Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
PLoS One ; 17(6): e0262060, 2022.
Article in English | MEDLINE | ID: mdl-35737933

ABSTRACT

Dermal wound healing is a complex process which requires the interaction of many cell types and mediators in a highly sophisticated temporal sequence. Myeloid cells which compose of a significant proportion of the inflammatory cells infiltrate to the to a wound site where they play important roles in clearance of damaged tissue and microorganisms. Myeloid cells have the capacity to be converted into fibroblast-like cells and endothelial cells during wound healing process. However, whether myeloid cells in wounds can convert into epithelial cells where they contribute to healing process is not clear. In this study, we performed double immunofluorescent staining with antibodies for hematopoietic cells and keratinocytes as well as cell tracing technique to investigate hematopoietic cell conversion. The result showed that during the healing process, some of the CD45-positive hematopoietic cells also expressed keratin 14, a marker for keratinocytes. Further, double immunofluorescent staining in dermal wounds, using CD11b and K14 antibodies indicated that CD11b-positive myeloid cells were the origin of newly generated epithelial cells. Through tracing injected labeled splenocyte-derived myeloid cells in skin, we confirmed that myeloid cells were able to convert into keratinocytes in repaired skin. Furthermore, our results from in vivo experiments provided new information on contribution of myeloid cells in hair follicle regeneration. In conclusion, this work highlights the myeloid cell contributions in wound repair and hair follicle regeneration through conversion of M-CSF-stimulated CD11b-positive myeloid cells into epithelial cells in a murine model.


Subject(s)
Hair Follicle , Re-Epithelialization , Animals , Endothelial Cells , Epithelial Cells , Macrophage Colony-Stimulating Factor/metabolism , Mice , Myeloid Cells , Regeneration , Skin/metabolism , Wound Healing
2.
Adv Wound Care (New Rochelle) ; 10(3): 113-122, 2021 03.
Article in English | MEDLINE | ID: mdl-32320360

ABSTRACT

Objective: Full-thickness burn wounds require immediate coverage, and the primary clinical approaches comprise of skin allografts and autografts. The use of allografts is often temporary due to the antigenicity of allografts. In contrast, the availability of skin autografts may be limited in large burn injuries. In such cases, skin autografts can be expanded through the use of a skin mesher, creating meshed split-thickness skin grafts (MSTSGs). MSTSGs have revolutionized the treatment of large full-thickness burn injuries since the 1960s. However, contractures and poor esthetic outcomes remain a problem. We previously formulated and prepared an in situ forming skin substitute, called MeshFill (MF), which can conform to complex shapes and contours of wounds. The objective of this study was to assess the esthetic and wound healing outcomes in full-thickness wounds treated with a combination of MF and MSTSG in a porcine model. Approach: Either MSTSGs or MSTSG+MF was applied to full-thickness excisional wounds in Yorkshire pigs. Wound healing outcomes were assessed using histology, immunohistochemistry, and wound surface area analysis from day 10 to 60. Clinical evaluation of wounds were utilized to assess esthetic outcomes. Results: The results demonstrated that the combination of MSTSGs and MF improved wound healing and esthetic outcomes. Innovation: Effects of MSTSGs and reconstitutable liquid MF in a full-thickness porcine model were investigated for the first time. Conclusion: MF provides promise as a combination therapeutic regimen to improve wound healing and esthetic outcomes.


Subject(s)
Burns/surgery , Skin Transplantation/methods , Wound Healing/physiology , Animals , Burns/pathology , Disease Models, Animal , Esthetics , Female , Skin, Artificial , Swine , Temperature
3.
J Investig Dermatol Symp Proc ; 20(1): S16-S21, 2020 11.
Article in English | MEDLINE | ID: mdl-33099378

ABSTRACT

Alopecia areata (AA), which is defined as an autoimmune hair loss disease, has a serious impact on the quality of life for patients with AA worldwide. In this study, to our knowledge, a previously unreported method of AA induction in C3H mice has been established and validated. Using this method, we showed that dermal injection of 1-3 million of a mixture of skin cells freshly isolated from AA-affected skin induces AA in more than 80% of healthy mice. Contrary to the previous protocol, the induction of AA by this approach does not need any surgical AA skin grafting, cell manipulation, or high number of activated T cells. We also showed that dermal injection of adherent myeloid cells (mainly CD11b+) in healthy mice is as potent as a mixture of none adherent CD3+ T cells and CD19+ B cells in the induction of AA. Interestingly, most of the mice (7 out of 8) that received non-adherent cells developed AA universalis, whereas most of the mice (5 out of 7) that received adherent cells developed patchy AA. Finally, we found a high number of stage-specific embryonic antigen-expressing cells whose expression in monocytes in an inflammatory disease causes the release of inflammatory cytokines, TNF-α and IL-1ß, from these cells in AA-affected skin.


Subject(s)
Alopecia Areata/metabolism , Alopecia Areata/pathology , Myeloid Cells/metabolism , Myeloid Cells/transplantation , Animals , Antigens, Tumor-Associated, Carbohydrate/metabolism , CD11b Antigen/metabolism , Cell Adhesion , Cells, Cultured , Disease Models, Animal , Female , Lewis X Antigen/metabolism , Mice , Mice, Inbred C3H , Stage-Specific Embryonic Antigens/metabolism
4.
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.

5.
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
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.
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
8.
Wound Repair Regen ; 24(4): 695-704, 2016 07.
Article in English | MEDLINE | ID: mdl-27197606

ABSTRACT

Skin transplantation provides an excellent potential model to investigate the immunology of allograft rejection and tolerance induction. Despite the theoretical ease of performing skin transplantation, as well as the potential of directly observing the reaction to the transplanted tissue, the poor reliability of skin transplantation in the mouse has largely precluded the use of this model. Furthermore, there is controversy regarding the most appropriate skin graft donor site due to poor success of back skin transplantation, as compared with the thinner ear or tail skin. This study demonstrates a reliable method to successfully perform skin grafts in a mouse model, as well as the clinical and histologic outcome of syngeneic grafts. A total of 287 grafts were performed (in 126 mice) utilizing donor skin from the ear, tail or back. No graft failure or postoperative mortality was observed. Comparison of this technique with two previously established protocols of skin transplantation (5.0 absorbable Suture + tissue glue technique and no-suture technique) demonstrates the significant improvement in the engraftment success of the new technique. In summary, a new technique for murine skin grafting demonstrates improved reliability across donor site locations and strains, increasing the potential for investigating interventions to alter the rejection process.


Subject(s)
Allografts/immunology , Graft Rejection/immunology , Graft Survival/immunology , Immune Tolerance , Skin Transplantation/methods , Wound Healing/physiology , Allografts/blood supply , Animals , Bandages , Disease Models, Animal , Graft Rejection/physiopathology , Graft Survival/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Reproducibility of Results
9.
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
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.
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
12.
Mol Cell Biochem ; 409(1-2): 213-24, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26298287

ABSTRACT

Methotrexate (MTX), an anti-metabolite and anti-inflammatory drug, has been used to effectively manage and prevent keloids, but its mechanism(s) of action has not been elucidated. Our study sought to evaluate the effect of MTX on the production of key extra cellular matrix components, collagen, and matrix metalloproteinase-1 (MMP-1), produced by fibroblasts and involved in development of fibrosis. The proliferation and viability of cultured human dermal fibroblasts in response to different concentrations of MTX were determined using cell counting and MTT assay, respectively. Western blot analysis was used to determine the levels of both intracellular and secreted type 1 collagen and MMP-1. The results showed no significant changes in the proliferation of fibroblasts treated with 50 ng/ml of MTX as compared to that of control. Under the same experimental conditions, the level of secreted and intracellular type I collagen was markedly reduced and, conversely, the level of MMP-1 increased in treated neonatal, adult, and hypertrophic scar fibroblasts as compared with those of controls. The possible involvement of MTX-induced extracellular signal-regulated kinase 1/2 (ERK1/2) pathway in MMP-1 production was also studied and the result showed an increase in phosphorylated ERK 1/2 in response to MTX treatment. In summary, the findings of this study revealed that MTX significantly reduced collagen production in different strains of fibroblasts derived from neonatal, adult, and hypertrophic scar tissues, while under the same experimental conditions, it increased the expression of MMP-1. As such, our findings validate and identify a potential mechanism through which MTX functions as an anti-fibrogenic factor in treating fibroproliferative disorders.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Cicatrix, Hypertrophic/metabolism , Collagen Type I/biosynthesis , Fibroblasts/metabolism , Matrix Metalloproteinase 1/biosynthesis , Methotrexate/pharmacology , Adult , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dermis/cytology , Dermis/metabolism , Extracellular Matrix/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/cytology , Humans , Infant, Newborn
13.
Transplantation ; 99(7): 1341-8, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25769070

ABSTRACT

BACKGROUND: We have previously shown that an immunomodulatory enzyme, indoleamine 2,3-dioxygenase (IDO) in dermal fibroblasts generates a tryptophan-deficient environment that selectively inhibits proliferation and induces apoptosis of bystander CD4+ and CD8+ T cells, but not pancreatic islets. Because these immune cells are involved in islet allograft rejection, we hypothesized that transplantation of islets embedded in a novel 3-dimensional composite scaffold within which stable IDO-expressing fibroblasts serve as source of local immunosuppression would lead to normoglycemia in a streptozotocin-induced diabetic mouse model. METHODS: Islet grafts were prepared by embedding stable IDO-expressing fibroblasts and allogeneic islets into a protease-resistant composite scaffold. Islets function and survival were evaluated in vitro using immunohistochemistry. Allografts were transplanted under the kidney capsule of streptozotocin-induced diabetic mice; viability, function, and criteria for graft take were evaluated. Flow cytometry was performed to determine specific intragraft, draining lymph nodes and spleen T-cell population, and splenocytes alloantigen responsiveness of graft recipients. RESULTS: The results of a series of in vitro experiments revealed that IDO-expressing fibroblasts do not compromise islet function or survival. The expression of IDO suppressed the proliferation of alloantigen-stimulated splenocytes. The in vivo experiments revealed that local IDO expression delivered by lentiviral vector prolonged islet allograft survival (51.0 ± 2.9 days) by increasing the population of FOXP3+ regulatory T cells at the graft site and graft-draining lymph nodes and preventing T-cell infiltration. CONCLUSIONS: This study shows that incorporation of islets within our novel matrix that is equipped with stable IDO-expressing fibroblasts prolongs allograft survival.


Subject(s)
Diabetes Mellitus, Experimental/therapy , Fibroblasts/transplantation , Genetic Therapy/methods , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Islets of Langerhans Transplantation/methods , Islets of Langerhans/surgery , Tissue Scaffolds , Animals , Biomarkers/blood , Blood Glucose/metabolism , Cell Proliferation , Cells, Cultured , Coculture Techniques , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/immunology , Fibroblasts/enzymology , Fibroblasts/immunology , Graft Rejection/immunology , Graft Rejection/prevention & control , Graft Survival , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Islets of Langerhans/enzymology , Islets of Langerhans/immunology , Lymph Nodes/enzymology , Lymph Nodes/immunology , Lymphocyte Activation , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Spleen/enzymology , Spleen/immunology , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , Time Factors , Tissue Culture Techniques , Transfection
14.
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
15.
Can J Diabetes ; 37(1): 27-35, 2013 Feb.
Article in English | MEDLINE | ID: mdl-24070745

ABSTRACT

INTRODUCTION: Islet transplantation is a promising strategy to restore efficient insulin regulation in type 1 diabetes mellitus patients. However, shortage of islet donors, poor islet survival and toxicity of immunosuppressants often reduce the graft functional lifetime. METHODS: We previously showed that a fibroblast populated-collagen matrix (CM) significantly improved engrafted islet viability/function. However, this composite was prone to gradual biodegradation and contraction. Moreover, to avoid use of systemic immunosuppressants, we proposed the use of a local immunosuppressive enzyme, indoleamine-2,3-dioxygenase (IDO). We developed a novel bioengineered crosslinked CM (CCM) to provide optimal matrix biomimetic. Viability and insulin secretory function of islets embedded within fibroblast populated CCM (FP-CCM) was evaluated in vitro and in vivo. IDO expression was transduced in fibroblasts by a lentiviral vector carrying IDO gene and islet viability was evaluated in the presence and absence of IDO producing cells. RESULTS: Islet survival/function markedly improved within FP-CCM. Furthermore, our data shows that local lentiviral induction of IDO delivered by FP-CCM is nontoxic to the embedded islets. CONCLUSIONS: This promising finding offers a new approach to improving islet transplant outcome.


Subject(s)
Cell Culture Techniques , Islets of Langerhans/cytology , Animals , Biomimetic Materials , Cell Proliferation , Cell Survival , Collagen , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Islets of Langerhans Transplantation/methods , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Tissue Scaffolds
16.
J Burn Care Res ; 34(3): e143-52, 2013.
Article in English | MEDLINE | ID: mdl-23128134

ABSTRACT

The healing of a burn injury is a dynamic biological process, and there must be a set of factors that gradually slow down and/or terminate this healing process upon epithelialization. We have previously demonstrated that some members of the 14-3-3 protein family including stratifin, which is mainly released by differentiated keratinocytes, have a potent matrix metalloproteinase-1 (MMP-1), -3, -8, and -24 stimulatory effect for fibroblasts. During the course of our studies, we discovered that human red blood cells (RBCs) contain a high level of different 14-3-3 isoforms. In this study, we evaluated the presence of these proteins in RBC and examined the efficacy of RBC in stimulating the expression of MMP-1 in dermal fibroblasts using Western blot analysis. The findings revealed the presence of all seven isoforms of 14-3-3 (η, γ, ε, σ, ß, ζ, and τ) in RBC lysate, though with different levels of expression. Interestingly, the levels of some of these proteins were higher in RBC from new born babies compared to those of adults. We also found that RBC lysate significantly increased the expression of MMP-1 in different strains of fibroblasts, and this effect has been abrogated by almost 20% when R18 was used to deplete the 14-3-3 proteins from lysate. However, when RBC lysate was passed through a 3-kDa filter, and then whole RBC lysate, retentate, and filtrate were used to treat three strains of fibroblasts, MMP-1 expression was increased significantly, indicating the presence of MMP-1-inducing factors smaller than 3kDa in RBC lysate as well. The MMP-1 stimulatory effect of RBC lysate for fibroblasts was stable at freeze/thawing as well as freezing for at least 1 year. In conclusion, the MMP-1 stimulatory effect of RBC lysates might play an important role in the regulation of extracellular matrix under physiological and pathological conditions. Because of its MMP-1 stimulating effect, the RBC lysate might be used as an antifibrogenic factor for the treatment of some fibroproliferative conditions such as hypertrophic scarring and keloid.


Subject(s)
14-3-3 Proteins/metabolism , Burns/metabolism , Dermis/metabolism , Erythrocytes/metabolism , Fibroblasts/metabolism , Matrix Metalloproteinase 1/metabolism , Wound Healing/physiology , Adult , Blotting, Western , Cells, Cultured , Dermis/cytology , Electrophoresis, Polyacrylamide Gel , Female , Fibroblasts/physiology , Humans , Infant, Newborn , Male , Middle Aged
17.
Mol Cell Biochem ; 370(1-2): 79-88, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22814701

ABSTRACT

During the early stage of wound healing process, blood clots can be served as a temporary extracellular matrix (ECM) to let skin cell migration and proliferation. The red blood cells are generally thought as inert bystanders in the early and inflammatory phase of wound healing. Here, we provide evidence that red blood cells (RBC) also play an important role in modulation of key ECM components such as type-I collagen, α-smooth muscle actin, fibronectin, and matrix metalloproteinases (MMPs). In this study, we used western blot analysis and showed a significant increase in the level of MMP-1, 2, 3. Furthermore, we found that RBC lysate significantly down-regulates type-I collagen and α-smooth muscle actin while up-regulates fibronectin expression in dermal fibroblasts. To further explore the mechanism by which RBC lysate modulates MMP-1 expression, the effect of inhibitors for three MAPK signaling pathways on RBC inducing MMP-1 expression by dermal fibroblasts were tested. The result showed that the inhibitor of ERK1/2 could abrogate the stimulatory effect of RBC lysate on MMP-1 expression in dermal fibroblasts. Consistently, RBC treatment results in an increase of ERK1/2 phosphorylation in dermal fibroblast. In conclusion, these findings suggest that RBC lysate can modulate the expression of MMPs and key ECM components which are important in healing process.


Subject(s)
Cell Extracts/pharmacology , Dermis/cytology , Erythrocytes/metabolism , Extracellular Matrix Proteins/metabolism , Fibroblasts/metabolism , Actins/genetics , Actins/metabolism , Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured , Collagen Type I/genetics , Collagen Type I/metabolism , Down-Regulation/drug effects , Extracellular Matrix Proteins/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/enzymology , Fibronectins/genetics , Fibronectins/metabolism , Humans , MAP Kinase Signaling System/drug effects , Matrix Metalloproteinases/metabolism
18.
PLoS One ; 7(5): e37747, 2012.
Article in English | MEDLINE | ID: mdl-22662207

ABSTRACT

Indoleamine 2,3-dioxygenase (IDO) can locally suppress T cell-mediated immune responses. It has been shown that defective self-tolerance in early prediabetic female non-obese diabetic (NOD) mice can be attributed to the impaired interferon-gamma (IFN-γ)- induced IDO expression in dendritic cells of these animals. As IFN-γ can induce IDO in both dendritic cells and fibroblasts, we asked the question of whether there exists a similar defect in IFN-γ-induced IDO expression in NOD mice dermal fibroblasts. To this end, we examined the effect of IFN-γ on expression of IDO and its enzymatic activity in NOD dermal fibroblasts. The results showed that fibroblasts from either prediabetic (8 wks of age) female or male, and diabetic female or male (12 and 24 wks of age respectively) NOD mice failed to express IDO in response to IFN-γ treatment. To find underlying mechanisms, we scrutinized the IFN- γ signaling pathway and investigated expression of other IFN-γ-modulated factors including major histocompatibility complex class I (MHC-I) and type I collagen (COL-I). The findings revealed a defect of signal transducer and activator of transcription 1 (STAT1) phosphorylation in NOD cells relative to that of controls. Furthermore, we found an increase in MHC-I and suppression of COL-I expression in fibroblasts from both NOD and control mice following IFN-γ treatment; indicating that the impaired response to IFN-γ in NOD fibroblasts is specific to IDO gene. Finally, we showed that an IFN-γ-independent IDO expression pathway i.e. lipopolysaccharide (LPS)-mediated-c-Jun kinase is operative in NOD mice fibroblast. In conclusion, the findings of this study for the first time indicate that IFN-γ fails to induce IDO expression in NOD dermal fibroblasts; this may partially be due to defective STAT1 phosphorylation in IFN-γ-induced-IDO signaling pathway.


Subject(s)
Diabetes Mellitus, Type 1/enzymology , Fibroblasts/drug effects , Fibroblasts/enzymology , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Interferon-gamma/pharmacology , Animals , Collagen/genetics , Dendritic Cells/metabolism , Diabetes Mellitus, Type 1/metabolism , Female , Fibroblasts/metabolism , Gene Expression/drug effects , Genes, MHC Class I , Lipopolysaccharides/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Phosphorylation/drug effects , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects , Skin/metabolism , Spleen/metabolism , Transduction, Genetic , Tryptophan/metabolism
20.
Mol Cell Biochem ; 360(1-2): 261-70, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21948273

ABSTRACT

Airway remodelling in asthma involves various mediators modulating the production/breakdown of collagen by lung fibroblasts. Matrix metalloproteinase-1 (MMP-1) plays an important role in collagen breakdown. We recently showed that epithelial cell-derived extracellular form of 14-3-3σ is an important inducer of MMP-1 expression in skin fibroblasts. Thus, we hypothesized that 14-3-3 proteins are important regulators of MMP-1 expression in the respiratory airway. We examined the presence of extracellular 14-3-3 proteins in conditioned media obtained from primary lung epithelial cells, A549 and HS24 cells, and their effect on MMP-1 expression by lung fibroblasts (IMR-90). In addition, we evaluated IMR-90 response to 14-3-3 proteins in the presence of transforming growth factor-ß(1) (TGF-ß(1)), a cytokine known to decrease MMP-1 expression by fibroblasts. Extracellular 14-3-3α/ß, but not -σ, is released by the human-derived lung epithelial cell lines, A549 and HS24. Unlike dermal fibroblasts, IMR-90 cells do not produce MMP-1 in response to 14-3-3σ. Conversely, MMP-1 production was induced following treatment of IMR-90 with recombinant or lung epithelial cell-derived 14-3-3α/ß. These findings were also confirmed using primary human bronchial epithelial cells and lung fibroblasts obtained from non-asthmatic patients. The MMP-1-inducing effect of 14-3-3α/ß on IMR-90 was not inhibited by TGF-ß(1). Lung epithelial cell-derived 14-3-3α/ß has a potent MMP-1-inducing effect on airway fibroblasts. Modulation of MMP-1 by 14-3-3α/ß, may be important in the alteration of collagenase production associated with airway remodelling in obstructive lung diseases. Our data indicate that 14-3-3 proteins may be potential targets for future therapeutic strategies aimed at modulating tissue remodelling in asthma.


Subject(s)
14-3-3 Proteins/physiology , Epithelial Cells/metabolism , Gene Expression , Lung/pathology , Matrix Metalloproteinase 1/metabolism , 14-3-3 Proteins/metabolism , Airway Remodeling , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Culture Media, Conditioned , Epithelial Cells/enzymology , Exonucleases/metabolism , Exoribonucleases , Fibroblasts/enzymology , Fibroblasts/metabolism , Gene Expression Regulation , Humans , Inflammation , L-Lactate Dehydrogenase/metabolism , Matrix Metalloproteinase 1/genetics , Primary Cell Culture , Protein Isoforms/metabolism , Transforming Growth Factor beta1/pharmacology , Transforming Growth Factor beta1/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...