epidermis and primary dermal fibroblasts from both DFS and NFS samples identified 5 miRs de-regulated in the epidermis of DFS though none reached statistical significance. MiR-31-5p and miR-31-3p were most profoundly induced. Although none were significantly regulated in diabetic fibroblasts, miR-29c-3p showed a trend of up-regulation, which was confirmed by qPCR in a prospective set of 20 skin samples. Gene expression profiling of full thickness biopsies identified 36 de-regulated genes in DFS (>2 fold-change, unadjusted p-value ≤ 0.05). Of this group, three out of seven tested genes were confirmed by qPCR: SERPINB3 was up-regulated whereas OR2A4 and LGR5 were down-regulated in DFS. However no morphological differences in histology, collagen deposition, and number of blood vessels or lymphocytes were found. No difference in proliferative capacity was observed by quantification of Ki67 positive cells in epidermis. These findings suggest DM causes only subtle changes to foot skin. Since morphology, mRNA and miR levels were not affected in a major way, additional factors, such as neuropathy, vascular complications, or duration of DM, may further compromise tissue's healing ability leading to development of DFUs.


Subject(s)
Dermis/pathology , Diabetic Foot/genetics , Diabetic Foot/pathology , Epidermis/pathology , Genomics , MicroRNAs/genetics , Fibroblasts/metabolism , Foot Ulcer/genetics , Foot Ulcer/pathology , Gene Expression Profiling , Humans , Transcription, Genetic
16.
Tissue Eng Part C Methods ; 21(5): 499-508, 2015 May.
Article in English | MEDLINE | ID: mdl-25343343

ABSTRACT

Diabetic foot ulcers (DFU) are a major, debilitating complication of diabetes mellitus. Unfortunately, many DFUs are refractory to existing treatments and frequently lead to amputation. The development of more effective therapies has been hampered by the lack of predictive in vitro methods to investigate the mechanisms underlying impaired healing. To address this need for realistic wound-healing models, we established patient-derived fibroblasts from DFUs and site-matched controls and used them to construct three-dimensional (3D) models of chronic wound healing. Incorporation of DFU-derived fibroblasts into these models accurately recapitulated the following key aspects of chronic ulcers: reduced stimulation of angiogenesis, increased keratinocyte proliferation, decreased re-epithelialization, and impaired extracellular matrix deposition. In addition to reflecting clinical attributes of DFUs, the wound-healing potential of DFU fibroblasts demonstrated in this suite of models correlated with in vivo wound closure in mice. Thus, the reported panel of 3D DFU models provides a more biologically relevant platform for elucidating the cell-cell and cell-matrix-related mechanisms responsible for chronic wound pathogenesis and may improve translation of in vitro findings into efficacious clinical applications.


Subject(s)
Diabetic Foot/physiopathology , Fibroblasts/cytology , Fibroblasts/pathology , Tissue Engineering/methods , Animals , Cell Culture Techniques , Cytokines/metabolism , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Humans , In Vitro Techniques , Keratinocytes/cytology , Male , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic , Wound Healing
17.
Epigenetics ; 9(10): 1339-49, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25437049

ABSTRACT

Diabetic foot ulcers (DFUs) are a serious complication of diabetes. Previous exposure to hyperglycemic conditions accelerates a decline in cellular function through metabolic memory despite normalization of glycemic control. Persistent, hyperglycemia-induced epigenetic patterns are considered a central mechanism that activates metabolic memory; however, this has not been investigated in patient-derived fibroblasts from DFUs. We generated a cohort of patient-derived lines from DFU fibroblasts (DFUF), and site- and age-matched diabetic foot fibroblasts (DFF) and non-diabetic foot fibroblasts (NFF) to investigate global and genome-wide DNA methylation patterns using liquid chromatography/mass spectrometry and the Illumina Infinium HumanMethylation450K array. DFFs and DFUFs demonstrated significantly lower global DNA methylation compared to NFFs (p = 0.03). Hierarchical clustering of differentially methylated probes (DMPs, p = 0.05) showed that DFFs and DFUFs cluster together and separately from NFFs. Twenty-five percent of the same probes were identified as DMPs when individually comparing DFF and DFUF to NFF. Functional annotation identified enrichment of DMPs associated with genes critical to wound repair, including angiogenesis (p = 0.07) and extracellular matrix assembly (p = 0.035). Identification of sustained DNA methylation patterns in patient-derived fibroblasts after prolonged passage in normoglycemic conditions demonstrates persistent metabolic memory. These findings suggest that epigenetic-related metabolic memory may also underlie differences in wound healing phenotypes and can potentially identify therapeutic targets.


Subject(s)
DNA Methylation , Diabetic Foot/genetics , Epigenesis, Genetic , Fibroblasts/metabolism , Adult , Aged , Cell Line , Computational Biology , Diabetic Foot/metabolism , Female , Humans , Male , Middle Aged
18.
Adv Wound Care (New Rochelle) ; 3(12): 742-750, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25493208

ABSTRACT

Significance: Human-induced pluripotent stem cells (iPSC) can be differentiated into patient-specific cells with a wide spectrum of cellular phenotypes and offer an alternative source of autologous cells for therapeutic use. Recent studies have shown that iPSC-derived fibroblasts display enhanced cellular functions suggesting that iPSC may eventually become an important source of stem cells for regenerative therapies. Recent Advances: The discovery of approaches to reprogram somatic cells into pluripotent cells opens exciting avenues for their use in personalized, regenerative therapies. The controlled differentiation of functional cell types from iPSC provides a replenishing source of fibroblasts. There is intriguing evidence that iPSC reprogramming and subsequent differentiation to fibroblast lineages may improve cellular functional properties. Augmenting the biological potency of iPSC-derived fibroblasts may enable the development of novel, personalized stem cell therapies to treat oral disease. Critical Issues: Numerous questions need to be addressed before iPSC-derived cells can be used as a practical oral therapy. This will include understanding why iPSC-derived cells are predisposed towards differentiation pathways along lineages related to their cell of origin, screening iPSC-derived cells to ensure their safety and phenotypic stability and developing engineered, three-dimensional tissue models to optimize their function and efficacy for future therapeutic transplantation. Future Directions: Future research will need to address how to develop efficient methods to deliver and integrate iPSC-derived fibroblasts into the oral mucosa. This will require an improved understanding of how to harness their biological potency for regenerative therapies that are specifically targeted to the oral mucosa.

19.
J Photochem Photobiol B ; 126: 17-25, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-23892186

ABSTRACT

UV exposure causes oxidative stress, inflammation, erythema, and skin cancer. α-Tocopherol (AT) and polyphenols (AP) present in almonds may serve as photoprotectants. Our objectives were to assess the feasibility of using a 3D human skin equivalent (HSE) in photoprotectant research and to determine photoprotection of AT and AP against UVA radiation. AT or AP was applied to medium (25 and 5µmol/L, respectively) or topically (1mg/cm(2) and 14µg/cm(2)), followed by UVA. Photodamage assessed 96h post UVA included HSE morphology, keratinocyte proliferation, apoptosis, and differentiation. UVA induced disorganization of basal layer, alteration of epidermal development, and fibroblast loss which were alleviated by all nutrient pretreatments. UVA significantly decreased keratinocyte proliferation compared to controls, and all pretreatments tended to negate the reduction though only the medium AT effect was statistically significant (p⩽0.05). UVA led to a significant 16-fold increase in apoptosis of fibroblasts compared to the control which was alleviated by topical AP pretreatment and completely negated by topical AT (p⩽0.05). In conclusion, we validated the feasibility of using HSE in evaluation of photoprotectants and found that AT and AP, applied to medium or topically, provided some degree of photoprotection against UVA.


Subject(s)
Polyphenols/pharmacology , Prunus/chemistry , Radiation-Protective Agents/pharmacology , Skin/drug effects , Skin/radiation effects , alpha-Tocopherol/pharmacology , Absorption , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Cattle , Cell Differentiation/drug effects , Cell Differentiation/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Diet , Humans , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/radiation effects , Pilot Projects , Polyphenols/metabolism , Radiation-Protective Agents/metabolism , Skin/cytology , Ultraviolet Rays/adverse effects , alpha-Tocopherol/metabolism
20.
Mol Aspects Med ; 34(4): 841-8, 2013.
Article in English | MEDLINE | ID: mdl-22982217

ABSTRACT

The controlled differentiation of induced pluripotent stem cells (iPSC) towards clinically-relevant cell types has benefitted from epigenetic profiling of lineage-specific markers to confirm the phenotype of iPSC-derived cells. Mapping epigenetic marks throughout the genome has identified unique changes which occur in the DNA methylation profile of cells as they differentiate to specific cell types. Beyond characterizing the development of cells derived from pluripotent stem cells, the process of reprogramming cells to iPSC resets lineage-specific DNA methylation marks established during differentiation to specific somatic cell types. This property of reprogramming has potential utility in reverting aberrant epigenetic alterations in nuclear organization that are linked to disease progression. Since DNA methylation marks are reset following reprogramming, and contribute to restarting developmental programs, it is possible that DNA methylation marks associated with the disease state may also be erased in these cells. The subsequent differentiation of such cells could result in cell progeny that will function effectively as therapeutically-competent cell types for use in regenerative medicine. This suggests that through reprogramming it may be possible to directly modify the epigenetic memory of diseased cells and help to normalize their cellular phenotype, while also broadening our understanding of disease pathogenesis.


Subject(s)
Cell Differentiation/genetics , Epigenesis, Genetic , Induced Pluripotent Stem Cells/physiology , Animals , Cellular Reprogramming , DNA Methylation , Embryonic Stem Cells/physiology , Humans , Regenerative Medicine , Stem Cell Transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...