Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Cells ; 13(19)2024 Sep 29.
Article in English | MEDLINE | ID: mdl-39404393

ABSTRACT

The mechanisms underlying the remarkable capacity of the liver to regenerate are still not completely understood. Particularly, the cross-talk between cytokines and cellular components of the process is of utmost importance because they represent potential avenues for diagnostics and therapeutics. WNT1-inducible-signaling pathway protein 1 (WISP1) is a cytokine member of the CCN family, a family of proteins that play many different roles in liver pathophysiology. WISP1 also belongs to the earliest and strongest upregulated genes in mouse livers after CCl4 intoxication and has recently been shown to be secreted by tumor cells and to bind to type 1 collagen to cause its linearization in vitro and in tumor tissue in vivo. We show that WISP1 expression is strongly induced by TGFß, a critical cytokine in wound healing processes. Additionally, secretion of WISP1 protein by hepatic stellate is increased in cells upon TGFß stimulation (~seven-fold increase). Furthermore, WISP1 facilitates the migration of mouse hepatic stellate cells through collagen in vitro. However, in WISP1 knockout mice, no difference in stellate cell accumulation in damaged liver tissue and no influence on fibrosis was obtained, probably because the knockout of WISP1 was compensated by other factors in vivo.


Subject(s)
CCN Intercellular Signaling Proteins , Cell Movement , Hepatic Stellate Cells , Liver Cirrhosis , Mice, Knockout , Proto-Oncogene Proteins , Transforming Growth Factor beta , CCN Intercellular Signaling Proteins/metabolism , CCN Intercellular Signaling Proteins/genetics , Animals , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Mice , Transforming Growth Factor beta/metabolism , Humans , Liver/metabolism , Liver/pathology , Mice, Inbred C57BL , Male
2.
J Geriatr Oncol ; 15(7): 101838, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39097500

ABSTRACT

INTRODUCTION: The phase 2 RAMONA study demonstrated that second-line nivolumab ± ipilimumab immunotherapy was feasible and effective in older patients with advanced esophageal squamous cell cancer (ESCC). Here, we presented results from functional status (FS) and quality-of-life (QoL) analyses. MATERIALS AND METHODS: Patients aged ≥65 years with advanced ESCC and disease progression following first-line therapy were enrolled for study treatment with nivolumab ± ipilimumab. Geriatric assessments (GA) consisting of G8 and GoGo/SlowGo evaluation, and quality of life (QoL) assessments with EORTC QLQ-C30 questionnaires were conducted at baseline and during the treatment. A post hoc analysis was performed to compare therapy efficacy, toxicity, and QoL between age groups (≥70 years vs. <70 years) and functionality groups (G8 > 14 vs. ≤14 and GoGo vs. SlowGo). RESULTS: In 66 treated patients with a median age of 70.5 years, older patients had non-inferior overall survival and tumor response compared to younger patients, with no increased treatment-related adverse events. Fitter patients (G8 > 14, GoGo) had a clinically, yet not statistically significant, survival advantage than less fit patients (G8 ≤ 14, SlowGo) patients. Moreover, FS by G8 and GoGo/SlowGo significantly correlated with QoL. Overall, QoL was impaired at baseline but remained stable in all scales over the course of immunotherapy. DISCUSSION: The administration of nivolumab ± ipilimumab second-line immunotherapy in older patients with ESCC did not show age-dependent effects and maintained QoL. GA could identify functional deficits and limitations of QoL and should be implemented in the context of immunotherapy. CLINICALTRIALS: gov: NCT03416244.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Functional Status , Ipilimumab , Nivolumab , Quality of Life , Humans , Aged , Male , Ipilimumab/therapeutic use , Ipilimumab/administration & dosage , Ipilimumab/adverse effects , Female , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/drug therapy , Nivolumab/therapeutic use , Nivolumab/adverse effects , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Geriatric Assessment
3.
Biochem Biophys Res Commun ; 732: 150409, 2024 Nov 05.
Article in English | MEDLINE | ID: mdl-39033550

ABSTRACT

INTRODUCTION: WNT1-inducible signalling pathway protein 1 (WISP1) promotes progression of several tumor entities often correlating with worse prognosis. Here its expression regulation and role in the progression of chronic liver diseases (CLD) was investigated. METHODS: WISP1 expression was analyzed in human HCC datasets, in biopsies and serum samples and an HCC patient tissue microarray (TMA) including correlation to clinicopathological parameters. Spatial distribution of WISP1 expression was determined using RNAscope analysis. Regulation of WISP1 expression was investigated in cytokine-stimulated primary mouse hepatocytes (PMH) by array analysis and qRT-PCR. Outcome of WISP1 stimulation was analyzed by IncuCyte S3-live cell imaging, qRT-PCR, and immunoblotting in murine AML12 cells. RESULTS: In a TMA, high WISP1 expression was positively correlated with early HCC stages and male sex. Highest WISP1 expression levels were detected in patients with cirrhosis as compared to healthy individuals, patients with early fibrosis, and non-cirrhotic HCC in liver biopsies, expression datasets and serum samples. WISP1 transcripts were predominantly detected in hepatocytes of cirrhotic rather than tumorous liver tissue. High WISP1 expression was associated with better survival. In PMH, AML12 and HepaRG, WISP1 was identified as a specific TGF-ß1 target gene. Accordingly, expression levels of both cytokines positively correlated in human HCC patient samples. WISP1-stimulation induced the expression of Bcl-xL, PCNA and p21 in AML12 cells. CONCLUSIONS: WISP1 expression is induced by TGF-ß1 in hepatocytes and is associated with cirrhotic liver disease. We propose a crucial role of WISP1 in balancing pro- and anti-tumorigenic effects during premalignant stages of CLD.


Subject(s)
CCN Intercellular Signaling Proteins , Carcinogenesis , Gene Expression Regulation, Neoplastic , Liver Cirrhosis , Liver Neoplasms , Tumor Microenvironment , CCN Intercellular Signaling Proteins/genetics , CCN Intercellular Signaling Proteins/metabolism , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Gene Expression Profiling , Survival Analysis , Humans , Male , Female , Animals , Mice , Hepatocytes/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Apoptosis/genetics , Cell Proliferation/genetics , Cell Cycle Checkpoints/genetics , Tumor Microenvironment/genetics , Carcinogenesis/genetics
4.
Sci Immunol ; 9(95): eadj7970, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38701193

ABSTRACT

Understanding the mechanisms that regulate T cell immunity is critical for the development of effective therapies for diseases associated with T cell dysfunction, including autoimmune diseases, chronic infections, and cancer. Co-inhibitory "checkpoint molecules," such as programmed cell death protein-1, balance excessive or prolonged immune activation by T cell-intrinsic signaling. Here, by screening for mediators of natural killer (NK) cell recognition on T cells, we identified the immunoglobulin superfamily ligand B7H6 to be highly expressed by activated T cells, including patient-infused CD19-targeting chimeric antigen receptor (CAR) T cells. Unlike other checkpoint molecules, B7H6 mediated NKp30-dependent recognition and subsequent cytolysis of activated T cells by NK cells. B7H6+ T cells were prevalent in the tissue and blood of several diseases, and their abundance in tumor tissue positively correlated with clinical response in a cohort of patients with immune checkpoint inhibitor-treated esophageal cancer. In humanized mouse models, NK cell surveillance via B7H6 limited the persistence and antitumor activity of CAR T cells, and its genetic deletion enhanced T cell proliferation and persistence. Together, we provide evidence of B7H6 protein expression by activated T cells and suggest the B7H6-NKp30 axis as a therapeutically actionable NK cell-dependent immune checkpoint that regulates human T cell function.


Subject(s)
B7 Antigens , Killer Cells, Natural , T-Lymphocytes , Humans , Killer Cells, Natural/immunology , Animals , Mice , B7 Antigens/immunology , T-Lymphocytes/immunology , Natural Cytotoxicity Triggering Receptor 3/immunology , Lymphocyte Activation/immunology , Female , Esophageal Neoplasms/immunology
5.
Int J Exp Pathol ; 105(2): 64-74, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38328944

ABSTRACT

Transforming growth factor (TGF)-ß and toll-like receptors (TLRs) have been shown to independently modulate the proliferation of hepatocellular carcinoma (HCC). Since a direct cross-talk between these two signalling pathways in HCC has not been clearly described before, we aimed here to explore the possibility of such interaction. A human HCC tissue array (n = 20 vs. four control samples), human HCC samples (n = 10) and steatohepatitis-driven murine HCC samples (control, NASH and HCC; n = 6/group) were immunostained for TGFßR1, pSMAD2, TRAF6, IRAK1 and PCNA. The results were confirmed by immunoblotting. Effects of constant activation of the SMAD pathway by constitutive expression of ALK5 or knockdown of mediators of TLR signalling, IRAK1 and MyD88, on HCC proliferation, were investigated in the HCC cell line (HUH-7) after treatment with TGFß1 cytokine or TGFßR1 kinase inhibitor (LY2157299) using PCNA and MTS assay. TGFßR1 expression is decreased in human and murine HCC and associated with downregulated pSMAD2, but increased IRAK1, TRAF6 and PCNA staining. TGFßR1 kinase inhibition abolished the cytostatic effects of TGFß1 and led to the induction of IRAK1, pIRAK1 and elevated mRNA levels of TLR-9. Overexpression of ALK5 and knockdown of MyD88 or IRAK1 augmented the cytostatic effects of TGFß1 on HUH-7. In another epithelial HCC cell line, that is, HepG2, TGFßR1 kinase inhibitor similarly elevated cellular proliferation. There is a balance between the canonical SMAD-driven tumour-suppressing arm and the non-canonical tumour-promoting arm of TGFß signalling. Disruption of this balance, by inhibition of the canonical pathway, induces HCC proliferation through TLR signalling.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Cytostatic Agents , Liver Neoplasms , Animals , Humans , Mice , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Liver Neoplasms/pathology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Proliferating Cell Nuclear Antigen/metabolism , TNF Receptor-Associated Factor 6/metabolism , Toll-Like Receptors/metabolism , Transforming Growth Factor beta/metabolism
6.
J Cancer Res Clin Oncol ; 149(8): 4579-4590, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36163558

ABSTRACT

BACKGROUND: Small bowel adenocarcinoma (SBA) remains a rare malignancy accounting for less than 5% of all the gastrointestinal tract cancers. However, only limited data and expert guidelines are available for this entity. As a result, treatment concepts are predominantly derived from colorectal cancer. METHODS: To substantiate data on the course of disease, diagnosis and treatment of SBA, we performed a population-based analysis from a Bavarian population of 2.2 million people. RESULTS: We identified 223 patients with SBA. Mean age at diagnosis was 67.8 years and patients were diagnosed rather late (34.5% UICC stage IV). Largest proportion of these patients were diagnosed with adenocarcinoma of the duodenum (132 patients, 59.2%) and most patients were diagnosed with late stage cancer, stage IV (70 patients, 31.4%). With respect to treatment, most patients underwent primary surgery (187 patients, 84.6%). Systemic therapy seemed to have an impact in UICC stage IV patients but not in UICC stage IIB or III. The 5-year survival rate was 29.0%. This was significantly less compared to colon cancer in the same cohort, which was 50.0%. Furthermore, median survival of patients with small bowel cancer was only 2.0 years (95% CI 1.4-2.5) compared to 4.9 years (95% CI 4.8-5.1) of patients with colon cancer. CONCLUSION: SBA showed a distinct epidemiology compared to colon cancer. Thus, data acquisition particularly on systemic treatment are paramount, with the objective to complement the available guidelines.


Subject(s)
Adenocarcinoma , Colonic Neoplasms , Duodenal Neoplasms , Ileal Neoplasms , Intestinal Neoplasms , Jejunal Neoplasms , Humans , Intestinal Neoplasms/epidemiology , Intestinal Neoplasms/therapy , Intestine, Small/pathology , Ileal Neoplasms/pathology , Ileal Neoplasms/therapy , Duodenal Neoplasms/epidemiology , Duodenal Neoplasms/therapy , Jejunal Neoplasms/pathology , Jejunal Neoplasms/therapy , Adenocarcinoma/epidemiology , Adenocarcinoma/therapy , Adenocarcinoma/diagnosis , Colonic Neoplasms/pathology
7.
Lancet Healthy Longev ; 3(6): e417-e427, 2022 06.
Article in English | MEDLINE | ID: mdl-36098320

ABSTRACT

BACKGROUND: The overall survival of patients with advanced and refractory oesophageal squamous cell carcinoma, mostly aged 65 years and older, is poor. Treatment with PD-1 antibodies showed improved progression-free survival and overall survival. We assessed the safety and efficacy of combined nivolumab and ipilimumab therapy in this population. METHODS: This multicentre, open-label, phase 2 trial done in 32 sites in Germany included patients aged 65 years and older with oesophageal squamous cell carcinoma and disease progression or recurrence following first-line therapy. Patients were treated with nivolumab (240 mg fixed dose once every 2 weeks, intravenously) in the safety run-in phase and continued with nivolumab and ipilimumab (nivolumab 240 mg fixed dose once every 2 weeks and ipilimumab 1 mg/kg once every 6 weeks, intravenously). The primary endpoint was overall survival, which was compared with a historical cohort receiving standard chemotherapy in the intention-to-treat population. This study is registered with ClinicalTrials.gov, NCT03416244. FINDINGS: Between March 2, 2018, and Aug 20, 2020, we screened 75 patients with advanced oesophageal squamous cell carcinoma. We enrolled 66 patients (50 [76%] men and 16 [24%] women; median age 70·5 years [IQR 67·0-76·0]), 44 (67%) of whom received combined nivolumab and ipilimumab therapy and 22 (33%) received nivolumab alone. Median overall survival time at the prespecified data cutoff was 7·2 months (95% CI 5·7-12·4) and significantly higher than in a historical cohort receiving standard chemotherapy (p=0·0063). The most common treatment-related adverse events were fatigue (12 [29%] of 42), nausea (11 [26%]), and diarrhoea (ten [24%]). Grade 3-5 treatment-related adverse events occurred in 13 (20%) of 66 patients. Treatment-related death occurred in one patient with bronchiolitis obliterans while on nivolumab and ipilimumab treatment. INTERPRETATION: Patients aged at least 65 years, with advanced oesophageal squamous cell carcinoma might benefit from combined nivolumab and ipilimumab therapy in second-line treatment. FUNDING: Bristol Myers Squibb.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Epithelial Cells , Esophageal Neoplasms/drug therapy , Esophageal Squamous Cell Carcinoma/drug therapy , Female , Humans , Ipilimumab/adverse effects , Male , Nivolumab/adverse effects
8.
Gut ; 69(9): 1677-1690, 2020 09.
Article in English | MEDLINE | ID: mdl-31992593

ABSTRACT

OBJECTIVE: TGF-ß2 (TGF-ß, transforming growth factor beta), the less-investigated sibling of TGF-ß1, is deregulated in rodent and human liver diseases. Former data from bile duct ligated and MDR2 knockout (KO) mouse models for human cholestatic liver disease suggested an involvement of TGF-ß2 in biliary-derived liver diseases. DESIGN: As we also found upregulated TGFB2 in liver tissue of patients with primary sclerosing cholangitis (PSC) and primary biliary cholangitis (PBC), we now fathomed the positive prospects of targeting TGF-ß2 in early stage biliary liver disease using the MDR2-KO mice. Specifically, the influence of TgfB2 silencing on the fibrotic and inflammatory niche was analysed on molecular, cellular and tissue levels. RESULTS: TgfB2-induced expression of fibrotic genes in cholangiocytes and hepatic stellate cellswas detected. TgfB2 expression in MDR2-KO mice was blunted using TgfB2-directed antisense oligonucleotides (AON). Upon AON treatment, reduced collagen deposition, hydroxyproline content and αSMA expression as well as induced PparG expression reflected a significant reduction of fibrogenesis without adverse effects on healthy livers. Expression analyses of fibrotic and inflammatory genes revealed AON-specific regulatory effects on Ccl3, Ccl4, Ccl5, Mki67 and Notch3 expression. Further, AON treatment of MDR2-KO mice increased tissue infiltration by F4/80-positive cells including eosinophils, whereas the number of CD45-positive inflammatory cells decreased. In line, TGFB2 and CD45 expression correlated positively in PSC/PBC patients and localised in similar areas of the diseased liver tissue. CONCLUSIONS: Taken together, our data suggest a new mechanistic explanation for amelioration of fibrogenesis by TGF-ß2 silencing and provide a direct rationale for TGF-ß2-directed drug development.


Subject(s)
Cholangitis, Sclerosing , Gene Silencing , Liver Cirrhosis, Biliary , Liver Cirrhosis , Oligonucleotides, Antisense , Transforming Growth Factor beta2/genetics , Transforming Growth Factor beta2/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , Animals , Cholangitis, Sclerosing/metabolism , Cholangitis, Sclerosing/pathology , Disease Models, Animal , Drug Discovery , Gene Expression Regulation , Hepatic Stellate Cells/metabolism , Humans , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/prevention & control , Liver Cirrhosis, Biliary/metabolism , Liver Cirrhosis, Biliary/pathology , Mice , Mice, Knockout , Up-Regulation , ATP-Binding Cassette Sub-Family B Member 4
9.
BMC Cancer ; 19(1): 231, 2019 Mar 14.
Article in English | MEDLINE | ID: mdl-30871493

ABSTRACT

BACKGROUND: Advanced esophageal squamous cell cancer (ESCC) is frequently diagnosed in elderly patients. The impact of 2nd line chemotherapy is poorly defined. Recent data demonstrated effectiveness of checkpoint inhibitors in different squamous cell carcinomas. Therefore, we assess combined nivolumab/ipilimumab as 2nd line therapy in elderly ESCC patients. METHODS: RAMONA is a multicenter open-label phase II trial. The primary objective is to demonstrate a significant survival benefit of nivolumab/ipilimumab in advanced ESCC compared to historical data of standard chemotherapy. Primary endpoint is therefore overall survival (OS). Major secondary objective is the evaluation of tolerability. Time to QoL deterioration will thus be determined as key secondary endpoint. Further secondary endpoints are tumor response, PFS and safety. We aim to recruit a total of n = 75 subjects that have to be > 65 years old. Eligibility is determined by the geriatric status (G8 screening and Deficit Accumulation Frailty Index (DAFI)). A safety assessment will be performed after a 3 cycle run-in phase of nivolumab (240 mg Q2W) to justify escalation for eligible patients to combined nivolumab (240 mg Q2W) and ipilimumab (1 mg/kg Q6W), while the other patients will remain on nivolumab only. RAMONA also includes translational research sub-studies to identify predictive biomarkers, including PD-1 and PD-L1 evaluation at different time points, establishment of organoid cultures and microbiome analyses for response prediction. DISCUSSION: The RAMONA trial aims to implement checkpoint inhibitors for elderly patients with advanced ESCC as second line therapy. Novel biomarkers for checkpoint-inhibitor response are analyzed in extensive translational sub-studies. TRIAL REGISTRATION: EudraCT Number: 2017-002056-86 ; NCT03416244 , registered: 31.1.2018.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Squamous Cell/drug therapy , Esophageal Neoplasms/drug therapy , Ipilimumab/administration & dosage , Nivolumab/administration & dosage , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B7-H1 Antigen/metabolism , Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/metabolism , Humans , Ipilimumab/therapeutic use , Nivolumab/therapeutic use , Programmed Cell Death 1 Receptor/metabolism , Survival Analysis , Treatment Outcome
11.
Oncotarget ; 7(15): 19499-518, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26799667

ABSTRACT

TGF-ß1 is a major player in chronic liver diseases promoting fibrogenesis and tumorigenesis through various mechanisms. The expression and function of TGF-ß2 have not been investigated thoroughly in liver disease to date. In this paper, we provide evidence that TGF-ß2 expression correlates with fibrogenesis and liver cancer development.Using quantitative realtime PCR and ELISA, we show that TGF-ß2 mRNA expression and secretion increased in murine HSCs and hepatocytes over time in culture and were found in the human-derived HSC cell line LX-2. TGF-ß2 stimulation of the LX-2 cells led to upregulation of the TGF-ß receptors 1, 2, and 3, whereas TGF-ß1 treatment did not alter or decrease their expression. In liver regeneration and fibrosis upon CCl4 challenge, the transient increase of TGF-ß2 expression was accompanied by TGF-ß1 and collagen expression. In bile duct ligation-induced fibrosis, TGF-ß2 upregulation correlated with fibrotic markers and was more prominent than TGF-ß1 expression. Accordingly, MDR2-KO mice showed significant TGF-ß2 upregulation within 3 to 15 months but minor TGF-ß1 expression changes. In 5 of 8 hepatocellular carcinoma (HCC)/hepatoblastoma cell lines, relatively high TGF-ß2 expression and secretion were observed, with some cell lines even secreting more TGF-ß2 than TGF-ß1. TGF-ß2 was also upregulated in tumors of TGFα/cMyc and DEN-treated mice. The analysis of publically available microarray data of 13 human HCC collectives revealed considerable upregulation of TGF-ß2 as compared to normal liver.Our study demonstrates upregulation of TGF-ß2 in liver disease and suggests TGF-ß2 as a promising therapeutic target for tackling fibrosis and HCC.


Subject(s)
Liver Diseases/genetics , Liver Neoplasms/genetics , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta2/genetics , Animals , Cell Line , Cell Line, Tumor , Cells, Cultured , Female , Gene Expression Regulation/drug effects , Hep G2 Cells , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Liver Diseases/metabolism , Liver Neoplasms/metabolism , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology , Transforming Growth Factor beta2/metabolism , Transforming Growth Factor beta2/pharmacology
12.
Clin Sci (Lond) ; 128(11): 761-74, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25602745

ABSTRACT

Transforming growth factor ß (TGF-ß) is cytostatic towards damage-induced compensatory hepatocyte proliferation. This function is frequently lost during hepatocarcinogenesis, thereby switching the TGF-ß role from tumour suppressor to tumour promoter. In the present study, we investigate Smad7 overexpression as a pathophysiological mechanism for cytostatic TGF-ß inhibition in liver damage and hepatocellular carcinoma (HCC). Transgenic hepatocyte-specific Smad7 overexpression in damaged liver of fumarylacetoacetate hydrolase (FAH)-deficient mice increased compensatory proliferation of hepatocytes. Similarly, modulation of Smad7 expression changed the sensitivity of Huh7, FLC-4, HLE and HLF HCC cell lines for cytostatic TGF-ß effects. In our cohort of 140 HCC patients, Smad7 transcripts were elevated in 41.4% of HCC samples as compared with adjacent tissue, with significant positive correlation to tumour size, whereas low Smad7 expression levels were significantly associated with worse clinical outcome. Univariate and multivariate analyses indicate Smad7 levels as an independent predictor for overall (P<0.001) and disease-free survival (P=0.0123). Delineating a mechanism for Smad7 transcriptional regulation in HCC, we identified cold-shock Y-box protein-1 (YB-1), a multifunctional transcription factor. YB-1 RNAi reduced TGF-ß-induced and endogenous Smad7 expression in Huh7 and FLC-4 cells respectively. YB-1 and Smad7 mRNA expression levels correlated positively (P<0.0001). Furthermore, nuclear co-localization of Smad7 and YB-1 proteins was present in cancer cells of those patients. In summary, the present study provides a YB-1/Smad7-mediated mechanism that interferes with anti-proliferative/tumour-suppressive TGF-ß actions in a subgroup of HCC cells that may facilitate aspects of tumour progression.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cell Proliferation , Hepatocytes/metabolism , Liver Diseases/genetics , Liver Neoplasms/genetics , Smad7 Protein/genetics , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cells, Cultured , Female , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver Diseases/metabolism , Liver Diseases/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice, 129 Strain , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Middle Aged , Multivariate Analysis , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Smad7 Protein/metabolism , Survival Analysis , Transforming Growth Factor beta/pharmacology , Y-Box-Binding Protein 1/genetics , Y-Box-Binding Protein 1/metabolism
13.
PLoS One ; 8(8): e72252, 2013.
Article in English | MEDLINE | ID: mdl-23991075

ABSTRACT

Hepatocellular carcinoma (HCC) is a major public health problem due to increased incidence, late diagnosis and limited treatment options. TGF-ß is known to provide cytostatic signals during early stages of liver damage and regeneration, but exerts tumor promoting effects in onset and progression of liver cancer. To understand the mechanistic background of such a switch, we systematically correlated loss of cytostatic TGF-ß effects with strength and dynamics of its downstream signaling in 10 HCC cell lines. We demonstrate that TGF-ß inhibits proliferation and induces apoptosis in cell lines with low endogenous levels of TGF-ß and Smad7 and strong transcriptional Smad3 activity (PLC/PRF/5, HepG2, Hep3B, HuH7), previously characterized to express early TGF-ß signatures correlated with better outcome in HCC patients. TGF-ß dependent cytostasis is blunted in another group of cell lines (HLE, HLF, FLC-4) expressing high amounts of TGF-ß and Smad7 and showing significantly reduced Smad3 signaling. Of those, HLE and HLF exhibit late TGF-ß signatures, which is associated with bad prognosis in HCC patients. RNAi with Smad3 blunted cytostatic effects in PLC/PRF/5, Hep3B and HuH7. HCC-M and HCC-T represent a third group of cell lines lacking cytostatic TGF-ß signaling despite strong and prolonged Smad3 phosphorylation and low Smad7 and TGF-ß expression. Inhibitory linker phosphorylation, as in HCC-T, may disrupt C-terminally phosphorylated Smad3 function. In summary, we assort 10 HCC cell lines in at least two clusters with respect to TGF-ß sensitivity. Cell lines responsive to the TGF-ß cytostatic program, which recapitulate early stage of liver carcinogenesis exhibit transcriptional Smad3 activity. Those with disturbed TGF-ß/Smad3 signaling are insensitive to TGF-ß dependent cytostasis and might represent late stage of the disease. Regulation of this switch remains complex and cell line specific. These features may be relevant to discriminate stage dependent TGF-ß functions for the design of efficient TGF-ß directed therapy in liver cancer.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Cycle , Liver Neoplasms/pathology , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Base Sequence , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , DNA Primers , Humans , Liver Neoplasms/metabolism , RNA Interference , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/biosynthesis
14.
Dig Dis ; 30(5): 514-23, 2012.
Article in English | MEDLINE | ID: mdl-23108308

ABSTRACT

Transforming growth factor (TGF)-ß is a central regulator in chronic liver disease, contributing to all stages of disease progression from initial liver injury through inflammation and fibrosis to cirrhosis and hepatocellular carcinoma. Liver damage-induced levels of active TGF-ß enhance hepatocyte destruction and mediate hepatic stellate cell and fibroblast activation resulting in a wound-healing response, including myofibroblast generation and extracellular matrix deposition. Further evidence points to a decisive role of cytostatic and apoptotic functions mediated on hepatocytes, which is critical for the control of liver mass, with loss of TGF-ß activities resulting in hyperproliferative disorders and cancer. This concept is based on studies that describe a bipartite role of TGF-ß with tumor suppressor functions at early stages of liver damage and regeneration, whereas during cancer progression TGF-ß may turn from a tumor suppressor into a tumor promoter that exacerbates invasive and metastatic behavior. We have delineated this molecular switch of the pathway from cytostatic to tumor promoting in further detail and identify activation of survival signaling pathways in hepatocytes as a most critical requirement. Targeting the TGF-ß signaling pathway has been explored to inhibit liver disease progression. While interfering with TGF-ß signaling in various short-term animal models has demonstrated promising results, liver disease progression in humans is a process of decades with different phases in which TGF-ß or its targeting may have both beneficial and adverse outcomes. We emphasize that, in order to achieve therapeutic effects, targeting TGF-ß signaling in the right cell type at the right time is required.


Subject(s)
Carcinoma, Hepatocellular/pathology , Disease Progression , Liver Neoplasms/pathology , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Animals , Apoptosis/physiology , Carcinoma, Hepatocellular/metabolism , Cell Transformation, Neoplastic/metabolism , Humans , Liver Neoplasms/metabolism , Mice , Rats
15.
World J Gastroenterol ; 16(41): 5211-24, 2010 Nov 07.
Article in English | MEDLINE | ID: mdl-21049555

ABSTRACT

AIM: To analyse the influence of Smad7, antagonist of transforming growth factor (TGF)-ß canonical signaling pathways on hepatic stellate cell (HSC) transdifferentiation in detail. METHODS: We systematically analysed genes regulated by TGF-ß/Smad7 in activated HSCs by microarray analysis and validated the results using real time polymerase chain reaction and Western blotting analysis. RESULTS: We identified 100 known and unknown targets underlying the regulation of Smad7 expression and delineated 8 gene ontology groups. Hk2, involved in glycolysis, was one of the most downregulated proteins, while BMP2, activator of the Smad1/5/8 pathway, was extremely upregulated by Smad7. However, BMP2 dependent Smad1 activation could be inhibited in vitro by Smad7 overexpression in HSCs. CONCLUSION: We conclude (1) the existence of a tight crosstalk of TGF-ß and BMP2 pathways in HSCs and (2) a Smad7 dependently decreased sugar metabolism ameliorates HSC activation probably by energy withdrawal.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Cell Transdifferentiation/physiology , Gene Expression Regulation , Hepatic Stellate Cells/physiology , Signal Transduction/physiology , Smad7 Protein/metabolism , Tissue Kallikreins/metabolism , Animals , Bone Morphogenetic Protein 2/genetics , Cells, Cultured , Female , Male , Microarray Analysis , Molecular Sequence Data , Rats , Rats, Sprague-Dawley , Rats, Wistar , Smad7 Protein/genetics , Tissue Kallikreins/genetics
16.
J Hepatol ; 52(3): 407-16, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20129692

ABSTRACT

BACKGROUND & AIMS: Adverse alcohol effects in the liver involve oxidative metabolism, fat deposition and release of fibrogenic mediators, including TGF-beta. The work presents an assessment of liver damaging cross-talk between ethanol and TGF-beta in hepatocytes. METHODS: To investigate TGF-beta effects on hepatocytes, microarray analyses were performed and validated by qRT-PCR, Western blot analysis and immunohistochemistry. The cellular state was determined by assessing lactate dehydrogenase, cellular glutathione, reactive oxygen species, lipid peroxidation and neutral lipid deposition. RNA interference was used for gene silencing in vitro. RESULTS: TGF-beta is induced in mouse livers after chronic ethanol insult, enhances ethanol induced oxidative stress and toxicity towards cultured hepatocytes plus induces lipid-, oxidative stress metabolism- and fibrogenesis-gene expression signatures. Interestingly, TGF-beta down-regulates alcohol metabolizing enzyme Adh1 mRNA in cultured hepatocytes and liver tissue from TGF-beta transgenic mice via the ALK5/Smad2/3 signalling branch, with Smad7 as a potent negative regulator. ADH1 deficiency is a determining factor for the increased lipid accumulation and Cyp2E1 dependent toxicity in liver cells upon alcohol challenge. Further, ADH1 expression was decreased during liver damage in an intragastric ethanol infusion mouse model. CONCLUSION: In the presence of ethanol, TGF-beta displays pro-steatotic action in hepatocytes via decreasing ADH1 expression. Low ADH1 levels are correlated with enhanced hepatocyte damage upon chronic alcohol consumption by favoring secondary metabolic pathways.


Subject(s)
Alcohol Dehydrogenase/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Down-Regulation/physiology , Ethanol/adverse effects , Hepatocytes/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cells, Cultured , Chemical and Drug Induced Liver Injury/pathology , Disease Models, Animal , Hepatocytes/pathology , Lipid Metabolism/physiology , Lipid Peroxidation/physiology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/physiology , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/physiology , Smad7 Protein/metabolism
17.
Front Biosci (Landmark Ed) ; 15(2): 740-9, 2010 01 01.
Article in English | MEDLINE | ID: mdl-20036843

ABSTRACT

Chronic alcohol consumption is a risk factor for the development of chronic liver disease. Ethanol exerts its detrimental effects by various means: Directly via toxic metabolites, and indirectly by affecting the gut barrier leading to elevated levels of endotoxins in the blood challenging the liver. These factors, together with the resulting inflammatory and profibrogenic cytokine production, drive the organ's response, characterized by activation of hepatic stellate cells. Recent evidence argues for other cell types besides hepatic stellate cells, including hepatocytes, as additional sources of fibroblasts producing extracellular matrix and to be responsible for scar formation. Besides mediating hepatocyte apoptosis, TGF-beta additionally induces fibroblastoid transdifferentiation. This process is accompanied with loss of epithelial marker proteins and upregulation of fibrosis related proteins. These findings challenge the current view of the passive role of hepatocytes in liver fibrosis. In line, hepatocyte-specific inhibition of the TGF-beta pathway prevents CCl4 induced liver injury. Hence, this review focuses on the interplay of TGF-beta and alcohol in chronic liver disease with special emphasis on the potential contribution of hepatocytes.


Subject(s)
Ethanol/pharmacology , Liver/drug effects , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Central Nervous System Depressants/pharmacology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Models, Biological
18.
Alcohol Clin Exp Res ; 34(1): 4-18, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19860812

ABSTRACT

Alcohol is recognized as a direct hepatotoxin, but the precise molecular pathways that are important for the initiation and progression of alcohol-induced tissue injury are not completely understood. The current understanding of alcohol toxicity to organs suggests that alcohol initiates injury by generation of oxidative and nonoxidative ethanol metabolites and via translocation of gut-derived endotoxin. These processes lead to cellular injury and stimulation of the inflammatory responses mediated through a variety of molecules. With continuing alcohol abuse, the injury progresses through impairment of tissue regeneration and extracellular matrix (ECM) turnover, leading to fibrogenesis and cirrhosis. Several cell types are involved in this process, the predominant being stellate cells, macrophages, and parenchymal cells. In response to alcohol, growth factors and cytokines activate many signaling cascades that regulate fibrogenesis. This mini-review brings together research focusing on the underlying mechanisms of alcohol-mediated injury in a number of organs. It highlights the various processes and molecules that are likely involved in inflammation, immune modulation, susceptibility to infection, ECM turnover and fibrogenesis in the liver, pancreas, and lung triggered by alcohol abuse.


Subject(s)
Alcoholism/metabolism , Extracellular Matrix/metabolism , Signal Transduction/physiology , Alcoholism/pathology , Animals , Ethanol/administration & dosage , Ethanol/metabolism , Extracellular Matrix/pathology , Humans , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology
19.
Proc Natl Acad Sci U S A ; 106(28): 11588-93, 2009 Jul 14.
Article in English | MEDLINE | ID: mdl-19581593

ABSTRACT

The insertion efficiency of transmembrane (TM) helices by the Sec61 translocon depends on helix amino acid composition, the positions of the amino acids within the helix, and helix length. We have used an in vitro expression system to examine systematically the insertion efficiency of short polyleucine segments (L(n), n = 4 ... 12) flanked at either end by 4-residue sequences of the form XXPX-L(n)-XPXX with X = G, N, D, or K. Except for X = K, insertion efficiency (p) is <10% for n < 8, but rises steeply to 100% for n = 12. For X = K, p is already close to 100% for n = 10. A similar pattern is observed for synthetic peptides incorporated into oriented phospholipid bilayer arrays, consistent with the idea that recognition of TM segments by the translocon critically involves physical partitioning of nascent peptide chains into the lipid bilayer. Molecular dynamics simulations suggest that insertion efficiency is determined primarily by the energetic cost of distorting the bilayer in the vicinity of the TM helix. Very short lysine-flanked leucine segments can reduce the energetic cost by extensive hydrogen bonding with water and lipid phosphate groups (snorkeling) and by partial unfolding.


Subject(s)
Lipid Bilayers/metabolism , Membrane Proteins/metabolism , Models, Biological , Peptides/metabolism , Computer Simulation , Hydrogen Bonding , SEC Translocation Channels
20.
J Gastroenterol Hepatol ; 23 Suppl 1: S122-7, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18336655

ABSTRACT

Currently, hepatic stellate cells (HSC) are thought to be the major fibrotic precursor cells that transdifferentiate to fibrogenic, extracellular matrix producing myofibroblasts in inflammatory liver tissue upon transforming growth factor-beta (TGF-beta) signaling, whereas hepatocytes are thought to respond with apoptosis to this cytokine. Starting out from in vitro experiments with primary hepatocyte cultures and immortalized AML-12 cells, TGF-beta signaling in this cell type was assessed and apoptosis was found to be only a minor effect. Instead, hepatocytes undergo epithelial mesenchymal transition (EMT), a physiological process in embryogenesis and of relevance for cancerous cell transformation. In injured liver, however, this process contributes to the promotion of fibrosis. Already after a few days of culture, hepatocytes lose their epithelial honeycomb-like shape towards a fibroblast-like phenotype. We could demonstrate by microarray analysis that stimulation of hepatocytes with TGF-beta regulates the expression of genes involved in EMT and fibrosis. Among these were, for example, Snail, a known mediator of EMT, and connective tissue growth factor (CTGF), a strong inducer of fibrosis. In a mouse model, hepatocyte-specific overexpression of Smad7 was able to blunt a fibrogenic response after CCl(4) intoxication. These results emphasize the dynamic nature of liver fibrosis, challenge the paradigm of HSC as a crucial source of liver myofibroblasts and hint towards a prominent role for hepatocytes in liver fibrogenesis.


Subject(s)
Cell Transdifferentiation/physiology , Hepatocytes/cytology , Liver Cirrhosis/etiology , Transforming Growth Factor beta/physiology , Animals , Epithelial Cells/cytology , Humans , Mesoderm/cytology
SELECTION OF CITATIONS
SEARCH DETAIL