218 results on '"Monga SP"'
Search Results
2. Role of leukocyte cell-derived chemotaxin 2 as a biomarker in hepatocellular carcinoma
- Author
-
Okabe, H, Delgado, E, Lee, JM, Yang, J, Kinoshita, H, Hayashi, H, Tsung, A, Behari, J, Beppu, T, Baba, H, Monga, SP, Okabe, H, Delgado, E, Lee, JM, Yang, J, Kinoshita, H, Hayashi, H, Tsung, A, Behari, J, Beppu, T, Baba, H, and Monga, SP
- Abstract
We sought to identify a secreted biomarker for β-catenin activation commonly seen in hepatocellular carcinoma (HCC). By examination of our previously published genearray of hepatocyte-specific β-catenin knockout (KO) livers, we identified secreted factors whose expression may be β-catenin-dependent. We verified expression and secretion of the leading factor in HCC cells transfected with mutated (Hep3BS33Y)-β- catenin. Serum levels of biomarker were next investigated in a mouse model of HCC with β-catenin gene (Ctnnb1) mutations and eventually in HCC patients. Leukocyte cell-derived chemotaxin-2 (LECT2) expression was decreased in KO livers. Hep3BS33Y expressed and secreted more LECT2 in media as compared to Hep3BWT. Mice developing HCC with Ctnnb1 mutations showed significantly higher serum LECT2 levels. However patients with CTNNB1 mutations showed LECT2 levels of 54.28±22.32 ng/mL (Mean ± SD; n = 8) that were insignificantly different from patients with non-neoplastic chronic liver disease (32.8±21.1 ng/mL; n = 15) or healthy volunteers (33.2±7.2 ng/mL; n = 11). Intriguingly, patients without β-catenin mutations showed significantly higher serum LECT2 levels (54.26 ± 22.25 ng/mL; n = 46). While β-catenin activation was evident in a subset of non-mutant β-catenin HCC group with high LECT2 expression, serum LECT2 was unequivocally similar between β-catenin-active and -normal group. Further analysis showed that LECT2 levels greater than 50 ng/ml diagnosed HCC in patients irrespective of β-catenin mutations with specificity of 96.1% and positive predictive value of 97.0%. Thus, LECT2 is regulated by β-catenin in HCC in both mice and men, but serum LECT2 reflects β-catenin activity only in mice. Serum LECT2 could be a potential biomarker of HCC in patients. © 2014 Okabe et al.
- Published
- 2014
3. Regucalcin mediated up‐regulation of Wnt/β‐catenin signaling
- Author
-
DAR, MOHD JAMAL, primary and MONGA, SP, additional
- Published
- 2011
- Full Text
- View/download PDF
4. Precision models in hepatocellular carcinoma.
- Author
-
Barcena-Varela M, Monga SP, and Lujambio A
- Abstract
Hepatocellular carcinoma (HCC) represents a global health challenge, and ranks among one of the most prevalent and deadliest cancers worldwide. Therapeutic advances have expanded the treatment armamentarium for patients with advanced HCC, but obstacles remain. Precision oncology, which aims to match specific therapies to patients who have tumours with particular features, holds great promise. However, its implementation has been hindered by the existence of numerous 'HCC influencers' that contribute to the high inter-patient heterogeneity. HCC influencers include tumour-related characteristics, such as genetic alterations, immune infiltration, stromal composition and aetiology, and patient-specific factors, such as sex, age, germline variants and the microbiome. This Review delves into the intricate world of HCC, describing the most innovative model systems that can be harnessed to identify precision and/or personalized therapies. We provide examples of how different models have been used to nominate candidate biomarkers, their limitations and strategies to optimize such models. We also highlight the importance of reproducing distinct HCC influencers in a flexible and modular way, with the aim of dissecting their relative contribution to therapy response. Next-generation HCC models will pave the way for faster discovery of precision therapies for patients with advanced HCC., Competing Interests: Competing interests: The authors declare no competing interests., (© 2024. Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
5. The Dark Side of Human Hepatocyte Plasticity.
- Author
-
Ko S and Monga SP
- Subjects
- Humans, Animals, Cell Differentiation, Hepatocytes, Cell Plasticity
- Published
- 2024
- Full Text
- View/download PDF
6. Context-Dependent Distinct Roles of SOX9 in Combined Hepatocellular Carcinoma-Cholangiocarcinoma.
- Author
-
Park Y, Hu S, Kim M, Oertel M, Singhi A, Monga SP, Liu S, and Ko S
- Subjects
- Animals, Humans, Mice, Cell Proliferation genetics, Carcinoma, Hepatocellular pathology, Carcinoma, Hepatocellular genetics, Carcinoma, Hepatocellular metabolism, Cholangiocarcinoma pathology, Cholangiocarcinoma genetics, Cholangiocarcinoma metabolism, Liver Neoplasms pathology, Liver Neoplasms genetics, Liver Neoplasms metabolism, SOX9 Transcription Factor metabolism, SOX9 Transcription Factor genetics
- Abstract
Combined hepatocellular carcinoma-cholangiocarcinoma (cHCC-CCA) is a challenging primary liver cancer subtype with limited treatment options and a devastating prognosis. Recent studies have underscored the context-dependent roles of SOX9 in liver cancer formation in a preventive manner. Here, we revealed that liver-specific developmental Sox9 elimination using Alb-Cre;Sox9
(flox/flox) (LKO) and CRISPR/Cas9 -based tumor-specific acute Sox9 elimination (CKO) in SB-HDTVI-based Akt-YAP1 (AY) and Akt-NRAS (AN) cHCC-CCA models showed contrasting responses. LKO abrogates the AY CCA region while stimulating poorly differentiated HCC proliferation, whereas CKO prevents AY and AN cHCC-CCA development irrespective of tumor cell fate. Additionally, AN, but not AY, tumor formation partially depends on the Sox9-Dnmt1 cascade. SOX9 is dispensable for AY-mediated, HC-derived, LPC-like immature CCA formation but is required for their maintenance and transformation into mature CCA. Therapeutic Sox9 elimination using the OPN-CreERT2 strain combined with inducible Sox9 iKO specifically reduces AY but not AN cHCC-CCA tumors. This necessitates the careful consideration of genetic liver cancer studies using developmental Cre and somatic mutants, particularly for genes involved in liver development. Our findings suggest that SOX9 elimination may hold promise as a therapeutic approach for a subset of cHCC-CCA and highlight the need for further investigation to translate these preclinical insights into personalized clinical applications.- Published
- 2024
- Full Text
- View/download PDF
7. Development of mutated β-catenin gene signature to identify CTNNB1 mutations from whole and spatial transcriptomic data in patients with HCC.
- Author
-
Lehrich BM, Tao J, Liu S, Hirsch TZ, Yasaka TM, Cao C, Delgado ER, Guan X, Lu S, Pan L, Liu Y, Singh S, Poddar M, Bell A, Singhi AD, Zucman-Rossi J, Wang Y, and Monga SP
- Abstract
Background & Aims: Patients with β-catenin (encoded by CTNNB1 )-mutated hepatocellular carcinoma (HCC) demonstrate heterogenous responses to first-line immune checkpoint inhibitors (ICIs). Precision-medicine based treatments for this subclass are currently in clinical development. Here, we report derivation of the Mutated β-catenin Gene Signature (MBGS) to predict CTNNB1 -mutational status in patients with HCC for future application in personalized medicine treatment regimens., Methods: Co-expression of mutant-Nrf2 and hMet ± mutant-β-catenin in murine livers in mice led to HCC development. The MBGS was derived using bulk RNA-seq and intersectional transcriptomic analysis of β-catenin-mutated and non-mutated HCC models. Integrated RNA/whole-exome-sequencing and spatial transcriptomic data from multiple cohorts of patients with HCC was assessed to address the ability of MBGS to detect CTNNB1 mutation, the tumor immune microenvironment, and/or predict therapeutic responses., Results: Bulk RNA-seq comparing HCC specimens in mutant β-catenin-Nrf2, β-catenin-Met and β-catenin-Nrf2-Met to Nrf2-Met HCC model yielded 95 common upregulated genes. In The Cancer Genome Atlas (TCGA)-LIHC dataset, differential gene expression analysis with false discovery rate (FDR) = 0.05 and log
2 (fold change) >1.5 on the 95 common genes comparing CTNNB1 -mutated vs. wild-type patients narrowed the gene panel to a 13-gene MBGS. MBGS predicted CTNNB1 -mutations in TCGA (n = 374) and French (n = 398) patient cohorts with AUCs of 0.90 and 0.94, respectively. Additionally, a higher MBGS expression score was associated with lack of significant improvement in overall survival or progression-free survival in the atezolizumab-bevacizumab arm vs. the sorafenib arm in the IMbrave150 cohort. MBGS performed comparable or superior to other CTNNB1 -mutant classifiers. MBGS overlapped with Hoshida S3, Boyault G5/G6, and Chiang CTNNB1 subclass tumors in TCGA and in HCC spatial transcriptomic datasets visually depicting these tumors to be situated in an immune excluded tumor microenvironment., Conclusions: MBGS will aid in patient stratification to guide precision medicine therapeutics for CTNNB1 -mutated HCC subclass as a companion diagnostic, as anti-β-catenin therapies become available., Impact and Implications: As precision medicine for liver cancer treatment becomes a reality, diagnostic tools are needed to help classify patients into groups for the best treatment choices. We have developed a molecular signature that could serve as a companion diagnostic and uses bulk or spatial transcriptomic data to identify a unique subclass of liver tumors. This subgroup of liver cancer patients derive limited benefit from the current standard of care and are expected to benefit from specialized directed therapies that are on the horizon., (© 2024 The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
8. Identification of a novel GSK3β inhibitor involved in abrogating KRas dependent pancreatic tumors in Wnt/beta-catenin and NF-kB dependent manner.
- Author
-
Ayaz MO, Bhat AQ, Akhter Z, Badsera N, Hossain MM, Showket F, Parveen S, Dar MS, Tiwari H, Kumari N, Bhardwaj M, Hussain R, Sharma A, Kumar M, Singh U, Nargorta A, Kshatri AS, Nandi U, Monga SP, Ramajayan P, Singh PP, and Dar MJ
- Subjects
- Humans, Animals, Mice, Cell Line, Tumor, Xenograft Model Antitumor Assays, Mice, Nude, Wnt Signaling Pathway drug effects, Female, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms pathology, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms genetics, Glycogen Synthase Kinase 3 beta metabolism, Glycogen Synthase Kinase 3 beta antagonists & inhibitors, NF-kappa B metabolism, beta Catenin metabolism, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, Apoptosis drug effects
- Abstract
Pancreatic cancer is an aggressive malignancy with a poor survival rate because it is difficult to diagnose the disease during its early stages. The currently available treatments, which include surgery, chemotherapy and radiation therapy, offer only limited survival benefit. Pharmacological interventions to inhibit Glycogen Synthase Kinase-3beta (GSK3β) activity is an important therapeutic strategy for the treatment of pancreatic cancer because GSK3β is one of the key factors involved in the onset, progression as well as in the acquisition of chemoresistance in pancreatic cancer. Here, we report the identification of MJ34 as a potent GSK3β inhibitor that significantly reduced growth and survival of human mutant KRas dependent pancreatic tumors. MJ34 mediated GSK3β inhibition was seen to induce apoptosis in a β-catenin dependent manner and downregulate NF-kB activity in MiaPaCa-2 cells thereby impeding cell survival and anti-apoptotic processes in these cells as well as in the xenograft model of pancreatic cancer. In vivo acute toxicity and in vitro cardiotoxicity studies indicate that MJ34 is well tolerated without any adverse effects. Taken together, we report the discovery of MJ34 as a potential drug candidate for the therapeutic treatment of mutant KRas-dependent human cancers through pharmacological inhibition of GSK3β., Competing Interests: Declaration of competing interest The authors declare that they have no conflict of interest., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
9. Loss of β-catenin reveals a role for glutathione in regulating oxidative stress during cholestatic liver disease.
- Author
-
Balogun O, Shao D, Carson M, King T, Kosar K, Zhang R, Zeng G, Cornuet P, Goel C, Lee E, Patel G, Brooks E, Monga SP, Liu S, and Nejak-Bowen K
- Subjects
- Female, Animals, Mice, Mice, Inbred C57BL, 1-Naphthylisothiocyanate toxicity, Mice, Knockout, Bile Acids and Salts metabolism, Glutathione Transferase metabolism, NF-E2-Related Factor 2 metabolism, Signal Transduction drug effects, Disease Models, Animal, Cell Proliferation drug effects, Liver metabolism, Liver pathology, Oxidative Stress drug effects, Glutathione metabolism, beta Catenin genetics, beta Catenin metabolism, Cholestasis, Intrahepatic genetics, Cholestasis, Intrahepatic metabolism, Cholestasis, Intrahepatic pathology
- Abstract
Background: Cholestasis is an intractable liver disorder that results from impaired bile flow. We have previously shown that the Wnt/β-catenin signaling pathway regulates the progression of cholestatic liver disease through multiple mechanisms, including bile acid metabolism and hepatocyte proliferation. To further explore the impact of these functions during intrahepatic cholestasis, we exposed mice to a xenobiotic that causes selective biliary injury., Methods: α-naphthylisothiocyanate (ANIT) was administered to liver-specific knockout (KO) of β-catenin and wild-type mice in the diet. Mice were killed at 6 or 14 days to assess the severity of cholestatic liver disease, measure the expression of target genes, and perform biochemical analyses., Results: We found that the presence of β-catenin was protective against ANIT, as KO mice had a significantly lower survival rate than wild-type mice. Although serum markers of liver damage and total bile acid levels were similar between KO and wild-type mice, the KO had minor histological abnormalities, such as sinusoidal dilatation, concentric fibrosis around ducts, and decreased inflammation. Notably, both total glutathione levels and expression of glutathione-S-transferases, which catalyze the conjugation of ANIT to glutathione, were significantly decreased in KO after ANIT. Nuclear factor erythroid-derived 2-like 2, a master regulator of the antioxidant response, was activated in KO after ANIT as well as in a subset of patients with primary sclerosing cholangitis lacking activated β-catenin. Despite the activation of nuclear factor erythroid-derived 2-like 2, KO livers had increased lipid peroxidation and cell death, which likely contributed to mortality., Conclusions: Loss of β-catenin leads to increased cellular injury and cell death during cholestasis through failure to neutralize oxidative stress, which may contribute to the pathology of this disease., (Copyright © 2024 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the American Association for the Study of Liver Diseases.)
- Published
- 2024
- Full Text
- View/download PDF
10. Quantitative radiomics and qualitative LI-RADS imaging descriptors for non-invasive assessment of β-catenin mutation status in hepatocellular carcinoma.
- Author
-
Arefan D, D'Ardenne NM, Iranpour N, Catania R, Yousef J, Chupetlovska K, Moghe A, Sholosh B, Thangasamy S, Borhani AA, Singhi AD, Monga SP, Furlan A, and Wu S
- Subjects
- Humans, Retrospective Studies, Female, Male, Middle Aged, Aged, Mutation, Adult, Liver diagnostic imaging, Radiology Information Systems, Radiomics, Liver Neoplasms diagnostic imaging, Liver Neoplasms genetics, beta Catenin genetics, Carcinoma, Hepatocellular diagnostic imaging, Carcinoma, Hepatocellular genetics, Magnetic Resonance Imaging methods, Tomography, X-Ray Computed methods
- Abstract
Purpose: Gain-of-function mutations in CTNNB1, gene encoding for β-catenin, are observed in 25-30% of hepatocellular carcinomas (HCCs). Recent studies have shown β-catenin activation to have distinct roles in HCC susceptibility to mTOR inhibitors and resistance to immunotherapy. Our goal was to develop and test a computational imaging-based model to non-invasively assess β-catenin activation in HCC, since liver biopsies are often not done due to risk of complications., Methods: This IRB-approved retrospective study included 134 subjects with pathologically proven HCC and available β-catenin activation status, who also had either CT or MR imaging of the liver performed within 1 year of histological assessment. For qualitative descriptors, experienced radiologists assessed the presence of imaging features listed in LI-RADS v2018. For quantitative analysis, a single biopsy proven tumor underwent a 3D segmentation and radiomics features were extracted. We developed prediction models to assess the β-catenin activation in HCC using both qualitative and quantitative descriptors., Results: There were 41 cases (31%) with β-catenin mutation and 93 cases (69%) without. The model's AUC was 0.70 (95% CI 0.60, 0.79) using radiomics features and 0.64 (0.52, 0.74; p = 0.468) using qualitative descriptors. However, when combined, the AUC increased to 0.88 (0.80, 0.92; p = 0.009). Among the LI-RADS descriptors, the presence of a nodule-in-nodule showed a significant association with β-catenin mutations (p = 0.015). Additionally, 88 radiomics features exhibited a significant association (p < 0.05) with β-catenin mutations., Conclusion: Combination of LI-RADS descriptors and CT/MRI-derived radiomics determine β-catenin activation status in HCC with high confidence, making precision medicine a possibility., (© 2024. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
11. Therapeutic potential of SOX9 dysruption in Combined Hepatocellular Carcinoma-Cholangiocarcinoma.
- Author
-
Park Y, Hu S, Kim M, Oertel M, Singhi A, Monga SP, Liu S, and Ko S
- Abstract
Combined hepatocellular carcinoma-cholangiocarcinoma (cHCC-CCA) represents a challenging subtype of primary liver cancer with limited treatment options and a poor prognosis. Recently, we and others have highlighted the context-dependent roles of the biliary-specific transcription factor SOX9 in the pathogenesis of liver cancers using various Cre applications in Sox9
(flox/flox) strains, to achieve elimination for exon 2 and 3 of the Sox9 gene locus as a preventive manner. Here, we reveal the contrasting responses of developmental Sox9 elimination using Alb-Cre;Sox9(flox/flox) ( Sox9 LKO) versus CRISPR/Cas9 -based tumor specific acute Sox9 CKO in SB-HDTVI-based Akt-YAP1 and Akt-NRAS cHCC-CCA formation. Sox9 LKO specifically abrogates the Akt-YAP1 CCA region while robustly stimulating the proliferation of remaining poorly differentiated HCC pertaining liver progenitor cell characteristics, whereas Sox9 CKO potently prevents Akt-YAP1 and Akt-NRAS cHCC-CCA development irrespective of fate of tumor cells compared to respective controls. Additionally, we find that Akt-NRAS , but not Akt-YAP1 , tumor formation is partially dependent on the Sox9-Dnmt1 cascade. Pathologically, SOX9 is indispensable for Akt-YAP1 -mediated HC-to-BEC/CCA reprogramming but required for the maintenance of CCA nodules. Lastly, therapeutic elimination of Sox9 using the OPN-CreERT2 strain combined with an inducible CRISPR/Cas9 -based Sox9 iKO significantly reduces Akt-YAP1 cHCC-CCA tumor burden, similar to Sox9 CKO. Thus, we contrast the outcomes of acute Sox9 deletion with developmental Sox9 knockout models, emphasizing the importance of considering adaptation mechanisms in therapeutic strategies. This necessitates the careful consideration of genetic liver cancer studies using developmental Cre and somatic mutant lines, particularly for genes involved in hepatic commitment during development. Our findings suggest that SOX9 elimination may hold promise as a therapeutic approach for cHCC-CCA and underscore the need for further investigation to translate these preclinical insights into clinical applications.- Published
- 2024
- Full Text
- View/download PDF
12. Selective Targeting of α 4 β 7 /MAdCAM-1 Axis Suppresses Fibrosis Progression by Reducing Proinflammatory T Cell Recruitment to the Liver.
- Author
-
Gupta B, Rai RP, Pal PB, Rossmiller D, Chaudhary S, Chiaro A, Seaman S, Singhi AD, Liu S, Monga SP, Iyer SS, and Raeman R
- Subjects
- Animals, Female, Humans, Male, Mice, Antibodies, Monoclonal pharmacology, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes immunology, Disease Models, Animal, Immunoglobulins metabolism, Inflammation pathology, Mice, Inbred C57BL, T-Lymphocytes immunology, T-Lymphocytes metabolism, Carbon Tetrachloride pharmacology, Carbon Tetrachloride toxicity, Cell Adhesion Molecules metabolism, Disease Progression, Integrins metabolism, Liver pathology, Liver metabolism, Liver Cirrhosis chemically induced, Liver Cirrhosis immunology, Liver Cirrhosis pathology, Mucoproteins metabolism
- Abstract
Integrin α
4 β7 + T cells perpetuate tissue injury in chronic inflammatory diseases, yet their role in hepatic fibrosis progression remains poorly understood. Here, we report increased accumulation of α4 β7 + T cells in the liver of people with cirrhosis relative to disease controls. Similarly, hepatic fibrosis in the established mouse model of CCl4 -induced liver fibrosis was associated with enrichment of intrahepatic α4 β7 + CD4 and CD8 T cells. Monoclonal antibody (mAb)-mediated blockade of α4 β7 or its ligand mucosal addressin cell adhesion molecule (MAdCAM)-1 attenuated hepatic inflammation and prevented fibrosis progression in CCl4 -treated mice. Improvement in liver fibrosis was associated with a significant decrease in the infiltration of α4 β7 + CD4 and CD8 T cells, suggesting that α4 β7 /MAdCAM-1 axis regulates both CD4 and CD8 T cell recruitment to the fibrotic liver, and α4 β7 + T cells promote hepatic fibrosis progression. Analysis of hepatic α4 β7 + and α4 β7 - CD4 T cells revealed that α4 β7 + CD4 T cells were enriched for markers of activation and proliferation, demonstrating an effector phenotype. The findings suggest that α4 β7 + T cells play a critical role in promoting hepatic fibrosis progression, and mAb-mediated blockade of α4 β7 or MAdCAM-1 represents a promising therapeutic strategy for slowing hepatic fibrosis progression in chronic liver diseases.- Published
- 2024
- Full Text
- View/download PDF
13. Therapeutic targeting at genome mutations of liver cancer by the insertion of HSV1 thymidine kinase through Cas9-mediated editing.
- Author
-
Kader M, Sun W, Ren BG, Yu YP, Tao J, Foley LM, Liu S, Monga SP, and Luo JH
- Subjects
- Humans, Animals, Mice, Thymidine Kinase genetics, CRISPR-Cas Systems genetics, Catenins, Mutation genetics, Herpesvirus 1, Human genetics, Liver Neoplasms genetics, Liver Neoplasms therapy
- Abstract
Background: Liver cancer is one of the most lethal malignancies for humans. The treatment options for advanced-stage liver cancer remain limited. A new treatment is urgently needed to reduce the mortality of the disease., Methods: In this report, we developed a technology for mutation site insertion of a suicide gene (herpes simplex virus type 1- thymidine kinase) based on type II CRISPR RNA-guided endonuclease Cas9-mediated genome editing to treat liver cancers., Results: We applied the strategy to 3 different mutations: S45P mutation of catenin beta 1, chromosome breakpoint of solute carrier family 45 member 2-alpha-methylacyl-CoA racemase gene fusion, and V235G mutation of SAFB-like transcription modulator. The results showed that the herpes simplex virus type 1-thymidine kinase insertion rate at the S45P mutation site of catenin beta 1 reached 77.8%, while the insertion rates at the breakpoint of solute carrier family 45 member 2 - alpha-methylacyl-CoA racemase gene fusion were 95.1%-98.7%, and the insertion at V235G of SAFB-like transcription modulator was 51.4%. When these targeting reagents were applied to treat mouse spontaneous liver cancer induced by catenin beta 1S45P or solute carrier family 45 member 2-alpha-methylacyl-CoA racemase, the mice experienced reduced tumor burden and increased survival rate. Similar results were also obtained for the xenografted liver cancer model: Significant reduction of tumor volume, reduction of metastasis rate, and improved survival were found in mice treated with the targeting reagent, in comparison with the control-treated groups., Conclusions: Our studies suggested that mutation targeting may hold promise as a versatile and effective approach to treating liver cancers., (Copyright © 2024 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the American Association for the Study of Liver Diseases.)
- Published
- 2024
- Full Text
- View/download PDF
14. Battle of the biopsies: Role of tissue and liquid biopsy in hepatocellular carcinoma.
- Author
-
Lehrich BM, Zhang J, Monga SP, and Dhanasekaran R
- Subjects
- Humans, Biomarkers, Tumor, Neoplastic Cells, Circulating, Carcinoma, Hepatocellular diagnosis, Carcinoma, Hepatocellular therapy, Carcinoma, Hepatocellular genetics, Liquid Biopsy methods, Liver Neoplasms diagnosis, Liver Neoplasms therapy, Liver Neoplasms genetics
- Abstract
The diagnosis and management of hepatocellular carcinoma (HCC) have improved significantly in recent years. With the introduction of immunotherapy-based combination therapy, there has been a notable expansion in treatment options for patients with unresectable HCC. Simultaneously, innovative molecular tests for early detection and management of HCC are emerging. This progress prompts a key question: as liquid biopsy techniques rise in prominence, will they replace traditional tissue biopsies, or will both techniques remain relevant? Given the ongoing challenges of early HCC detection, including issues with ultrasound sensitivity, accessibility, and patient adherence to surveillance, the evolution of diagnostic techniques is more relevant than ever. Furthermore, the accurate stratification of HCC is limited by the absence of reliable biomarkers which can predict response to therapies. While the advantages of molecular diagnostics are evident, their potential has not yet been fully harnessed, largely because tissue biopsies are not routinely performed for HCC. Liquid biopsies, analysing components such as circulating tumour cells, DNA, and extracellular vesicles, provide a promising alternative, though they are still associated with challenges related to sensitivity, cost, and accessibility. The early results from multi-analyte liquid biopsy panels are promising and suggest they could play a transformative role in HCC detection and management; however, comprehensive clinical validation is still ongoing. In this review, we explore the challenges and potential of both tissue and liquid biopsy, highlighting that these diagnostic methods, while distinct in their approaches, are set to jointly reshape the future of HCC management., (Copyright © 2023 European Association for the Study of the Liver. Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
15. Matrix viscoelasticity promotes liver cancer progression in the pre-cirrhotic liver.
- Author
-
Fan W, Adebowale K, Váncza L, Li Y, Rabbi MF, Kunimoto K, Chen D, Mozes G, Chiu DK, Li Y, Tao J, Wei Y, Adeniji N, Brunsing RL, Dhanasekaran R, Singhi A, Geller D, Lo SH, Hodgson L, Engleman EG, Charville GW, Charu V, Monga SP, Kim T, Wells RG, Chaudhuri O, and Török NJ
- Subjects
- Animals, Humans, beta Catenin metabolism, Cell Proliferation, Collagen chemistry, Collagen metabolism, Computer Simulation, Diabetes Mellitus, Type 2 complications, Diabetes Mellitus, Type 2 metabolism, Glycation End Products, Advanced metabolism, Integrin beta1 metabolism, Neoplasm Invasiveness, Viscosity, YAP-Signaling Proteins metabolism, Carcinoma, Hepatocellular complications, Carcinoma, Hepatocellular metabolism, Carcinoma, Hepatocellular pathology, Elasticity, Extracellular Matrix metabolism, Liver Neoplasms complications, Liver Neoplasms metabolism, Liver Neoplasms pathology, Disease Progression, Liver Cirrhosis complications, Liver Cirrhosis metabolism, Liver Cirrhosis pathology
- Abstract
Type 2 diabetes mellitus is a major risk factor for hepatocellular carcinoma (HCC). Changes in extracellular matrix (ECM) mechanics contribute to cancer development
1,2 , and increased stiffness is known to promote HCC progression in cirrhotic conditions3,4 . Type 2 diabetes mellitus is characterized by an accumulation of advanced glycation end-products (AGEs) in the ECM; however, how this affects HCC in non-cirrhotic conditions is unclear. Here we find that, in patients and animal models, AGEs promote changes in collagen architecture and enhance ECM viscoelasticity, with greater viscous dissipation and faster stress relaxation, but not changes in stiffness. High AGEs and viscoelasticity combined with oncogenic β-catenin signalling promote HCC induction, whereas inhibiting AGE production, reconstituting the AGE clearance receptor AGER1 or breaking AGE-mediated collagen cross-links reduces viscoelasticity and HCC growth. Matrix analysis and computational modelling demonstrate that lower interconnectivity of AGE-bundled collagen matrix, marked by shorter fibre length and greater heterogeneity, enhances viscoelasticity. Mechanistically, animal studies and 3D cell cultures show that enhanced viscoelasticity promotes HCC cell proliferation and invasion through an integrin-β1-tensin-1-YAP mechanotransductive pathway. These results reveal that AGE-mediated structural changes enhance ECM viscoelasticity, and that viscoelasticity can promote cancer progression in vivo, independent of stiffness., (© 2024. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)- Published
- 2024
- Full Text
- View/download PDF
16. shinyDeepDR: A user-friendly R Shiny app for predicting anti-cancer drug response using deep learning.
- Author
-
Wang LJ, Ning M, Nayak T, Kasper MJ, Monga SP, Huang Y, Chen Y, and Chiu YC
- Abstract
Advancing precision oncology requires accurate prediction of treatment response and accessible prediction models. To this end, we present shinyDeepDR, a user-friendly implementation of our innovative deep learning model, DeepDR, for predicting anti-cancer drug sensitivity. The web tool makes DeepDR more accessible to researchers without extensive programming experience. Using shinyDeepDR, users can upload mutation and/or gene expression data from a cancer sample (cell line or tumor) and perform two main functions: "Find Drug," which predicts the sample's response to 265 approved and investigational anti-cancer compounds, and "Find Sample," which searches for cell lines in the Cancer Cell Line Encyclopedia (CCLE) and tumors in The Cancer Genome Atlas (TCGA) with genomics profiles similar to those of the query sample to study potential effective treatments. shinyDeepDR provides an interactive interface to interpret prediction results and to investigate individual compounds. In conclusion, shinyDeepDR is an intuitive and free-to-use web tool for in silico anti-cancer drug screening., Competing Interests: The authors declare no competing interests., (© 2023 The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
17. Exploring the Impact of the β-Catenin Mutations in Hepatocellular Carcinoma: An In-Depth Review.
- Author
-
Idrissi YA, Rajabi MR, Beumer JH, Monga SP, and Saeed A
- Subjects
- Humans, Wnt Signaling Pathway genetics, Animals, beta Catenin genetics, beta Catenin metabolism, Carcinoma, Hepatocellular genetics, Carcinoma, Hepatocellular pathology, Liver Neoplasms genetics, Liver Neoplasms pathology, Mutation
- Abstract
Liver cancer, primarily hepatocellular carcinoma, represents a major global health issue with significant clinical, economic, and psychological impacts. Its incidence continues to rise, driven by risk factors such as hepatitis B and C infections, nonalcoholic steatohepatitis, and various environmental influences. The Wnt/β-Catenin signaling pathway, frequently dysregulated in HCC, emerges as a promising therapeutic target. Critical genetic alterations, particularly in the CTNNB1 gene, involve mutations at key phosphorylation sites on β-catenin's N-terminal domain (S33, S37, T41, and S45) and in armadillo repeat domains (K335I and N387 K). These mutations impede β-catenin degradation, enhancing its oncogenic potential. In addition to genetic alterations, molecular and epigenetic mechanisms, including DNA methylation, histone modifications, and noncoding RNAs, further influence β-catenin signaling and tumor progression. However, β-catenin activation alone is insufficient for hepatocarcinogenesis; additional genetic "hits" are required for tumor initiation. Mutations or alterations in genes such as Ras, c-Met, NRF2, and LKB1, when combined with β-catenin activation, significantly contribute to HCC development and progression. Understanding these cooperative mutations provides crucial insights into the disease and reveals potential therapeutic strategies. The complex interplay between genetic variations and the tumor microenvironment, coupled with novel therapeutic approaches targeting the Wnt/β-Catenin pathway, offers promise for improved treatment of HCC. Despite advances, translating preclinical findings into clinical practice remains a challenge. Future research should focus on elucidating how specific β-catenin mutations and additional genetic alterations contribute to HCC pathogenesis, leveraging genetically clengineered mouse models to explore distinct signaling impacts, and identifying downstream targets. Relevant clinical trials will be essential for advancing personalized therapies and enhancing patient outcomes. This review provides a comprehensive analysis of β-Catenin signaling in HCC, highlighting its role in pathogenesis, diagnosis, and therapeutic targeting, and identifies key research directions to improve understanding and clinical outcomes., Competing Interests: Declaration of Conflicting InterestsThe author(s) declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: Anwaar Saeed reports consulting or advisory board role with AstraZeneca, Bristol-Myers Squibb, Merck, Exelixis, Pfizer, Xilio therapeutics, Taiho, Amgen, Autem therapeutics, KAHR medical, Autem therapeutics and Daiichi Sankyo; institutional research funding from AstraZeneca, Bristol-Myers Squibb, Merck, Clovis, Exelixis, Actuate therapeutics, Incyte Corporation, Daiichi Sankyo, Five prime therapeutics, Amgen, Innovent biologics, Dragonfly therapeutics, Oxford Biotherapeutics, Arcus therapeutics, and KAHR medical; and participation as a data safety monitoring board chair for Arcus therapeutics. Satdarshan Monga is on scientific advisory board or is a consultant for Vicero Inc., Mermaid Bio, UbiquiTx, and Alnylam Pharmaceuticals. He also has or has had sponsored research grants over last year from Alnylam and Fog Pharmaceuticals.
- Published
- 2024
- Full Text
- View/download PDF
18. Learning human liver biology in humanized mice.
- Author
-
Lehrich BM and Monga SP
- Subjects
- Humans, Mice, Animals, Biology, Liver, Hepatocytes
- Published
- 2024
- Full Text
- View/download PDF
19. Restoring glucose balance: Conditional HMGB1 knockdown mitigates hyperglycemia in a Streptozotocin induced mouse model.
- Author
-
Liu Z, Annarapu G, Yazdani HO, Wang Q, Liu S, Luo JH, Yu YP, Ren B, Neal MD, Monga SP, and Mota Alvidrez RI
- Abstract
Diabetes mellitus (DM) poses a significant global health burden, with hyperglycemia being a primary contributor to complications and high morbidity associated with this disorder. Existing glucose management strategies have shown suboptimal effectiveness, necessitating alternative approaches. In this study, we explored the role of high mobility group box 1 (HMGB1) in hyperglycemia, a protein implicated in initiating inflammation and strongly correlated with DM onset and progression. We hypothesized that HMGB1 knockdown will mitigate hyperglycemia severity and enhance glucose tolerance. To test this hypothesis, we utilized a novel inducible HMGB1 knockout (iHMGB1 KO) mouse model exhibiting systemic HMGB1 knockdown. Hyperglycemic phenotype was induced using low dose streptozotocin (STZ) injections, followed by longitudinal glucose measurements and oral glucose tolerance tests to evaluate the effect of HMGB1 knockdown on glucose metabolism. Our findings showed a substantial reduction in glucose levels and enhanced glucose tolerance in HMGB1 knockdown mice. Additionally, we performed RNA sequencing analyses, which identified potential alternations in genes and molecular pathways within the liver and skeletal muscle tissue that may account for the in vivo phenotypic changes observed in hyperglycemic mice following HMGB1 knockdown. In conclusion, our present study delivers the first direct evidence of a causal relationship between systemic HMGB1 knockdown and hyperglycemia in vivo, an association that had remained unexamined prior to this research. This discovery positions HMGB1 knockdown as a potentially efficacious therapeutic target for addressing hyperglycemia and, by extension, the DM epidemic. Furthermore, we have revealed potential underlying mechanisms, establishing the essential groundwork for subsequent in-depth mechanistic investigations focused on further elucidating and harnessing the promising therapeutic potential of HMGB1 in DM management., Competing Interests: The authors declare the following financial interests/personal relationships which may be considered as potential., (© 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
20. Wnt-β-catenin in hepatobiliary homeostasis, injury, and repair.
- Author
-
Nejak-Bowen K and Monga SP
- Subjects
- Wnt Signaling Pathway, Liver Regeneration, Homeostasis, beta Catenin metabolism, Liver pathology
- Abstract
Wnt-β-catenin signaling has emerged as an important regulatory pathway in the liver, playing key roles in zonation and mediating contextual hepatobiliary repair after injuries. In this review, we will address the major advances in understanding the role of Wnt signaling in hepatic zonation, regeneration, and cholestasis-induced injury. We will also touch on some important unanswered questions and discuss the relevance of modulating the pathway to provide therapies for complex liver pathologies that remain a continued unmet clinical need., (Copyright © 2023 American Association for the Study of Liver Diseases.)
- Published
- 2023
- Full Text
- View/download PDF
21. Defining spatiotemporal gene modules in liver regeneration using Analytical Dynamic Visual Spatial Omics Representation (ADViSOR).
- Author
-
Singh-Varma A, Shah AM, Liu S, Zamora R, Monga SP, and Vodovotz Y
- Subjects
- Humans, beta Catenin genetics, beta Catenin metabolism, Gene Regulatory Networks genetics, Wnt Signaling Pathway genetics, Liver Regeneration genetics, Focal Nodular Hyperplasia
- Abstract
Background: The liver is the only organ with the ability to regenerate following surgical or toxicant insults, and partial hepatectomy serves as an experimental model of liver regeneration (LR). Dynamic changes in gene expression occur from the periportal to pericentral regions of the liver following partial hepatectomy; thus, spatial transcriptomics, combined with a novel computational pipeline (ADViSOR [Analytic Dynamic Visual Spatial Omics Representation]), was employed to gain insights into the spatiotemporal molecular underpinnings of LR., Methods: ADViSOR, comprising Time-Interval Principal Component Analysis and sliding dynamic hypergraphs, was applied to spatial transcriptomics data on 100 genes assayed serially through LR, including key components of the Wnt/β-catenin pathway at critical timepoints after partial hepatectomy., Results: This computational pipeline identified key functional modules demonstrating cell signaling and cell-cell interactions, inferring shared regulatory mechanisms. Specifically, ADViSOR analysis suggested that macrophage-mediated inflammation is a critical component of early LR and confirmed prior studies showing that Ccnd1, a hepatocyte proliferative gene, is regulated by the Wnt/β-catenin pathway. These findings were subsequently validated through protein localization, which provided further confirmation and novel insights into the spatiotemporal changes in the Wnt/β-catenin pathway during LR., Conclusions: Thus, ADViSOR may yield novel insights in other complex, spatiotemporal contexts., (Copyright © 2023 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the American Association for the Study of Liver Diseases.)
- Published
- 2023
- Full Text
- View/download PDF
22. Huntingtin loss in hepatocytes is associated with altered metabolism, adhesion, and liver zonation.
- Author
-
Bragg RM, Coffey SR, Cantle JP, Hu S, Singh S, Legg SR, McHugh CA, Toor A, Zeitlin SO, Kwak S, Howland D, Vogt TF, Monga SP, and Carroll JB
- Subjects
- Animals, Mice, Acetaminophen, Phenotype, Liver, Hepatocytes
- Abstract
Huntington's disease arises from a toxic gain of function in the huntingtin ( HTT ) gene. As a result, many HTT-lowering therapies are being pursued in clinical studies, including those that reduce HTT RNA and protein expression in the liver. To investigate potential impacts, we characterized molecular, cellular, and metabolic impacts of chronic HTT lowering in mouse hepatocytes. Lifelong hepatocyte HTT loss is associated with multiple physiological changes, including increased circulating bile acids, cholesterol and urea, hypoglycemia, and impaired adhesion. HTT loss causes a clear shift in the normal zonal patterns of liver gene expression, such that pericentral gene expression is reduced. These alterations in liver zonation in livers lacking HTT are observed at the transcriptional, histological, and plasma metabolite levels. We have extended these phenotypes physiologically with a metabolic challenge of acetaminophen, for which the HTT loss results in toxicity resistance. Our data reveal an unexpected role for HTT in regulating hepatic zonation, and we find that loss of HTT in hepatocytes mimics the phenotypes caused by impaired hepatic β-catenin function., (© 2023 Bragg et al.)
- Published
- 2023
- Full Text
- View/download PDF
23. A Novel Transgenic Mouse Model Implicates Sirt2 as a Promoter of Hepatocellular Carcinoma.
- Author
-
Schmidt AV, Monga SP, Prochownik EV, and Goetzman ES
- Subjects
- Animals, Mice, Mice, Transgenic, Sirtuin 2 genetics, Carcinogens, Disease Models, Animal, Carcinoma, Hepatocellular genetics, Liver Neoplasms genetics
- Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer deaths globally. Incidence rates are steadily increasing, creating an unmet need for new therapeutic options. Recently, the inhibition of sirtuin-2 ( Sirt2 ) was proposed as a potential treatment for HCC, despite contradictory findings of its role as both a tumor promoter and suppressor in vitro. Sirt2 functions as a lysine deacetylase enzyme. However, little is known about its biological influence, despite its implication in several age-related diseases. This study evaluated Sirt2 's role in HCC in vivo using an inducible c-MYC transgene in Sirt2
+ / + and Sirt2-/- mice. Sirt2-/- HCC mice had smaller, less proliferative, and more differentiated liver tumors, suggesting that Sirt2 functions as a tumor promoter in this context. Furthermore, Sirt2-/- HCCs had significantly less c-MYC oncoprotein and reduction in c-MYC nuclear localization. The RNA-seq showed that only three genes were significantly dysregulated due to loss of Sirt2 , suggesting the underlying mechanism is due to Sirt2 -mediated changes in the acetylome, and that the therapeutic inhibition of Sirt2 would not perturb the oncogenic transcriptome. The findings of this study suggest that Sirt2 inhibition could be a promising molecular target for slowing HCC growth.- Published
- 2023
- Full Text
- View/download PDF
24. TMEM16A partners with mTOR to influence pathways of cell survival, proliferation, and migration in cholangiocarcinoma.
- Author
-
Kulkarni S, Li Q, Singhi AD, Liu S, Monga SP, and Feranchak AP
- Subjects
- Humans, Bile Ducts, Intrahepatic metabolism, Bile Ducts, Intrahepatic pathology, Cell Line, Tumor, Cell Movement, Cell Proliferation genetics, Cell Survival, Signal Transduction, Sirolimus metabolism, TOR Serine-Threonine Kinases metabolism, Bile Duct Neoplasms metabolism, Cholangiocarcinoma pathology
- Abstract
Expression of transmembrane protein 16 A (TMEM16A), a calcium activated chloride channel, is elevated in some human cancers and impacts tumor cell proliferation, metastasis, and patient outcome. Evidence presented here uncovers a molecular synergy between TMEM16A and mechanistic/mammalian target of rapamycin (mTOR), a serine-threonine kinase that is known to promote cell survival and proliferation in cholangiocarcinoma (CCA), a lethal cancer of the secretory cells of bile ducts. Analysis of gene and protein expression in human CCA tissue and CCA cell line detected elevated TMEM16A expression and Cl
- channel activity. The Cl- channel activity of TMEM16A impacted the actin cytoskeleton and the ability of cells to survive, proliferate, and migrate as revealed by pharmacological inhibition studies. The basal activity of mTOR, too, was elevated in the CCA cell line compared with the normal cholangiocytes. Molecular inhibition studies provided further evidence that TMEM16A and mTOR were each able to influence the regulation of the other's activity or expression respectively. Consistent with this reciprocal regulation, combined TMEM16A and mTOR inhibition produced a greater loss of CCA cell survival and migration than their individual inhibition alone. Together these data reveal that the aberrant TMEM16A expression and cooperation with mTOR contribute to a certain advantage in CCA. NEW & NOTEWORTHY This study points to the dysregulation of transmembrane protein 16 A (TMEM16A) expression and activity in cholangiocarcinoma (CCA), the inhibition of which has functional consequences. Dysregulated TMEM16A exerts an influence on the regulation of mechanistic/mammalian target of rapamycin (mTOR) activity. Moreover, the reciprocal regulation of TMEM16A by mTOR demonstrates a novel connection between these two protein families. These findings support a model in which TMEM16A intersects the mTOR pathway to regulate cell cytoskeleton, survival, proliferation, and migration in CCA.- Published
- 2023
- Full Text
- View/download PDF
25. Lysosomal mitochondrial interaction promotes tumor growth in squamous cell carcinoma of the head and neck.
- Author
-
Gopalkrishnan A, Wang N, Cruz-Rangel S, Kassab AY, Shiva S, Kurukulasuriya C, Monga SP, DeBerardinis RJ, Kiselyov K, and Duvvuri U
- Abstract
Tumor growth and proliferation are regulated by numerous mechanisms. Communication between intracellular organelles has recently been shown to regulate cellular proliferation and fitness. The way lysosomes and mitochondria communicate with each other (lysosomal/mitochondrial interaction) is emerging as a major determinant of tumor proliferation and growth. About 30% of squamous carcinomas (including squamous cell carcinoma of the head and neck, SCCHN) overexpress TMEM16A, a calcium-activated chloride channel, which promotes cellular growth and negatively correlates with patient survival. TMEM16A has recently been shown to drive lysosomal biogenesis, but its impact on mitochondrial function is unclear. Here, we show that (1) patients with high TMEM16A SCCHN display increased mitochondrial content specifically complex I; (2) In vitro and in vivo models uniquely depend on mitochondrial complex I activity for growth and survival; (3) β-catenin/NRF2 signaling is a critical linchpin that drives mitochondrial biogenesis, and (4) mitochondrial complex I and lysosomal function are codependent for proliferation. Taken together, our data demonstrate that LMI drives tumor proliferation and facilitates a functional interaction between lysosomes and mitochondria. Therefore, inhibition of LMI may serve as a therapeutic strategy for patients with SCCHN.
- Published
- 2023
- Full Text
- View/download PDF
26. Western diet unmasks transient low-level vinyl chloride-induced tumorigenesis; potential role of the (epi-)transcriptome.
- Author
-
Liu S, He L, Bannister OB, Li J, Schnegelberger RD, Vanderpuye CM, Althouse AD, Schopfer FJ, Wahlang B, Cave MC, Monga SP, Zhang X, Arteel GE, and Beier JI
- Subjects
- Mice, Animals, Transcriptome, Diet, Western, Liver metabolism, Carcinogenesis metabolism, Cell Transformation, Neoplastic metabolism, Vinyl Chloride toxicity, Vinyl Chloride metabolism, Carcinoma, Hepatocellular pathology, Liver Neoplasms chemically induced, Liver Neoplasms genetics, Liver Neoplasms metabolism, Non-alcoholic Fatty Liver Disease metabolism
- Abstract
Background & Aims: Vinyl chloride (VC) monomer is a volatile organic compound commonly used in industry. At high exposure levels, VC causes liver cancer and toxicant-associated steatohepatitis. However, lower exposure levels (i.e., sub-regulatory exposure limits) that do not directly damage the liver, enhance injury caused by Western diet (WD). It is still unknown if the long-term impact of transient low-concentration VC enhances the risk of liver cancer development. This is especially a concern given that fatty liver disease is in and of itself a risk factor for the development of liver cancer., Methods: C57Bl/6 J mice were fed WD or control diet (CD) for 1 year. During the first 12 weeks of feeding only, mice were also exposed to VC via inhalation at sub-regulatory limit concentrations (<1 ppm) or air for 6 h/day, 5 days/week., Results: Feeding WD for 1 year caused significant hepatic injury, which was exacerbated by VC. Additionally, VC increased the number of tumors which ranged from moderately to poorly differentiated hepatocellular carcinoma (HCC). Transcriptomic analysis demonstrated VC-induced changes in metabolic but also ribosomal processes. Epitranscriptomic analysis showed a VC-induced shift of the modification pattern that has been associated with metabolic disease, mitochondrial dysfunction, and cancer., Conclusions: These data indicate that VC sensitizes the liver to other stressors (e.g., WD), resulting in enhanced tumorigenesis. These data raise concerns about potential interactions between VC exposure and WD. It also emphasizes that current safety restrictions may be insufficient to account for other factors that can influence hepatotoxicity., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Juliane Beier reports financial support was provided by National Institutes of Health., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
27. Differential requirement of Hippo cascade during CTNNB1 or AXIN1 mutation-driven hepatocarcinogenesis.
- Author
-
Liang B, Wang H, Qiao Y, Wang X, Qian M, Song X, Zhou Y, Zhang Y, Shang R, Che L, Chen Y, Huang Z, Wu H, Monga SP, Zeng Y, Calvisi DF, Chen X, and Chen X
- Subjects
- Humans, Mice, Animals, beta Catenin genetics, Carcinogenesis genetics, Mutation, Wnt Signaling Pathway genetics, Protein Serine-Threonine Kinases metabolism, Tumor Suppressor Proteins metabolism, Axin Protein genetics, Carcinoma, Hepatocellular pathology, Liver Neoplasms pathology
- Abstract
Background and Aims: Gain-of-function (GOF) mutations of CTNNB1 and loss-of-function (LOF) mutations of AXIN1 are recurrent genetic alterations in hepatocellular carcinoma (HCC). We aim to investigate the functional contribution of Hippo/YAP/TAZ in GOF CTNNB1 or LOF AXIN1 mutant HCCs., Approach and Results: The requirement of YAP/TAZ in c-Met/β-Catenin and c-Met/sgAxin1-driven HCC was analyzed using conditional Yap , Taz , and Yap;Taz knockout (KO) mice. Mechanisms of AXIN1 in regulating YAP/TAZ were investigated using AXIN1 mutated HCC cells. Hepatocyte-specific inducible TTR-CreER T2KO system was applied to evaluate the role of Yap;Taz during tumor progression. Cabozantinib and G007-LK combinational treatment were tested in vitro and in vivo . Nuclear YAP/TAZ was strongly induced in c-Met/sgAxin1 mouse HCC cells. Activation of Hippo via overexpression of Lats2 or concomitant deletion of Yap and Taz significantly inhibited c-Met/sgAxin1 driven HCC development, whereas the same approaches had mild effects in c-Met/β-Catenin HCCs. YAP is the major Hippo effector in c-Met/β-Catenin HCCs, and both YAP and TAZ are required for c-Met/sgAxin1-dependent hepatocarcinogenesis. Mechanistically, AXIN1 binds to YAP/TAZ in human HCC cells and regulates YAP/TAZ stability. Genetic deletion of YAP/TAZ suppresses already formed c-Met/sgAxin1 liver tumors, supporting the requirement of YAP/TAZ during tumor progression. Importantly, tankyrase inhibitor G007-LK, which targets Hippo and Wnt pathways, synergizes with cabozantinib, a c-MET inhibitor, leading to tumor regression in the c-Met/sgAxin1 HCC model., Conclusions: Our studies demonstrate that YAP/TAZ are major signaling molecules downstream of LOF AXIN1 mutant HCCs, and targeting YAP/TAZ is an effective treatment against AXIN1 mutant human HCCs., (Copyright © 2023 American Association for the Study of Liver Diseases.)
- Published
- 2023
- Full Text
- View/download PDF
28. Selective targeting of α 4 β 7 /MAdCAM-1 axis suppresses liver fibrosis by reducing proinflammatory T cell recruitment to the liver.
- Author
-
Gupta B, Rai RP, Pal PB, Chaudhary S, Chiaro A, Seaman S, Singhi AD, Monga SP, Iyer SS, and Raeman R
- Abstract
Integrin α
4 β7 + T cells perpetuate tissue injury in chronic inflammatory diseases, yet their role in hepatic fibrosis progression remains poorly understood. Here we report increased accumulation of α4 β7 + T cells in the liver of people with cirrhosis relative to disease controls. Similarly, hepatic fibrosis in the established mouse model of CCl4 -induced liver fibrosis was associated with enrichment of intrahepatic α4 β7 + CD4 and CD8 T cells. Monoclonal antibody (mAb)-mediated blockade of α4 β7 or its ligand mucosal addressin cell adhesion molecule (MAdCAM)-1 attenuated hepatic inflammation and prevented fibrosis progression in CCl4 treated mice. Improvement in liver fibrosis was associated with a significant decrease in the infiltration of α4 β7 + CD4 and CD8 T cells suggesting that α4 β7 /MAdCAM-1 axis regulates both CD4 and CD8 T cell recruitment to the fibrotic liver, and α4 β7 + T cells promote hepatic fibrosis progression. Analysis of hepatic α4 β7 + and α4 β7 -CD4 T cells revealed that α4 β7 + CD4 T cells enriched for markers of activation and proliferation demonstrating an effector phenotype. Notably, blockade of α4 β7 or MAdCAM-1 did not affect the recruitment of Foxp3+ regulatory T cells, demonstrating the specificity of α4 β7 /MAdCAM-1 axis in regulating effector T cell recruitment to the liver. The findings suggest that α4 β7 + T cells play a critical role in promoting hepatic fibrosis progression, and mAb-mediated blockade of α4 β7 or MAdCAM-1 represents a promising therapeutic strategy for slowing hepatic fibrosis progression in chronic liver diseases.- Published
- 2023
- Full Text
- View/download PDF
29. Hepatic stellate cell stearoyl co-A desaturase activates leukotriene B4 receptor 2 - β-catenin cascade to promote liver tumorigenesis.
- Author
-
Sinha S, Aizawa S, Nakano Y, Rialdi A, Choi HY, Shrestha R, Pan SQ, Chen Y, Li M, Kapelanski-Lamoureux A, Yochum G, Sher L, Monga SP, Lazaris A, Machida K, Karin M, Guccione E, and Tsukamoto H
- Subjects
- Animals, Male, Mice, beta Catenin metabolism, Carcinogenesis genetics, Carcinogenesis metabolism, Fatty Acid Desaturases, Hepatic Stellate Cells metabolism, Receptors, Leukotriene B4 genetics, Receptors, Leukotriene B4 metabolism, Tumor Microenvironment, Carcinoma, Hepatocellular metabolism, Liver Neoplasms metabolism
- Abstract
Hepatocellular carcinoma (HCC) is the 3
rd most deadly malignancy. Activated hepatic stellate cells (aHSC) give rise to cancer-associated fibroblasts in HCC and are considered a potential therapeutic target. Here we report that selective ablation of stearoyl CoA desaturase-2 (Scd2) in aHSC globally suppresses nuclear CTNNB1 and YAP1 in tumors and tumor microenvironment and prevents liver tumorigenesis in male mice. Tumor suppression is associated with reduced leukotriene B4 receptor 2 (LTB4R2) and its high affinity oxylipin ligand, 12-hydroxyheptadecatrienoic acid (12-HHTrE). Genetic or pharmacological inhibition of LTB4R2 recapitulates CTNNB1 and YAP1 inactivation and tumor suppression in culture and in vivo. Single cell RNA sequencing identifies a subset of tumor-associated aHSC expressing Cyp1b1 but no other 12-HHTrE biosynthetic genes. aHSC release 12-HHTrE in a manner dependent on SCD and CYP1B1 and their conditioned medium reproduces the LTB4R2-mediated tumor-promoting effects of 12-HHTrE in HCC cells. CYP1B1-expressing aHSC are detected in proximity of LTB4R2-positive HCC cells and the growth of patient HCC organoids is blunted by LTB4R2 antagonism or knockdown. Collectively, our findings suggest aHSC-initiated 12-HHTrE-LTB4R2-CTNNB1-YAP1 pathway as a potential HCC therapeutic target., (© 2023. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)- Published
- 2023
- Full Text
- View/download PDF
30. Reply.
- Author
-
Hu S and Monga SP
- Published
- 2023
- Full Text
- View/download PDF
31. Hepatocyte β-catenin loss is compensated by Insulin-mTORC1 activation to promote liver regeneration.
- Author
-
Hu S, Cao C, Poddar M, Delgado E, Singh S, Singh-Varma A, Stolz DB, Bell A, and Monga SP
- Subjects
- Mice, Animals, Mechanistic Target of Rapamycin Complex 1 metabolism, Insulin metabolism, Hepatocytes metabolism, TOR Serine-Threonine Kinases metabolism, Wnt Signaling Pathway physiology, Mice, Knockout, Cell Proliferation, Sirolimus pharmacology, Liver Regeneration physiology, beta Catenin metabolism
- Abstract
Background and Aims: Liver regeneration (LR) following partial hepatectomy (PH) occurs via activation of various signaling pathways. Disruption of a single pathway can be compensated by activation of another pathway to continue LR. The Wnt-β-catenin pathway is activated early during LR and conditional hepatocyte loss of β-catenin delays LR. Here, we study mechanism of LR in the absence of hepatocyte-β-catenin., Approach and Results: Eight-week-old hepatocyte-specific Ctnnb1 knockout mice (β-catenin ΔHC ) were subjected to PH. These animals exhibited decreased hepatocyte proliferation at 40-120 h and decreased cumulative 14-day BrdU labeling of <40%, but all mice survived, suggesting compensation. Insulin-mediated mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) activation was uniquely identified in the β-catenin ΔHC mice at 72-96 h after PH. Deletion of hepatocyte regulatory-associated protein of mTOR (Raptor), a critical mTORC1 partner, in the β-catenin ΔHC mice led to progressive hepatic injury and mortality by 30 dys. PH on early stage nonmorbid Raptor ΔHC -β-catenin ΔHC mice led to lethality by 12 h. Raptor ΔHC mice showed progressive hepatic injury and spontaneous LR with β-catenin activation but died by 40 days. PH on early stage nonmorbid Raptor ΔHC mice was lethal by 48 h. Temporal inhibition of insulin receptor and mTORC1 in β-catenin ΔHC or controls after PH was achieved by administration of linsitinib at 48 h or rapamycin at 60 h post-PH and completely prevented LR leading to lethality by 12-14 days., Conclusions: Insulin-mTORC1 activation compensates for β-catenin loss to enable LR after PH. mTORC1 signaling in hepatocytes itself is critical to both homeostasis and LR and is only partially compensated by β-catenin activation. Dual inhibition of β-catenin and mTOR may have notable untoward hepatotoxic side effects., (Copyright © 2023 American Association for the Study of Liver Diseases.)
- Published
- 2023
- Full Text
- View/download PDF
32. A Spatial Atlas of Wnt Receptors in Adult Mouse Liver.
- Author
-
Gayden J, Hu S, Joseph PN, Delgado E, Liu S, Bell A, Puig S, Monga SP, and Freyberg Z
- Subjects
- Mice, Animals, In Situ Hybridization, Fluorescence, Endothelial Cells metabolism, Frizzled Receptors genetics, Frizzled Receptors metabolism, Liver metabolism, Wnt Signaling Pathway, RNA, Messenger genetics, RNA, Messenger metabolism, beta Catenin metabolism, Receptors, Wnt genetics, Receptors, Wnt metabolism, Wnt Proteins genetics
- Abstract
Hepatic zonation is critical for most metabolic functions in liver. Wnt signaling plays an important role in establishing and maintaining liver zonation. Yet, the anatomic expression of Wnt signaling components, especially all 10 Frizzled (Fzd) receptors, has not been characterized in adult liver. To address this, the spatial expression of Fzd receptors was quantitatively mapped in adult mouse liver via multiplex fluorescent in situ hybridization. Although all 10 Fzd receptors were expressed within a metabolic unit, Fzd receptors 1, 4, and 6 were the highest expressed. Although most Wnt signaling occurs in zone 3, expression of most Fzd receptors was not zonated. In contrast, Fzd receptor 6 was preferentially expressed in zone 1. Wnt2 and Wnt9b expression was highly zonated and primarily found in zone 3. Therefore, the current results suggest that zonated Wnt/β-catenin signaling at baseline occurs primarily due to Wnt2 and Wnt9b rather than zonation of Fzd mRNA expression. Finally, the study showed that Fzd receptors and Wnts are not uniformly expressed by all hepatic cell types. Instead, there is broad distribution among both hepatocytes and nonparenchymal cells, including endothelial cells. Overall, this establishment of a definitive mRNA expression atlas, especially of Fzd receptors, opens the door to future functional characterization in healthy and diseased liver states., (Copyright © 2023 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
33. Western diet dampens T regulatory cell function to fuel hepatic inflammation in nonalcoholic fatty liver disease.
- Author
-
Chaudhary S, Rai R, Pal PB, Tedesco D, Singhi AD, Monga SP, Grakoui A, Iyer SS, and Raeman R
- Abstract
Background and Aims: The immunosuppressive T regulatory cells (Tregs) regulate immune responses and maintain immune homeostasis, yet their functions in nonalcoholic fatty liver disease (NAFLD) pathogenesis remains controversial., Methods: Mice were fed a normal diet (ND) or a western diet (WD) for 16 weeks to induce NAFLD. Diphtheria toxin injection to deplete Tregs in Foxp3
DTR mice or Treg induction therapy in WT mice to augment Treg numbers was initiated at twelve and eight weeks, respectively. Liver tissues from mice and NASH human subjects were analyzed by histology, confocal imaging, and qRT-PCR., Results: WD triggered accumulation of adaptive immune cells, including Tregs and effector T cells, within the liver parenchyma. This pattern was also observed in NASH patients, where an increase in intrahepatic Tregs was noted. In the absence of adaptive immune cells in Rag1 KO mice, WD promoted accumulation of intrahepatic neutrophils and macrophages and exacerbated hepatic inflammation and fibrosis. Similarly, targeted Treg depletion exacerbated WD-induced hepatic inflammation and fibrosis. In Treg-depleted mice, hepatic injury was associated with increased accumulation of neutrophils, macrophages, and activated T cells within the liver. Conversely, induction of Tregs using recombinant IL2/αIL2 mAb cocktail reduced hepatic steatosis, inflammation, and fibrosis in WD-fed mice. Analysis of intrahepatic Tregs from WD-fed mice revealed a phenotypic signature of impaired Treg function in NAFLD. Ex vivo functional studies showed that glucose and palmitate, but not fructose, impaired the immunosuppressive ability of Treg cells., Conclusions: Our findings indicate that the liver microenvironment in NAFLD impairs ability of Tregs to suppress effector immune cell activation, thus perpetuating chronic inflammation and driving NAFLD progression. These data suggest that targeted approaches aimed at restoring Treg function may represent a potential therapeutic strategy for treating NAFLD., Lay Summary: In this study, we elucidate the mechanisms contributing to the perpetuation of chronic hepatic inflammation in nonalcoholic fatty liver disease (NAFLD). We show that dietary sugar and fatty acids promote chronic hepatic inflammation in NAFLD by impairing immunosuppressive function of regulatory T cells. Finally, our preclinical data suggest that targeted approaches aimed at restoring T regulatory cell function have the potential to treat NAFLD.- Published
- 2023
- Full Text
- View/download PDF
34. Author Correction: Defective HNF4alpha-dependent gene expression as a driver of hepatocellular failure in alcoholic hepatitis.
- Author
-
Argemi J, Latasa MU, Atkinson SR, Blokhin IO, Massey V, Gue JP, Cabezas J, Lozano JJ, Van Booven D, Bell A, Cao S, Vernetti LA, Arab JP, Ventura-Cots M, Edmunds LR, Fondevila C, Stärkel P, Dubuquoy L, Louvet A, Odena G, Gomez JL, Aragon T, Altamirano J, Caballeria J, Jurczak MJ, Taylor DL, Berasain C, Wahlestedt C, Monga SP, Morgan MY, Sancho-Bru P, Mathurin P, Furuya S, Lackner C, Rusyn I, Shah VH, Thursz MR, Mann J, Avila MA, and Bataller R
- Published
- 2023
- Full Text
- View/download PDF
35. Inhibition of Heat Shock Factor 1 Signaling Decreases Hepatoblastoma Growth via Induction of Apoptosis.
- Author
-
Hurley EH, Tao J, Liu S, Krutsenko Y, Singh S, and Monga SP
- Subjects
- Animals, Mice, DNA-Binding Proteins metabolism, Heat Shock Transcription Factors, Apoptosis, Heat-Shock Response, Hepatoblastoma, Liver Neoplasms
- Abstract
Although rare compared with adult liver cancers, hepatoblastoma (HB) is the most common pediatric liver malignancy, and its incidence is increasing. Currently, the treatment includes surgical resection with or without chemotherapy, and in severe cases, liver transplantation in children. The effort to develop more targeted, HB-specific therapies has been stymied by the lack of fundamental knowledge about HB biology. Heat shock factor 1 (HSF1), a transcription factor, is a canonical inducer of heat shock proteins, which act as chaperone proteins to prevent or undo protein misfolding. Recent work has shown a role for HSF1 in cancer beyond the canonical heat shock response. The current study found increased HSF1 signaling in HB versus normal liver. It showed that less differentiated, more embryonic tumors had higher levels of HSF1 than more differentiated, more fetal-appearing tumors. Most strikingly, HSF1 expression levels correlated with mortality. This study used a mouse model of HB to test the effect of inhibiting HSF1 early in tumor development on cancer growth. HSF1 inhibition resulted in fewer and smaller tumors, suggesting HSF1 is needed for aggressive tumor growth. Moreover, HSF1 inhibition also increased apoptosis in tumor foci. These data suggest that HSF1 may be a viable pharmacologic target for HB treatment., (Copyright © 2023 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
36. Ductular Reaction and Liver Regeneration: Fulfilling the Prophecy of Prometheus!
- Author
-
Monga SP and Nejak-Bowen K
- Subjects
- Humans, Hyperplasia pathology, Liver Regeneration, Liver pathology
- Published
- 2023
- Full Text
- View/download PDF
37. Single-cell spatial transcriptomics reveals a dynamic control of metabolic zonation and liver regeneration by endothelial cell Wnt2 and Wnt9b.
- Author
-
Hu S, Liu S, Bian Y, Poddar M, Singh S, Cao C, McGaughey J, Bell A, Blazer LL, Adams JJ, Sidhu SS, Angers S, and Monga SP
- Subjects
- Humans, beta Catenin genetics, Endothelial Cells metabolism, Transcriptome, Wnt Proteins genetics, Acetaminophen metabolism, Wnt2 Protein genetics, Liver Regeneration genetics, Focal Nodular Hyperplasia metabolism
- Abstract
The conclusive identity of Wnts regulating liver zonation (LZ) and regeneration (LR) remains unclear despite an undisputed role of β-catenin. Using single-cell analysis, we identified a conserved Wnt2 and Wnt9b expression in endothelial cells (ECs) in zone 3. EC-elimination of Wnt2 and Wnt9b led to both loss of β-catenin targets in zone 3, and re-appearance of zone 1 genes in zone 3, unraveling dynamicity in the LZ process. Impaired LR observed in the knockouts phenocopied models of defective hepatic Wnt signaling. Administration of a tetravalent antibody to activate Wnt signaling rescued LZ and LR in the knockouts and induced zone 3 gene expression and LR in controls. Administration of the agonist also promoted LR in acetaminophen overdose acute liver failure (ALF) fulfilling an unmet clinical need. Overall, we report an unequivocal role of EC-Wnt2 and Wnt9b in LZ and LR and show the role of Wnt activators as regenerative therapy for ALF., Competing Interests: Declaration of interests S.P.M. is a consultant for Surrozen. L.L.B., J.J.A., S.S.S., and S.A. are shareholders of AntlerA Therapeutics., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
38. β-Catenin Sustains and Is Required for YES-associated Protein Oncogenic Activity in Cholangiocarcinoma.
- Author
-
Zhang Y, Xu H, Cui G, Liang B, Chen X, Ko S, Affo S, Song X, Liao Y, Feng J, Wang P, Wang H, Xu M, Wang J, Pes GM, Ribback S, Zeng Y, Singhi A, Schwabe RF, Monga SP, Evert M, Tang L, Calvisi DF, and Chen X
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Animals, Bile Ducts, Intrahepatic pathology, Carcinogenesis, Humans, Mice, Proto-Oncogene Proteins c-akt metabolism, Transcription Factors genetics, Transcription Factors metabolism, Bile Duct Neoplasms genetics, Bile Duct Neoplasms pathology, Cholangiocarcinoma genetics, Cholangiocarcinoma pathology, YAP-Signaling Proteins genetics, YAP-Signaling Proteins metabolism, beta Catenin genetics, beta Catenin metabolism
- Abstract
Background & Aims: YES-associated protein (YAP) aberrant activation is implicated in intrahepatic cholangiocarcinoma (iCCA). Transcriptional enhanced associate domain (TEAD)-mediated transcriptional regulation is the primary signaling event downstream of YAP. The role of Wnt/β-Catenin signaling in cholangiocarcinogenesis remains undetermined. Here, we investigated the possible molecular interplay between YAP and β-Catenin cascades in iCCA., Methods: Activated AKT (Myr-Akt) was coexpressed with YAP (YapS127A) or Tead2VP16 via hydrodynamic tail vein injection into mouse livers. Tumor growth was monitored, and liver tissues were collected and analyzed using histopathologic and molecular analysis. YAP, β-Catenin, and TEAD interaction in iCCAs was investigated through coimmunoprecipitation. Conditional Ctnnb1 knockout mice were used to determine β-Catenin function in murine iCCA models. RNA sequencing was performed to analyze the genes regulated by YAP and/or β-Catenin. Immunostaining of total and nonphosphorylated/activated β-Catenin staining was performed in mouse and human iCCAs., Results: We discovered that TEAD factors are required for YAP-dependent iCCA development. However, transcriptional activation of TEADs did not fully recapitulate YAP's activities in promoting cholangiocarcinogenesis. Notably, β-Catenin physically interacted with YAP in human and mouse iCCA. Ctnnb1 ablation strongly suppressed human iCCA cell growth and Yap-dependent cholangiocarcinogenesis. Furthermore, RNA-sequencing analysis revealed that YAP/ transcriptional coactivator with PDZ-binding motif (TAZ) regulate a set of genes significantly overlapping with those controlled by β-Catenin. Importantly, activated/nonphosphorylated β-Catenin was detected in more than 80% of human iCCAs., Conclusion: YAP induces cholangiocarcinogenesis via TEAD-dependent transcriptional activation and interaction with β-Catenin. β-Catenin binds to YAP in iCCA and is required for YAP full transcriptional activity, revealing the functional crosstalk between YAP and β-Catenin pathways in cholangiocarcinogenesis., (Copyright © 2022 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
39. NOTCH-YAP1/TEAD-DNMT1 Axis Drives Hepatocyte Reprogramming Into Intrahepatic Cholangiocarcinoma.
- Author
-
Hu S, Molina L, Tao J, Liu S, Hassan M, Singh S, Poddar M, Bell A, Sia D, Oertel M, Raeman R, Nejak-Bowen K, Singhi A, Luo J, Monga SP, and Ko S
- Subjects
- Bile Ducts, Intrahepatic pathology, Hepatocytes metabolism, Humans, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction genetics, YAP-Signaling Proteins, Bile Duct Neoplasms pathology, Cholangiocarcinoma pathology, Cholestasis pathology
- Abstract
Background & Aims: Intrahepatic cholangiocarcinoma (ICC) is a devastating liver cancer with extremely high intra- and inter-tumoral molecular heterogeneity, partly due to its diverse cellular origins. We investigated clinical relevance and the molecular mechanisms underlying hepatocyte (HC)-driven ICC development., Methods: Expression of ICC driver genes in human diseased livers at risk for ICC development were examined. The sleeping beauty and hydrodynamic tail vein injection based Akt-NICD/YAP1 ICC model was used to investigate pathogenetic roles of SRY-box transcription factor 9 (SOX9) and yes-associated protein 1 (YAP1) in HC-driven ICC. We identified DNA methyltransferase 1 (DNMT1) as a YAP1 target, which was validated by loss- and gain-of-function studies, and its mechanism addressed by chromatin immunoprecipitation sequencing., Results: Co-expression of AKT and Notch intracellular domain (NICD)/YAP1 in HC yielded ICC that represents 13% to 29% of clinical ICC. NICD independently regulates SOX9 and YAP1 and deletion of either, significantly delays ICC development. Yap1 or TEAD inhibition, but not Sox9 deletion, impairs HC-to-biliary epithelial cell (BEC) reprogramming. DNMT1 was discovered as a novel downstream effector of YAP1-TEAD complex that directs HC-to-BEC/ICC fate switch through the repression of HC-specific genes regulated by master regulators for HC differentiation, including hepatocyte nuclear factor 4 alpha, hepatocyte nuclear factor 1 alpha, and CCAAT/enhancer-binding protein alpha/beta. DNMT1 loss prevented NOTCH/YAP1-dependent HC-driven cholangiocarcinogenesis, and DNMT1 re-expression restored ICC development following TEAD repression. Co-expression of DNMT1 with AKT was sufficient to induce tumor development including ICC. DNMT1 was detected in a subset of HCs and dysplastic BECs in cholestatic human livers prone to ICC development., Conclusion: We identified a novel NOTCH-YAP1/TEAD-DNMT1 axis essential for HC-to-BEC/ICC conversion, which may be relevant in cholestasis-to-ICC pathogenesis in the clinic., (Copyright © 2022 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
40. Myeloid FoxO1 depletion attenuates hepatic inflammation and prevents nonalcoholic steatohepatitis.
- Author
-
Lee S, Usman TO, Yamauchi J, Chhetri G, Wang X, Coudriet GM, Zhu C, Gao J, McConnell R, Krantz K, Rajasundaram D, Singh S, Piganelli J, Ostrowska A, Soto-Gutierrez A, Monga SP, Singhi AD, Muzumdar R, Tsung A, and Dong HH
- Subjects
- Animals, Diet, High-Fat adverse effects, Disease Models, Animal, Fibrosis, Inflammation metabolism, Liver metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Forkhead Box Protein O1 genetics, Forkhead Box Protein O1 metabolism, Non-alcoholic Fatty Liver Disease chemically induced, Non-alcoholic Fatty Liver Disease genetics, Non-alcoholic Fatty Liver Disease prevention & control, Overnutrition pathology
- Abstract
Hepatic inflammation is culpable for the evolution of asymptomatic steatosis to nonalcoholic steatohepatitis (NASH). Hepatic inflammation results from abnormal macrophage activation. We found that FoxO1 links overnutrition to hepatic inflammation by regulating macrophage polarization and activation. FoxO1 was upregulated in hepatic macrophages, correlating with hepatic inflammation, steatosis, and fibrosis in mice and patients with NASH. Myeloid cell conditional FoxO1 knockout skewed macrophage polarization from proinflammatory M1 to the antiinflammatory M2 phenotype, accompanied by a reduction in macrophage infiltration in liver. These effects mitigated overnutrition-induced hepatic inflammation and insulin resistance, contributing to improved hepatic metabolism and increased energy expenditure in myeloid cell FoxO1-knockout mice on a high-fat diet. When fed a NASH-inducing diet, myeloid cell FoxO1-knockout mice were protected from developing NASH, culminating in a reduction in hepatic inflammation, steatosis, and fibrosis. Mechanistically, FoxO1 counteracts Stat6 to skew macrophage polarization from M2 toward the M1 signature to perpetuate hepatic inflammation in NASH. FoxO1 appears to be a pivotal mediator of macrophage activation in response to overnutrition and a therapeutic target for ameliorating hepatic inflammation to stem the disease progression from benign steatosis to NASH.
- Published
- 2022
- Full Text
- View/download PDF
41. A Quantitative Systems Pharmacology Platform Reveals NAFLD Pathophysiological States and Targeting Strategies.
- Author
-
Lefever DE, Miedel MT, Pei F, DiStefano JK, Debiasio R, Shun TY, Saydmohammed M, Chikina M, Vernetti LA, Soto-Gutierrez A, Monga SP, Bataller R, Behari J, Yechoor VK, Bahar I, Gough A, Stern AM, and Taylor DL
- Abstract
Non-alcoholic fatty liver disease (NAFLD) has a high global prevalence with a heterogeneous and complex pathophysiology that presents barriers to traditional targeted therapeutic approaches. We describe an integrated quantitative systems pharmacology (QSP) platform that comprehensively and unbiasedly defines disease states, in contrast to just individual genes or pathways, that promote NAFLD progression. The QSP platform can be used to predict drugs that normalize these disease states and experimentally test predictions in a human liver acinus microphysiology system (LAMPS) that recapitulates key aspects of NAFLD. Analysis of a 182 patient-derived hepatic RNA-sequencing dataset generated 12 gene signatures mirroring these states. Screening against the LINCS L1000 database led to the identification of drugs predicted to revert these signatures and corresponding disease states. A proof-of-concept study in LAMPS demonstrated mitigation of steatosis, inflammation, and fibrosis, especially with drug combinations. Mechanistically, several structurally diverse drugs were predicted to interact with a subnetwork of nuclear receptors, including pregnane X receptor (PXR; NR1I2), that has evolved to respond to both xenobiotic and endogenous ligands and is intrinsic to NAFLD-associated transcription dysregulation. In conjunction with iPSC-derived cells, this platform has the potential for developing personalized NAFLD therapeutic strategies, informing disease mechanisms, and defining optimal cohorts of patients for clinical trials.
- Published
- 2022
- Full Text
- View/download PDF
42. Chronic Activation of LXRα Sensitizes Mice to Hepatocellular Carcinoma.
- Author
-
Xie Y, Sun R, Gao L, Guan J, Wang J, Bell A, Zhu J, Zhang M, Xu M, Lu P, Cai X, Ren S, Xu P, Monga SP, Ma X, Yang D, Liu Y, Lu B, and Xie W
- Subjects
- Animals, Cell Transformation, Neoplastic genetics, Interleukin-6, Mice, Mice, Transgenic, Carcinoma, Hepatocellular chemically induced, Liver Neoplasms chemically induced, Oxysterols
- Abstract
The oxysterol receptor liver X receptor (LXR) is a nuclear receptor best known for its function in the regulation of lipid and cholesterol metabolism. LXRs, both the α and β isoforms, have been suggested as potential therapeutic targets for several cancer types. However, there was a lack of report on whether and how LXRα plays a role in the development of hepatocellular carcinoma (HCC). In the current study, we found that systemic activation of LXRα in the VP-LXRα knock-in (LXRαKI) mice or hepatocyte-specific activation of LXRα in the VP-LXRα transgenic mice sensitized mice to liver tumorigenesis induced by the combined treatment of diethylnitrosamine (DEN) and 3,3',5,5'-tetrachloro-1,4-bis (pyridyloxy) benzene (TCPOBOP). Mechanistically, the LXRα-responsive up-regulation of interleukin-6 (IL-6)/signal transducer and activator of transcription 3 (STAT3) signaling pathway and the complement system, and down-regulation of bile acid metabolism, may have contributed to increased tumorigenesis. Accumulations of secondary bile acids and oxysterols were found in both the serum and liver tissue of LXRα activated mice. We also observed an induction of monocytic myeloid-derived suppressor cells accompanied by down-regulation of dendritic cells and cytotoxic T cells in DEN/TCPOBOP-induced liver tumors, indicating that chronic activation of LXRα may have led to the activation of innate immune suppression. The HCC sensitizing effect of LXRα activation was also observed in the c-MYC driven HCC model. Conclusion: Our results indicated that chronic activation of LXRα promotes HCC, at least in part, by promoting innate immune suppressor as a result of accumulation of oxysterols, as well as up-regulation of the IL-6/Janus kinase/STAT3 signaling and complement pathways., (© 2022 The Authors. Hepatology Communications published by Wiley Periodicals, Inc., on behalf of the American Association for the Study of Liver Diseases.)
- Published
- 2022
- Full Text
- View/download PDF
43. Inhibition of p53 Sulfoconjugation Prevents Oxidative Hepatotoxicity and Acute Liver Failure.
- Author
-
Xu P, Xi Y, Wang P, Luka Z, Xu M, Tung HC, Wang J, Ren S, Feng D, Gao B, Singhi AD, Monga SP, York JD, Ma X, Huang Z, and Xie W
- Subjects
- Acetaminophen toxicity, Animals, Humans, Liver metabolism, Mammals metabolism, Mice, Mice, Inbred C57BL, NF-E2-Related Factor 2 genetics, NF-E2-Related Factor 2 metabolism, Oxidative Stress, Tumor Suppressor Protein p53 metabolism, Chemical and Drug Induced Liver Injury etiology, Chemical and Drug Induced Liver Injury metabolism, Chemical and Drug Induced Liver Injury prevention & control, Liver Failure, Acute chemically induced, Liver Failure, Acute metabolism, Liver Failure, Acute prevention & control
- Abstract
Background & Aims: Sulfoconjugation of small molecules or protein peptides is a key mechanism to ensure biochemical and functional homeostasis in mammals. The PAPS synthase 2 (PAPSS2) is the primary enzyme to synthesize the universal sulfonate donor 3'-phosphoadenosine 5'-phosphosulfate (PAPS). Acetaminophen (APAP) overdose is the leading cause of acute liver failure (ALF), in which oxidative stress is a key pathogenic event, whereas sulfation of APAP contributes to its detoxification. The goal of this study was to determine whether and how PAPSS2 plays a role in APAP-induced ALF., Methods: Gene expression was analyzed in APAP-induced ALF in patients and mice. Liver-specific Papss2-knockout mice using Alb-Cre (Papss2
ΔHC ) or AAV8-TBG-Cre (Papss2iΔHC ) were created and subjected to APAP-induced ALF. Primary human and mouse hepatocytes were used for in vitro mechanistic analysis., Results: The hepatic expression of PAPSS2 was decreased in APAP-induced ALF in patients and mice. Surprisingly, Papss2ΔHC mice were protected from APAP-induced hepatotoxicity despite having a decreased APAP sulfation, which was accompanied by increased hepatic antioxidative capacity through the activation of the p53-p2-Nrf2 axis. Treatment with a sulfation inhibitor also ameliorated APAP-induced hepatotoxicity. Gene knockdown experiments showed that the hepatoprotective effect of Papss2ΔHC was Nrf2, p53, and p21 dependent. Mechanistically, we identified p53 as a novel substrate of sulfation. Papss2 ablation led to p53 protein accumulation by preventing p53 sulfation, which disrupts p53-MDM2 interaction and p53 ubiquitination and increases p53 protein stability., Conclusions: We have uncovered a previously unrecognized and p53-mediated role of PAPSS2 in controlling oxidative response. Inhibition of p53 sulfation may be explored for the clinical management of APAP overdose., (Copyright © 2022. Published by Elsevier Inc.)- Published
- 2022
- Full Text
- View/download PDF
44. LiverClear: A versatile protocol for mouse liver tissue clearing.
- Author
-
Molina LM, Krutsenko Y, Jenkins NEC, Smith MC, Tao J, Wheeler TB, Watkins SC, Watson AM, and Monga SP
- Subjects
- Animals, Mice, Staining and Labeling, Liver diagnostic imaging
- Abstract
Although there are numerous tissue clearing protocols, most are inadequate for clearing liver tissue. Here we present a flexible protocol for mouse liver tissue; we combine strategies from several previously published protocols for delipidation, decolorization, staining, and refractive index matching. LiverClear is sufficiently versatile to allow clearing of healthy and diseased mouse liver followed by immunofluorescence staining and imaging to visualize intact 3D structures such as bile ducts and hepatocyte canaliculi. We also adapted this protocol for clearing human livers. For complete details on the use and execution of this protocol, please refer to Molina et al. (2021)., Competing Interests: The authors declare no competing interests., (© 2022 The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
45. Mitigation of portal fibrosis and cholestatic liver disease in ANKS6-deficient livers by macrophage depletion.
- Author
-
Airik M, McCourt B, Ozturk TT, Huynh AB, Zhang X, Tometich JT, Topaloglu R, Ozen H, Orhan D, Nejak-Bowen K, Monga SP, Hand TW, Ozaltin F, and Airik R
- Subjects
- Animals, Disease Models, Animal, Gene Expression physiology, Inflammation metabolism, Inflammation pathology, Liver pathology, Liver Cirrhosis pathology, Macrophages pathology, Mice, Mice, Inbred C57BL, Mice, Knockout, Monocytes metabolism, Monocytes pathology, Carrier Proteins metabolism, Cholestasis pathology, Liver metabolism, Liver Cirrhosis metabolism, Macrophages metabolism
- Abstract
Congenital hepatic fibrosis (CHF) is a developmental liver disease that is caused by mutations in genes that encode ciliary proteins and is characterized by bile duct dysplasia and portal fibrosis. Recent work has demonstrated that mutations in ANKS6 can cause CHF due to its role in bile duct development. Here, we report a novel ANKS6 mutation, which was identified in an infant presenting with neonatal jaundice due to underlying biliary abnormalities and liver fibrosis. Molecular analysis revealed that ANKS6 liver pathology is associated with the infiltration of inflammatory macrophages to the periportal fibrotic tissue and ductal epithelium. To further investigate the role of macrophages in CHF pathophysiology, we generated a novel liver-specific Anks6 knockout mouse model. The mutant mice develop biliary abnormalities and rapidly progressing periportal fibrosis reminiscent of human CHF. The development of portal fibrosis in Anks6 KO mice coincided with the accumulation of inflammatory monocytes and macrophages in the mutant liver. Gene expression and flow cytometric analysis demonstrated the preponderance of M1- over M2-like macrophages at the onset of fibrosis. A critical role for macrophages in promoting peribiliary fibrosis was demonstrated by depleting the macrophages with clodronate liposomes which effectively reduced inflammatory gene expression and fibrosis, and ameliorated tissue histology and biliary function in Anks6 KO livers. Together, this study demonstrates that macrophages play an important role in the initiation of liver fibrosis in ANKS6-deficient livers and their therapeutic elimination may provide an avenue to mitigate CHF in patients., (© 2022 Federation of American Societies for Experimental Biology.)
- Published
- 2022
- Full Text
- View/download PDF
46. Role of YAP1 Signaling in Biliary Development, Repair, and Disease.
- Author
-
Molina L, Nejak-Bowen K, and Monga SP
- Subjects
- Bile Ducts, Intrahepatic metabolism, Bile Ducts, Intrahepatic pathology, Child, Humans, Liver metabolism, Transcription Factors metabolism, YAP-Signaling Proteins, Biliary Tract, Biological Phenomena
- Abstract
Yes-associated protein 1 (YAP1) is a transcriptional coactivator that activates transcriptional enhanced associate domain transcription factors upon inactivation of the Hippo signaling pathway, to regulate biological processes like proliferation, survival, and differentiation. YAP1 is most prominently expressed in biliary epithelial cells (BECs) in normal adult livers and during development. In the current review, we will discuss the multiple roles of YAP1 in the development and morphogenesis of bile ducts inside and outside the liver, as well as in orchestrating the cholangiocyte repair response to biliary injury. We will review how biliary repair can occur through the process of hepatocyte-to-BEC transdifferentiation and how YAP1 is pertinent to this process. We will also discuss the liver's capacity for metabolic reprogramming as an adaptive mechanism in extreme cholestasis, such as when intrahepatic bile ducts are absent due to YAP1 loss from hepatic progenitors. Finally, we will discuss the roles of YAP1 in the context of pediatric pathologies afflicting bile ducts, such as Alagille syndrome and biliary atresia. In conclusion, we will comprehensively discuss the spatiotemporal roles of YAP1 in biliary development and repair after biliary injury while describing key interactions with other well-known developmental pathways., Competing Interests: None declared., (Thieme. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
47. Role and Regulation of Wnt/β-Catenin in Hepatic Perivenous Zonation and Physiological Homeostasis.
- Author
-
Goel C, Monga SP, and Nejak-Bowen K
- Subjects
- Animals, Gene Expression Regulation, Heme biosynthesis, Humans, Xenobiotics metabolism, Homeostasis, Liver metabolism, Wnt Signaling Pathway genetics
- Abstract
Metabolic heterogeneity or functional zonation is a key characteristic of the liver that allows different metabolic pathways to be spatially regulated within the hepatic system and together contribute to whole body homeostasis. These metabolic pathways are segregated along the portocentral axis of the liver lobule into three hepatic zones: periportal, intermediate or midzonal, and perivenous. The liver performs complementary or opposing metabolic functions within different hepatic zones while synergistic functions are regulated by overlapping zones, thereby maintaining the overall physiological stability. The Wnt/β-catenin signaling pathway is well known for its role in liver growth, development, and regeneration. In addition, the Wnt/β-catenin pathway plays a fundamental and dominant role in hepatic zonation and signals to orchestrate various functions of liver metabolism and pathophysiology. The β-catenin protein is the central player in the Wnt/β-catenin signaling cascade, and its activation is crucial for metabolic patterning of the liver. However, dysregulation of Wnt/β-catenin signaling is also implicated in different liver pathologies, including those associated with metabolic syndrome. β-Catenin is preferentially localized in the central region of the hepatic lobule surrounding the central vein and regulates multiple functions of this region. This review outlines the role of Wnt/β-catenin signaling pathway in controlling the different metabolic processes surrounding the central vein and its relation to liver homeostasis and dysfunction., (Copyright © 2022 American Society for Investigative Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
48. YAP1 activation and Hippo pathway signaling in the pathogenesis and treatment of intrahepatic cholangiocarcinoma.
- Author
-
Ko S, Kim M, Molina L, Sirica AE, and Monga SP
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Signal Transducing metabolism, Bile Ducts, Intrahepatic metabolism, Bile Ducts, Intrahepatic pathology, Hippo Signaling Pathway, Humans, Tumor Microenvironment, YAP-Signaling Proteins, Bile Duct Neoplasms pathology, Cholangiocarcinoma pathology
- Abstract
Intrahepatic cholangiocarcinoma (iCCA), the second most common primary liver cancer, is a highly lethal epithelial cell malignancy exhibiting features of cholangiocyte differentiation. iCCAs can potentially develop from multiple cell types of origin within liver, including immature or mature cholangiocytes, hepatic stem cells/progenitor cells, and from transdifferentiation of hepatocytes. Understanding the molecular mechanisms and genetic drivers that diversely drive specific cell lineage pathways leading to iCCA has important biological and clinical implications. In this context, activation of the YAP1-TEAD dependent transcription, driven by Hippo-dependent or -independent diverse mechanisms that lead to the stabilization of YAP1 is crucially important to biliary fate commitment in hepatobiliary cancer. In preclinical models, YAP1 activation in hepatocytes or cholangiocytes is sufficient to drive their malignant transformation into iCCA. Moreover, nuclear YAP1/TAZ is highly prevalent in human iCCA irrespective of the varied etiology, and significantly correlates with poor prognosis in iCCA patients. Based on the ubiquitous expression and diverse physiologic roles for YAP1/TAZ in the liver, recent studies have further revealed distinct functions of active YAP1/TAZ in regulating tumor metabolism, as well as the tumor immune microenvironment. In the current review, we discuss our current understanding of the various roles of the Hippo-YAP1 signaling in iCCA pathogenesis, with a specific focus on the roles played by the Hippo-YAP1 pathway in modulating biliary commitment and oncogenicity, iCCA metabolism, and immune microenvironment. We also discuss the therapeutic potential of targeting the YAP1/TAZ-TEAD transcriptional machinery in iCCA, its current limitations, and what future studies are needed to facilitate clinical translation., Competing Interests: Conflict of interest statement None of the authors have any interests to declare related to this study., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
49. β-Catenin-NF-κB-CFTR interactions in cholangiocytes regulate inflammation and fibrosis during ductular reaction.
- Author
-
Hu S, Russell JO, Liu S, Cao C, McGaughey J, Rai R, Kosar K, Tao J, Hurley E, Poddar M, Singh S, Bell A, Shin D, Raeman R, Singhi AD, Nejak-Bowen K, Ko S, and Monga SP
- Subjects
- Animals, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Epithelial Cells metabolism, Fibrosis immunology, Inflammation immunology, Mice, Mice, Transgenic, NF-kappa B metabolism, beta Catenin metabolism, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Fibrosis genetics, Inflammation genetics, NF-kappa B genetics, beta Catenin genetics
- Abstract
Expansion of biliary epithelial cells (BECs) during ductular reaction (DR) is observed in liver diseases including cystic fibrosis (CF), and associated with inflammation and fibrosis, albeit without complete understanding of underlying mechanism. Using two different genetic mouse knockouts of β-catenin, one with β-catenin loss is hepatocytes and BECs (KO1), and another with loss in only hepatocytes (KO2), we demonstrate disparate long-term repair after an initial injury by 2-week choline-deficient ethionine-supplemented diet. KO2 show gradual liver repopulation with BEC-derived β-catenin-positive hepatocytes and resolution of injury. KO1 showed persistent loss of β-catenin, NF-κB activation in BECs, progressive DR and fibrosis, reminiscent of CF histology. We identify interactions of β-catenin, NFκB, and CF transmembranous conductance regulator (CFTR) in BECs. Loss of CFTR or β-catenin led to NF-κB activation, DR, and inflammation. Thus, we report a novel β-catenin-NFκB-CFTR interactome in BECs, and its disruption may contribute to hepatic pathology of CF., Competing Interests: SH, JR, SL, CC, JM, RR, KK, JT, EH, MP, SS, AB, DS, RR, AS, KN, SK, SM No competing interests declared, (© 2021, Hu et al.)
- Published
- 2021
- Full Text
- View/download PDF
50. Silvio O. Conte Digestive Disease Research Core Centers-Connecting People, Creating Opportunities, Developing Careers.
- Author
-
Sandler RS, Davidson NO, Monga SP, and Rockey DC
- Subjects
- Cooperative Behavior, Humans, Interinstitutional Relations, Mentors, Academies and Institutes organization & administration, Biomedical Research organization & administration, Career Mobility, Gastroenterologists organization & administration, Gastroenterology organization & administration, Interpersonal Relations, Staff Development
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.