Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Clin J Gastroenterol ; 14(1): 330-335, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33038000

ABSTRACT

We report a case of primary sclerosing cholangitis (PSC) with autoimmune hemolytic anemia (AIHA). A 47-year-old woman was diagnosed with PSC. One year later, she was admitted to our hospital for jaundice and fatigue. Magnetic resonance cholangiopancreatography (MRCP) showed worsening of the biliary stricture, and rapidly progressive anemia developed simultaneously. Based on the various laboratory findings, she was diagnosed with AIHA. The administration of prednisolone improved not only the anemia but also the biliary stricture. This case is impactful, as there are few case reports of PSC with AIHA. In addition, we were able to observe the changes in imaging findings using MRCP over time.


Subject(s)
Anemia, Hemolytic, Autoimmune , Cholangitis, Sclerosing , Anemia, Hemolytic, Autoimmune/complications , Anemia, Hemolytic, Autoimmune/drug therapy , Cholangiopancreatography, Magnetic Resonance , Cholangitis, Sclerosing/complications , Cholangitis, Sclerosing/drug therapy , Female , Humans , Middle Aged , Prednisolone/therapeutic use
3.
Int J Mol Med ; 46(1): 427-438, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32377696

ABSTRACT

The cytokine transforming growth factor­ß (TGF­ß) serves a key role in hepatic fibrosis and has cytostatic effects on hepatocytes. The present study investigated the anti­fibrogenic and regenerative effects of the TGF­ß receptor type I kinase inhibitor galunisertib (LY2157299) in mice with carbon tetrachloride (CCl4)­induced liver cirrhosis and in vitro. Mice were intraperitoneally treated with CCl4 for 8 weeks. At week 5, the mice were divided randomly into four treatment groups: Vehicle­treated; and treated with low­; middle­; and high­dose galunisertib, which was administered from weeks 5­8. The mice were sacrificed after 8 weeks of CCl4 treatment. Liver fibrosis, as evaluated by histology and determination of hydroxyproline content, progressed during week 4­8 of CCl4 treatment in the vehicle­treated mice. Galunisertib treatment dose­dependently prevented liver fibrosis, as demonstrated by the direct inhibition of α­smooth muscle actin­positive activated hepatic stellate cells (HSCs) after 8 weeks of CCl4 treatment. The levels of active matrix metalloproteinase (MMP)­9 in galunisertib­treated livers were significantly increased compared with the vehicle­treated livers. In the high­dose group, the number of PCNA­positive hepatocytes and endothelial cells markedly increased compared with the vehicle group. Reverse transcription­quantitative PCR analysis verified that interleukin­6 and epiregulin expression levels were significantly increased in livers from the group treated with high­dose galunisertib compared with the vehicle­treated group. Galunisertib inhibited the proliferation of activated HSCs and collagen synthesis in addition to restoring MMP activity. Moreover, galunisertib promoted liver remodeling by proliferating hepatocytes and vascular endothelial cells, while significantly increasing liver weight. These results are consistent with the cytostatic action of TGF­ß that negatively regulates liver regeneration, and demonstrated that galunisertib inhibited TGF­ß signaling, halted liver fibrosis progression and promoted hepatic regeneration. The results of the present study suggest that galunisertib may be an effective treatment for liver cirrhosis.


Subject(s)
Carbon Tetrachloride/toxicity , Liver Regeneration/drug effects , Pyrazoles/therapeutic use , Quinolines/therapeutic use , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Animals , Cell Line , Hep G2 Cells , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice
4.
Med Mol Morphol ; 52(4): 198-208, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30805710

ABSTRACT

Cancer cells surviving in ascites exhibit cancer stem cell (CSC)-like features. This study analyzed the expression of the CSC marker CD133 in the ascites-derived exosomes obtained from patients with unresectable pancreatic cancer. In addition, inverse correlation of CD133 expression with prognosis was examined. Of the 133 consecutive patients, 19 patients were enrolled in the study. Exosomes derived from the malignant ascites demonstrated higher density and wider variation in size than those from non-malignant ascites. Western blot revealed enhanced expression of CD133 in exosomes obtained from patients with pancreatic cancer compared to those obtained from patients with gastric cancer or liver cirrhosis. A xenograft mouse model with malignant ascites was established by intraperitoneal inoculation of human pancreatic cancer cells in nude mice. Results obtained from the human study were reproduced in the mouse model. Statistically significant equilateral correlation was identified between the band intensity of CD133 in western blot and overall survival of patients. Lectin microarray analyses revealed glycosylation of CD133 by sialic acids as the major glycosylation among diverse others responsible for the glycosylation of exosomal CD133. These findings suggest that highly glycosylated CD133 in ascites-derived exosomes as a potential biomarker for better prognosis of patients with advanced pancreatic cancer.


Subject(s)
AC133 Antigen/metabolism , Ascites/metabolism , Biomarkers, Tumor/metabolism , Exosomes/metabolism , Pancreatic Neoplasms/metabolism , Animals , Cell Line, Tumor , Glycosylation , HeLa Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/metabolism , PC-3 Cells , Prognosis , Stomach Neoplasms/metabolism
5.
Cancer Sci ; 109(9): 2801-2810, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29981246

ABSTRACT

CD44v9 is expressed in cancer stem cells (CSC) and stabilizes the glutamate-cystine transporter xCT on the cytoplasmic membrane, thereby decreasing intracellular levels of reactive oxygen species (ROS). This mechanism confers ROS resistance to CSC and CD44v9-expressing cancer cells. The aims of the present study were to assess: (i) expression status of CD44v9 and xCT in hepatocellular carcinoma (HCC) tissues, including those derived from patients treated with hepatic arterial infusion chemoembolization (HAIC) therapy with cisplatin (CDDP); and (ii) whether combination of CDDP with sulfasalazine (SASP), an inhibitor of xCT, was more effective on tumor cells than CDDP alone by inducing ROS-mediated apoptosis. Twenty non-pretreated HCC tissues and 7 HCC tissues administered HAIC therapy with CDDP before surgical resection were subjected to immunohistochemistry analysis of CD44v9 and xCT expression. Human HCC cell lines HAK-1A and HAK-1B were used in this study; the latter was also used for xenograft experiments in nude mice to assess in vivo efficacy of combination treatment. CD44v9 positivity was significantly higher in HAIC-treated tissues (5/7) than in non-pretreated tissues (2/30), suggesting the involvement of CD44v9 in the resistance to HAIC. xCT was significantly expressed in poorly differentiated HCC tissues. Combination treatment effectively killed the CD44v9-harboring HAK-1B cells through ROS-mediated apoptosis and significantly decreased xenografted tumor growth. In conclusion, the xCT inhibitor SASP augmented ROS-mediated apoptosis in CDDP-treated HCC cells, in which the CD44v9-xCT system functioned. As CD44v9 is typically expressed in HAIC-resistant HCC cells, combination treatment with SASP with CDDP may overcome such drug resistance.


Subject(s)
Amino Acid Transport System y+/physiology , Carcinoma, Hepatocellular/drug therapy , Hyaluronan Receptors/physiology , Liver Neoplasms/drug therapy , Sulfasalazine/pharmacology , Aged , Aged, 80 and over , Amino Acid Transport System y+/analysis , Animals , Apoptosis/drug effects , Carcinoma, Hepatocellular/chemistry , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Female , Hep G2 Cells , Humans , Hyaluronan Receptors/analysis , Liver Neoplasms/chemistry , Male , Mice , Mice, Inbred BALB C , Middle Aged , Reactive Oxygen Species/metabolism
6.
Cell Metab ; 28(1): 104-117.e5, 2018 Jul 03.
Article in English | MEDLINE | ID: mdl-29861385

ABSTRACT

Intrinsic and evasive antiangiogenic drug (AAD) resistance is frequently developed in cancer patients, and molecular mechanisms underlying AAD resistance remain largely unknown. Here we describe AAD-triggered, lipid-dependent metabolic reprogramming as an alternative mechanism of AAD resistance. Unexpectedly, tumor angiogenesis in adipose and non-adipose environments is equally sensitive to AAD treatment. AAD-treated tumors in adipose environment show accelerated growth rates in the presence of a minimal number of microvessels. Mechanistically, AAD-induced tumor hypoxia initiates the fatty acid oxidation metabolic reprogramming and increases uptake of free fatty acid (FFA) that stimulates cancer cell proliferation. Inhibition of carnitine palmitoyl transferase 1A (CPT1) significantly compromises the FFA-induced cell proliferation. Genetic and pharmacological loss of CPT1 function sensitizes AAD therapeutic efficacy and enhances its anti-tumor effects. Together, we propose an effective cancer therapy concept by combining drugs that target angiogenesis and lipid metabolism.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Carnitine O-Palmitoyltransferase/metabolism , Drug Resistance, Neoplasm , Fatty Acids/metabolism , Neoplasms , Neovascularization, Pathologic/metabolism , Adipose Tissue/metabolism , Angiogenesis Inhibitors/therapeutic use , Animals , Carnitine O-Palmitoyltransferase/genetics , Cell Line, Tumor , Lipid Metabolism , Mice , Mice, Inbred C57BL , Neoplasms/drug therapy , Neoplasms/pathology
7.
J Exp Med ; 215(2): 611-626, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29305395

ABSTRACT

Angiogenesis plays an instrumental role in the modulation of adipose tissue mass and metabolism. Targeting adipose vasculature provides an outstanding opportunity for treatment of obesity and metabolic disorders. Here, we report the physiological functions of VEGFR1 in the modulation of adipose angiogenesis, obesity, and global metabolism. Pharmacological inhibition and genetic deletion of endothelial VEGFR1 augmented adipose angiogenesis and browning of subcutaneous white adipose tissue, leading to elevated thermogenesis. In a diet-induced obesity model, endothelial-VEGFR1 deficiency demonstrated a potent anti-obesity effect by improving global metabolism. Along with metabolic changes, fatty liver and insulin sensitivity were also markedly improved in VEGFR1-deficient high fat diet (HFD)-fed mice. Together, our data indicate that targeting of VEGFR1 provides an exciting new opportunity for treatment of obesity and metabolic diseases, such as liver steatosis and type 2 diabetes.


Subject(s)
Adipose Tissue/blood supply , Adipose Tissue/metabolism , Endothelium, Vascular/metabolism , Metabolic Diseases/therapy , Vascular Endothelial Growth Factor Receptor-1/deficiency , Adipose Tissue, Brown/blood supply , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/blood supply , Adipose Tissue, White/metabolism , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Metabolic Diseases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic , Obesity/etiology , Obesity/metabolism , Obesity/therapy , Thermogenesis , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism
8.
World J Gastroenterol ; 23(37): 6833-6844, 2017 Oct 07.
Article in English | MEDLINE | ID: mdl-29085226

ABSTRACT

AIM: To elucidate the role of STAT3 in hepatocarcinogenesis and biliary ductular proliferation following chronic liver injury. METHODS: We investigated thioacetamide (TAA)-induced liver injury, compensatory hepatocyte proliferation, and hepatocellular carcinoma (HCC) development in hepatic STAT3-deficient mice. In addition, we evaluated TAA-induced biliary ductular proliferation and analyzed the activation of sex determining region Y-box9 (SOX9) and Yes-associated protein (YAP), which regulate the transdifferentiation of hepatocytes to cholangiocytes. RESULTS: Both compensatory hepatocyte proliferation and HCC formation were significantly decreased in hepatic STAT3-deficient mice as compared with control mice. STAT3 deficiency resulted in augmentation of hepatic necrosis and fibrosis. On the other hand, biliary ductular proliferation increased in hepatic STAT3-deficient livers as compared with control livers. SOX9 and YAP were upregulated in hepatic STAT3-deficient hepatocytes. CONCLUSION: STAT3 may regulate hepatocyte proliferation as well as transdifferentiation into cholangiocytes and serve as a therapeutic target for HCC inhibition and biliary regeneration.


Subject(s)
Biliary Tract/physiology , Carcinogenesis/pathology , Carcinoma, Hepatocellular/pathology , Chemical and Drug Induced Liver Injury/pathology , Liver Neoplasms, Experimental/pathology , Liver Neoplasms/pathology , Regeneration , STAT3 Transcription Factor/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Biliary Tract/cytology , Carcinoma, Hepatocellular/chemically induced , Cell Cycle Proteins , Cell Proliferation , Cell Transdifferentiation , Chemical and Drug Induced Liver Injury/etiology , Hepatocytes/physiology , Liver/drug effects , Liver/pathology , Liver Neoplasms/chemically induced , Liver Neoplasms, Experimental/chemically induced , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphoproteins/metabolism , Phosphorylation , SOX9 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Thioacetamide/toxicity , Up-Regulation , YAP-Signaling Proteins
9.
Dig Dis Sci ; 62(6): 1527-1536, 2017 06.
Article in English | MEDLINE | ID: mdl-28365916

ABSTRACT

BACKGROUND AND AIMS: Pigment epithelium-derived factor (PEDF) has been shown to be a potent inhibitor of inflammation through its anti-oxidative property. Since oxidative response is considered to play the pivotal role of the development and progression of nonalcoholic steatohepatitis (NASH), it is conceivable that PEDF may play a protective role against NASH. In this study, we examined whether administration of PEDF slowed the progression of NASH in mice models. METHODS: Mice were fed methionine- and choline-deficient (MCD) diet with or without intramuscular administration of adenovirus-expressing PEDF (Ad-PEDF). Effects of PEDF administration on NASH were histologically and biochemically evaluated. RESULTS: Administration of Ad-PEDF significantly decreased hepatic fat storage as well as serum levels of ALT in MCD diet-fed mice. Dihydroethidium staining showed that MCD diet-triggered oxidative stress was reduced in the liver of Ad-PEDF-administered mice compared to that of PBS- or Ad-LacZ-administered mice. Activation of Kupffer cells and hepatic fibrosis was also inhibited by Ad-PEDF administration. Quantitative real-time RT-PCR revealed that MCD diet up-regulated expressions of TNF-α, IL-1ß, IL-6, TGF-ß, collagen-1, and collagen-3 mRNA, which were also attenuated with Ad-PEDF administration, whereas MCD diet-induced down-regulation of expressions of PPAR-γ mRNA was restored with Ad-PEDF administration. Furthermore, immunoblotting analysis showed that MCD diet-induced up-regulation of NADPH oxidase components was significantly decreased in Ad-PEDF-administered mice. CONCLUSIONS: The present results demonstrated for the first time that PEDF could slow the development and progression of steatohepatitis through the suppression of steatosis and inflammatory response in MCD diet-fed mice. Our study suggests that PEDF supplementation may be a novel therapeutic strategy for the treatment of NASH.


Subject(s)
Adipose Tissue/pathology , Eye Proteins/pharmacology , Fatty Liver/metabolism , Fatty Liver/prevention & control , Nerve Growth Factors/pharmacology , Protease Inhibitors/pharmacology , RNA, Messenger/metabolism , Serpins/pharmacology , Adenoviridae/genetics , Alanine Transaminase/blood , Animals , Choline Deficiency/complications , Collagen Type I/genetics , Collagen Type III/genetics , Diet , Disease Models, Animal , Down-Regulation , Eye Proteins/genetics , Fatty Liver/genetics , Fatty Liver/pathology , Injections, Intramuscular , Interleukin-1beta/genetics , Interleukin-6/genetics , Kupffer Cells , Liver Cirrhosis/prevention & control , Male , Methionine/administration & dosage , Methionine/deficiency , Mice , Mice, Inbred C57BL , NADPH Oxidases/metabolism , Nerve Growth Factors/genetics , Oxidative Stress , PPAR gamma/genetics , Serpins/genetics , Tumor Necrosis Factor-alpha/genetics , Up-Regulation
10.
Mol Cancer Res ; 15(6): 744-752, 2017 06.
Article in English | MEDLINE | ID: mdl-28179411

ABSTRACT

Doublecortin-like kinase 1 (DCLK1), a marker for intestinal and pancreatic cancer stem cells, is highly expressed in neuroblastomas. This study was conducted to assess DCLK1 expression levels in pancreatic neuroendocrine tumor (PNET) tissues and to explore the roles of this molecule in clinical tissue from multiple PNET patients, cells (BON1, QGP1, and CM) and tumor xenografts. Immunohistochemically, all PNET tissues highly and diffusely expressed DCLK1 as a full-length isoform, identical to that detected in primary liver NETs. A DCLK1-overexpressing PNET cell line (QGP1-DCLK1) exhibited epithelial-mesenchymal transition (EMT)-related gene signatures, and robust upregulation of Slug (SNAI2), N-Cadherin (CDH2), and Vimentin (VIM) was validated by real-time PCR and immunoblotting. QGP1-DCLK1 cells had increased cell migration in a wound-healing assay and formed significantly larger xenograft tumors in nude mice. The factors involved in the formation of the fast-growing tumors included p-FAK (on Tyr925), p-ERK1/2, p-AKT, Paxillin, and Cyclin D1, which upon knockdown or pharmacologic inhibition of DCLK1 abolished the expression of these molecules. In conclusion, robust and ubiquitous expression of DCLK1 was first demonstrated here in human PNET tissue specimens and cells. DCLK1 characterized the PNET cell behavior, inducing p-FAK/SLUG-mediated EMT. These findings suggest the possibility of developing novel therapeutic strategies against PNETs by targeting DCLK1.Implications: Evidence here reveals that human PNETs diffusely and robustly express the cancer stem cell marker DCLK1, which drives SLUG-mediated EMT, and suggests that NETs share biological features for druggable targets with other tumors, including neuroblastoma that also highly expresses DCLK1. Mol Cancer Res; 15(6); 744-52. ©2017 AACR.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Biomarkers, Tumor/genetics , Cell Line, Tumor , Doublecortin-Like Kinases , Female , Focal Adhesion Kinase 1/metabolism , Humans , Male , Mice, Inbred BALB C , Middle Aged , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neuroendocrine Tumors/genetics , Pancreatic Neoplasms/genetics , Tyrosine/metabolism , Xenograft Model Antitumor Assays
11.
Nat Commun ; 7: 12680, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27580750

ABSTRACT

The impact of discontinuation of anti-VEGF cancer therapy in promoting cancer metastasis is unknown. Here we show discontinuation of anti-VEGF treatment creates a time-window of profound structural changes of liver sinusoidal vasculatures, exhibiting hyper-permeability and enlarged open-pore sizes of the fenestrated endothelium and loss of VE-cadherin. The drug cessation caused highly leaky hepatic vasculatures permit tumour cell intravasation and extravasation. Discontinuation of an anti-VEGF antibody-based drug and sunitinib markedly promotes liver metastasis. Mechanistically, host hepatocyte, but not tumour cell-derived vascular endothelial growth factor (VEGF), is responsible for cancer metastasis. Deletion of hepatocyte VEGF markedly ablates the 'off-drug'-induced metastasis. These findings provide mechanistic insights on anti-VEGF cessation-induced metastasis and raise a new challenge for uninterrupted and sustained antiangiogenic therapy for treatment of human cancers.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Indoles/therapeutic use , Liver/blood supply , Liver/pathology , Neoplasms/pathology , Pyrroles/therapeutic use , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Female , Humans , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Metastasis/pathology , Neoplasms/drug therapy , Sunitinib , Vascular Endothelial Growth Factor A/genetics
12.
Neoplasia ; 18(7): 413-24, 2016 07.
Article in English | MEDLINE | ID: mdl-27435924

ABSTRACT

BACKGROUND & AIM: Aflibercept known as ziv-aflibercept in the United States is a soluble decoy receptor of both vascular endothelial growth factor (VEGF) receptor-1 and -2 known to inhibit the binding of VEGF and placental growth factor (PlGF) to VEGF receptor-1 and -2. Here, we analyzed the mechanisms of the antitumor effects of aflibercept in mouse hepatoma models. METHODS: In in vitro studies, we determined the effects of aflibercept on human umbilical vein cell (HUVEC) proliferation and bone marrow (BM) cell differentiation to endothelial progenitor cells (EPCs). In in vivo experiments, aflibercept was injected intraperitoneally in hepatoma cell tumor-bearing mice, and its inhibitory effects on tumor growth and BM cell migration to tumor tissues were evaluated. RESULTS: Aflibercept suppressed phosphorylation of VEGF receptor-1 and -2 in HUVEC and dose-dependently inhibited VEGF-induced HUVEC proliferation. It suppressed the differentiation of BM cells to EPCs and migration of BM cells to tumor tissues. It also suppressed tumor growth and prolonged survival time of tumor-bearing mice without side effects. In tumor tissues, aflibercept upregulated the expression of hypoxia inducible factor1-α, VEGF, PlGF, fibroblast growth factor-2, platelet derived growth factor-BB, and transforming growth factor-α and reduced microvascular density. It also reduced sinusoidal density in noncancerous liver tissues. CONCLUSIONS: Our results demonstrated potent antitumor activity for aflibercept in a mouse model of hepatocellular carcinoma. These effects were mediated through inhibition of neovascularization, caused by inhibition of endothelial cell proliferation, EPC differentiation, and BM cell migration to tumor tissues.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Recombinant Fusion Proteins/pharmacology , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Bone Marrow Cells/cytology , Carcinoma, Hepatocellular/pathology , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Endothelial Progenitor Cells/cytology , Human Umbilical Vein Endothelial Cells/cytology , Humans , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Receptors, Vascular Endothelial Growth Factor
13.
Mol Ther Methods Clin Dev ; 3: 16025, 2016.
Article in English | MEDLINE | ID: mdl-27162932

ABSTRACT

Ex vivo expansion of autologous cells is indispensable for cell transplantation therapy of patients with liver cirrhosis. The aim of this study was to investigate the efficacy of human ex vivo-expanded CD34(+) cells for treatment of cirrhotic rat liver. Recipient rats were intraperitoneally injected with CCl4 twice weekly for 3 weeks before administration of CD34(+) cells. CCl4 was then re-administered twice weekly for 3 more weeks, and the rats were sacrificed. Saline, nonexpanded or expanded CD34(+) cells were injected via the spleen. After 7 days, CD34(+) cells were effectively expanded in a serum-free culture medium. Expanded CD34(+) cells were also increasingly positive for cell surface markers of VE-cadherin, VEGF receptor-2, and Tie-2. The expression of proangiogenic growth factors and adhesion molecules in expanded CD34(+) cells increased compared with nonexpanded CD34(+) cells. Expanded CD34(+) cell transplantation reduced liver fibrosis, with a decrease of αSMA(+) cells. Assessments of hepatocyte and sinusoidal endothelial cell proliferative activity indicated the superior potency of expanded CD34(+) cells over non-expanded CD34(+) cells. The inhibition of integrin αvß3 and αvß5 disturbed the engraftment of transplanted CD34(+) cells and aggravated liver fibrosis. These findings suggest that expanded CD34(+) cells enhanced the preventive efficacy of cell transplantation in a cirrhotic model.

14.
Oncol Lett ; 10(4): 2015-2020, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26622789

ABSTRACT

Doublecortin-like kinase 1 (DCLK1), a microtubule-associated protein, is known to regulate neuronal differentiation, migration and neurogenesis. Recent evidence suggests that the protein is a putative marker for intestinal and pancreatic stem cells, including their cancer stem cell counterparts. The present study conducted immunohistochemical analyses for DCLK1 and the stemness marker, NANOG, in human intestinal neuroendocrine tumors (NETs), as their expression had not been previously investigated in these tumors. Eighteen patients with endoscopically resected rectal NETs were enrolled in the study. The mean age of the patients was 51 years old. The mean diameter of the resected tumors was 5.2 mm, and a histological diagnosis of NET grade G1 was formed for all tumors. Immunohistochemical analysis was performed not only for DCLK1, but also for the known NET markers, synaptophysin, chromogranin A and cluster of differentiation (CD)56. The intensity and distribution of staining were scored on a scale of 0-3 and 0-2, respectively. The sum of the scores was calculated for each specimen. Co-expression of DCLK1 and NANOG was also examined. The mean scores for DCLK1 and synaptophysin were significantly higher than those for chromogranin A (P<0.0001) and CD56 (P<0.01). There were no significant differences in the scores between DCLK1 and synaptophysin or between chromogranin A and CD56. Notably, NANOG was expressed in high quantities in all the tumor tissues studied, showing clear co-expression with DCLK1. In conclusion, DCLK1 may be a novel marker for rectal NET, potentially indicating the presence of the stemness gene product, NANOG.

15.
Mol Ther Oncolytics ; 2: 15020, 2015.
Article in English | MEDLINE | ID: mdl-27119112

ABSTRACT

"Angiogenic switch off" is one of the ideal therapeutic concepts in the treatment of cancer. However, the specific molecules which can induce "angiogenic switch off" in tumor have not been identified yet. In this study, we focused on von Hippel-Lindau protein (pVHL) in hepatocellular carcinoma (HCC) and investigated the effects of sulfoquinovosyl-acylpropanediol (SQAP), a novel synthetic sulfoglycolipid, for HCC. We examined mutation ratio of VHL gene in HCC using 30 HCC samples and we treated the HCC-implanted mice with SQAP. Thirty clinical samples showed no VHL genetic mutation in HCC. SQAP significantly inhibited tumor growth by inhibiting angiogenesis in a hepatoma mouse model. SQAP induced tumor "angiogenic switch off" by decreasing hypoxia-inducible factor (HIF)-1, 2α protein via pVHL upregulation. pVHL upregulation decreased HIFα protein levels through different multiple mechanisms: (i) increasing pVHL-dependent HIFα protein degradation; (ii) decreasing HIFα synthesis with decrease of NF-κB expression; and (iii) decrease of tumor hypoxia by vascular normalization. We confirmed these antitumor effects of SQAP by the loss-of-function experiments. We found that SQAP directly bound to and inhibited transglutaminase 2. This study provides evidence that upregulation of tumor pVHL is a promising target, which can induce "angiogenic switch off" in HCC.

16.
J Gastroenterol Hepatol ; 29(10): 1830-8, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24731186

ABSTRACT

BACKGROUND AND AIM: Preclinical studies in rodent models of chronic liver fibrosis have shown that transplantation of peripheral blood (PB) CD34(+) cells leads to hepatic regeneration and a reduction of liver fibrosis by suppressing hepatic stellate cell activity and increasing matrix metalloproteinase activity. The aim of this study was to examine the safety and clinical efficacy of intrahepatic transplantation of autologous granulocyte colony-stimulating factor (G-CSF)-mobilized PB-CD34(+) cells in patients with decompensated liver cirrhosis. METHODS: PB-CD34(+) cells were isolated from G-CSF-mobilized apheresis products. Ten patients were treated with G-CSF-mobilized PB-CD34(+) cells (treatment group) and seven patients were treated with standard medical therapy. For mobilization, patients in the treatment group received subcutaneous injections of 10 µg G-CSF/kg/day for 5 days. The cells were then injected at three different doses (5 × 10(5) , 1 × 10(6) and 2 × 10(6) cells/kg) through the hepatic artery. Thereafter, all patients were followed up for 24 months. RESULTS: G-CSF treatment and leukapheresis were well tolerated, and no serious adverse events were observed. Patients in the treatment group had a significant but transient splenomegaly. After 24 weeks, serum albumin was significantly increased in patients who had received middle or high doses of CD34(+) cells compared with baseline. Doppler ultrasound showed a significant increase in hepatic blood flow velocity and blood flow volume after CD34(+) cell therapy. The hepatic vein pressure gradient decreased in two patients who received high-dose CD34(+) cells at week 16. CONCLUSIONS: CD34(+) cell therapy is feasible, safe and effective in slowing the decline of hepatic reserve function.


Subject(s)
Antigens, CD34 , Cell- and Tissue-Based Therapy/methods , Granulocyte Colony-Stimulating Factor/administration & dosage , Liver Cirrhosis/therapy , Peripheral Blood Stem Cell Transplantation/methods , Aged , Autografts , Feasibility Studies , Female , Follow-Up Studies , Granulocyte Colony-Stimulating Factor/pharmacology , Hepatic Artery , Hepatic Stellate Cells/parasitology , Hepatic Veins/physiopathology , Humans , Injections, Subcutaneous , Liver Circulation , Liver Cirrhosis/enzymology , Liver Cirrhosis/pathology , Liver Cirrhosis/physiopathology , Liver Regeneration , Male , Matrix Metalloproteinases/metabolism , Middle Aged , Prospective Studies , Therapeutics , Time Factors , Venous Pressure
17.
J Gastroenterol Hepatol ; 28(1): 168-78, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22849788

ABSTRACT

BACKGROUND AND AIM: In cirrhosis, sinusoidal endothelial cell injury results in increased endothelin-1 (ET-1) and decreased nitric oxide synthase (NOS) activity, leading to portal hypertension. However, the effects of transplanted endothelial progenitor cells (EPCs) on the cirrhotic liver have not yet been clarified. We investigated whether EPC transplantation reduces portal hypertension. METHODS: Cirrhotic rats were created by the administration of carbon tetrachloride (CCl(4) ) twice weekly for 10 weeks. From week 7, rat bone marrow-derived EPCs were injected via the tail vein in this model once a week for 4 weeks. Endothelial NOS (eNOS), vascular endothelial growth factor (VEGF) and caveolin expressions were examined by Western blots. Hepatic tissue ET-1 was measured by a radioimmunoassay (RIA). Portal venous pressure, mean aortic pressure, and hepatic blood flow were measured. RESULTS: Endothelial progenitor cell transplantation reduced liver fibrosis, α-smooth muscle actin-positive cells, caveolin expression, ET-1 concentration and portal venous pressure. EPC transplantation increased hepatic blood flow, protein levels of eNOS and VEGF. Immunohistochemical analyses of eNOS and isolectin B4 demonstrated that the livers of EPC-transplanted animals had markedly increased vascular density, suggesting reconstitution of sinusoidal blood vessels with endothelium. CONCLUSIONS: Transplantation of EPCs ameliorates vascular dysfunction and portal hypertension, suggesting this treatment may provide a new approach in the therapy of portal hypertension with liver cirrhosis.


Subject(s)
Blood Vessels/physiopathology , Endothelial Cells/transplantation , Endothelium/metabolism , Hypertension, Portal/physiopathology , Hypertension, Portal/therapy , Liver Cirrhosis, Experimental/physiopathology , Stem Cell Transplantation , Animals , Carbon Tetrachloride , Caveolins/metabolism , Cell Proliferation , Endothelial Cells/physiology , Endothelin-1/metabolism , Endothelium/enzymology , Hypertension, Portal/etiology , Liver Circulation , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/complications , Male , Nitric Oxide Synthase Type III/metabolism , Portal Pressure , Rats , Rats, Wistar , Vascular Endothelial Growth Factor A/metabolism
18.
J Gastroenterol ; 48(2): 277-85, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22790351

ABSTRACT

BACKGROUND: Glucose intolerance in patients with liver cirrhosis (LC), known as hepatogenous diabetes, is thought to be distinct from type 2 diabetes (T2DM) in some aspects. Hyperinsulinemia and/or insulin resistance in liver disease is associated with hepatocarcinogenesis, growth of hepatocellular carcinoma, and poor prognosis. However, the pathophysiological processes in islets that are responsible for hyperinsulinemia in LC are still not precisely known. Therefore, we investigated the histopathological differences in islets of Langerhans cells between LC and T2DM. METHODS: A total of 35 human autopsy pancreatic tissue samples were used in this study (control, n = 18; T2DM, n = 6; LC, n = 11). The expression of insulin, glucagon, somatostatin, pancreatic duodenal homeobox-1 (PDX-1), proliferating cell nuclear antigen (PCNA), and Ki-67 was examined using immunohistochemistry and quantitated by image analysis. RESULTS: Islet hypertrophy and a significant increase in PCNA-positive cells in islets were observed in the tissues from LC cases. The insulin-positive areas in islets were significantly decreased in LC cases compared with control and T2DM cases (P = 0.001, P = 0.035, respectively), whereas the PDX-1-positive area was significantly increased in LC cases (P = 0.001) compared with the control. Furthermore, disorganization of pancreatic endocrine cells and nucleocytoplasmic translocation of PDX-1 were both seen in the LC subjects. CONCLUSIONS: In LC, islets undergo hypertrophy and exhibit paradoxical expression of insulin and PDX-1. In the subjects autopsied, insulin expression was decreased, whereas expression of the pancreatic transcription factor PDX-1 was increased in LC. These results point to important distinctions between LC and T2DM.


Subject(s)
Homeodomain Proteins/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Liver Cirrhosis/metabolism , Trans-Activators/metabolism , Aged , Biopsy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Female , Glucagon/metabolism , Humans , Hypertrophy/metabolism , Islets of Langerhans/pathology , Ki-67 Antigen/metabolism , Liver Cirrhosis/pathology , Male , Middle Aged , Proliferating Cell Nuclear Antigen/metabolism , Retrospective Studies , Somatostatin/metabolism
19.
Clin Cancer Res ; 18(14): 3924-33, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22611027

ABSTRACT

PURPOSE: VEGF, EGF, and TGF-α are expressed in hepatocellular carcinomas (HCC) and play a role in its growth. Vandetanib, a multikinase inhibitor, suppresses the phosphorylation of VEGF receptor 2 (VEGFR-2) and EGF receptor (EGFR). The aim of this study was to clarify the antitumor effect of vandetanib in mouse HCCs. EXPERIMENTAL DESIGN: We evaluated the effects of vandetanib on proliferation of human umbilical vein endothelial cells (HUVEC) and three hepatoma cell lines, as well as the phosphorylation of VEGFR-2 and EGFR in these cells. Mice were implanted with hepatoma cells subcutaneously or orthotopically in the liver and treated with 50 or 75 mg/kg vandetanib. We analyzed the effects of treatment on tumor cell proliferation and apoptosis, vessel density, phosphorylation of VEGFR-2 and EGFR, and production of VEGF, TGF-α, and EGF in tumor tissues. Adverse events on vandetanib administration were also investigated. RESULTS: Vandetanib suppressed phosphorylation of VEGFR-2 in HUVECs and EGFR in hepatoma cells and inhibited cell proliferation. In tumor-bearing mice, vandetanib suppressed phosphorylation of VEGFR-2 and EGFR in tumor tissues, significantly reduced tumor vessel density, enhanced tumor cell apoptosis, suppressed tumor growth, improved survival, reduced number of intrahepatic metastases, and upregulated VEGF, TGF-α, and EGF in tumor tissues. Treatment with vandetanib was not associated with serious adverse events, including alanine aminotransferase abnormality, bone marrow suppression, or body weight loss. CONCLUSIONS: The antitumor effects of vandetanib in mice suggest that it is a potentially suitable and safe chemotherapeutic agent for HCCs.


Subject(s)
ErbB Receptors , Liver Neoplasms , Neoplasms, Experimental , Piperidines/administration & dosage , Quinazolines/administration & dosage , Vascular Endothelial Growth Factor Receptor-2 , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Mice , Mice, Nude , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Phosphorylation/drug effects , Prognosis , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism
20.
J Histochem Cytochem ; 60(6): 475-83, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22473941

ABSTRACT

Mallory-Denk bodies (MDBs) are hepatocyte cytoplasmic inclusions found in several liver diseases and consist primarily of the cytoskeletal proteins, keratins 8 and 18 (K8/K18). Recent evidence indicates that the extent of stress-induced protein misfolding, a K8>K18 overexpression state, and transglutaminase-2 activation promote MDB formation. In addition, the genetic background and gender play an important role in mouse MDB formation, but the effect of aging on this process is unknown. Given that oxidative stress increases with aging, the authors hypothesized that aging predisposes to MDB formation. They used an established mouse MDB model-namely, feeding non-transgenic male FVB/N mice (1, 3, and 8 months old) with 3,5 diethoxycarbonyl-1,4-dihydrocollidine for 2 months. MDB formation was assessed using immunofluorescence staining and biochemically by demonstrating keratin and ubiquitin-containing crosslinks generated by transglutaminase-2. Immunofluorescence staining showed that old mice had a significant increase in MDB formation compared with young mice. MDB formation paralleled the generation of high molecular weight ubiquitinated keratin-containing complexes and induction of p62. Old mouse livers had increased oxidative stress. In addition, 20S proteasome activity and autophagy were decreased, and endoplasmic reticulum stress was increased in older livers. Therefore, aging predisposes to experimental MDB formation, possibly by decreased activity of protein degradation machinery.


Subject(s)
Aging/pathology , Liver/pathology , Mallory Bodies/pathology , Oxidative Stress , Aging/metabolism , Animals , Autophagy , Endoplasmic Reticulum Stress , Fluorescent Antibody Technique , Hypertrophy , Liver/metabolism , Male , Mice , Proteasome Endopeptidase Complex/metabolism , Proteolysis
SELECTION OF CITATIONS
SEARCH DETAIL
...