Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Exp Dermatol ; 31(5): 715-724, 2022 05.
Article in English | MEDLINE | ID: mdl-34816490

ABSTRACT

Lack of matrix deposition is one of the main factors that complicates the healing process of wounds. The aim of this study was to test the efficacy and safety of a liquid dermal scaffold, referred to as MeshFill (MF) that can fill the complex network of tunnels and cavities which are usually found in chronic wounds and hence improve the healing process. We evaluated in vitro and in vivo properties of a novel liquid dermal scaffold in a delayed murine full-thickness wound model. We also compared this scaffold with two commercially available granular collagen-based products (GCBP). Liquid dermal scaffold accelerated wound closure significantly compared with no-treated control and collagen-based injectable composites in a delayed splinted wound model. When we compared cellular composition and count between MF, no treatment and GCBP at the histology level, it was found that MF was the most analogous and consistent with the normal anatomy of the skin. These findings were matched with the clinical outcome observation. The flowable in situ forming scaffold is liquid at cold temperature and gels after application to the wound site. Therefore, it would conform to the topography of the wound when liquid and provides adequate tensile strength when solidified. This patient-ready gelling dermal scaffold also contains the nutritional ingredients and therefore supports cell growth. Applying an injectable liquid scaffold that can fill wound gaps and generate a matrix to promote keratinocytes and fibroblasts migration, can result in improvement of the healing process of complex wounds.


Subject(s)
Skin, Artificial , Wound Healing , Animals , Collagen , Disease Models, Animal , Humans , Mice , Skin/injuries
2.
Exp Dermatol ; 31(4): 475-484, 2022 04.
Article in English | MEDLINE | ID: mdl-34932841

ABSTRACT

Tissue repair is a very complex event and involves a continuously orchestrated sequence of signals and responses from platelets, fibroblasts, epithelial, endothelial and immune cells. The details of interaction between these signals, which are mainly growth factors and cytokines, have been widely discussed. However, it is still not clear how activated cells at wound sites lessen their activities after epithelialization is completed. Termination of the wound healing process requires a fine balance between extracellular matrix (ECM) deposition and degradation. Maintaining this balance requires highly accurate epithelial-mesenchymal communication and correct information exchange between keratinocytes and fibroblasts. As it has been reported in the literature, a disruption in epithelialization during the process of wound healing increases the frequency of developing chronic wounds or fibrotic conditions, as seen in a variety of clinical cases. Conversely, the potential stop signal for wound healing should have a regulatory role on both ECM synthesis and degradation to reach a successful wound healing outcome. This review briefly describes the potential roles of growth factors and cytokines in controlling the early phase of wound healing and predominantly explores the role of releasable factors from epithelial-mesenchymal interaction in controlling during and the late stage of the healing process. Emphasis will be given on the crosstalk between keratinocytes and fibroblasts in ECM modulation and the healing outcome following a brief discussion of the wound healing initiation mechanism. In particular, we will review the termination of acute dermal wound healing, which frequently leads to the development of hypertrophic scarring.


Subject(s)
Keratinocytes , Wound Healing , Cell Communication/physiology , Cytokines/metabolism , Fibroblasts/physiology , Keratinocytes/metabolism , Wound Healing/physiology
3.
Adv Wound Care (New Rochelle) ; 8(2): 58-70, 2019 Feb 01.
Article in English | MEDLINE | ID: mdl-31737409

ABSTRACT

Background and Objective: Despite the effectiveness of skin autotransplantation, the high degree of immunogenicity of the skin precludes the use of allografts and systemic immunosuppression is generally inappropriate for isolated skin grafts. Indoleamine 2,3 dioxygenase (IDO) is a potent immunoregulatory factor with allo- and autoimmune suppression and tolerance induction properties. This study examines the potential use of locally expressed IDO to prolong the allogeneic skin graft take in a mouse model. Approach: Syngeneic-fibroblasts were transfected with noncompetent IDO viral vector and the level of Kynurenine (Kyn) in conditioned medium was measured as an index for IDO activity. Either 1 or 3 × 106 IDO-fibroblasts were introduced intra/hypo-dermally to the mouse skin. The expression, localization, and functionality of IDO were then evaluated. The cell-injected areas were harvested and grafted on the back of allogeneic mice. The graft survival, immune-cells infiltration, and interaction with dendritic cells were evaluated. Results: The results showed a significant improvement in allogeneic graft take injected with 1 × 106 IDO-fibroblasts (18.4 ± 3.3 days) compared with control (12.2 ± 1.9 days). This duration increased to 35.4 ± 4.7 days in grafts injected with 3 × 106 IDO-expressing cells. This observation might be due to a significantly lower T cells infiltration within the IDO-grafts. Further, the result of a flow cytometric analysis showed that the expression of PD-L1/PD-L2 on CD11c+/eFluor+ cells in the regional lymph nodes of injected skin areas was significantly higher in IDO groups compared with control. Conclusion: These data suggest that allogeneic skin graft survival outcome can be prolonged significantly by local overexpression of IDO without any systemic effect.

4.
J Burn Care Res ; 40(6): 727-733, 2019 10 16.
Article in English | MEDLINE | ID: mdl-31314104

ABSTRACT

Autologous split thickness skin graft is necessary for the survival of patients with large burns and skin defects. It is not clear how a thin split thickness skin graft becomes remarkably thicker within a few weeks following transplantation. Here, we hypothesized that growth of split thickness graft should be from bottom up probably through conversion of immune cells into collagen producing skin cells. We tested this hypothesis in a preclinical porcine model by grafting split thickness meshed skin (0.508 mm thickness, meshed at 3:1 ratio) on full thickness wounds in pigs. New tissue formation was evaluated on days 10 and 20 postoperation through histological analysis and co-staining for immune cell markers (CD45) and type I collagen. The findings revealed that a split thickness graft grew from bottom up and reached to almost the same level as uninjured skin within 60 days postoperation. The result of immune-staining identified a large number of cells, which co-expressed immune cell marker (CD45) and collagen on day 10 postoperation. Interestingly, as the number of these cells reduced on day 20, most of these cells became positive for collagen production. In another set of experiments, we tested whether immune cells can convert to collagen producing cells in vitro. The results showed that mouse adherent immune cells started to express type 1 procollagen and α-smooth muscle actin when cultured in the presence of fibroblast conditioned media. In conclusion, the early thickening of split thickness graft is likely happening through a major contribution of infiltrated immune cells that convert into mainly collagen producing fibroblasts in large skin injuries.


Subject(s)
Regeneration , Skin Physiological Phenomena , Skin Transplantation , Skin/injuries , Wound Healing/physiology , Actins/metabolism , Animals , Autografts , Cell Culture Techniques , Cell Differentiation , Collagen Type I/metabolism , Fibroblasts/physiology , Leukocyte Common Antigens/metabolism , Leukocytes, Mononuclear/physiology , Mice, Inbred C57BL , Models, Animal , Skin/cytology , Skin/metabolism , Swine , Wounds and Injuries/surgery
5.
J Diabetes Metab Disord ; 18(2): 657-664, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31890690

ABSTRACT

PURPOSE: Childhood overweight/ obesity is one of critical public health concern. It has been suggested that there is a link between breakfast skipping and obesity. However, results are conflicting. The aim of the present study was to summarize the association between breakfast skipping and overweight/obesity in children and adolescent. METHODS: We performed a literature search using Pubmed/Medline, Scopus, Web of Science and EMBASE electronic databases from 2000 through 28 February 2018 without language limitation. Observational studies in which risk measures were reported regarding the link between breakfast skipping and obesity in children and adolescent were included. Studies with at least the score of 5 from Newcastle-Ottawa Scale were considered as low risk of bias. Random effect model was used for data synthesis. RESULTS: Of 3276 publications, finally 16 studies (14 cross-sectional studies, 2 cohort studies) were included for meta-analysis. Based on cross-sectional studies, we found a positive association between breakfast skipping and obesity (Odd ratio (OR) trim & fill: 1.43; 95%CI: 1.32, 1.54), while cohort studies showed no significant link (OR:1.01, 95%CI: 0.93, 1.11; I2: 48%, p = 0.14). Subgroup analysis in cross-sectional studies showed that the association between breakfast skipping and the risk of obesity in boys was OR: 1.64; 95% CI: 1.38, 1.95; I2: 38.3%, p = 0.18, while it was 1.56 (95% CI: 1.38, 1.77, I2: 0.0%, p = 0.49) in girls. CONCLUSION: The risk of obesity in children and adolescents who skipped breakfast was 43% greater than those who ate breakfast regularly in cross-sectional studies, while no significant link was found in cohort studies. However, due to high heterogeneity and limited cohort studies, findings should be interpreted by caution.

6.
Cell Transplant ; 27(6): 994-1004, 2018 06.
Article in English | MEDLINE | ID: mdl-29871523

ABSTRACT

Alopecia areata (AA) is an autoimmune hair loss disease with infiltration of proinflammatory cells into hair follicles. Current therapeutic regimens are unsatisfactory mainly because of the potential for side effects and/or limited efficacy. Here we report that cultured, transduced fibroblasts, which express the immunomodulatory molecule indoleamine 2,3-dioxygenase (IDO), can be applied to prevent hair loss in an experimental AA model. A single intraperitoneal (IP) injection of IDO-expressing primary dermal fibroblasts was given to C3H/HeJ mice at the time of AA induction. While 60-70% of mice that received either control fibroblasts or vehicle injections developed extensive AA, none of the IDO-expressing fibroblast-treated mice showed new hair loss up to 20 weeks post injection. IDO cell therapy significantly reduced infiltration of CD4+ and CD8+ T cells into hair follicles and resulted in decreased expression of TNF-α, IFN-γ and IL-17 in the skin. Skin draining lymph nodes of IDO fibroblast-treated mice were significantly smaller, with more CD4+ CD25+ FoxP3+ regulatory T cells and fewer Th17 cells than those of control fibroblast and vehicle-injected mice. These findings indicate that IP injected IDO-expressing dermal fibroblasts can control inflammation and thereby prevent AA hair loss.


Subject(s)
Alopecia Areata/therapy , Fibroblasts/transplantation , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Alopecia Areata/pathology , Animals , Cell- and Tissue-Based Therapy , Cells, Cultured , Cytokines/analysis , Disease Models, Animal , Fibroblasts/cytology , Fibroblasts/metabolism , Mice, Inbred C3H , Transduction, Genetic
7.
J Burn Care Res ; 39(2): 175-182, 2018 02 20.
Article in English | MEDLINE | ID: mdl-28486306

ABSTRACT

Burn injuries are one of the most common sources of trauma globally that comprise a significant drain on long-term personal and healthcare cost. Large surface area burn wounds are difficult to manage and may result in significant physiologic and psychologic sequelae. The goal of burn wound healing research is to fully repair and restore skin's original structure and functionality while minimizing problems such as hypertrophic scarring and contracture. One of the ways this can be achieved is through augmentation of the skin's natural healing process using the regenerative capability of stem cells. In this review, the authors highlight some recent developments in treatment of burn wounds employing stem cells. We compare and contrast the benefits and drawbacks to various sources of stem cells and techniques of delivery into damaged tissues that have been the focus of established and ongoing research, and avenues of exploration this burgeoning arena offers for the future.


Subject(s)
Burns/therapy , Stem Cell Transplantation , Stem Cells/physiology , Humans
8.
Clin Immunol ; 174: 1-9, 2017 01.
Article in English | MEDLINE | ID: mdl-27989895

ABSTRACT

Indoleamine 2,3-dioxygenase (IDO) is an immunosuppressive enzyme with tolerogenic effects on different immune cells. Our group has previously shown that co-transplantation of IDO-expressing fibroblasts with donor tissues can delay immune rejection by inducing local immunosuppression. In this study, we have employed a systemic approach to improve allograft survival without using any immunosuppressive medication. To achieve this, 10 million lentiviral transduced IDO-expressing donor derived fibroblasts were injected into the peritoneal cavity of allograft recipients. We showed that IDO-fibroblast therapy increases the survival of both islets and skin allografts and decreases the infiltration of immune cells in subcutaneous transplanted skins. Indirect pathway of allo-reactive T cell activation was suppressed more than the direct pathway. Injected IDO-fibroblasts were found in peritoneal cavity and mesenteric lymph nodes of the recipient mice. In conclusion, IDO-expressing fibroblast therapy proved to be a novel approach in improving the allogeneic graft survival.


Subject(s)
Fibroblasts/transplantation , Graft Survival , Indoleamine-Pyrrole 2,3,-Dioxygenase , Animals , Blood Glucose/analysis , Diabetes Mellitus, Experimental/blood , Female , Injections, Intraperitoneal , Islets of Langerhans Transplantation , Male , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Skin/cytology , Skin/immunology , T-Lymphocytes, Regulatory/immunology , Transplantation, Homologous
9.
PLoS One ; 11(1): e0146970, 2016.
Article in English | MEDLINE | ID: mdl-26765526

ABSTRACT

Type 1 diabetes (T1D) results from autoimmune destruction of insulin producing ß cells of the pancreatic islets. Curbing autoimmunity at the initiation of T1D can result in recovery of residual ß cells and consequently remission of diabetes. Here we report a cell-based therapy for autoimmune diabetes in non-obese diabetic (NOD) mice using dermal fibroblasts. This was achieved by a single injection of fibroblasts, expressing the immunoregulatory molecule indoleamine 2,3 dioxygenase (IDO), into peritoneal cavity of NOD mice shortly after the onset of overt hyperglycemia. Mice were then monitored for reversal of hyperglycemia and changes in inflammatory/regulatory T cell profiles. Blood glucose levels dropped into the normal range in 82% of NOD mice after receiving IDO-expressing fibroblasts while all control mice remained diabetic. We found significantly reduced islet inflammation, increased regulatory T cells, and decreased T helper 17 cells and ß cell specific autoreactive CD8+ T cells following IDO cell therapy. We further showed that some of intraperitoneal injected fibroblasts migrated to local lymph nodes and expressed co-inhibitory molecules. These findings suggest that IDO fibroblasts therapy can reinstate self-tolerance and alleviate ß cell autoreactivity in NOD mice, resulting in remission of autoimmune diabetes.


Subject(s)
Cell- and Tissue-Based Therapy , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Fibroblasts/metabolism , Animals , Autoimmunity/genetics , Autoimmunity/immunology , Cell Movement/genetics , Cell Movement/immunology , Cell- and Tissue-Based Therapy/methods , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , Gene Expression , Hyperglycemia/genetics , Hyperglycemia/metabolism , Hyperglycemia/therapy , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans/immunology , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphocyte Count , Mice , Mice, Inbred NOD , Receptors, CCR7/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
10.
J Cell Physiol ; 231(9): 1964-73, 2016 09.
Article in English | MEDLINE | ID: mdl-26743772

ABSTRACT

Indoleamine 2,3-dioxygenase (IDO) induces immunological tolerance in physiological and pathological conditions. Therefore, we used dermal fibroblasts with stable IDO expression as a cell therapy to: (i) Investigate the factors determining the efficacy of this cell therapy for autoimmune diabetes in non-obese diabetic (NOD) mice; (ii) Scrutinize the potential immunological mechanisms. Newly diabetic NOD mice were randomly injected with either 10 × 10(6) (10M) or 15 × 10(6) (15M) IDO-expressing dermal fibroblasts. Blood glucose levels (BGLs), body weight, plasma kynurenine levels, insulitis severity, islet beta cell function, autoreactive CD8(+) T cells, Th17 cells and regulatory T cells (Tregs) were then investigated in these mice. IL-1ß and cleaved caspase-3 levels were assessed in islets co-cultured with IDO-expressing fibroblasts. BGLs in 83% mice treated with 15M IDO-expressing fibroblasts recovered to normal up to 120 days. However, only 17% mice treated with 10M IDO-expressing cells were reversed to normoglycemia. A 15M IDO-expressing fibroblasts significantly reduced infiltrated immune cells in islets and recovered the functionality of remaining islet beta cells in NOD mice. Additionally, they successfully inhibited autoreactive CD8(+) T cells and Th17 cells as well as increased Tregs in different organs of NOD mice. Islet beta cells co-cultured with IDO-expressing fibroblasts had reduced IL-1ß levels and cell apoptosis. Both cell number and IDO enzymatic activity contributes to the efficiency of IDO cell therapy. Optimized IDO-expressing fibroblasts successfully reverse the progression of diabetes in NOD mice through induction of Tregs as well as inhibition of beta cell specific autoreactive CD8(+) T cells and Th17 cells. J. Cell. Physiol. 231: 1964-1973, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Fibroblasts/enzymology , Hyperglycemia/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Insulin-Secreting Cells/immunology , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Female , Hyperglycemia/immunology , Insulin-Secreting Cells/enzymology , Male , Mice, Inbred C57BL , Mice, Inbred NOD , T-Lymphocytes, Regulatory/immunology
11.
Immunology ; 148(1): 22-33, 2016 May.
Article in English | MEDLINE | ID: mdl-26789277

ABSTRACT

There is controversy about the immunomodulatory effect of fibroblasts on dendritic cells (DCs). To clarify this issue, in this study, we have evaluated different features of fibroblast-primed DCs including their ability to express co-inhibitory and co-stimulatory molecules, pro-inflammatory and anti-inflammatory cytokines and their ability to induce T-cell proliferation. We also examined migratory capacity of DCs to lymphatic tissues and present fibroblast-derived antigens after encountering fibroblasts. The results of our in vitro study showed that both co-inhibitory (programmed death ligand 1 and ligand 2 and B7H4) and co-stimulatory (CD86) molecules were up-regulated when DCs were co-cultured with fibroblasts. In an animal model, we showed that intra- peritoneal injection (IP) of both syngeneic and allogeneic fibroblasts significantly increased both total DC count and expression level of co-inhibitory and co-stimulatory molecules on DCs. Priming of DCs with syngeneic and allogeneic fibroblasts reduced the proliferation of CD4(+) and CD8(+) T cells. Even activation of fibroblast- primed DCs failed to restore their ability to induce T-cell proliferation. Likewise, priming of DCs with fibroblasts blocked the ability of ovalbumin-pulsed DCs to induce proliferation of ovalbumin-specific CD4(+) T cells. Compared with non-activated DCs, fibroblast-primed DCs had significantly higher expression levels of interleukin-10 and indoleamine 2, 3 dioxygenase. Fibroblast-primed DCs had a significantly reduced interleukin-12 expression level compared with that of activated DCs. After priming with fibroblasts, DCs were able to migrate to lymphatic tissues and present fibroblast-derived antigens (ovalbumin). In conclusion, after priming with fibroblasts, DCs gain tolerogenic features. This finding suggests the potential role of fibroblasts in the maintenance of immune tolerance.


Subject(s)
Dendritic Cells/immunology , Fibroblasts/physiology , Immune Tolerance , Animals , Antigen Presentation , Cells, Cultured , Coculture Techniques , Cytokines/analysis , Female , Lymphocyte Activation , Mice , Mice, Inbred C3H , Mice, Inbred C57BL
12.
Tissue Eng Part A ; 21(5-6): 1085-94, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25412924

ABSTRACT

Wound repair requires a sequential series of biological events that begins with the deposition of a temporary scaffold within which cells can repair the skin. Without a scaffold, repair is essentially impossible. Aberrant wound healing, such as hypertrophic scarring or nonhealing, has a tremendous burden on healthcare and quality of life. Timely wound closure dramatically reduces the risk of infection and scarring. Cellular skin substitutes are opportune to meet this need. Our goal was to create an in-situ forming scaffold that can be easily combined with cells to rapidly form a dermal substitute within the wound bed. In this study, we evaluated the application of a polyvinyl alcohol-collagen-glycosaminoglycan-based biohybrid scaffold system in full-thickness wounds on a rabbit fibrotic ear model. Punch wounds (6 mm) were either untreated or filled with an acellular scaffold, a scaffold containing xenofibroblasts, or a scaffold containing xenofibroblasts expressing indoleamine 2,3-dioxygenase (IDO). Results demonstrated that (1) both acellular and IDO-expressing fibroblast in-situ forming scaffolds significantly reduced scar elevation index (1.24±0.05 and 1.25±0.03; p<0.05) and improved overall healing quality compared with xenofibroblast scaffolds and untreated wounds; (2) IDO-expressing fibroblast scaffolds significantly reduced T-cell infiltration into the scaffold-engrafted area (p<0.05); and (3) both IDO-expressing and acellular in-situ forming scaffolds demonstrated increased vessel-like and nerve-like structures (p<0.05). The results demonstrated that the use of the in-situ forming scaffold, and even more so when delivering IDO-expressing cells, improved healing outcome in full-thickness hypertrophic rabbit ear wounds.


Subject(s)
Skin, Artificial , Wound Healing , Adult , Animals , Biocompatible Materials/pharmacology , CD3 Complex/metabolism , Cell Shape/drug effects , Cell Survival/drug effects , Child , Cicatrix, Hypertrophic , Collagen Type I/metabolism , Disease Models, Animal , Ear/pathology , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibroblasts/drug effects , Fibroblasts/pathology , Humans , Hydrogel, Polyethylene Glycol Dimethacrylate/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Male , Matrix Metalloproteinase 1/metabolism , Neovascularization, Physiologic/drug effects , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Rabbits , Skin/blood supply , Skin/drug effects , Skin/innervation , T-Lymphocytes/cytology , Tissue Scaffolds/chemistry , Wound Healing/drug effects
13.
J Invest Dermatol ; 134(3): 643-650, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23877570

ABSTRACT

We previously demonstrated that the formation of hypertrophic scarring on the wounds of a rabbit ear fibrotic model was significantly reduced by grafting a bilayer skin substitute expressing indoleamine 2,3-dioxygenase (IDO). Here, we hypothesize that the improved healing quality is due to extracellular matrix modulatory effect of IDO-mediated tryptophan metabolites. To test this hypothesis, a series of in vitro and in vivo experiments were conducted and the findings revealed a significant increase in the expression of matrix metalloproteinase 1 (MMP-1) in fibroblasts either transduced with human IDO gene or cultured with conditioned media obtained from IDO-expressing cells. Consistent with this finding, kynurenine (Kyn) treatment markedly increased the levels of MMP-1 and MMP-3 expression through activation of the MEK (mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) kinase)-ERK1/2 MAPK signaling pathway. On the other hand, Kyn significantly suppressed the expression of type I collagen in fibroblasts as compared with that of control. To test the anti-fibrogenic effect of Kyn in an in vivo model, rabbit ear fibrotic wounds were topically treated with cream containing 50 µg Kyn per l00 µl of cream per wound. The result showed a marked improvement in scar formation relative to the controls. These findings collectively suggest that Kyn can potentially be used as an anti-fibrogenic agent for treating hypertrophic scarring.


Subject(s)
Cicatrix, Hypertrophic/drug therapy , Dermatologic Agents/pharmacology , Dermis/drug effects , Kynurenine/pharmacology , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 3/genetics , Animals , Cells, Cultured , Cicatrix, Hypertrophic/metabolism , Cicatrix, Hypertrophic/pathology , Dermis/cytology , Dermis/enzymology , Disease Models, Animal , Ear, External/pathology , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/enzymology , Foreskin/cytology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Male , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 3/metabolism , Rabbits , Tryptophan/metabolism
14.
Am J Pathol ; 181(4): 1296-305, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22902430

ABSTRACT

Islet transplantation provides a promising approach for treatment of type 1 diabetes mellitus. Amyloid formation and loss of extracellular matrix are two nonimmune factors contributing to death of isolated human islets. We tested the effects of two types of three-dimensional scaffolds, collagen matrix (CM) and fibroblast-populated collagen matrix (FPCM), on amyloid formation, viability, and function of isolated islets. Islets from cadaveric donors were cultured in FPCM, CM, or two-dimensional plate (2D) for 7 days. After 7 days, compared with the 2D culture condition, CM and FPCM markedly reduced amyloid formation of cultured islets and decreased apoptotic ß-cell rate by ∼75%. IL-1ß and Fas levels were also reduced in scaffold-embedded islets. Furthermore, ß/α cell ratios were increased by ∼18% and ∼36% in CM- and FPCM-embedded islets, respectively. Insulin content and insulin response to elevated glucose were also enhanced by both three-dimensional scaffolds. Moreover, culture in CM and FPCM (but not 2D) preserved insulin, GLUT-2, and PDX-1 mRNA expression. FPCM-embedded islets had significantly higher insulin response and lower amyloid formation than CM-embedded islets. These findings suggest that three-dimensional scaffolds reduce amyloid formation and improve viability and function of human islets in vitro, and that CM and fibroblasts have additive effects in enhancing islet function and reducing amyloid formation. Using this strategy is likely to improve outcome in human islet transplantation.


Subject(s)
Amyloid/metabolism , Islets of Langerhans/metabolism , Tissue Culture Techniques/methods , Tissue Scaffolds/chemistry , Tissue Survival , Apoptosis , Caspase 3/metabolism , Cell Count , Enzyme Activation , Gene Expression Regulation , Glucagon-Secreting Cells/metabolism , Glucagon-Secreting Cells/pathology , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Insulin/genetics , Insulin/metabolism , Insulin-Secreting Cells/enzymology , Insulin-Secreting Cells/pathology , Interleukin-1beta/metabolism , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , fas Receptor/metabolism
15.
J Cell Biochem ; 113(8): 2622-32, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22422640

ABSTRACT

We previously suggested that keratinocyte releasable factors might modulate the wound healing process by regulating the expression of key extracellular matrix components such as collagenase (matrix metalloproteinase-1) and type I collagen in fibroblasts. The first one, we called it keratinocyte-derived anti-fibrogenic factor (KDAF), identified as stratifin (SFN) also named 14-3-3σ, revealing a strong collagenase activity. However, the second factor, which we named keratinocyte-derived collagen-inhibiting factor(s) (KD-CIF) that has shown to control the synthesis of type I collagen, was not known. Upon conducting a series of systematic protein purification methods followed by mass spectroscopy, two proteins: secreted protein acidic rich in cystein (SPARC) and SFN were identified in keratinocyte-conditioned media. Using co-immunoprecipitation and 3D modeling, we determined that SFN and SPARC form a complex thereby controlling the type I collagen synthesis and expression in fibroblasts. The levels of these proteins in fibrotic tissues (animal and human) were also evaluated and a differential expression of these proteins between normal and fibrotic tissue confirmed their potential role in development of fibrotic condition. In conclusion, this study describes for the first time an interaction between SPARC and SFN that may have implications for the regulation of matrix deposition and prevention of dermal fibrotic conditions such as hypertrophic scars and keloid.


Subject(s)
14-3-3 Proteins/metabolism , Biomarkers, Tumor/metabolism , Collagen Type I/metabolism , Exonucleases/metabolism , Fibroblasts/metabolism , Osteonectin/metabolism , Skin/cytology , 14-3-3 Proteins/genetics , Biomarkers, Tumor/genetics , Cells, Cultured , Collagen Type I/genetics , Exonucleases/genetics , Exoribonucleases , Humans , Immunoprecipitation , Infant, Newborn , Keratinocytes/metabolism , Osteonectin/genetics , Protein Binding
17.
Invest New Drugs ; 30(4): 1361-70, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21678129

ABSTRACT

Due to the poor prognosis and limited therapeutic options for adult patients with acute lymphoblastic leukemia (ALL), development of novel therapies is much needed to prolong patient survival and increase the efficacy of their treatment. Malignant T cells need high levels of nutrients to maintain their proliferation rate. Borrelidin, a small molecule nitrile-containing macrolide, is an inhibitor of bacterial and eukaryal threonyl-tRNA synthetase. Borrelidin-mediated inhibition of aminoacyl-tRNA synthesis, leads to an induction in the levels of uncharged tRNA, nutritional stress and ultimately inhibition of protein synthesis. The aim of the present study was to investigate whether borrelidin treatment inhibits the proliferation of malignant ALL cell lines, Jurkat and CEM cells, and study the mechanism by which this drug acts. Our results show that borrelidin was able to potently inhibit the proliferation of ALL cell lines with a half maximal inhibitory concentration of 50 ng/ml. Borrelidin showed a greater inhibitory effect on ALL cell lines compared to primary fibroblasts. Flow cytometry and western blot analysis indicated that borrelidin was able to increase the level of apoptosis and cause G(1) arrest in ALL cell lines. Activation of the general control nonderepressible-2 (GCN2) kinase stress responsive pathway and induction of CHOP protein was significantly higher in ALL cell lines treated with borrelidin. These findings collectively suggest for the first time that borrelidin targets ALL cell lines by inducing apoptosis and mediating G(1) arrest and that borrelidin treatment in ALL cell lines is correlated with activation of the GCN2 kinase pathway.


Subject(s)
Apoptosis/drug effects , Nitriles/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Small Molecule Libraries/pharmacology , Threonine-tRNA Ligase/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Fatty Alcohols/pharmacology , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Macrolides/pharmacology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Threonine-tRNA Ligase/metabolism , Transcription Factor CHOP
18.
J Cell Physiol ; 226(7): 1813-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21506112

ABSTRACT

Islet transplantation represents a viable treatment for type 1 diabetes. However, due to loss of substantial mass of islets early after transplantation, islets from two or more donors are required to achieve insulin independence. Islet-extracellular matrix disengagement, which occurs during islet isolation process, leads to subsequent islet cell apoptosis and is an important contributing factor to early islet loss. In this study, we developed a fibroblast populated collagen matrix (FPCM) as a novel scaffold to improve islet cell viability and function post-transplantation. FPCM was developed by embedding fibroblasts within type-I collagen and used as scaffold for islet grafts. Viability and insulin secretory function of islets embedded within FPCM was evaluated in vitro and in a syngeneic murine islet transplantation model. Islets embedded within acellular matrix or naked islets were used as control. Islet cell survival and function was markedly improved particularly after embedding within FPCM. The composite scaffold significantly promoted islet isograft survival and reduced the critical islet mass required for diabetes reversal by half (from 200 to 100 islets per recipient). Fibroblast embedded within FPCM produced fibronectin and growth factors and induced islet cell proliferation. No evidence of fibroblast over-growth within composite grafts was noticed. These results confirm that FPCM significantly promotes islet viability and functionality, enhances engraftment of islet grafts and decreases the critical islet mass needed to reverse hyperglycemia. This promising finding offers a new approach to reducing the number of islet donors per recipient and improving islet transplant outcome.


Subject(s)
Collagen Type I/metabolism , Diabetes Mellitus, Experimental/surgery , Fibroblasts/transplantation , Graft Survival , Islets of Langerhans Transplantation , Islets of Langerhans/surgery , Tissue Scaffolds , Animals , Apoptosis , Blood Glucose/metabolism , Cell Proliferation , Cells, Cultured , Coculture Techniques , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Fibroblasts/metabolism , Fibronectins/metabolism , Insulin/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred C57BL , Time Factors , Tissue Culture Techniques
19.
Wound Repair Regen ; 18(6): 614-23, 2010.
Article in English | MEDLINE | ID: mdl-20955343

ABSTRACT

Engineered skin substitutes (ESSs) comprising both keratinocytes and fibroblasts can afford many advantages over the use of autologous keratinocyte grafts for the treatment of full-thickness and partial-thickness burns. In this study, we investigated the efficacy of a novel ESS containing both genetically altered fibroblasts that express the immunosuppressive factor indoleamine 2,3-dioxygenase (IDO) and primary keratinocytes from a nonautologous source to confer immune protection of xenogeneic cells cultured in a bilayer ESS. The results show that engraftment of IDO expressing skin substitutes on the back of rats significantly improves healing progression over 7 days compared with both nontreated and non-IDO-expressing skin substitutes (p<0.001). Immuno-staining of CD3 and CD31 suggests that IDO-expressing skin substitutes significantly suppress T cell infiltration (p<0.001) and improve neovascularization by four-fold (12.6±1.2 vs. 3.0±1.0 vessel-like structure/high power field), respectively. In conclusion, we found that IDO expression can improve the efficacy of nonautologous ESS for the purpose of wound healing by mitigating T-cell infiltration as well as promoting vascularization of the graft.


Subject(s)
Fibroblasts/metabolism , Graft Rejection/immunology , Immunosuppressive Agents/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Skin, Artificial , T-Lymphocytes/metabolism , Adenoviridae/genetics , Animals , Cell Proliferation , Cells, Cultured , Genetic Vectors , Immunosuppressive Agents/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/pharmacology , Neovascularization, Physiologic , Rats , Rats, Sprague-Dawley , Tissue Engineering , Transfection , Wound Healing
20.
J Cell Physiol ; 225(1): 196-205, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20648630

ABSTRACT

Indoleamine 2,3-dioxygenase (IDO), a tryptophan degrading enzyme, is a potent immunomodulatory factor. IDO expression in fibroblasts selectively induces apoptosis in immune cells but not in primary skin cells. However, the mechanism(s) of this selective effect of IDO-induced low tryptophan environment is not elucidated. The aim of present study was to investigate whether the activity of general control non-derepressible-2(GCN2) kinase stress-responsive pathway and its known inhibitor, protein IMPACT homolog, in immune and skin cells are differentially regulated in response to IDO-induced low tryptophan environment. IDO-expressing human fibroblasts were co-cultured with Jurkat cells, human T cells, fibroblasts, or keratinocytes. Activation of GCN2 pathway was significantly higher in immune cells exposed to IDO-expressing environment relative to that of skin cells. In contrast, IMPACT was highly and constitutively expressed in skin cells while its expression was very low in stimulated T cells and undetectable in Jurkat cells. A significant IDO-induced suppressive as well as apoptotic effect was demonstrated in IMPACT knocked down fibroblasts co-cultured with IDO-expressing fibroblasts. Proliferation of Jurkat cells, stably transduced with IMPACT-expressing vector, was rescued significantly in tryptophan-deficient but not IDO-expressing environment. This may be due to the ability of IMPACT to recover the effects of IDO-mediated tryptophan depletion (GCN2 dependent) but not the effects of IDO-generated cytotoxic metabolites. These findings collectively suggest for the first time that high expression of protein IMPACT homolog in non-immune cells such as skin cells acts as a protective mechanism against IDO-induced GCN2 activation, therefore, makes them resistant to the amino acid-deprived environment caused by IDO.


Subject(s)
Cell Death/drug effects , Fibroblasts/drug effects , Indoleamine-Pyrrole 2,3,-Dioxygenase/pharmacology , Proteins/metabolism , Animals , Antiviral Agents/pharmacology , Cells, Cultured , Coculture Techniques , Culture Media, Conditioned/chemistry , Fibroblasts/cytology , Fibroblasts/physiology , Humans , Interferon-gamma/pharmacology , Intracellular Signaling Peptides and Proteins , Jurkat Cells , Keratinocytes/cytology , Keratinocytes/physiology , Kynurenine/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteins/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction/physiology , T-Lymphocytes/cytology , T-Lymphocytes/physiology , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Tryptophan/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...