Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Commun Biol ; 5(1): 85, 2022 01 21.
Article in English | MEDLINE | ID: mdl-35064244

ABSTRACT

Notch signaling is one of the most common drivers of carcinogenesis in many types of cancers, including hepatocellular carcinoma (HCC); however, it occasionally suppresses tumor progression. Moreover, it is virtually unknown how different sets of Notch ligands and receptors regulate the HCC development. In this study, we demonstrate that the expression of the Notch ligands, Delta-like 4 (Dll4) and Jagged-1 (Jag1), is upregulated during diethylnitrosamine-induced hepatocarcinogenesis. Dll4 is detected in the preneoplastic hepatocytes and HCC cells, but not in the normal hepatocytes, while Jag1 is expressed in the desmin-positive mesenchymal cells. Hepatocyte-specific Dll4 knockout abolishes the Notch1 signaling and suppresses the tumor progression. In contrast, Jag1 deletion induces the ectopic expression of Dll4 in hepatocytes along with the loss of Notch2 signaling, leading to the tumor progression. These results indicate that the two distinct Notch signals, Dll4/Notch1 and Jag1/Notch2, are antagonistic to each other, exerting opposite effects on HCC progression.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Calcium-Binding Proteins/metabolism , Carcinogenesis/metabolism , Jagged-1 Protein/metabolism , Liver Neoplasms/metabolism , Receptor, Notch1/metabolism , Receptor, Notch2/metabolism , Animals , Carcinogenesis/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Gene Expression Regulation , Genetic Predisposition to Disease , Jagged-1 Protein/genetics , Liver Neoplasms/pathology , Mice , Mice, Knockout , Receptor, Notch1/genetics , Receptor, Notch2/genetics
2.
Biochem Biophys Res Commun ; 583: 93-99, 2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34735885

ABSTRACT

Exosomes are secreted from a variety of cells and transmit parental cell-derived biomolecules, such as nucleic acids and proteins, to recipient cells in distant organs. In addition to their important roles in both physiological and pathological conditions, exosomes are expected to serve as natural drug carriers without any cytotoxicity, immunogenicity, or tumorigenicity. However, the use of exosomes as drug delivery tools is limited due to the low uptake efficiency of the target cells, insufficient release of the contents from the endosome to the cytosol, and possible adverse effects caused by the delivery to non-target cells. In the present study, we examined the effects of the modification of exosomes with carbonate apatite or a lactose-carrying polymer. Using newly generated monitoring exosomes that contain either firefly luciferase or fused mCherry/enhanced green fluorescent protein, we demonstrated that the modification of exosomes with carbonate apatite improved their release from the endosome into the cytosol in recipient cells. Meanwhile, the modification of exosomes with a lactose-carrying polymer enhanced the selective delivery to parenchymal hepatocytes. These modified exosomes may provide an efficient strategy for macromolecule therapy for incurable diseases that cannot be treated with conventional small-molecule compounds.

3.
Regen Ther ; 18: 223-230, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34377752

ABSTRACT

INTRODUCTION: Artificial dermis is an effective therapeutic method for full-thickness dermal defects. However, the currently available artificial dermis made of porcine or bovine type I collagen has several limitations such as incomplete epithelialization and delayed migration of fibrogenic and angiogenic cells into the graft. We previously developed a composite dermal graft containing a mixture of moon jellyfish collagen and porcine type I collagen, and reported its stimulatory effect on both the re-epithelialization of the epidermis and the migration of fibrogenic and angiogenic cells into the graft. In the present study, we examined whether the same effect was observed by administering jellyfish collagen solution externally onto an artificial dermal graft made of bovine type I collagen. METHODS: We used a 6 mm full-thickness wound defect model. Moon jellyfish collagen was prepared as a concentrated 0.5% solution and dripped externally onto a transplanted artificial dermal graft made of bovine type I collagen. Wound repair and long-term dermal tissue remodeling were compared between mice administered jellyfish collagen solution on the bovine collagen graft and those transplanted with a composite dermal graft containing the same amounts of jellyfish and bovine collagens. The stimulatory effect of jellyfish collagen solution was also evaluated using diabetic dB/dB mice. RESULTS: External administration of jellyfish collagen solution onto the bovine collagen graft significantly accelerated wound closure compared to control saline. It also decreased the number of inflammatory cells infiltrating the wound and suppressed absorption of the transplanted graft, as well as reduced subsequent scar formation. Furthermore, external administration of jellyfish collagen solution onto the bovine collagen graft improved the delayed wound healing in diabetic model mice, and this effect was superior to that of the currently used basic fibroblast growth factor. CONCLUSIONS: External administration of moon jellyfish collagen solution onto a bovine collagen graft significantly accelerated physiological wound healing and prevented excessive scar formation. It also improved wound closure in diabetic model mice, confirming its therapeutic application for intractable skin ulcers caused by impaired wound healing.

4.
Biochem Biophys Res Commun ; 528(3): 420-425, 2020 07 30.
Article in English | MEDLINE | ID: mdl-32505348

ABSTRACT

Parenchymal hepatocytes are responsible for most of the metabolic functions of the liver, but exhibit distinct functional properties depending on their localization within the hepatic lobule. Cytochrome P450 oxidases represent a family of drug-metabolizing enzymes, which are expressed predominantly in hepatocytes localized in the centrilobular area (zone 3). The present study describes a unique transgenic mouse strain that distinguishes zone 3 hepatocytes from periportal zone 1 hepatocytes by the intensity of EGFP fluorescence. Both zone 1 and zone 3 hepatocytes isolated from these mice showed the same zone-specific gene expression patterns as in liver tissue in vivo. Experiments using primary cultures of hepatocytes indicated that a combination of low oxygen concentration and activation of Wnt/ß-catenin signaling maintained the expression of zone 3-specific P450 drug-metabolizing enzymes, which was characterized by their susceptibility to acetaminophen-induced mitochondrial dysfunction. These zone-specific hepatocytes provide a useful system in the research area of liver pathophysiology and drug development.


Subject(s)
Hepatocytes/metabolism , NADPH-Ferrihemoprotein Reductase/metabolism , Acetaminophen/toxicity , Animals , Cell Separation , Cells, Cultured , Flow Cytometry , Gene Expression , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hepatocytes/cytology , Hepatocytes/drug effects , Liver/anatomy & histology , Liver/cytology , Liver/metabolism , Mice , Mice, Transgenic , Mitochondria, Liver/metabolism , NADPH-Ferrihemoprotein Reductase/genetics , Oxygen/metabolism , Wnt Signaling Pathway/drug effects
5.
Adv Wound Care (New Rochelle) ; 9(6): 295-311, 2020 06.
Article in English | MEDLINE | ID: mdl-32286206

ABSTRACT

Background and Objective: Impaired dermal wound healing represents a major medical issue in today's aging populations. Granulation tissue formation in the dermis and reepithelization of the epidermis are both important and necessary for proper wound healing. Although a number of artificial dermal grafts have been used to treat full-thickness dermal loss in humans, they do not induce reepithelization of the wound, requiring subsequent epithelial transplantation. In the present study, we sought a novel biomaterial that accelerates the wound healing process. Approach: We prepared a composite biomaterial made of jellyfish and porcine collagens and developed a hybrid-type dermal graft that composed of the upper layer film and the lower layer sponge made of this composite biomaterial. Its effect on dermal wound healing was examined using a full-thickness excisional wound model. Structural properties of the dermal graft and histological features of the regenerating skin tissue were characterized by electron microscopic observation and immunohistological examination, respectively. Results: The composite biomaterial film stimulated migration of keratinocytes, leading to prompt reepithelization. The regenerating epithelium consisted of two distinct cell populations: keratin 5-positive basal keratinocytes and more differentiated cells expressing tight junction proteins such as claudin-1 and occludin. At the same time, the sponge made of the composite biomaterial possessed a significantly enlarged intrinsic space and enhanced infiltration of inflammatory cells and fibroblasts, accelerating granulation tissue formation. Innovation: This newly developed composite biomaterial may serve as a dermal graft that accelerates wound healing in various pathological conditions. Conclusion: We have developed a novel dermal graft composed of jellyfish and porcine collagens that remarkably accelerates the wound healing process.


Subject(s)
Biocompatible Materials/pharmacology , Collagen/pharmacology , Dermis/pathology , Granulation Tissue/physiology , Wound Healing/drug effects , Animals , Biocompatible Materials/administration & dosage , Cell Differentiation , Cell Movement/physiology , Collagen/metabolism , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/physiology , Female , Fibroblasts , Keratin-5/metabolism , Keratinocytes/drug effects , Keratinocytes/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Electron/methods , Regeneration , Scyphozoa , Skin/growth & development , Skin/ultrastructure , Skin Transplantation/methods , Swine , Tight Junction Proteins/metabolism
6.
Stem Cells ; 37(1): 89-101, 2019 01.
Article in English | MEDLINE | ID: mdl-30270488

ABSTRACT

Granulocyte colony stimulating factor (G-CSF) has been reported to ameliorate impaired liver function in patients with advanced liver diseases through mobilization and proliferation of hepatic progenitor cells (HPCs). However, the underlying mechanisms remain unknown. We previously showed that G-CSF treatment increased the number of bone marrow (BM)-derived cells migrating to the fibrotic liver following repeated carbon tetrachloride (CCl4 ) injections into mice. In this study, we identified opioid growth factor receptor-like 1 (OGFRL1) as a novel BM cell-derived accelerator of fibrotic liver regeneration in response to G-CSF treatment. Endogenous Ogfrl1 was highly expressed in the hematopoietic organs such as the BM and spleen, whereas the liver contained a relatively small amount of Ogfrl1 mRNA. Among the peripheral blood cells, monocytes were the major sources of OGFRL1. Endogenous Ogfrl1 expression in both the peripheral blood monocytes and the liver was decreased following repeated CCl4 injections. An intrasplenic injection of cells overexpressing OGFRL1 into CCl4 -treated fibrotic mice increased the number of HPC and stimulated proliferation of hepatic parenchymal cells after partial resection of the fibrotic liver. Furthermore, overexpression of OGFRL1 in cultured HPC accelerated their differentiation as estimated by increased expression of liver-specific genes such as hepatocyte nuclear factor 4α, cytochrome P450, and fatty acid binding protein 1, although it did not affect the colony forming ability of HPC. These results indicate a critical role of OGFRL1 in the mobilization and differentiation of HPC in the fibrotic liver, and administration of OGFRL1-expressing cells may serve as a potential regenerative therapy for advanced liver fibrosis. Stem Cells 2019;37:89-101.


Subject(s)
Hematopoietic Stem Cell Mobilization/methods , Liver Cirrhosis/genetics , Liver Cirrhosis/therapy , Liver Regeneration/genetics , Regenerative Medicine/methods , Stem Cells/metabolism , Animals , Cell Differentiation , Humans , Male , Mice , Transfection
7.
Hepatol Commun ; 1(3): 215-229, 2017 05.
Article in English | MEDLINE | ID: mdl-29404455

ABSTRACT

The liver is well known to possess high regenerative capacity in response to partial resection or tissue injury. However, liver regeneration is often impaired in the case of advanced liver fibrosis/cirrhosis when mature hepatocytes can hardly self-proliferate. Hepatic progenitor cells have been implicated as a source of hepatocytes in regeneration of the fibrotic liver. Although alpha-fetoprotein (AFP) is known as a clinical marker of progenitor cell induction in injured/fibrotic adult liver, the origin and features of such AFP-producing cells are not fully understood. Here, we demonstrate a unique and distinct AFP-expressing cell population that is induced by the Jagged1/Notch2 signal in murine fibrotic liver. Following repeated carbon tetrachloride injections, a significant number of AFP-positive cells with high proliferative ability were observed along the fibrous septa depending on the extent of liver fibrosis. These AFP-positive cells exhibited features of immature hepatocytes that were stained positively for hepatocyte-lineage markers, such as albumin and hepatocyte nuclear factor 4 alpha, and a stem/progenitor cell marker Sox9. A combination of immunohistological examination of fibrotic liver tissues and coculture experiments with primary hepatocytes and hepatic stellate cells indicated that increased Jagged1 expression in activated hepatic stellate cells stimulated Notch2 signaling and up-regulated AFP expression in adjacent hepatocytes. The mobilization and proliferation of AFP-positive cells in fibrotic liver were further enhanced after partial hepatectomy, which was significantly suppressed in Jagged1-conditional knockout mice. Finally, forced expression of the intracellular domain of Notch2 in normal liver induced a small number of AFP-expressing hepatocytes in vivo. Conclusion: Insight is provided into a novel pathophysiological role of Jagged1/Notch2 signaling in the induction of AFP-positive cells in fibrotic liver through the interaction between hepatocytes and activated hepatic stellate cells. (Hepatology Communications 2017;1:215-229).

8.
PLoS One ; 11(1): e0146592, 2016.
Article in English | MEDLINE | ID: mdl-26745268

ABSTRACT

Mitochondrial oxidative stress is considered as a key accelerator of fibrosis in various organs including the liver. However, the production of oxidative stress and progression of liver fibrosis may merely represent the independent consequences of hepatocellular injury caused by the primary disease. Because of a lack of appropriate experimental models to evaluate the sole effects of oxidative stress, it is virtually unknown whether this stress is causatively linked to the progression of liver fibrosis. Here, we examined the direct effects of mitochondrial reactive oxygen species (ROS) on the progression of high fat/calorie diet-induced steatohepatitis using Tet-mev-1 mice, in which a mutated succinate dehydrogenase transgene impairs the mitochondrial electron transport and generates an excess amount of ROS in response to doxycycline administration. Wild type and Tet-mev-1 mice that had been continuously given doxycycline-containing water were subsequently fed either normal chow or a cholesterol-free high-fat/high-sucrose diet for 4 months at approximately 1 or 2 years of age. Histopathological examinations indicated that neither the mitochondrial ROS induced in Tet-mev-1 mice nor the feeding of wild type animals with high-fat/high-sucrose diet alone caused significant liver fibrosis. Only when the Tet-mev-1 mice were fed a high-fat/high-sucrose diet, it induced lipid peroxidation in hepatocytes and enhanced hepatic CC chemokine expression. These events were accompanied by increased infiltration of CCR5-positive cells and activation of myofibroblasts, resulting in extensive liver fibrosis. Interestingly, this combinatorial effect of mitochondrial ROS and excess fat/calorie intake on liver fibrosis was observed only in 2-year-old Tet-mev-1 mice, not in the 1-year-old animals. Collectively, these results indicate that mitochondrial ROS in combination with excess fat/calorie intake accelerates liver fibrosis by enhancing CC chemokine production in aged animals. We have provided a good experimental model to explore how high fat/calorie intake increases the susceptibility to nonalcoholic steatohepatitis in aged individuals who have impaired mitochondrial adaptation.


Subject(s)
Chemokines/biosynthesis , Non-alcoholic Fatty Liver Disease/metabolism , Oxidative Stress , Animals , Cells, Cultured , Diet, High-Fat/adverse effects , Disease Progression , Energy Intake , Gene Ontology , Hepatocytes/metabolism , Lipid Peroxidation , Liver/immunology , Liver/metabolism , Macrophages/metabolism , Male , Membrane Potential, Mitochondrial , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Annotation , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/immunology , Non-alcoholic Fatty Liver Disease/pathology , Reactive Oxygen Species/metabolism , Receptors, CCR5/metabolism
9.
J Dermatol Sci ; 74(3): 204-13, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24702853

ABSTRACT

BACKGROUND: Impaired wound healing in skin ulcer is one of the major medical issues in the aged society. Wound healing is a complex process orchestrated by a number of humoral factors and cellular components. TGF-ß is known to stimulate collagen production in dermal fibroblasts while inhibiting proliferation of epidermal keratinocyte. A screening of small compounds that suppress type I collagen production in fibroblasts has identified HSc025 that antagonizes the TGF-ß/Smad signal. OBJECTIVE: We examined the effects of HSc025 on dermal wound healing and elucidated the underlying mechanisms. METHODS: Effects of HSc025 on the wound closure process were evaluated in a murine full-thickness excisional wound healing model. Cell proliferation and migration were estimated using primary cultures of human keratinocytes and fibroblasts. Comprehensive analyses of gene expression profiles were performed using untreated and HSc025-treated fibroblasts. RESULTS: Oral HSc025 administration suppressed macrophage infiltration and accelerated wound closure as early as at day 2 after the dermal excision. Treatment of cultured keratinocytes with HSc025 counteracted the inhibitory effects of TGF-ß on cell proliferation and migration. On the other hand, HSc025 stimulated migration, but not proliferation, of dermal fibroblasts independently of TGF-ß. Experiments using an artificial dermis graft revealed that HSc025 stimulated migration of collagen-producing cells into the graft tissue. A cDNA microarray analysis of untreated and HSc025-treated fibroblasts identified pirin as a critical mediator accelerating fibroblast migration. CONCLUSION: HSc025 accelerates wound healing by modifying infiltration, proliferation and migration of distinct cellular components, which provides a novel insight into the therapy for intractable skin ulcer.


Subject(s)
Alkadienes/therapeutic use , Skin Ulcer/drug therapy , Wound Healing/drug effects , Alkadienes/pharmacology , Animals , Carrier Proteins/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Collagen Type I/genetics , Collagen Type I/metabolism , Dioxygenases , Drug Evaluation, Preclinical , Female , Fibroblasts/drug effects , Granulation Tissue/cytology , Humans , Keratinocytes/drug effects , Mice , Nuclear Proteins/metabolism , Transforming Growth Factor beta
10.
J Invest Dermatol ; 131(2): 529-36, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20962852

ABSTRACT

Recent studies show that bone marrow (BM)-derived cells migrating into a dermal wound promote healing by producing collagen type I. However, their contribution to the repair process has not been fully verified yet. It is also unclear whether BM-derived cells participate in dermal fibrogenesis. We have addressed these issues using transgenic mice that harbor tissue-specific enhancer/promoter sequences of α2(I) collagen gene linked to either enhanced green fluorescent protein (COL/EGFP) or the luciferase (COL/LUC) reporter gene. Following dermal excision or subcutaneous bleomycin administration, a large number of EGFP-positive collagen-producing cells appeared in the dermis of COL/EGFP reporter mice. When wild-type mice were transplanted with BM cells from transgenic COL/EGFP animals and subjected to dermal excision, no EGFP-positive BM-derived collagen-producing cells were detected throughout the repair process. Luciferase assays of dermal tissues from COL/LUC recipient mice also excluded collagen production by BM-derived cells during dermal excision healing. In contrast, a limited but significant number of CD45-positive collagen-producing cells migrated from BM following bleomycin injection. These results indicate that resident cells in the skin are the major source of de novo collagen deposition in both physiological and pathological conditions, whereas BM-derived cells participate, in part, in collagen production during dermal fibrogenesis.


Subject(s)
Bone Marrow Cells/cytology , Collagen/metabolism , Dermis/pathology , Wound Healing/physiology , Animals , Bone Marrow Transplantation , Cell Movement/physiology , Collagen Type I , Dermis/injuries , Dermis/metabolism , Fibrosis/physiopathology , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Promoter Regions, Genetic/physiology
11.
Gastroenterology ; 137(4): 1459-66.e1, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19596008

ABSTRACT

BACKGROUND & AIMS: Recent studies have reported that bone marrow (BM)-derived cells migrating into fibrotic liver tissue exhibit a myofibroblast-like phenotype and may participate in the progression of liver fibrosis. However, their contribution to collagen production has not been fully verified yet. We revisited this issue by using 2 mechanistically distinct liver fibrosis models introduced into transgenic collagen reporter mice and their BM recipients. METHODS: BM of wild-type mice was replaced by cells obtained from transgenic animals harboring tissue-specific enhancer/promoter sequences of alpha2(I) collagen gene (COL1A2) linked to enhanced green fluorescent protein (EGFP) or firefly luciferase (LUC) gene. Liver fibrosis was introduced into those mice by repeated carbon tetrachloride injections or ligation of the common bile duct. Activation of COL1A2 promoter was assessed by confocal microscopic examination detecting EGFP signals and luciferase assays of liver homogenates. RESULTS: The tissue-specific COL1A2 enhancer/promoter was activated in hepatic stellate cells following a single carbon tetrachloride injection or during primary culture on plastic. A large number of EGFP-positive collagen-expressing cells were observed in liver tissue of transgenic COL1A2/EGFP mice in both liver fibrosis models. In contrast, there were few EGFP-positive BM-derived collagen-producing cells detected in fibrotic liver tissue of COL1A2/EGFP recipients. Luciferase assays of liver tissues from COL1A2/LUC-recipient mice further indicated that BM-derived cells produced little collagen in response to fibrogenic stimuli. CONCLUSIONS: By using a specific and sensitive experimental system, which detects exclusively BM-derived collagen-producing cells, we conclude an unexpectedly limited role of BM-derived cells in collagen production during hepatic fibrogenesis.


Subject(s)
Bone Marrow Cells/metabolism , Collagen/metabolism , Hepatic Stellate Cells/metabolism , Liver Cirrhosis, Experimental/metabolism , Liver/metabolism , Animals , Bone Marrow Transplantation , Carbon Tetrachloride , Cell Differentiation , Cell Movement , Cells, Cultured , Collagen/genetics , Collagen Type I , Common Bile Duct/surgery , Disease Progression , Genes, Reporter , Green Fluorescent Proteins/genetics , Hepatic Stellate Cells/pathology , Ligation , Liver/pathology , Liver Cirrhosis, Experimental/etiology , Liver Cirrhosis, Experimental/pathology , Luciferases, Firefly/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , Time Factors
12.
Life Sci ; 83(15-16): 531-9, 2008 Oct 10.
Article in English | MEDLINE | ID: mdl-18771671

ABSTRACT

AIMS: Glycyrrhizin has been widely used for the treatment of chronic hepatitis C. It decreases the serum levels of aminotransferases, and suppresses progression of liver fibrosis as well as subsequent occurrence of hepatocellular carcinoma. Although previous studies have shown that glycyrrhizin and its metabolite inhibit collagen gene expression, its underlying mechanisms are virtually unknown. This study was aimed to explore molecular mechanisms responsible for the inhibitory effect of glycyrrhizin on type I collagen gene transcription. MAIN METHODS: Effects of glycyrrhizin and its metabolite, glycyrrhetinic acid, on collagen promoter activity were examined by using transgenic reporter mice harboring alpha2(I) collagen gene (COL1A2) promoter. Their effects on the TGF-beta/Smad signaling pathway were studied by cell transfection assays and immunofluorescence studies using cultured hepatic stellate cells. KEY FINDINGS: Administration of glycyrrhizin or its metabolite, glycyrrhetinic acid, significantly suppressed COL1A2 promoter activation and progression of liver fibrosis induced by repeated carbon tetrachloride injections. In cultured hepatic stellate cells, glycyrrhetinic acid, but not glycyrrhizin, inhibited type I collagen synthesis mostly at the level of gene transcription. This inhibitory effect of glycyrrhetinic acid was abolished by a mutation introduced into a Smad3-binding region within the COL1A2 promoter. Glycyrrhetinic acid did not affect gene expression of TGF-beta receptors or Smad proteins, but inhibited nuclear accumulation of Smad3 in activated hepatic stellate cells. In addition to those direct inhibitory effects on COL1A2 transcription, glycyrrhetinic acid also suppressed activation of quiescent hepatic stellate cells in primary culture. SIGNIFICANCE: The results provide a molecular basis for the anti-fibrotic effect of glycyrrhizin treatment.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Collagen Type I/genetics , Glycyrrhizic Acid/pharmacology , Liver Cirrhosis/prevention & control , Smad3 Protein/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/metabolism , Blotting, Western , Carbon Tetrachloride Poisoning/pathology , Carbon Tetrachloride Poisoning/prevention & control , Carcinoma, Hepatocellular/prevention & control , Cells, Cultured , Fluorescent Antibody Technique , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Glycyrrhizic Acid/metabolism , Humans , Indicators and Reagents , Luciferases/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , Mutant Chimeric Proteins/metabolism , Promoter Regions, Genetic/drug effects , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Transcription, Genetic
13.
Gastroenterology ; 134(4): 1180-90, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18395096

ABSTRACT

BACKGROUND & AIMS: Hepatocyte growth factor (HGF) and transforming growth factor-beta (TGF-beta) regulate diversified cellular functions and often act antagonistically against each other. For example, TGF-beta is the most potent factor accelerating liver fibrosis, whereas HGF treatment prevents its progression. Here, we propose a novel molecular mechanism by which HGF counter represses TGF-beta-stimulated profibrogenic signal transduction. METHODS: Effects of HGF on TGF-beta-responsive gene transcription of type I collagen, the major matrix component of fibrotic liver, were examined by using cultured hepatic stellate cells (HSC) and transgenic mice harboring alpha2(I) collagen gene (COL1A2) promoter. Expression and subcellular localization of Smad3 were determined by Western blot analyses and immunofluorescence staining, respectively. A mass spectrometric analysis was employed to identify immunoprecipitated proteins with antiphospho-Smad2/3 antibodies. RESULTS: Over expression of HGF inhibited COL1A2 transcription in cultured HSC and suppressed activation of COL1A2 promoter in liver tissue induced by carbon tetrachloride administration. A mass spectrometric analysis identified galectin-7 as one of the immunoprecipitated proteins with antiphospho-Smad2/3 antibodies following HGF treatment. HGF accelerated nuclear export of Smad3 by enhancing its interaction with galectin-7. Transfection of cells with galectin-7 small interfering RNA inhibited nuclear export of Smad3 and abolished suppressive effect of HGF on expression of TGF-beta-responsive genes such as COL1A2 and plasminogen activator inhibitor-1. On the other hand, over expression of galectin-7 suppressed TGF-beta-stimulated expression of those target genes. CONCLUSIONS: These results reveal a novel function of intracellular galectin-7 as a transcriptional regulator via its interaction with Smad3 and provide a molecular basis for the antifibrotic effect of HGF.


Subject(s)
Galectins/genetics , Hepatocyte Growth Factor/therapeutic use , Liver Cirrhosis, Experimental/prevention & control , Signal Transduction/drug effects , Smad3 Protein/genetics , Animals , Antibodies/analysis , Blotting, Western , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cells, Cultured , Collagen/biosynthesis , Collagen/genetics , Collagen Type I , Disease Progression , Enzyme Activation/drug effects , Flavonoids/pharmacology , Fluorescent Antibody Technique , Galectins/biosynthesis , Gene Expression , Hepatocyte Growth Factor/biosynthesis , Hepatocyte Growth Factor/genetics , Immunoprecipitation , Liver Cirrhosis, Experimental/genetics , Liver Cirrhosis, Experimental/metabolism , Mass Spectrometry , Mice , Mice, Transgenic , Microscopy, Confocal , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Plasminogen Activator Inhibitor 1/biosynthesis , Plasminogen Activator Inhibitor 1/genetics , Promoter Regions, Genetic/drug effects , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction , Smad3 Protein/biosynthesis , Smad3 Protein/immunology , Transcription, Genetic/drug effects
14.
Hepatology ; 45(1): 213-22, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17187438

ABSTRACT

UNLABELLED: Liver fibrosis is usually progressive, but it can occasionally be reversible if the causative agents are adequately removed or if patients are treated effectively. However, molecular mechanisms responsible for this reversibility of liver fibrosis have been poorly understood. To reveal the contribution of bone marrow (BM)-derived cells to the spontaneous regression of liver fibrosis, mice were treated with repeated carbon tetrachloride injections after hematopoietic reconstitution with enhanced green fluorescent protein (EGFP)-expressing BM cells. The distribution and characteristics of EGFP-positive (EGFP(+)) cells present in fibrotic liver tissue were examined at different time points after cessation of carbon tetrachloride intoxication. A large number of EGFP(+) cells were observed in liver tissue at peak fibrosis, which decreased during the recovery from liver fibrosis. Some of them, as well as EGFP-negative (EGFP(-)) liver resident cells, expressed matrix metalloproteinase (MMP)-13 and MMP-9. Whereas MMP-13 was transiently expressed mainly in the cells clustering in the periportal areas, MMP-9 expression and enzymatic activity were detected over the resolution process in several different kinds of cells located in the portal areas and along the fibrous septa. Therapeutic recruitment of BM cells by granulocyte colony-stimulating factor (G-CSF) treatment significantly enhanced migration of BM-derived cells into fibrotic liver and accelerated the regression of liver fibrosis. Experiments using transgenic mice overexpressing hepatocyte growth factor (HGF) indicated that G-CSF and HGF synergistically increased MMP-9 expression along the fibrous septa. CONCLUSION: Autologous BM cells contribute to the spontaneous regression of liver fibrosis, and their therapeutic derivation could be a new treatment strategy for intractable liver fibrosis.


Subject(s)
Bone Marrow Cells/metabolism , Liver Cirrhosis/metabolism , Liver/metabolism , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 9/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Transplantation/methods , Carbon Tetrachloride , Cell Differentiation , Cell Movement , Cell- and Tissue-Based Therapy/methods , Fluorescent Antibody Technique , Gene Expression Regulation/drug effects , Granulocyte Colony-Stimulating Factor/pharmacology , Green Fluorescent Proteins , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Liver/pathology , Liver/physiopathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Liver Cirrhosis/therapy , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Remission, Spontaneous
SELECTION OF CITATIONS
SEARCH DETAIL
...