44 results on '"Swiderska-Syn M"'
Search Results
2. Osteopontin neutralisation abrogates the liver progenitor cell response and fibrogenesis in mice
- Author
-
Coombes, J D, Swiderska-Syn, M, Dollé, L, Reid, D, Eksteen, B, Claridge, L, Briones-Orta, M A, Shetty, S, Oo, Y H, Riva, A, Chokshi, S, Papa, S, Mi, Z, Kuo, P C, Williams, R, Canbay, A, Adams, D H, Diehl, A M, van Grunsven, L A, Choi, S S, and Syn, W K
- Published
- 2015
- Full Text
- View/download PDF
3. Myofibroblastic cells function as progenitors to regenerate murine livers after partial hepatectomy
- Author
-
Swiderska-Syn, M, Syn, W K, Xie, G, Krüger, L, Machado, M V, Karaca, G, Michelotti, G A, Choi, S S, Premont, R T, and Diehl, A M
- Published
- 2014
- Full Text
- View/download PDF
4. Thyroid hormone (T3) regulates liver progenitor cell response
- Author
-
Manka, Paul, Coombes, J. D., Wang, C., Swiderska-Syn, M., Briones, M. A., Sydor, S., Williams, R., Gerken, Guido, Canbay, Aliekber, Gauthier, K., Bechmann, Lars Peter, and Syn, W. K.
- Subjects
Medizin - Published
- 2017
5. Cholangiocyte chemokine secretion and macrophage accumulation is mediated by osteopontin in murine liver models
- Author
-
Coombes, J. D., Manka, P. P., Swiderska-Syn, M., Reid, D., Riva, A., Claridge, L. C., Dolle, L., Younis, R., Briones, M. A., Kitamura, N., Mehta, K., Mi, Z. Y., Kuo, P. C., Williams, R., Diehl, A. M., van Grunsven, L. A., Chokshi, S., Canbay, Aliekber, Flamant, F., Gauthier, K., Eksteen, B., Syn, W. K., King‘s College London, Duke University, University of Calgary, Leeds Teaching Hospital (NHS LTHN), Vrije Universiteit [Brussels] (VUB), Loyola University of Chicago, Essen University Hospital, Institut de Génomique Fonctionnelle de Lyon (IGFL), École normale supérieure - Lyon (ENS Lyon)-Institut National de la Recherche Agronomique (INRA)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS), École normale supérieure - Lyon (ENS Lyon), Vrije Universiteit Brussel (VUB), École normale supérieure - Lyon (ENS Lyon)-Institut National de la Recherche Agronomique (INRA)-Centre National de la Recherche Scientifique (CNRS)-Université Claude Bernard Lyon 1 (UCBL), and Université de Lyon-Université de Lyon
- Subjects
stomatognathic system ,[SDV]Life Sciences [q-bio] ,Medizin - Abstract
Background and aims Progression of chronic liver disease involves accumulation of inflammatory cells towards the peri-portal regions during a ductular inflammatory response. Osteopontin (OPN), an effector of Hh signalling, contributes to liver fibrogenesis and ductular inflammation via activation of hepatic stellate and progenitor cells. In tissue injury, OPN regulates macrophage functions via pro-inflammatory chemoattractant properties. In liver, however, the role of OPN in macrophage activation and recruitment remains unclear. We investigated the role of OPN in cholangiocyte chemokine secretion and macrophage recruitment using in vivo, in vitro, and in silico approaches. Methods In MCD and DDC murine models of liver fibrosis, total liver chemokine expression was measured by qRTPCR and macrophages detected by FACS (CD 11b, F4/80, CCR2, Ly6C). Lentiviral-mediated shRNA (shOPN) was used for OPN knockdown in murine 603B cholangiocytes, and secreted OPN neutralized by specific aptamers. Cholangiocyte chemokine secretion was measured by cytometric bead array and mRNA by qRTPCR. Macrophage migration was assessed in transwells using RAW264.7 cells. To obtain a global perspective of genes affected by OPN, next-generation RNA sequencing was used to compare control and shOPN cholan- giocytes. Transcripts were assessed in DESeq and gene ontologies and pathways by GOrilla, DAVID, and ReviGO software. Results Liver fibrosis in MCD and DDC was accompanied by upregulated total liver OPN, TGF-β, Ccl2, Ccl5, and Cxcll mRNA, and accumulation of liver CDl1b/F4/80(+) CCR2(hi) macrophages. Mice treated with OPN-aptamers had less fibrosis, reduced Ccl2, Ccl5, and Cxcll mRNA, and reduced accumulation of liver CD11b/F4/80(+) CCR2(hi) macrophages and the Ly6C(hi) inflammatory monocyte subset. In shOPN cholangiocytes, RNAseq detected 670 affected genes (Ben- jamini-Hochsberg padj
- Published
- 2016
6. Osteopontin promotes cholangiocyte chemokine secretion and macrophage accumulation in mice
- Author
-
Coombes, J. D., Manka, P. P., Swiderska-Syn, M., Riva, A., Reid, D., Claridge, L. C., Dolle, L., Younis, R., Briones-Orta, M. A., Kitamura, N., Mi, Z. Y., Kuo, P. C., Williams, R., Diehl, A. M., Chokshi, S., Eksteen, B., Canbay, Aliekber, and Syn, W. K.
- Subjects
Medizin - Published
- 2015
7. Thyroid Hormone receptor regulates hepatic stellate cell activation
- Author
-
Manka, P.P., primary, Coombes, J.D., additional, Bechmann, L.P., additional, Dollé, L., additional, Swiderska-Syn, M., additional, Briones-Orta, M.A., additional, Williams, R., additional, van Grunsven, L., additional, Canbay, A., additional, Flamant, F., additional, Gauthier, K., additional, and Syn, W.-K., additional
- Published
- 2017
- Full Text
- View/download PDF
8. Thyroid Hormone Receptor (TR): a regulator in Liver Fibrogenesis
- Author
-
Manka, PP, additional, Coombes, JD, additional, Bechmann, L, additional, Swiderska-Syn, M, additional, Reid, D, additional, Claridge, LC, additional, Younis, R, additional, Mehta, K, additional, Briones, MA, additional, Kitamura, N, additional, Mi, Z, additional, Kuo, PC, additional, Williams, R, additional, Eksteen, B, additional, Diehl, AM, additional, Gerken, G, additional, Canbay, A, additional, Flamant, F, additional, Gauthier, K, additional, and Syn, WK, additional
- Published
- 2016
- Full Text
- View/download PDF
9. Osteopontin mediates fibrogenic effects of leptin and enhances NASH progression
- Author
-
Coombes, J. D., Choi, S. S., Manka, P. P., Briones-Orta, M. A., Swiderska-Syn, M., Kitamura, N., Younis, R., Bitencourt, S., Dolle, L., Williams, R., van Grunsven, L. A., Diehl, A. M., Canbay, Aliekber, and Syn, W. K.
- Subjects
Medizin - Published
- 2014
10. Osteopontin Promotes Cholangiocyte Chemokine Secretion and Macrophage Accumulation in Murine Liver Fibrosis
- Author
-
Coombes, J.D., primary, Manka, P.P., additional, Swiderska-Syn, M., additional, Reid, D., additional, Riva, A., additional, Claridge, L.C., additional, Dolle, L., additional, Younis, R., additional, Briones-Orta, M.A., additional, Kitamura, N., additional, Mehta, K., additional, Mi, Z., additional, Kuo, P.C., additional, Williams, R., additional, Diehl, A.M., additional, van Grunsven, L.A., additional, Chokshi, S., additional, Eksteen, B., additional, Canbay, A., additional, and Syn, W.-K., additional
- Published
- 2016
- Full Text
- View/download PDF
11. P0469 : Markers of tissue repair and cellular aging are increased in liver tissue of patients with dual chronic HIV/HCV infections
- Author
-
Swiderska-Syn, M., primary, Choi, S., additional, Lan, A., additional, Ferrari, G., additional, Diehl, A.M., additional, and Naggie, S., additional
- Published
- 2015
- Full Text
- View/download PDF
12. Reduced lipoapoptosis, hedgehog pathway activation and fibrosis in caspase-2 deficient mice with non-alcoholic steatohepatitis
- Author
-
Machado, M V, primary, Michelotti, G A, additional, Pereira, T de Almeida, additional, Boursier, J, additional, Kruger, L, additional, Swiderska-Syn, M, additional, Karaca, G, additional, Xie, G, additional, Guy, C D, additional, Bohinc, B, additional, Lindblom, K R, additional, Johnson, E, additional, Kornbluth, S, additional, and Diehl, A M, additional
- Published
- 2014
- Full Text
- View/download PDF
13. Osteopontin neutralisation abrogates the liver progenitor cell response and fibrogenesis in mice
- Author
-
Coombes, J D, primary, Swiderska-Syn, M, additional, Dollé, L, additional, Reid, D, additional, Eksteen, B, additional, Claridge, L, additional, Briones-Orta, M A, additional, Shetty, S, additional, Oo, Y H, additional, Riva, A, additional, Chokshi, S, additional, Papa, S, additional, Mi, Z, additional, Kuo, P C, additional, Williams, R, additional, Canbay, A, additional, Adams, D H, additional, Diehl, A M, additional, van Grunsven, L A, additional, Choi, S S, additional, and Syn, W K, additional
- Published
- 2014
- Full Text
- View/download PDF
14. SAT-058 - Thyroid Hormone receptor regulates hepatic stellate cell activation
- Author
-
Manka, P.P., Coombes, J.D., Bechmann, L.P., Dollé, L., Swiderska-Syn, M., Briones-Orta, M.A., Williams, R., van Grunsven, L., Canbay, A., Flamant, F., Gauthier, K., and Syn, W.-K.
- Published
- 2017
- Full Text
- View/download PDF
15. P612 OSTEOPONTIN NEUTRALIZATION ABROGATES THE LIVER PROGENITOR CELL RESPONSE AND FIBROGENIC OUTCOMES IN MICE
- Author
-
Coombes, J.D., primary, Swiderska-Syn, M., additional, Dollé, L., additional, Reid, D.T., additional, Eksteen, B., additional, Claridge, L.C., additional, Briones-Orta, M.A., additional, Shishir, S., additional, Oo, Y.H., additional, Riva, A., additional, Chokshi, S., additional, Papa, S., additional, Mi, Z., additional, Kuo, P.C., additional, Williams, R., additional, Canbay, A., additional, Adams, D.H., additional, van Grunsven, L.A., additional, Diehl, A.M., additional, Choi, S.S., additional, and Syn, W.-K., additional
- Published
- 2014
- Full Text
- View/download PDF
16. FRI-230 - Osteopontin Promotes Cholangiocyte Chemokine Secretion and Macrophage Accumulation in Murine Liver Fibrosis
- Author
-
Coombes, J.D., Manka, P.P., Swiderska-Syn, M., Reid, D., Riva, A., Claridge, L.C., Dolle, L., Younis, R., Briones-Orta, M.A., Kitamura, N., Mehta, K., Mi, Z., Kuo, P.C., Williams, R., Diehl, A.M., van Grunsven, L.A., Chokshi, S., Eksteen, B., Canbay, A., and Syn, W.-K.
- Published
- 2016
- Full Text
- View/download PDF
17. Myofibroblastic cells function as progenitors to regenerate murine livers after partial hepatectomy
- Author
-
Swiderska-Syn, M, primary, Syn, W K, additional, Xie, G, additional, Krüger, L, additional, Machado, M V, additional, Karaca, G, additional, Michelotti, G A, additional, Choi, S S, additional, Premont, R T, additional, and Diehl, A M, additional
- Published
- 2013
- Full Text
- View/download PDF
18. Intraduodenal fecal microbiota transplantation ameliorates gut atrophy and cholestasis in a novel parenteral nutrition piglet model.
- Author
-
Manithody C, Denton C, Mehta S, Carter J, Kurashima K, Bagwe A, Swiderska-Syn M, Guzman M, Besmer S, Jain S, McHale M, Qureshi K, Nazzal M, Caliskan Y, Long J, Lin CJ, Hutchinson C, Ericsson AC, and Jain AK
- Subjects
- Animals, Swine, Atrophy, Disease Models, Animal, Liver pathology, Liver metabolism, Fecal Microbiota Transplantation methods, Cholestasis therapy, Cholestasis microbiology, Gastrointestinal Microbiome, Parenteral Nutrition, Total methods
- Abstract
Total parenteral nutrition (TPN) provides lifesaving nutritional support intravenously; however, it is associated with significant side effects. Given gut microbial alterations noted with TPN, we hypothesized that transferring fecal microbiota from healthy controls would restore gut-systemic signaling in TPN and mitigate injury. Using our novel ambulatory model (US Patent: US 63/136,165), 31 piglets were randomly allocated to enteral nutrition (EN), TPN only, TPN + antibiotics (TPN-A), or TPN + intraduodenal fecal microbiota transplant (TPN + FMT) for 14 days. Gut, liver, and serum were assessed through histology, biochemistry, and qPCR. Stool samples underwent 16 s rRNA sequencing. Permutational multivariate analysis of variance, Jaccard, and Bray-Curtis metrics were performed. Significant bilirubin elevation in TPN and TPN-A versus EN ( P < 0.0001) was prevented with FMT. IFN-G, TNF-α, IL-β, IL-8, and lipopolysaccharide (LPS) were significantly higher in TPN ( P = 0.009, P = 0.001, P = 0.043, P = 0.011, P < 0.0001), with preservation upon FMT. Significant gut atrophy by villous-to-crypt ratio in TPN ( P < 0.0001) and TPN-A ( P = 0.0001) versus EN was prevented by FMT ( P = 0.426 vs. EN). Microbiota profiles using principal coordinate analysis demonstrated significant FMT and EN overlap, with the largest separation in TPN-A followed by TPN, driven primarily by Firmicutes and Fusobacteria. TPN-altered gut barrier was preserved upon FMT; upregulated cholesterol 7 α-hydroxylase and bile salt export pump in TPN and TPN-A and downregulated fibroblast growth factor receptor 4, EGF, farnesoid X receptor, and Takeda G Protein-coupled Receptor 5 (TGR5) versus EN was prevented by FMT. This study provides novel evidence of prevention of gut atrophy, liver injury, and microbial dysbiosis with intraduodenal FMT, challenging current paradigms into TPN injury mechanisms and underscores the importance of gut microbes as prime targets for therapeutics and drug discovery. NEW & NOTEWORTHY Intraduodenal fecal microbiota transplantation presents a novel strategy to mitigate complications associated with total parenteral nutrition (TPN), highlighting gut microbiota as a prime target for therapeutic and diagnostic approaches. These results from a highly translatable model provide hope for TPN side effect mitigation for thousands of chronically TPN-dependent patients.
- Published
- 2024
- Full Text
- View/download PDF
19. Osteopontin Promotes Cholangiocyte Secretion of Chemokines to Support Macrophage Recruitment and Fibrosis in MASH.
- Author
-
Coombes JD, Manka PP, Swiderska-Syn M, Vannan DT, Riva A, Claridge LC, Moylan C, Suzuki A, Briones-Orta MA, Younis R, Kitamura N, Sydor S, Bittencourt S, Mi Z, Kuo PC, Diehl AM, van Grunsven LA, Chokshi S, Canbay A, Abdelmalek MF, Aspichueta P, Papa S, Eksteen B, and Syn WK
- Abstract
Background and Aims: Osteopontin (OPN) promotes the ductular reaction and is a major driver of chronic liver disease (CLD) progression. Although CLD is characterised by the accumulation of inflammatory cells including macrophages around the peri-portal regions, the influence of OPN on recruitment is unclear. We investigated the role of OPN in cholangiocyte chemokine production and macrophage recruitment by combining in vivo, in vitro, and in silico approaches., Methods: The effects of OPN on cholangiocyte chemokine production and macrophage migration were assessed in culture, alongside RNA-sequencing to identify genes and pathways affected by OPN depletion. Murine liver injury models were used to assess liver chemokine expression and liver macrophage/monocyte recruitment. OPN and chemokine expression were analysed in liver tissue and plasma from biopsy-proven metabolic dysfunction-associated alcoholic steatohepatitis (MASH) patients., Results: OPN-knockdown in cholangiocytes reduced chemokine secretion. RNA-sequencing showed OPN-related effects clustered around immunity, chemotaxis and chemokine production. Macrophage exposure to cholangiocyte-conditioned media showed OPN-supported migration via chemokines chemokine (C-C motif) ligand (CCL)2, CCL5 and chemokine (C-X-C motif) ligand (CXCL)1. These effects were related to NF-κB signalling. Murine liver fibrosis was accompanied by upregulated liver OPN, CCL2, CCL5 and CXCL1 mRNA, and accumulation of liver cluster of differentiation (CD)11b/F4/80
+ CC chemokine receptors (CCR2)high macrophages but treatment with OPN-specific neutralising aptamers reduced fibrosis, chemokine mRNAs and accumulation of liver CD11b/F4/80+ CCR2high /lymphocyte antigen 6 complexhigh inflammatory monocytes. In human MASH, liver OPN correlated with chemokines CCL2 and IL8 in association with portal injury and fibrosis. Plasma OPN, serum CCL2 and IL8 also increased with fibrosis stage., Conclusions: OPN promotes cholangiocyte chemokine secretion and the accumulation of pro-inflammatory monocytes. These data support neutralisation of OPN as an anti-inflammatory and anti-fibrotic strategy., (© 2024 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)- Published
- 2024
- Full Text
- View/download PDF
20. Triptolide and its prodrug Minnelide target high-risk MYC-amplified medulloblastoma in preclinical models.
- Author
-
Rodriguez-Blanco J, Salvador AD, Suter RK, Swiderska-Syn M, Palomo-Caturla I, Kliebe V, Shahani P, Peterson K, Turos-Cabal M, Vieira ME, Wynn DT, Howell AJ, Yang F, Ban Y, McCrea HJ, Zindy F, Danis E, Vibhakar R, Jermakowicz A, Martin V, Coss CC, Harris BT, de Cubas A, Chen XS, Barnoud T, Roussel MF, Ayad NG, and Robbins DJ
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Prodrugs pharmacology, Cerebellar Neoplasms drug therapy, Cerebellar Neoplasms genetics, Cerebellar Neoplasms pathology, Cerebellar Neoplasms metabolism, Female, Gene Expression Regulation, Neoplastic drug effects, Organophosphates, Phenanthrenes pharmacology, Diterpenes pharmacology, Epoxy Compounds pharmacology, Medulloblastoma drug therapy, Medulloblastoma genetics, Medulloblastoma pathology, Medulloblastoma metabolism, Proto-Oncogene Proteins c-myc genetics, Proto-Oncogene Proteins c-myc metabolism, Xenograft Model Antitumor Assays
- Abstract
Most children with medulloblastoma (MB) achieve remission, but some face very aggressive metastatic tumors. Their dismal outcome highlights the critical need to advance therapeutic approaches that benefit such high-risk patients. Minnelide, a clinically relevant analog of the natural product triptolide, has oncostatic activity in both preclinical and early clinical settings. Despite its efficacy and tolerable toxicity, this compound has not been evaluated in MB. Utilizing a bioinformatic data set that integrates cellular drug response data with gene expression, we predicted that Group 3 (G3) MB, which has a poor 5-year survival, would be sensitive to triptolide/Minnelide. We subsequently showed that both triptolide and Minnelide attenuate the viability of G3 MB cells ex vivo. Transcriptomic analyses identified MYC signaling, a pathologically relevant driver of G3 MB, as a downstream target of this class of drugs. We validated this MYC dependency in G3 MB cells and showed that triptolide exerts its efficacy by reducing both MYC transcription and MYC protein stability. Importantly, Minnelide acted on MYC to reduce tumor growth and leptomeningeal spread, which resulted in improved survival of G3 MB animal models. Moreover, Minnelide improved the efficacy of adjuvant chemotherapy, further highlighting its potential for the treatment of MYC-driven G3 MB.
- Published
- 2024
- Full Text
- View/download PDF
21. Gut Microbiota Modulation of Short Bowel Syndrome and the Gut-Brain Axis.
- Author
-
Carter J, Bettag J, Morfin S, Manithody C, Nagarapu A, Jain A, Nazzal H, Prem S, Unes M, McHale M, Lin CJ, Hutchinson C, Trello G, Jain A, Portz E, Verma A, Swiderska-Syn M, Goldenberg D, and Kurashima K
- Subjects
- Humans, Brain-Gut Axis, Intestine, Small microbiology, Bacteria, Dysbiosis microbiology, Short Bowel Syndrome complications, Gastrointestinal Microbiome physiology
- Abstract
Short bowel syndrome (SBS) is a condition that results from a reduction in the length of the intestine or its functional capacity. SBS patients can have significant side effects and complications, the etiology of which remains ill-defined. Thus, facilitating intestinal adaptation in SBS remains a major research focus. Emerging data supports the role of the gut microbiome in modulating disease progression. There has been ongoing debate on defining a "healthy" gut microbiome, which has led to many studies analyzing the bacterial composition and shifts that occur in gastrointestinal disease states such as SBS and the resulting systemic effects. In SBS, it has also been found that microbial shifts are highly variable and dependent on many factors, including the anatomical location of bowel resection, length, and structure of the remnant bowel, as well as associated small intestinal bacterial overgrowth (SIBO). Recent data also notes a bidirectional communication that occurs between enteric and central nervous systems called the gut-brain axis (GBA), which is regulated by the gut microbes. Ultimately, the role of the microbiome in disease states such as SBS have many clinical implications and warrant further investigation. The focus of this review is to characterize the role of the gut microbiota in short bowel syndrome and its impact on the GBA, as well as the therapeutic potential of altering the microbiome.
- Published
- 2023
- Full Text
- View/download PDF
22. A Druggable UHRF1/DNMT1/GLI Complex Regulates Sonic Hedgehog-Dependent Tumor Growth.
- Author
-
Yang F, Rodriguez-Blanco J, Long J, Swiderska-Syn M, Wynn DT, Li B, Shen C, Nayak A, Ban Y, Sun X, Suter RK, McCrea HJ, Capobianco AJ, Ayad NG, and Robbins DJ
- Subjects
- Humans, Hedgehog Proteins metabolism, Smoothened Receptor genetics, Smoothened Receptor metabolism, Signal Transduction genetics, CCAAT-Enhancer-Binding Proteins genetics, CCAAT-Enhancer-Binding Proteins metabolism, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism, Medulloblastoma drug therapy, Medulloblastoma genetics, Medulloblastoma metabolism, Cerebellar Neoplasms metabolism
- Abstract
Dysregulation of Sonic hedgehog (SHH) signaling drives the growth of distinct cancer subtypes, including medulloblastoma (MB). Such cancers have been treated in the clinic with a number of clinically relevant SHH inhibitors, the majority of which target the upstream SHH regulator, Smoothened (SMO). Despite considerable efficacy, many of these patients develop resistance to these drugs, primarily due to mutations in SMO. Therefore, it is essential to identify druggable, signaling components downstream of SMO to target in SMO inhibitor resistant cancers. We utilized an integrated functional genomics approach to identify epigenetic regulators of SHH signaling and identified a novel complex of Ubiquitin-like with PHD and RING finger domains 1 (UHRF1), DNA methyltransferase 1 (DNMT1), and GLI proteins. We show that this complex is distinct from previously described UHRF1/DNMT1 complexes, suggesting that it works in concert to regulate GLI activity in SHH driven tumors. Importantly, we show that UHRF1/DNMT1/GLI complex stability is targeted by a repurposed FDA-approved therapy, with a subsequent reduction in the growth of SHH-dependent MB ex vivo and in vivo., Implications: This work describes a novel, druggable UHRF1/DNMT1/GLI complex that regulates SHH-dependent tumor growth, and highlights an FDA-approved drug capable of disrupting this complex to attenuate tumor growth., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
23. Noncanonical activation of GLI signaling in SOX2 + cells drives medulloblastoma relapse.
- Author
-
Swiderska-Syn M, Mir-Pedrol J, Oles A, Schleuger O, Salvador AD, Greiner SM, Seward C, Yang F, Babcock BR, Shen C, Wynn DT, Sanchez-Mejias A, Gershon TR, Martin V, McCrea HJ, Lindsey KG, Krieg C, and Rodriguez-Blanco J
- Subjects
- Child, Hedgehog Proteins metabolism, Humans, Neoplasm Recurrence, Local, SOXB1 Transcription Factors genetics, SOXB1 Transcription Factors metabolism, Signal Transduction, Zinc Finger Protein GLI1 metabolism, Brain Neoplasms, Cerebellar Neoplasms genetics, Medulloblastoma genetics, Medulloblastoma pathology
- Abstract
SRY (sex determining region Y)-box 2 (SOX2)-labeled cells play key roles in chemoresistance and tumor relapse; thus, it is critical to elucidate the mechanisms propagating them. Single-cell transcriptomic analyses of the most common malignant pediatric brain tumor, medulloblastoma (MB), revealed the existence of astrocytic Sox2
+ cells expressing sonic hedgehog (SHH) signaling biomarkers. Treatment with vismodegib, an SHH inhibitor that acts on Smoothened (Smo), led to increases in astrocyte-like Sox2+ cells. Using SOX2-enriched MB cultures, we observed that SOX2+ cells required SHH signaling to propagate, and unlike in the proliferative tumor bulk, the SHH pathway was activated in these cells downstream of Smo in an MYC-dependent manner. Functionally different GLI inhibitors depleted vismodegib-resistant SOX2+ cells from MB tissues, reduced their ability to further engraft in vivo, and increased symptom-free survival. Our results emphasize the promise of therapies targeting GLI to deplete SOX2+ cells and provide stable tumor remission.- Published
- 2022
- Full Text
- View/download PDF
24. Phase I Trial Characterizing the Pharmacokinetic Profile of N-803, a Chimeric IL-15 Superagonist, in Healthy Volunteers.
- Author
-
Rubinstein MP, Williams C, Mart C, Beall J, MacPherson L, Azar J, Swiderska-Syn M, Manca P, Gibney BC, Robinson MD, Krieg C, Hill EG, Taha SA, Rock AD, Lee JH, Soon-Shiong P, and Wrangle J
- Subjects
- Healthy Volunteers, Humans, Recombinant Fusion Proteins, CD8-Positive T-Lymphocytes, Interleukin-15
- Abstract
The oncotherapeutic promise of IL-15, a potent immunostimulant, is limited by a short serum t
1/2 The fusion protein N-803 is a chimeric IL-15 superagonist that has a >20-fold longer in vivo t1/2 versus IL-15. This phase 1 study characterized the pharmacokinetic (PK) profile and safety of N-803 after s.c. administration to healthy human volunteers. Volunteers received two doses of N-803, and after each dose, PK and safety were assessed for 9 d. The primary endpoint was the N-803 PK profile, the secondary endpoint was safety, and immune cell levels and immunogenicity were measures of interest. Serum N-803 concentrations peaked 4 h after administration and declined with a t1/2 of ∼20 h. N-803 did not cause treatment-emergent serious adverse events (AEs) or grade ≥3 AEs. Injection site reactions, chills, and pyrexia were the most common AEs. Administration of N-803 was well tolerated and accompanied by proliferation of NK cells and CD8+ T cells and sustained increases in the number of NK cells. Our results suggest that N-803 administration can potentiate antitumor immunity., (Copyright © 2022 by The American Association of Immunologists, Inc.)- Published
- 2022
- Full Text
- View/download PDF
25. Nocturnal Hypoxia Activation of the Hedgehog Signaling Pathway Affects Pediatric Nonalcoholic Fatty Liver Disease Severity.
- Author
-
Sundaram SS, Swiderska-Syn M, Sokol RJ, Halbower AC, Capocelli KE, Pan Z, Robbins K, Graham B, and Diehl AM
- Abstract
Chronic intermittent hypoxia and hedgehog (Hh) pathway dysregulation are associated with nonalcoholic fatty liver disease (NAFLD) progression. In this study, we determined the relationship between obstructive sleep apnea (OSA)/nocturnal hypoxia and Hh signaling in pediatric NAFLD. Adolescents with histologic NAFLD (n = 31) underwent polysomnogram testing, laboratory testing, and Sonic Hh (SHh), Indian hedgehog (IHh), glioblastoma-associated oncogene 2 (Gli2), keratin 7 (K7), α-smooth muscle actin (α-SMA), and hypoxia-inducible factor 1α (HIF-1α) immunohistochemistry. Aspartate aminotransferase (AST) correlated with SHh, r = 0.64; Gli2, r = 0.4; α-SMA, r = 0.55; and K7, r = 0.45 ( P < 0.01), as did alanine aminotransferase (ALT) (SHh, r = 0.51; Gli2, r = 0.43; α-SMA, r = 0.51; P < 0.02). SHh correlated with NAFLD activity score ( r = 0.39), whereas IHh correlated with inflammation ( r = -0.478) and histologic grade ( r = -0.43); P < 0.03. Subjects with OSA/hypoxia had higher SHh (4.0 ± 2.9 versus 2.0 ± 1.5), Gli2 (74.2 ± 28.0 versus 55.8 ± 11.8), and α-SMA (6.2 ± 3.3 versus 4.3 ± 1.2); compared to those without ( P < 0.03). OSA severity correlated with SHh ( r = 0.31; P = 0.09) and Gli2 ( r = 0.37; P = 0.04) as did hypoxia severity, which was associated with increasing SHh ( r = -0.53), Gli2 ( r = -0.52), α-SMA ( r = -0.61), and K7 ( r = -0.42); P < 0.02. Prolonged O
2 desaturations <90% also correlated with SHh ( r = 0.55) and Gli2 ( r = 0.61); P < 0.05. Conclusion : The Hh pathway is activated in pediatric patients with NAFLD with nocturnal hypoxia and relates to disease severity. Tissue hypoxia may allow for functional activation of HIF-1α, with induction of genes important in epithelial-mesenchymal transition, including SHh, and NAFLD progression.- Published
- 2019
- Full Text
- View/download PDF
26. Liver regeneration requires Yap1-TGFβ-dependent epithelial-mesenchymal transition in hepatocytes.
- Author
-
Oh SH, Swiderska-Syn M, Jewell ML, Premont RT, and Diehl AM
- Subjects
- Animals, Cell Cycle Proteins, Cell Proliferation, Mice, Mice, Knockout, Signal Transduction, YAP-Signaling Proteins, Adaptor Proteins, Signal Transducing metabolism, Epithelial-Mesenchymal Transition physiology, Hepatocytes physiology, Liver Regeneration physiology, Phosphoproteins metabolism, Transforming Growth Factor beta metabolism
- Abstract
Background & Aims: Chronic failure of mechanisms that promote effective regeneration of dead hepatocytes causes replacement of functional hepatic parenchyma with fibrous scar tissue, ultimately resulting in cirrhosis. Therefore, defining and optimizing mechanisms that orchestrate effective regeneration might prevent cirrhosis. We hypothesized that effective regeneration of injured livers requires hepatocytes to evade the growth-inhibitory actions of TGFβ, since TGFβ signaling inhibits mature hepatocyte growth but drives cirrhosis pathogenesis., Methods: Wild-type mice underwent 70% partial hepatectomy (PH); TGFβ expression and signaling were evaluated in intact tissue and primary hepatocytes before, during, and after the period of maximal hepatocyte proliferation that occurs from 24-72 h after PH. To determine the role of Yap1 in regulating TGFβ signaling in hepatocytes, studies were repeated after selectively deleting Yap1 from hepatocytes of Yap1
flox/flox mice., Results: TGFβ expression and hepatocyte nuclear accumulation of pSmad2 and Yap1 increased in parallel with hepatocyte proliferative activity after PH. Proliferative hepatocytes also upregulated Snai1, a pSmad2 target gene that promotes epithelial-to-mesenchymal transition (EMT), suppressed epithelial genes, induced myofibroblast markers, and produced collagen 1α1. Deleting Yap1 from hepatocytes blocked their nuclear accumulation of pSmad2 and EMT-like response, as well as their proliferation., Conclusion: Interactions between the TGFβ and Hippo-Yap signaling pathways stimulate hepatocytes to undergo an EMT-like response that is necessary for them to grow in a TGFβ-enriched microenvironment and regenerate injured livers., Lay Summary: The adult liver has an extraordinary ability to regenerate after injury despite the accumulation of scar-forming factors that normally block the proliferation and reduce the survival of residual liver cells. We discovered that liver cells manage to escape these growth-inhibitory influences by transiently becoming more like fibroblasts themselves. They do this by reactivating programs that are known to drive tissue growth during fetal development and in many cancers. Understanding how the liver can control programs that are involved in scarring and cancer may help in the development of new treatments for cirrhosis and liver cancer., (Copyright © 2018 European Association for the Study of the Liver. Published by Elsevier B.V. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
27. Markers of Tissue Repair and Cellular Aging Are Increased in the Liver Tissue of Patients With HIV Infection Regardless of Presence of HCV Coinfection.
- Author
-
Naggie S, Swiderska-Syn M, Choi S, Lusk S, Lan A, Ferrari G, Syn WK, Guy CD, and Diehl AM
- Abstract
Liver disease is a leading cause of HIV-related mortality. Hepatitis C virus (HCV)-related fibrogenesis is accelerated in the setting of HIV coinfection, yet the mechanisms underlying this aggressive pathogenesis are unclear. We identified formalin-fixed paraffin-embedded liver tissue for HIV-infected patients, HCV-infected patients, HIV/HCV-coinfected patients, and controls at Duke University Medical Center. De-identified sections were stained for markers against the wound repair Hedgehog (Hh) pathway, resident T-lymphocytes, and immune activation and cellular aging. HIV infection was independently associated with Hh activation and markers of immune dysregulation in the liver tissue.
- Published
- 2018
- Full Text
- View/download PDF
28. ALT-803, an IL-15 superagonist, in combination with nivolumab in patients with metastatic non-small cell lung cancer: a non-randomised, open-label, phase 1b trial.
- Author
-
Wrangle JM, Velcheti V, Patel MR, Garrett-Mayer E, Hill EG, Ravenel JG, Miller JS, Farhad M, Anderton K, Lindsey K, Taffaro-Neskey M, Sherman C, Suriano S, Swiderska-Syn M, Sion A, Harris J, Edwards AR, Rytlewski JA, Sanders CM, Yusko EC, Robinson MD, Krieg C, Redmond WL, Egan JO, Rhode PR, Jeng EK, Rock AD, Wong HC, and Rubinstein MP
- Subjects
- Aged, Antineoplastic Agents, Immunological adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Carcinoma, Non-Small-Cell Lung secondary, Female, Humans, Lung Neoplasms pathology, Male, Middle Aged, Neoplasm Staging, Nivolumab adverse effects, Proteins adverse effects, Recombinant Fusion Proteins, Time Factors, Treatment Outcome, United States, Antineoplastic Agents, Immunological administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Carcinoma, Non-Small-Cell Lung drug therapy, Lung Neoplasms drug therapy, Nivolumab administration & dosage, Proteins administration & dosage
- Abstract
Background: Immunotherapy with PD-1 or PD-L1 blockade fails to induce a response in about 80% of patients with unselected non-small cell lung cancer (NSCLC), and many of those who do initially respond then develop resistance to treatment. Agonists that target the shared interleukin-2 (IL-2) and IL-15Rβγ pathway have induced complete and durable responses in some cancers, but no studies have been done to assess the safety or efficacy of these agonists in combination with anti-PD-1 immunotherapy. We aimed to define the safety, tolerability, and activity of this drug combination in patients with NSCLC., Methods: In this non-randomised, open-label, phase 1b trial, we enrolled patients (aged ≥18 years) with previously treated histologically or cytologically confirmed stage IIIB or IV NSCLC from three academic hospitals in the USA. Key eligibility criteria included measurable disease, eligibility to receive anti-PD-1 immunotherapy, and an Eastern Cooperative Oncology Group performance status of 0 or 1. Patients received the anti-PD-1 monoclonal antibody nivolumab intravenously at 3 mg/kg (then 240 mg when US Food and Drug Administration [FDA]-approved dosing changed) every 14 days (either as new treatment or continued treatment at the time of disease progression) and the IL-15 superagonist ALT-803 subcutaneously once per week on weeks 1-5 of four 6-week cycles for 6 months. ALT-803 was administered at one of four escalating dose concentrations: 6, 10, 15, or 20 μg/kg. The primary endpoint was to define safety and tolerability and to establish a recommended phase 2 dose of ALT-803 in combination with nivolumab. Analyses were per-protocol and included any patients who received at least one dose of study treatment. This trial is registered with ClinicalTrials.gov, number NCT02523469; phase 2 enrolment of patients is ongoing., Findings: Between Jan 18, 2016, and June 28, 2017, 23 patients were enrolled and 21 were treated at four dose levels of ALT-803 in combination with nivolumab. Two patients did not receive treatment because of the development of inter-current illness during enrolment, one patient due to leucopenia and one patient due to pulmonary dysfunction. No dose-limiting toxicities were recorded and the maximum tolerated dose was not reached. The most common adverse events were injection-site reactions (in 19 [90%] of 21 patients) and flu-like symptoms (15 [71%]). The most common grade 3 adverse events, occurring in two patients each, were lymphocytopenia and fatigue. A grade 3 myocardial infarction occurred in one patient. No grade 4 or 5 adverse events were recorded. The recommended phase 2 dose of ALT-803 is 20 μg/kg given once per week subcutaneously in combination with 240 mg intravenous nivolumab every 2 weeks., Interpretation: ALT-803 in combination with nivolumab can be safely administered in an outpatient setting. The promising clinical activity observed with the addition of ALT-803 to the regimen of patients with PD-1 monoclonal antibody relapsed and refractory disease shows evidence of anti-tumour activity for a new class of agents in NSCLC., Funding: Altor BioScience (a NantWorks company), National Institutes of Health, and Medical University of South Carolina Hollings Cancer Center., (Copyright © 2018 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
29. Hedgehog-YAP Signaling Pathway Regulates Glutaminolysis to Control Activation of Hepatic Stellate Cells.
- Author
-
Du K, Hyun J, Premont RT, Choi SS, Michelotti GA, Swiderska-Syn M, Dalton GD, Thelen E, Rizi BS, Jung Y, and Diehl AM
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Animals, Case-Control Studies, Cell Cycle Proteins, Cell Proliferation, Cell Transdifferentiation, Cells, Cultured, Cellular Reprogramming, Gene Expression Regulation, Glutaminase metabolism, Hedgehog Proteins genetics, Hepatic Stellate Cells pathology, Humans, Ketoglutaric Acids metabolism, Liver pathology, Liver Cirrhosis genetics, Liver Cirrhosis pathology, Liver Cirrhosis, Experimental genetics, Liver Cirrhosis, Experimental metabolism, Liver Cirrhosis, Experimental pathology, Mice, Inbred C57BL, Mice, Knockout, Mitochondria, Liver metabolism, Mitochondria, Liver pathology, Myofibroblasts pathology, Phenotype, Phosphoproteins genetics, RNA Interference, Rats, Signal Transduction, Smoothened Receptor genetics, Smoothened Receptor metabolism, Time Factors, Transcription Factors, Transfection, YAP-Signaling Proteins, Adaptor Proteins, Signal Transducing metabolism, Energy Metabolism, Glutamine metabolism, Hedgehog Proteins metabolism, Hepatic Stellate Cells metabolism, Liver metabolism, Liver Cirrhosis metabolism, Myofibroblasts metabolism, Phosphoproteins metabolism
- Abstract
Background & Aims: Cirrhosis results from accumulation of myofibroblasts derived from quiescent hepatic stellate cells (Q-HSCs); it regresses when myofibroblastic HSCs are depleted. Hedgehog signaling promotes transdifferentiation of HSCs by activating Yes-associated protein 1 (YAP1 or YAP) and inducing aerobic glycolysis. However, increased aerobic glycolysis alone cannot meet the high metabolic demands of myofibroblastic HSCs. Determining the metabolic processes of these cells could lead to strategies to prevent progressive liver fibrosis, so we investigated whether glutaminolysis (conversion of glutamine to alpha-ketoglutarate) sustains energy metabolism and permits anabolism when Q-HSCs become myofibroblastic, and whether this is controlled by hedgehog signaling to YAP., Methods: Primary HSCs were isolated from C57BL/6 or Smo
flox/flox mice; we also performed studies with rat and human myofibroblastic HSCs. We measured changes of glutaminolytic genes during culture-induced primary HSC transdifferentiation. Glutaminolysis was disrupted in cells by glutamine deprivation or pathway inhibitors (bis-2-[5-phenylacetamido-1,2,4-thiadiazol-2-yl] ethyl sulfide, CB-839, epigallocatechin gallate, and aminooxyacetic acid), and effects on mitochondrial respiration, cell growth and migration, and fibrogenesis were measured. Hedgehog signaling to YAP was disrupted in cells by adenovirus expression of Cre-recombinase or by small hairpin RNA knockdown of YAP. Hedgehog and YAP activity were inhibited by incubation of cells with cyclopamine or verteporfin, and effects on glutaminolysis were measured. Acute and chronic liver fibrosis were induced in mice by intraperitoneal injection of CCl4 or methionine choline-deficient diet. Some mice were then given injections of bis-2-[5-phenylacetamido-1,2,4-thiadiazol-2-yl] ethyl sulfide to inhibit glutaminolysis, and myofibroblast accumulation was measured. We also performed messenger RNA and immunohistochemical analyses of percutaneous liver biopsies from healthy human and 4 patients with no fibrosis, 6 patients with mild fibrosis, and 3 patients with severe fibrosis., Results: Expression of genes that regulate glutaminolysis increased during transdifferentiation of primary Q-HSCs into myofibroblastic HSCs, and inhibition of glutaminolysis disrupted transdifferentiation. Blocking glutaminolysis in myofibroblastic HSCs suppressed mitochondrial respiration, cell growth and migration, and fibrogenesis; replenishing glutaminolysis metabolites to these cells restored these activities. Knockout of the hedgehog signaling intermediate smoothened or knockdown of YAP inhibited expression of glutaminase, the rate-limiting enzyme in glutaminolysis. Hedgehog and YAP inhibitors blocked glutaminolysis and suppressed myofibroblastic activities in HSCs. In livers of patients and of mice with acute or chronic fibrosis, glutaminolysis was induced in myofibroblastic HSCs. In mice with liver fibrosis, inhibition of glutaminase blocked accumulation of myofibroblasts and fibrosis progression., Conclusions: Glutaminolysis controls accumulation of myofibroblast HSCs in mice and might be a therapeutic target for cirrhosis., (Copyright © 2018 AGA Institute. Published by Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
30. Loss of pericyte smoothened activity in mice with genetic deficiency of leptin.
- Author
-
Xie G, Swiderska-Syn M, Jewell ML, Machado MV, Michelotti GA, Premont RT, and Diehl AM
- Subjects
- Adipocytes cytology, Adipocytes metabolism, Adipose Tissue cytology, Animals, Cell Differentiation, Cell Movement, Cell Proliferation, Cells, Cultured, Hedgehog Proteins physiology, Hepatic Stellate Cells cytology, Hepatic Stellate Cells metabolism, Leptin deficiency, Leptin metabolism, Mesenchymal Stem Cells cytology, Mesenchymal Stem Cells metabolism, Mesenchymal Stem Cells physiology, Mice, Mice, Obese, Myofibroblasts cytology, Myofibroblasts metabolism, Obesity genetics, Paracrine Communication genetics, Receptors, Leptin metabolism, Smoothened Receptor agonists, Hepatic Stellate Cells physiology, Leptin genetics, Obesity physiopathology, Smoothened Receptor genetics, Smoothened Receptor metabolism
- Abstract
Background: Obesity is associated with multiple diseases, but it is unclear how obesity promotes progressive tissue damage. Recovery from injury requires repair, an energy-expensive process that is coupled to energy availability at the cellular level. The satiety factor, leptin, is a key component of the sensor that matches cellular energy utilization to available energy supplies. Leptin deficiency signals energy depletion, whereas activating the Hedgehog pathway drives energy-consuming activities. Tissue repair is impaired in mice that are obese due to genetic leptin deficiency. Tissue repair is also blocked and obesity enhanced by inhibiting Hedgehog activity. We evaluated the hypothesis that loss of leptin silences Hedgehog signaling in pericytes, multipotent leptin-target cells that regulate a variety of responses that are often defective in obesity, including tissue repair and adipocyte differentiation., Results: We found that pericytes from liver and white adipose tissue require leptin to maintain expression of the Hedgehog co-receptor, Smoothened, which controls the activities of Hedgehog-regulated Gli transcription factors that orchestrate gene expression programs that dictate pericyte fate. Smoothened suppression prevents liver pericytes from being reprogrammed into myofibroblasts, but stimulates adipose-derived pericytes to become white adipocytes. Progressive Hedgehog pathway decay promotes senescence in leptin-deficient liver pericytes, which, in turn, generate paracrine signals that cause neighboring hepatocytes to become fatty and less proliferative, enhancing vulnerability to liver damage., Conclusions: Leptin-responsive pericytes evaluate energy availability to inform tissue construction by modulating Hedgehog pathway activity and thus, are at the root of progressive obesity-related tissue pathology. Leptin deficiency inhibits Hedgehog signaling in pericytes to trigger a pericytopathy that promotes both adiposity and obesity-related tissue damage.
- Published
- 2017
- Full Text
- View/download PDF
31. Hypoxia of the growing liver accelerates regeneration.
- Author
-
Schadde E, Tsatsaris C, Swiderska-Syn M, Breitenstein S, Urner M, Schimmer R, Booy C, Z'graggen BR, Wenger RH, Spahn DR, Hertl M, Knechtle S, Diehl AM, Schläpfer M, and Beck-Schimmer B
- Subjects
- Animals, Cell Hypoxia physiology, Hepatectomy, Ligation, Male, Models, Animal, Rats, Rats, Wistar, Liver blood supply, Liver pathology, Liver Regeneration physiology, Portal Vein
- Abstract
Background: After portal vein ligation of 1 side of the liver, the other side regenerates at a slow rate. This slow growth may be accelerated to rapid growth by adding a transection between the 2 sides, i.e., performing portal vein ligation and parenchymal transection. We found that in patients undergoing portal vein ligation and parenchymal transection, portal vein hyperflow in the regenerating liver causes a significant reduction of arterial flow due to the hepatic arterial buffer response. We postulated that the reduction of arterial flow induces hypoxia in the regenerating liver and used a rat model to assess hypoxia and its impact on kinetic growth., Methods: A rat model of rapid (portal vein ligation and parenchymal transection) and slow regeneration (portal vein ligation) was established. Portal vein flow and pressure data were collected. Liver regeneration was assessed in rats using computed tomography, proliferation with Ki-67, and hypoxia with pimonidazole and HIF-1α staining., Results: The rat model confirmed acceleration of regeneration in portal vein ligation and parenchymal transection as well as the portal vein hyperflow seen in patients. Additionally, tissue hypoxia was observed after portal vein ligation and parenchymal transection, while little hypoxia staining was detected after portal vein ligation. To determine if hypoxia is a consequence or an inciting stimulus of rapid liver regeneration, we used a prolyl-hydroxylase blocker to activate hypoxia signaling pathways in the slow model. This clearly accelerated slow to rapid liver regeneration. Inversely, abrogation of hypoxia led to a blunting of rapid growth to slow growth. The topical application of prolyl-hydroxylase inhibitors on livers in rats induced spontaneous areas of regeneration., Conclusion: This study shows that pharmacologically induced hypoxic signaling accelerates liver regeneration similar to portal vein ligation and parenchymal transection. Hypoxia is likely an accelerator of liver regeneration. Also, prolyl-hydroxylase inhibitors may be used to enhance liver regeneration pharmaceutically., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
32. Hedgehog regulates yes-associated protein 1 in regenerating mouse liver.
- Author
-
Swiderska-Syn M, Xie G, Michelotti GA, Jewell ML, Premont RT, Syn WK, and Diehl AM
- Subjects
- Animals, Cell Cycle Proteins, Cell Dedifferentiation, Cell Proliferation, Hepatectomy, Hepatocytes physiology, Male, Mice, Inbred C57BL, Mice, Transgenic, Paracrine Communication, Up-Regulation, YAP-Signaling Proteins, Adaptor Proteins, Signal Transducing metabolism, Hedgehog Proteins metabolism, Hepatic Stellate Cells metabolism, Liver Regeneration, Phosphoproteins metabolism
- Abstract
Unlabelled: Adult liver regeneration requires induction and suppression of proliferative activity in multiple types of liver cells. The mechanisms that orchestrate the global changes in gene expression that are required for proliferative activity to change within individual liver cells, and that coordinate proliferative activity among different types of liver cells, are not well understood. Morphogenic signaling pathways that are active during fetal development, including Hedgehog and Hippo/Yes-associated protein 1 (Yap1), regulate liver regeneration in adulthood. Cirrhosis and liver cancer result when these pathways become dysregulated, but relatively little is known about the mechanisms that coordinate and control morphogenic signaling during effective liver regeneration. We evaluated the hypothesis that the Hedgehog pathway controls Yap1 activation during liver regeneration by studying intact mice and cultured liver cells. In cultured hepatic stellate cells (HSCs), disrupting Hedgehog signaling blocked activation of Yap1, and knocking down Yap1 inhibited induction of both Yap1- and Hedgehog-regulated genes that enable HSC to become myofibroblasts (MFs). In mice, disrupting Hedgehog signaling in MFs inhibited liver regeneration after partial hepactectomy (PH). Reduced proliferative activity in the liver epithelial compartment resulted from loss of stroma-derived paracrine signals that activate Yap1 and the Hedgehog pathway in hepatocytes. This prevented hepatocytes from up-regulating Yap1- and Hedgehog-regulated transcription factors that normally promote their proliferation., Conclusions: Morphogenic signaling in HSCs is necessary to reprogram hepatocytes to regenerate the liver epithelial compartment post-PH. This discovery identifies novel molecules that might be targeted to correct defective repair during cirrhosis and liver cancer. (Hepatology 2016;64:232-244)., (© 2016 by the American Association for the Study of Liver Diseases.)
- Published
- 2016
- Full Text
- View/download PDF
33. Pleiotrophin regulates the ductular reaction by controlling the migration of cells in liver progenitor niches.
- Author
-
Michelotti GA, Tucker A, Swiderska-Syn M, Machado MV, Choi SS, Kruger L, Soderblom E, Thompson JW, Mayer-Salman M, Himburg HA, Moylan CA, Guy CD, Garman KS, Premont RT, Chute JP, and Diehl AM
- Subjects
- Animals, Biomarkers blood, Blotting, Western, Cell Differentiation physiology, Immunohistochemistry, Mice, Mice, Knockout, Phosphoproteins metabolism, RNA analysis, Real-Time Polymerase Chain Reaction, Receptor-Like Protein Tyrosine Phosphatases, Class 5 metabolism, Signal Transduction, Bile Ducts pathology, Carrier Proteins physiology, Cell Movement physiology, Cytokines physiology, Liver Diseases pathology
- Abstract
Objective: The ductular reaction (DR) involves mobilisation of reactive-appearing duct-like cells (RDC) along canals of Hering, and myofibroblastic (MF) differentiation of hepatic stellate cells (HSC) in the space of Disse. Perivascular cells in stem cell niches produce pleiotrophin (PTN) to inactivate the PTN receptor, protein tyrosine phosphatase receptor zeta-1 (PTPRZ1), thereby augmenting phosphoprotein-dependent signalling. We hypothesised that the DR is regulated by PTN/PTPRZ1 signalling., Design: PTN-GFP, PTN-knockout (KO), PTPRZ1-KO, and wild type (WT) mice were examined before and after bile duct ligation (BDL) for PTN, PTPRZ1 and the DR. RDC and HSC from WT, PTN-KO, and PTPRZ1-KO mice were also treated with PTN to determine effects on downstream signaling phosphoproteins, gene expression, growth, and migration. Liver biopsies from patients with DRs were also interrogated., Results: Although quiescent HSC and RDC lines expressed PTN and PTPRZ1 mRNAs, neither PTN nor PTPRZ1 protein was demonstrated in healthy liver. BDL induced PTN in MF-HSC and increased PTPRZ1 in MF-HSC and RDC. In WT mice, BDL triggered a DR characterised by periportal accumulation of collagen, RDC and MF-HSC. All aspects of this DR were increased in PTN-KO mice and suppressed in PTPRZ1-KO mice. In vitro studies revealed PTN-dependent accumulation of phosphoproteins that control cell-cell adhesion and migration, with resultant inhibition of cell migration. PTPRZ1-positive cells were prominent in the DRs of patients with ductal plate defects and adult cholestatic diseases., Conclusions: PTN, and its receptor, PTPRZ1, regulate the DR to liver injury by controlling the migration of resident cells in adult liver progenitor niches., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/)
- Published
- 2016
- Full Text
- View/download PDF
34. Osteopontin is a proximal effector of leptin-mediated non-alcoholic steatohepatitis (NASH) fibrosis.
- Author
-
Coombes JD, Choi SS, Swiderska-Syn M, Manka P, Reid DT, Palma E, Briones-Orta MA, Xie G, Younis R, Kitamura N, Della Peruta M, Bitencourt S, Dollé L, Oo YH, Mi Z, Kuo PC, Williams R, Chokshi S, Canbay A, Claridge LC, Eksteen B, Diehl AM, and Syn WK
- Subjects
- Animals, Cell Line, Cells, Cultured, Gene Deletion, Hepatocytes metabolism, Hepatocytes pathology, Leptin genetics, Liver metabolism, Liver Cirrhosis genetics, Liver Cirrhosis pathology, Male, Mice, Inbred C57BL, Non-alcoholic Fatty Liver Disease genetics, Non-alcoholic Fatty Liver Disease pathology, Osteopontin genetics, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, Rats, Sprague-Dawley, Signal Transduction, Up-Regulation, Leptin metabolism, Liver pathology, Liver Cirrhosis metabolism, Non-alcoholic Fatty Liver Disease metabolism, Osteopontin metabolism
- Abstract
Introduction: Liver fibrosis develops when hepatic stellate cells (HSC) are activated into collagen-producing myofibroblasts. In non-alcoholic steatohepatitis (NASH), the adipokine leptin is upregulated, and promotes liver fibrosis by directly activating HSC via the hedgehog pathway. We reported that hedgehog-regulated osteopontin (OPN) plays a key role in promoting liver fibrosis. Herein, we evaluated if OPN mediates leptin-profibrogenic effects in NASH., Methods: Leptin-deficient (ob/ob) and wild-type (WT) mice were fed control or methionine-choline deficient (MCD) diet. Liver tissues were assessed by Sirius-red, OPN and αSMA IHC, and qRT-PCR for fibrogenic genes. In vitro, HSC with stable OPN (or control) knockdown were treated with recombinant (r)leptin and OPN-neutralizing or sham-aptamers. HSC response to OPN loss was assessed by wound healing assay. OPN-aptamers were also added to precision-cut liver slices (PCLS), and administered to MCD-fed WT (leptin-intact) mice to determine if OPN neutralization abrogated fibrogenesis., Results: MCD-fed WT mice developed NASH-fibrosis, upregulated OPN, and accumulated αSMA+ cells. Conversely, MCD-fed ob/ob mice developed less fibrosis and accumulated fewer αSMA+ and OPN+ cells. In vitro, leptin-treated HSC upregulated OPN, αSMA, collagen 1α1 and TGFβ mRNA by nearly 3-fold, but this effect was blunted by OPN loss. Inhibition of PI3K and transduction of dominant negative-Akt abrogated leptin-mediated OPN induction, while constitutive active-Akt upregulated OPN. Finally, OPN neutralization reduced leptin-mediated fibrogenesis in both PCLS and MCD-fed mice., Conclusion: OPN overexpression in NASH enhances leptin-mediated fibrogenesis via PI3K/Akt. OPN neutralization significantly reduces NASH fibrosis, reinforcing the potential utility of targeting OPN in the treatment of patients with advanced NASH., (Copyright © 2015 Elsevier B.V. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
35. Ductal metaplasia in oesophageal submucosal glands is associated with inflammation and oesophageal adenocarcinoma.
- Author
-
Garman KS, Kruger L, Thomas S, Swiderska-Syn M, Moser BK, Diehl AM, and McCall SJ
- Subjects
- Adenocarcinoma surgery, Adult, Aged, Aged, 80 and over, Esophageal Neoplasms surgery, Esophagectomy, Esophagus surgery, Female, Humans, Inflammation surgery, Male, Metaplasia pathology, Metaplasia surgery, Middle Aged, Adenocarcinoma pathology, Esophageal Neoplasms pathology, Esophagus pathology, Inflammation pathology
- Abstract
Aims: Recent studies have suggested that oesophageal submucosal gland (ESMG) ducts harbour progenitor cells that may contribute to oesophageal metaplasia. Our objective was to determine whether histological differences exist between the ESMGs of individuals with and without oesophageal adenocarcinoma (EAC)., Methods and Results: We performed histological assessment of 343 unique ESMGs from 30 control patients, 24 patients with treatment-naïve high-grade columnar dysplasia (HGD) or EAC, and 23 non-EAC oesophagectomy cases. A gastrointestinal pathologist assessed haematoxylin and eosin-stained ESMG images by using a scoring system that assigns individual ESMG acini to five histological types (mucous, serous, oncocytic, dilated, or ductal metaplastic). In our model, ductal metaplastic acini were more common in patients with HGD/EAC (12.7%) than in controls (3.5%) (P = 0.006). We also identified greater proportions of acini with dilation (21.9%, P < 0.001) and, to a lesser extent, ductal metaplasia (4.3%, P = 0.001) in non-EAC oesophagectomy cases than in controls. Ductal metaplasia tended to occur in areas of mucosal ulceration or tumour., Conclusions: We found a clear association between ductal metaplastic ESMG acini and HGD/EAC. Non-EAC cases had dilated acini and some ductal dilation. Because ESMGs and ducts harbour putative progenitor cells, these associations could have significance for understanding the pathogenesis of EAC., (© 2015 John Wiley & Sons Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
36. Role of Fn14 in acute alcoholic steatohepatitis in mice.
- Author
-
Karaca G, Xie G, Moylan C, Swiderska-Syn M, Guy CD, Krüger L, Machado MV, Choi SS, Michelotti GA, Burkly LC, and Diehl AM
- Subjects
- Acute Disease, Animals, Apoptosis, Carbon Tetrachloride, Cell Proliferation, Disease Models, Animal, Ethanol, Fatty Liver, Alcoholic etiology, Fatty Liver, Alcoholic genetics, Fatty Liver, Alcoholic pathology, Hydroxyproline metabolism, Inflammation Mediators metabolism, Liver pathology, Liver Cirrhosis, Alcoholic etiology, Liver Cirrhosis, Alcoholic metabolism, Liver Cirrhosis, Alcoholic pathology, Male, Mice, Inbred C57BL, Mice, Knockout, Receptors, Tumor Necrosis Factor deficiency, Receptors, Tumor Necrosis Factor genetics, Signal Transduction, TWEAK Receptor, Time Factors, Tumor Necrosis Factor-alpha metabolism, Wound Healing, Fatty Liver, Alcoholic metabolism, Liver metabolism, Receptors, Tumor Necrosis Factor metabolism
- Abstract
TNF-like weak inducer of apoptosis (TWEAK) is a growth factor for bipotent liver progenitors that express its receptor, fibroblast growth factor-inducible 14 (Fn14), a TNF receptor superfamily member. Accumulation of Fn14(+) progenitors occurs in severe acute alcoholic steatohepatitis (ASH) and correlates with acute mortality. In patients with severe ASH, inhibition of TNF-α increases acute mortality. The aim of this study was to determine whether deletion of Fn14 improves the outcome of liver injury in alcohol-consuming mice. Wild-type (WT) and Fn14 knockout (KO) mice were fed control high-fat Lieber deCarli diet or high-fat Lieber deCarli diet with 2% alcohol (ETOH) and injected intraperitoneally with CCl₄ for 2 wk to induce liver injury. Mice were euthanized 3 or 10 days after CCl₄ treatment. Survival was assessed. Liver tissues were analyzed for cell death, inflammation, proliferation, progenitor accumulation, and fibrosis by quantitative RT-PCR, immunoblot, hydroxyproline content, and quantitative immunohistochemistry. During liver injury, Fn14 expression, apoptosis, inflammation, hepatocyte replication, progenitor and myofibroblast accumulation, and fibrosis increased in WT mice fed either diet. Mice fed either diet expressed similar TWEAK/Fn14 levels, but ETOH-fed mice had higher TNF-α expression. The ETOH-fed group developed more apoptosis, inflammation, fibrosis, and regenerative responses. Fn14 deletion did not reduce hepatic TNF-α expression but improved all injury parameters in mice fed the control diet. In ETOH-fed mice, Fn14 deletion inhibited TNF-α induction and increased acute mortality, despite improvement in liver injury. Fn14 mediates wound-healing responses that are necessary to survive acute liver injury during alcohol exposure.
- Published
- 2015
- Full Text
- View/download PDF
37. Repair-related activation of hedgehog signaling in stromal cells promotes intrahepatic hypothyroidism.
- Author
-
Bohinc BN, Michelotti G, Xie G, Pang H, Suzuki A, Guy CD, Piercy D, Kruger L, Swiderska-Syn M, Machado M, Pereira T, Zavacki AM, Abdelmalek M, and Diehl AM
- Subjects
- Animals, Case-Control Studies, Cells, Cultured, Gene Expression Regulation, Hedgehog Proteins metabolism, Humans, Hypothyroidism metabolism, Liver pathology, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Non-alcoholic Fatty Liver Disease genetics, Non-alcoholic Fatty Liver Disease metabolism, Non-alcoholic Fatty Liver Disease physiopathology, Rats, Rats, Sprague-Dawley, Signal Transduction genetics, Stromal Cells pathology, Thyroid Hormones metabolism, Wound Healing physiology, Hedgehog Proteins genetics, Hypothyroidism etiology, Liver metabolism, Liver Regeneration physiology, Stromal Cells metabolism
- Abstract
Thyroid hormone (TH) is important for tissue repair because it regulates cellular differentiation. Intrahepatic TH activity is controlled by both serum TH levels and hepatic deiodinases. TH substrate (T4) is converted into active hormone (T3) by deiodinase 1 (D1) but into inactive hormone (rT3) by deiodinase 3 (D3). Although the relative expressions of D1 and D3 are known to change during liver injury, the cell types and signaling mechanisms involved are unclear. We evaluated the hypothesis that changes in hepatic deiodinases result from repair-related activation of the Hedgehog pathway in stromal cells. We localized deiodinase expression, assessed changes during injury, and determined how targeted manipulation of Hedgehog signaling in stromal cells impacted hepatic deiodinase expression, TH content, and TH action in rodents. Humans with chronic liver disease were also studied. In healthy liver, hepatocytes strongly expressed D1 and stromal cells weakly expressed D3. During injury, hepatocyte expression of D1 decreased, whereas stromal expression of D3 increased, particularly in myofibroblasts. Conditionally disrupting Hedgehog signaling in myofibroblasts normalized deiodinase expression. Repair-related changes in deiodinases were accompanied by reduced hepatic TH content and TH-regulated gene expression. In patients, this was reflected by increased serum rT3. Moreover, the decreases in the free T3 to rT3 and free T4 to rT3 ratios distinguished advanced from mild fibrosis, even in individuals with similar serum levels of TSH and free T4. In conclusion, the Hedgehog-dependent changes in liver stromal cells drive repair-related changes in hepatic deiodinase expression that promote intrahepatic hypothyroidism, thereby limiting exposure to T3, an important factor for cellular differentiation.
- Published
- 2014
- Full Text
- View/download PDF
38. Osteopontin is up-regulated in chronic hepatitis C and is associated with cellular permissiveness for hepatitis C virus replication.
- Author
-
Choi SS, Claridge LC, Jhaveri R, Swiderska-Syn M, Clark P, Suzuki A, Pereira TA, Mi Z, Kuo PC, Guy CD, Pereira FE, Diehl AM, Patel K, and Syn WK
- Subjects
- Adult, Base Sequence, DNA Primers, Female, Humans, Male, Middle Aged, Osteopontin blood, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Hepacivirus physiology, Hepatitis C, Chronic physiopathology, Osteopontin physiology, Up-Regulation, Virus Replication
- Abstract
OPN (osteopontin)) is a Hh (Hedgehog)-regulated cytokine that is up-regulated during chronic liver injury and directly promotes fibrosis. We have reported that Hh signalling enhances viral permissiveness and replication in HCV (hepatitis C virus)-infected cells. Hence we hypothesized that OPN directly promotes HCV replication, and that targeting OPN could be beneficial in HCV. In the present study, we compared the expression of OPN mRNA and protein in HCV (JFH1)-infected Huh7 and Huh7.5 cells, and evaluated whether modulating OPN levels using exogenous OPN ligands (up-regulate OPN) or OPN-specific RNA-aptamers (neutralize OPN) leads to changes in HCV expression. Sera and livers from patients with chronic HCV were analysed to determine whether OPN levels were associated with disease severity or response to therapy. Compared with Huh7 cells, Huh7.5 cells support higher levels of HCV replication (15-fold) and expressed significantly more OPN mRNA (30-fold) and protein. Treating Huh7 cells with OPN ligands led to a dose-related increase in HCV (15-fold) and OPN (8-fold) mRNA. Conversely, treating Huh7.5 cells with OPN-specific RNA aptamers inhibited HCV RNA and protein by >50% and repressed OPN mRNA to basal levels. Liver OPN expression was significantly higher (3-fold) in patients with advanced fibrosis. Serum OPN positively correlated with fibrosis-stage (P=0.009), but negatively correlated with ETBCR (end-of-treatment biochemical response), ETVR (end-of-treatment virological response), SBCR (sustained biochemical response) and SVR (sustained virological response) (P=0.007). The OPN fibrosis score (serum OPN and presence of fibrosis ≥F2) may be a predictor of SVR. In conclusion, OPN is up-regulated in the liver and serum of patients with chronic hepatitis C, and supports increased viral replication. OPN neutralization may be a novel therapeutic strategy in chronic hepatitis C.
- Published
- 2014
- Full Text
- View/download PDF
39. Alcohol activates the hedgehog pathway and induces related procarcinogenic processes in the alcohol-preferring rat model of hepatocarcinogenesis.
- Author
-
Chan IS, Guy CD, Machado MV, Wank A, Kadiyala V, Michelotti G, Choi S, Swiderska-Syn M, Karaca G, Pereira TA, Yip-Schneider MT, Max Schmidt C, and Diehl AM
- Subjects
- Animals, Epithelial-Mesenchymal Transition, Liver drug effects, Liver metabolism, Liver pathology, Random Allocation, Rats, Carcinogenesis drug effects, Central Nervous System Depressants adverse effects, Ethanol adverse effects, Hedgehog Proteins metabolism, Liver Neoplasms, Experimental chemically induced
- Abstract
Background: Alcohol consumption promotes hepatocellular carcinoma (HCC). The responsible mechanisms are not well understood. Hepatocarcinogenesis increases with age and is enhanced by factors that impose a demand for liver regeneration. Because alcohol is hepatotoxic, habitual alcohol ingestion evokes a recurrent demand for hepatic regeneration. The alcohol-preferring (P) rat model mimics the level of alcohol consumption by humans who habitually abuse alcohol. Previously, we showed that habitual heavy alcohol ingestion amplified age-related hepatocarcinogenesis in P rats, with over 80% of alcohol-consuming P rats developing HCCs after 18 months of alcohol exposure, compared with only 5% of water-drinking controls., Methods: Herein, we used quantitative real-time PCR and quantitative immunocytochemistry to compare liver tissues from alcohol-consuming P rats and water-fed P rat controls after 6, 12, or 18 months of drinking. We aimed to identify potential mechanisms that might underlie the differences in liver cancer formation and hypothesized that chronic alcohol ingestion would activate Hedgehog (HH), a regenerative signaling pathway that is overactivated in HCC., Results: Chronic alcohol ingestion amplified age-related degenerative changes in hepatocytes, but did not cause appreciable liver inflammation or fibrosis even after 18 months of heavy drinking. HH signaling was also enhanced by alcohol exposure, as evidenced by increased levels of mRNAs encoding HH ligands, HH-regulated transcription factors, and HH target genes. Immunocytochemistry confirmed increased alcohol-related accumulation of HH ligand-producing cells and HH-responsive target cells. HH-related regenerative responses were also induced in alcohol-exposed rats. Three of these processes (i.e., deregulated progenitor expansion, the reverse Warburg effect, and epithelial-to-mesenchymal transitions) are known to promote cancer growth in other tissues., Conclusions: Alcohol-related changes in Hedgehog signaling and resultant deregulation of liver cell replacement might promote hepatocarcinogenesis., (Copyright © 2013 by the Research Society on Alcoholism.)
- Published
- 2014
- Full Text
- View/download PDF
40. TWEAK/Fn14 signaling is required for liver regeneration after partial hepatectomy in mice.
- Author
-
Karaca G, Swiderska-Syn M, Xie G, Syn WK, Krüger L, Machado MV, Garman K, Choi SS, Michelotti GA, Burkly LC, Ochoa B, and Diehl AM
- Subjects
- Animals, Antibodies pharmacology, Cell Proliferation drug effects, Cytokine TWEAK, Epithelial Cells cytology, Epithelial Cells drug effects, Gene Deletion, Hepatocytes cytology, Hepatocytes drug effects, Hepatocytes metabolism, Liver cytology, Mice, Mice, Inbred C57BL, Mice, Knockout, Mitogens metabolism, TWEAK Receptor, Tumor Necrosis Factor Inhibitors, Hepatectomy, Liver metabolism, Liver surgery, Liver Regeneration drug effects, Receptors, Tumor Necrosis Factor metabolism, Signal Transduction drug effects, Tumor Necrosis Factors metabolism
- Abstract
Background & Aims: Pro-inflammatory cytokines are important for liver regeneration after partial hepatectomy (PH). Expression of Fibroblast growth factor-inducible 14 (Fn14), the receptor for TNF-like weak inducer of apoptosis (TWEAK), is induced rapidly after PH and remains elevated throughout the period of peak hepatocyte replication. The role of Fn14 in post-PH liver regeneration is uncertain because Fn14 is expressed by liver progenitors and TWEAK-Fn14 interactions stimulate progenitor growth, but replication of mature hepatocytes is thought to drive liver regeneration after PH., Methods: To clarify the role of TWEAK-Fn14 after PH, we compared post-PH regenerative responses in wild type (WT) mice, Fn14 knockout (KO) mice, TWEAK KO mice, and WT mice treated with anti-TWEAK antibodies., Results: In WT mice, rare Fn14(+) cells localized with other progenitor markers in peri-portal areas before PH. PH rapidly increased proliferation of Fn14(+) cells; hepatocytic cells that expressed Fn14 and other progenitor markers, such as Lgr5, progressively accumulated from 12-8 h post-PH and then declined to baseline by 96 h. When TWEAK/Fn14 signaling was disrupted, progenitor accumulation, induction of pro-regenerative cytokines, hepatocyte and cholangiocyte proliferation, and over-all survival were inhibited, while post-PH liver damage and bilirubin levels were increased. TWEAK stimulated proliferation and increased Lgr5 expression in cultured liver progenitors, but had no effect on either parameter in cultured primary hepatocytes., Conclusions: TWEAK-FN14 signaling is necessary for the healthy adult liver to regenerate normally after acute partial hepatectomy.
- Published
- 2014
- Full Text
- View/download PDF
41. Cross-talk between Notch and Hedgehog regulates hepatic stellate cell fate in mice.
- Author
-
Xie G, Karaca G, Swiderska-Syn M, Michelotti GA, Krüger L, Chen Y, Premont RT, Choi SS, and Diehl AM
- Subjects
- Animals, Calcium-Binding Proteins metabolism, Cell Lineage, Dipeptides pharmacology, Genotype, Hepatic Stellate Cells cytology, Intercellular Signaling Peptides and Proteins metabolism, Male, Membrane Proteins metabolism, Mice, Mice, Inbred C57BL, Myofibroblasts physiology, Phenotype, Serrate-Jagged Proteins, Hedgehog Proteins physiology, Hepatic Stellate Cells physiology, Receptors, Notch physiology, Signal Transduction physiology
- Abstract
Unlabelled: Liver repair involves phenotypic changes in hepatic stellate cells (HSCs) and reactivation of morphogenic signaling pathways that modulate epithelial-to-mesenchymal/mesenchymal-to-epithelial transitions, such as Notch and Hedgehog (Hh). Hh stimulates HSCs to become myofibroblasts (MFs). Recent lineage tracing studies in adult mice with injured livers showed that some MFs became multipotent progenitors to regenerate hepatocytes, cholangiocytes, and HSCs. We studied primary HSC cultures and two different animal models of fibrosis to evaluate the hypothesis that activating the Notch pathway in HSCs stimulates them to become (and remain) MFs through a mechanism that involves an epithelial-to-mesenchymal-like transition and requires cross-talk with the canonical Hh pathway. We found that when cultured HSCs transitioned into MFs, they activated Hh signaling, underwent an epithelial-to-mesenchymal-like transition, and increased Notch signaling. Blocking Notch signaling in MFs/HSCs suppressed Hh activity and caused a mesenchymal-to-epithelial-like transition. Inhibiting the Hh pathway suppressed Notch signaling and also induced a mesenchymal-to-epithelial-like transition. Manipulating Hh and Notch signaling in a mouse multipotent progenitor cell line evoked similar responses. In mice, liver injury increased Notch activity in MFs and Hh-responsive MF progeny (i.e., HSCs and ductular cells). Conditionally disrupting Hh signaling in MFs of bile-duct-ligated mice inhibited Notch signaling and blocked accumulation of both MF and ductular cells., Conclusions: The Notch and Hedgehog pathways interact to control the fate of key cell types involved in adult liver repair by modulating epithelial-to-mesenchymal-like/mesenchymal-to-epithelial-like transitions., (© 2013 by the American Association for the Study of Liver Diseases.)
- Published
- 2013
- Full Text
- View/download PDF
42. Hedgehog pathway and pediatric nonalcoholic fatty liver disease.
- Author
-
Swiderska-Syn M, Suzuki A, Guy CD, Schwimmer JB, Abdelmalek MF, Lavine JE, and Diehl AM
- Subjects
- Actins metabolism, Adolescent, Age Factors, Biopsy, Child, Cross-Sectional Studies, Fatty Liver metabolism, Female, Humans, Keratin-7 metabolism, Kruppel-Like Transcription Factors metabolism, Liver metabolism, Liver pathology, Male, Non-alcoholic Fatty Liver Disease, Nuclear Proteins metabolism, Portal System metabolism, Portal System pathology, Sex Factors, Vimentin metabolism, Zinc Finger Protein Gli2, Fatty Liver physiopathology, Hedgehog Proteins physiology, Liver physiopathology, Signal Transduction physiology
- Abstract
Unlabelled: It is unclear why the histology of pediatric and adult nonalcoholic fatty liver disease (NAFLD) sometimes differs. In adults, severity of portal inflammation and fibrosis correlate with Hedgehog pathway activity. Hedgehog (Hh) signaling regulates organogenesis, but is silent in adult livers until injury reinduces Hh ligand production. During adolescence, liver development is completed and children's livers normally lose cells that produce and/or respond to Hh ligands. We postulated that fatty liver injury interferes with this process by increasing Hh ligand production, and theorized that hepatic responses to Hh ligands might differ among children according to age, gender, and/or puberty status. Using unstained liver biopsy slides from 56 children with NAFLD, we performed immunohistochemistry to assess Hh pathway activation and correlated the results with clinical information obtained at biopsy. Fibrosis stage generally correlated with Hh pathway activity, as demonstrated by the numbers of Hh-ligand-producing cells (P < 0.0001) and Hh-responsive (glioma-associated oncogene 2-positive [Gli2]) cells (P = 0.0013). The numbers of Gli2(+) cells also correlated with portal inflammation grade (P = 0.0012). Two distinct zonal patterns of Hh-ligand production, portal/periportal versus lobular, were observed. Higher portal/periportal Hh-ligand production was associated with male gender. Male gender and prepuberty were also associated with ductular proliferation (P < 0.05), increased numbers of portal Gli2(+) cells (P < 0.017) and portal fibrosis., Conclusion: The portal/periportal (progenitor) compartment of prepubescent male livers exhibits high Hh pathway activity. This may explain the unique histologic features of pediatric NAFLD because Hh signaling promotes the fibroductular response., (Copyright © 2013 American Association for the Study of Liver Diseases.)
- Published
- 2013
- Full Text
- View/download PDF
43. Paracrine Hedgehog signaling drives metabolic changes in hepatocellular carcinoma.
- Author
-
Chan IS, Guy CD, Chen Y, Lu J, Swiderska-Syn M, Michelotti GA, Karaca G, Xie G, Krüger L, Syn WK, Anderson BR, Pereira TA, Choi SS, Baldwin AS, and Diehl AM
- Subjects
- Animals, Carcinoma, Hepatocellular complications, Carcinoma, Hepatocellular pathology, Cells, Cultured, Fatty Liver complications, Fatty Liver metabolism, Fatty Liver pathology, Glycolysis physiology, Hedgehog Proteins metabolism, Hep G2 Cells, Humans, Lactic Acid metabolism, Lipogenesis physiology, Liver Neoplasms complications, Liver Neoplasms pathology, Mice, Mice, Knockout, Myofibroblasts metabolism, Myofibroblasts pathology, Non-alcoholic Fatty Liver Disease, Carcinoma, Hepatocellular metabolism, Hedgehog Proteins physiology, Liver Neoplasms metabolism, Paracrine Communication physiology
- Abstract
Hepatocellular carcinoma (HCC) typically develops in cirrhosis, a condition characterized by Hedgehog (Hh) pathway activation and accumulation of Hh-responsive myofibroblasts. Although Hh signaling generally regulates stromal-epithelial interactions that support epithelial viability, the role of Hh-dependent myofibroblasts in hepatocarcinogenesis is unknown. Here, we used human HCC samples, a mouse HCC model, and hepatoma cell/myofibroblast cocultures to examine the hypothesis that Hh signaling modulates myofibroblasts' metabolism to generate fuels for neighboring malignant hepatocytes. The results identify a novel paracrine mechanism whereby malignant hepatocytes produce Hh ligands to stimulate glycolysis in neighboring myofibroblasts, resulting in release of myofibroblast-derived lactate that the malignant hepatocytes use as an energy source. This discovery reveals new diagnostic and therapeutic targets that might be exploited to improve the outcomes of cirrhotic patients with HCCs.
- Published
- 2012
- Full Text
- View/download PDF
44. Hedgehog controls hepatic stellate cell fate by regulating metabolism.
- Author
-
Chen Y, Choi SS, Michelotti GA, Chan IS, Swiderska-Syn M, Karaca GF, Xie G, Moylan CA, Garibaldi F, Premont R, Suliman HB, Piantadosi CA, and Diehl AM
- Subjects
- Actins genetics, Actins metabolism, Animals, Bile Ducts, Carbon Tetrachloride, Cells, Cultured, Gene Expression Profiling, Glycolysis genetics, Hepatic Stellate Cells cytology, Hepatic Stellate Cells enzymology, Humans, Hypoxia-Inducible Factor 1, alpha Subunit, Lactic Acid metabolism, Ligation, Liver Cirrhosis chemically induced, Liver Cirrhosis genetics, Liver Cirrhosis metabolism, Male, Mice, Mice, Inbred C57BL, Mitochondria, Myofibroblasts enzymology, Pyruvate Kinase genetics, Pyruvate Kinase metabolism, RNA, Messenger metabolism, Rats, Time Factors, Cell Transdifferentiation genetics, Gene Expression Regulation, Hedgehog Proteins genetics, Hedgehog Proteins metabolism, Hepatic Stellate Cells metabolism, Myofibroblasts metabolism, Signal Transduction genetics
- Abstract
Background & Aims: The pathogenesis of cirrhosis, a disabling outcome of defective liver repair, involves deregulated accumulation of myofibroblasts derived from quiescent hepatic stellate cells (HSCs), but the mechanisms that control transdifferentiation of HSCs are poorly understood. We investigated whether the Hedgehog (Hh) pathway controls the fate of HSCs by regulating metabolism., Methods: Microarray, quantitative polymerase chain reaction, and immunoblot analyses were used to identify metabolic genes that were differentially expressed in quiescent vs myofibroblast HSCs. Glycolysis and lactate production were disrupted in HSCs to determine if metabolism influenced transdifferentiation. Hh signaling and hypoxia-inducible factor 1α (HIF1α) activity were altered to identify factors that alter glycolytic activity. Changes in expression of genes that regulate glycolysis were quantified and localized in biopsy samples from patients with cirrhosis and liver samples from mice following administration of CCl(4) or bile duct ligation. Mice were given systemic inhibitors of Hh to determine if they affect glycolytic activity of the hepatic stroma; Hh signaling was also conditionally disrupted in myofibroblasts to determine the effects of glycolytic activity., Results: Transdifferentiation of cultured, quiescent HSCs into myofibroblasts induced glycolysis and caused lactate accumulation. Increased expression of genes that regulate glycolysis required Hh signaling and involved induction of HIF1α. Inhibitors of Hh signaling, HIF1α, glycolysis, or lactate accumulation converted myofibroblasts to quiescent HSCs. In diseased livers of animals and patients, numbers of glycolytic stromal cells were associated with the severity of fibrosis. Conditional disruption of Hh signaling in myofibroblasts reduced numbers of glycolytic myofibroblasts and liver fibrosis in mice; similar effects were observed following administration of pharmacologic inhibitors of Hh., Conclusions: Hedgehog signaling controls the fate of HSCs by regulating metabolism. These findings might be applied to diagnosis and treatment of patients with cirrhosis., (Copyright © 2012 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.