39 results on '"Evason KJ"'
Search Results
2. The KEAP1-NRF2 pathway regulates TFEB/TFE3-dependent lysosomal biogenesis
- Author
-
Ong, AJS, Bladen, CE, Tigani, TA, Karamalakis, AP, Evason, KJ, Brown, KK, Cox, AG, Ong, AJS, Bladen, CE, Tigani, TA, Karamalakis, AP, Evason, KJ, Brown, KK, and Cox, AG
- Abstract
The maintenance of redox and metabolic homeostasis is integral to embryonic development. Nuclear factor erythroid 2-related factor 2 (NRF2) is a stress-induced transcription factor that plays a central role in the regulation of redox balance and cellular metabolism. Under homeostatic conditions, NRF2 is repressed by Kelch-like ECH-associated protein 1 (KEAP1). Here, we demonstrate that Keap1 deficiency induces Nrf2 activation and postdevelopmental lethality. Loss of viability is preceded by severe liver abnormalities characterized by an accumulation of lysosomes. Mechanistically, we demonstrate that loss of Keap1 promotes aberrant activation of transcription factor EB (TFEB)/transcription factor binding to IGHM Enhancer 3 (TFE3)-dependent lysosomal biogenesis. Importantly, we find that NRF2-dependent regulation of lysosomal biogenesis is cell autonomous and evolutionarily conserved. These studies identify a role for the KEAP1-NRF2 pathway in the regulation of lysosomal biogenesis and suggest that maintenance of lysosomal homeostasis is required during embryonic development.
- Published
- 2023
3. Yap reprograms glutamine metabolism to increase nucleotide biosynthesis and enable liver growth
- Author
-
Cox, AG, Hwang, KL, Brown, KK, Evason, KJ, Beltz, S, Tsomides, A, O'Connor, K, Galli, GG, Yimlamai, D, Chhangawala, S, Yuan, M, Lien, EC, Wucherpfennig, J, Nissim, S, Minami, A, Cohen, DE, Camargo, FD, Asara, JM, Houvras, Y, Stainier, DYR, Goessling, W, Cox, AG, Hwang, KL, Brown, KK, Evason, KJ, Beltz, S, Tsomides, A, O'Connor, K, Galli, GG, Yimlamai, D, Chhangawala, S, Yuan, M, Lien, EC, Wucherpfennig, J, Nissim, S, Minami, A, Cohen, DE, Camargo, FD, Asara, JM, Houvras, Y, Stainier, DYR, and Goessling, W
- Abstract
The Hippo pathway is an important regulator of organ size and tumorigenesis. It is unclear, however, how Hippo signalling provides the cellular building blocks required for rapid growth. Here, we demonstrate that transgenic zebrafish expressing an activated form of the Hippo pathway effector Yap1 (also known as YAP) develop enlarged livers and are prone to liver tumour formation. Transcriptomic and metabolomic profiling identify that Yap1 reprograms glutamine metabolism. Yap1 directly enhances glutamine synthetase (glul) expression and activity, elevating steady-state levels of glutamine and enhancing the relative isotopic enrichment of nitrogen during de novo purine and pyrimidine biosynthesis. Genetic or pharmacological inhibition of GLUL diminishes the isotopic enrichment of nitrogen into nucleotides, suppressing hepatomegaly and the growth of liver cancer cells. Consequently, Yap-driven liver growth is susceptible to nucleotide inhibition. Together, our findings demonstrate that Yap1 integrates the anabolic demands of tissue growth during development and tumorigenesis by reprogramming nitrogen metabolism to stimulate nucleotide biosynthesis.
- Published
- 2016
4. Cardiolipin deficiency disrupts CoQ redox state and induces steatohepatitis.
- Author
-
Brothwell MJ, Cao 曹国燊 G, Maschek JA, Poss AM, Peterlin AD, Wang 汪立平 L, Baker TB, Shahtout JL, Siripoksup P, Pearce QJ, Johnson JM, Finger FM, Prola A, Pellizzari SA, Hale GL, Manuel AM, Watanabe 渡邉真也 S, Miranda ER, Affolter KE, Tippetts TS, Nikolova LS, Choi 崔蘭煕 RH, Decker ST, Patil M, Catrow JL, Holland WL, Nowinski SM, Lark DS, Fisher-Wellman KH, Mimche PN, Evason KJ, Cox JE, Summers SA, Gerhart-Hines Z, and Funai 船井勝彦 K
- Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a progressive disorder marked by lipid accumulation, leading to steatohepatitis (MASH). A key feature of the transition to MASH involves oxidative stress resulting from defects in mitochondrial oxidative phosphorylation (OXPHOS). Here, we show that pathological alterations in the lipid composition of the inner mitochondrial membrane (IMM) directly instigate electron transfer inefficiency to promote oxidative stress. Specifically, cardiolipin (CL) was downregulated across four mouse models of MASLD. Hepatocyte-specific CL synthase knockout (CLS-LKO) led to spontaneous MASH with elevated mitochondrial electron leak. Loss of CL interfered with the ability of coenzyme Q (CoQ) to transfer electrons, promoting leak primarily at sites II
F and IIIQ0 . Data from human liver biopsies revealed a highly robust correlation between mitochondrial CL and CoQ, co-downregulated with MASH. Thus, reduction in mitochondrial CL promotes oxidative stress and contributes to pathogenesis of MASH.- Published
- 2024
- Full Text
- View/download PDF
5. HAF prevents hepatocyte apoptosis and progression to MASH and HCC through transcriptional regulation of the NF-κB pathway.
- Author
-
Acuña-Pilarte K, Reichert EC, Green YS, Halberg LM, Golkowski M, Maguire KM, Mimche PN, Kamdem SD, Hu PA, Wright J, Ducker GS, Voth WP, O'Connell RM, McFarland SA, Egal ESA, Chaix A, Summers SA, Reelitz JW, Maschek JA, Cox JE, Evason KJ, and Koh MY
- Abstract
Background and Aims: HCC incidence is increasing worldwide due to the obesity epidemic, which drives metabolic dysfunction-associated steatohepatitis (MASH) that can lead to HCC. However, the molecular pathways driving MASH-HCC are poorly understood. We have previously reported that male mice with haploinsufficiency of hypoxia-associated factor (HAF) ( SART1+/ - ) spontaneously develop MASH-HCC. However, the cell type(s) responsible for HCC associated with HAF loss are unclear., Approach and Results: We generated SART1 -floxed mice, which were crossed with mice expressing Cre recombinase within hepatocytes (Alb-Cre; hepS -/- ) or myeloid cells (LysM-Cre, macS -/- ). HepS - / - mice (both male and female) developed HCC associated with profound inflammatory and lipid dysregulation, suggesting that HAF protects against HCC primarily within hepatocytes. HAF-deficient hepatocytes showed decreased P-p65 and P-p50 in many components of the NF-κB pathway, which was recapitulated using HAF small interfering RNA in vitro. HAF depletion also triggered apoptosis, suggesting that HAF protects against HCC by suppressing hepatocyte apoptosis. We show that HAF regulates NF-κB activity by regulating the transcription of TRADD and RIPK1 . Mice fed a high-fat diet showed marked suppression of HAF, P-p65, and TRADD within their livers after 26 weeks but showed profound upregulation of these proteins after 40 weeks, implicating deregulation of the HAF-NF-κB axis in the progression to MASH. In humans, HAF was significantly decreased in livers with simple steatosis but significantly increased in HCC compared with normal liver., Conclusions: HAF is a novel transcriptional regulator of the NF-κB pathway and is a key determinant of cell fate during progression to MASH and MASH-HCC., (Copyright © 2024 The Author(s). Published by Wolters Kluwer Health, Inc.)
- Published
- 2024
- Full Text
- View/download PDF
6. Phospholipid isotope tracing suggests β-catenin-driven suppression of phosphatidylcholine metabolism in hepatocellular carcinoma.
- Author
-
VanSant-Webb C, Low HK, Kuramoto J, Stanley CE, Qiang H, Su AY, Ross AN, Cooper CG, Cox JE, Summers SA, Evason KJ, and Ducker GS
- Subjects
- Humans, Animals, Lipid Metabolism genetics, Animals, Genetically Modified, Phospholipids metabolism, Cell Line, Tumor, Lipidomics methods, beta Catenin metabolism, beta Catenin genetics, Carcinoma, Hepatocellular metabolism, Carcinoma, Hepatocellular genetics, Carcinoma, Hepatocellular pathology, Zebrafish metabolism, Zebrafish genetics, Phosphatidylcholines metabolism, Liver Neoplasms metabolism, Liver Neoplasms genetics, Liver Neoplasms pathology
- Abstract
Activating mutations in the CTNNB1 gene encoding β-catenin are among the most frequently observed oncogenic alterations in hepatocellular carcinoma (HCC). Profound alterations in lipid metabolism, including increases in fatty acid oxidation and transformation of the phospholipidome, occur in HCC with CTNNB1 mutations, but it is unclear what mechanisms give rise to these changes. We employed untargeted lipidomics and targeted isotope tracing to measure phospholipid synthesis activity in an inducible human liver cell line expressing mutant β-catenin, as well as in transgenic zebrafish with activated β-catenin-driven HCC. In both models, activated β-catenin expression was associated with large changes in the lipidome including conserved increases in acylcarnitines and ceramides and decreases in triglycerides. Lipid isotope tracing analysis in human cells revealed a reduction in phosphatidylcholine (PC) production rates as assayed by choline incorporation. We developed lipid isotope tracing analysis for zebrafish tumors and observed reductions in phosphatidylcholine synthesis by both the CDP-choline and PEMT pathways. The observed changes in the β-catenin-driven HCC phospholipidome suggest that zebrafish can recapitulate conserved features of HCC lipid metabolism and may serve as a model for identifying future HCC-specific lipid metabolic targets., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
7. Hepatocyte vitamin D receptor functions as a nutrient sensor that regulates energy storage and tissue growth in zebrafish.
- Author
-
Freeburg SH, Shwartz A, Kemény LV, Smith CJ, Weeks O, Miller BM, PenkoffLidbeck N, Fisher DE, Evason KJ, and Goessling W
- Subjects
- Animals, Zebrafish Proteins metabolism, Zebrafish Proteins genetics, Liver metabolism, Nutrients metabolism, Signal Transduction, Lipid Metabolism, Homeostasis, Fatty Acids metabolism, Zebrafish metabolism, Receptors, Calcitriol metabolism, Energy Metabolism, Hepatocytes metabolism
- Abstract
Vitamin D receptor (VDR) has been implicated in fatty liver pathogenesis, but its role in the regulation of organismal energy usage remains unclear. Here, we illuminate the evolutionary function of VDR by demonstrating that zebrafish Vdr coordinates hepatic and organismal energy homeostasis through antagonistic regulation of nutrient storage and tissue growth. Hepatocyte-specific Vdr impairment increases hepatic lipid storage, partially through acsl4a induction, while simultaneously diminishing fatty acid oxidation and liver growth. Importantly, Vdr impairment exacerbates the starvation-induced hepatic storage of systemic fatty acids, indicating that loss of Vdr signaling elicits hepatocellular energy deficiency. Strikingly, hepatocyte Vdr impairment diminishes diet-induced systemic growth while increasing hepatic and visceral fat in adult fish, revealing that hepatic Vdr signaling is required for complete adaptation to food availability. These data establish hepatocyte Vdr as a regulator of organismal energy expenditure and define an evolutionary function for VDR as a transcriptional effector of environmental nutrient supply., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. TNF Promoter Hypomethylation Is Associated With Mucosal Inflammation in IBD and Anti-TNF Response.
- Author
-
Levic DS, Niedzwiecki D, Kandakatla A, Karlovich NS, Juneja A, Park J, Stolarchuk C, Adams S, Willer JR, Schaner MR, Lian G, Beasley C, Marjoram L, Flynn AD, Valentine JF, Onken JE, Sheikh SZ, Davis EE, Evason KJ, Garman KS, and Bagnat M
- Abstract
Background and Aims: Inflammatory bowel diseases (IBDs) are chronic inflammatory conditions influenced heavily by environmental factors. DNA methylation is a form of epigenetic regulation linking environmental stimuli to gene expression changes and inflammation. Here, we investigated how DNA methylation of the tumor necrosis factor (TNF) promoter differs between inflamed and uninflamed mucosa of IBD patients, including anti-TNF responders and nonresponders., Methods: We obtained mucosal biopsies from 200 participants (133 IBDs and 67 controls) and analyzed TNF promoter methylation using bisulfite sequencing, comparing inflamed with uninflamed segments, in addition to paired inflamed/uninflamed samples from individual patients. We conducted similar analyses on purified intestinal epithelial cells from bowel resections. We also compared TNF methylation levels of inflamed and uninflamed mucosa from a separate cohort of 15 anti-TNF responders and 17 nonresponders. Finally, we sequenced DNA methyltransferase genes to identify rare variants in IBD patients and functionally tested them using rescue experiments in a zebrafish genetic model of DNA methylation deficiency., Results: TNF promoter methylation levels were decreased in inflamed mucosa of IBD patients and correlated with disease severity. Isolated intestinal epithelial cells from inflamed tissue showed proportional decreases in TNF methylation. Anti-TNF nonresponders showed lower levels of TNF methylation than responders in uninflamed mucosa. Our sequencing analysis revealed 2 missense variants in DNA methyltransferase 1, 1 of which had reduced function in vivo., Conclusion: Our study reveals an association of TNF promoter hypomethylation with mucosal inflammation, suggesting that IBD patients may be particularly sensitive to inflammatory environmental insults affecting DNA methylation. Together, our analyses indicate that TNF promoter methylation analysis may aid in the characterization of IBD status and evaluation of anti-TNF therapy response., (© 2024 The Authors.)
- Published
- 2024
- Full Text
- View/download PDF
9. Long-term ketogenic diet causes hyperlipidemia, liver dysfunction, and glucose intolerance from impaired insulin trafficking and secretion in mice.
- Author
-
Gallop MR, Vieira RFL, Matsuzaki ET, Mower PD, Liou W, Smart FE, Roberts S, Evason KJ, Holland WL, and Chaix A
- Abstract
A ketogenic diet (KD) is a very low-carbohydrate, very high-fat diet proposed to treat obesity and type 2 diabetes. While KD grows in popularity, its effects on metabolic health are understudied. Here we show that, in male and female mice, while KD protects against weight gain and induces weight loss, over long-term, mice develop hyperlipidemia, hepatic steatosis, and severe glucose intolerance. Unlike high fat diet-fed mice, KD mice are not insulin resistant and have low levels of insulin. Hyperglycemic clamp and ex vivo GSIS revealed cell-autonomous and whole-body impairments in insulin secretion. Major ER/Golgi stress and disrupted ER-Golgi protein trafficking was indicated by transcriptomic profiling of KD islets and confirmed by electron micrographs showing a dilated Golgi network likely responsible for impaired insulin granule trafficking and secretion. Overall, our results suggest long-term KD leads to multiple aberrations of metabolic parameters that caution its systematic use as a health promoting dietary intervention.
- Published
- 2024
- Full Text
- View/download PDF
10. Oct4 redox sensitivity potentiates reprogramming and differentiation.
- Author
-
Shen Z, Wu Y, Manna A, Yi C, Cairns BR, Evason KJ, Chandrasekharan MB, and Tantin D
- Subjects
- Animals, Mice, Embryonic Stem Cells cytology, Embryonic Stem Cells metabolism, Tretinoin pharmacology, Tretinoin metabolism, Gene Expression Regulation, Developmental genetics, Humans, Octamer Transcription Factor-3 metabolism, Octamer Transcription Factor-3 genetics, Oxidation-Reduction, Cell Differentiation genetics, Cellular Reprogramming genetics
- Abstract
The transcription factor Oct4/Pou5f1 is a component of the regulatory circuitry governing pluripotency and is widely used to induce pluripotency from somatic cells. Here we used domain swapping and mutagenesis to study Oct4's reprogramming ability, identifying a redox-sensitive DNA binding domain, cysteine residue (Cys48), as a key determinant of reprogramming and differentiation. Oct4 Cys48 sensitizes the protein to oxidative inhibition of DNA binding activity and promotes oxidation-mediated protein ubiquitylation. Pou5f1
C48S point mutation has little effect on undifferentiated embryonic stem cells (ESCs) but upon retinoic acid (RA) treatment causes retention of Oct4 expression, deregulated gene expression, and aberrant differentiation. Pou5f1C48S ESCs also form less differentiated teratomas and contribute poorly to adult somatic tissues. Finally, we describe Pou5f1C48S ( Janky ) mice, which in the homozygous condition are severely developmentally restricted after E4.5. Rare animals bypassing this restriction appear normal at birth but are sterile. Collectively, these findings uncover a novel Oct4 redox mechanism involved in both entry into and exit from pluripotency., (© 2024 Shen et al.; Published by Cold Spring Harbor Laboratory Press.)- Published
- 2024
- Full Text
- View/download PDF
11. HAF Prevents Hepatocyte Apoptosis and Hepatocellular Carcinoma through Transcriptional Regulation of the NF-κB pathway.
- Author
-
Pilarte KA, Reichert EC, Green YS, Halberg LM, McFarland SA, Mimche PN, Golkowski M, Kamdem SD, Maguire KM, Summers SA, Maschek JA, Reelitz JW, Cox JE, Evason KJ, and Koh MY
- Abstract
Background: Hepatocellular carcinoma (HCC) incidence is increasing worldwide due to the obesity epidemic, which drives metabolic dysfunction-associated steatohepatitis (MASH) that can lead to HCC. However, the molecular pathways that lead to MASH-HCC are poorly understood. We have previously reported that male mice with global haploinsufficiency of hypoxia-associated factor, HAF ( SART1
+/ - ) spontaneously develop MASH/HCC. However, the cell type(s) responsible for HCC associated with HAF loss are unclear., Results: SART1 -floxed mice were crossed with mice expressing Cre-recombinase within hepatocytes (Alb-Cre; hepS-/- ) or macrophages (LysM-Cre, macS-/- ). Only hepS-/- mice (both male and female) developed HCC suggesting that HAF protects against HCC primarily within hepatocytes. HAF-deficient macrophages showed decreased P-p65 and P-p50 and in many major components of the NF-κB pathway, which was recapitulated using HAF siRNA in vitro . HAF depletion increased apoptosis both in vitro and in vivo , suggesting that HAF mediates a tumor suppressor role by suppressing hepatocyte apoptosis. We show that HAF regulates NF-κB activity by controlling transcription of TRADD and RIPK1 . Mice fed a high-fat diet (HFD) showed marked suppression of HAF, P-p65 and TRADD within their livers after 26 weeks, but manifest profound upregulation of HAF, P-65 and TRADD within their livers after 40 weeks of HFD, implicating deregulation of the HAF-NF-κB axis in the progression to MASH. In humans, HAF was significantly decreased in livers with simple steatosis but significantly increased in HCC compared to normal liver., Conclusions: HAF is novel transcriptional regulator of the NF-κB pathway that protects against hepatocyte apoptosis and is a key determinant of cell fate during progression to MASH and MASH-HCC.- Published
- 2024
- Full Text
- View/download PDF
12. Phospholipid isotope tracing reveals β-catenin-driven suppression of phosphatidylcholine metabolism in hepatocellular carcinoma.
- Author
-
VanSant-Webb C, Low HK, Kuramoto J, Stanley CE, Qiang H, Su A, Ross AN, Cooper CG, Cox JE, Summers SA, Evason KJ, and Ducker GS
- Abstract
Background and Aims: Activating mutations in the CTNNB1 gene encoding β-catenin are among the most frequently observed oncogenic alterations in hepatocellular carcinoma (HCC). HCC with CTNNB1 mutations show profound alterations in lipid metabolism including increases in fatty acid oxidation and transformation of the phospholipidome, but it is unclear how these changes arise and whether they contribute to the oncogenic program in HCC., Methods: We employed untargeted lipidomics and targeted isotope tracing to quantify phospholipid production fluxes in an inducible human liver cell line expressing mutant β-catenin, as well as in transgenic zebrafish with activated β-catenin-driven HCC., Results: In both models, activated β-catenin expression was associated with large changes in the lipidome including conserved increases in acylcarnitines and ceramides and decreases in triglycerides. Lipid flux analysis in human cells revealed a large reduction in phosphatidylcholine (PC) production rates as assayed by choline tracer incorporation. We developed isotope tracing lipid flux analysis for zebrafish and observed similar reductions in phosphatidylcholine synthesis flux accomplished by sex-specific mechanisms., Conclusions: The integration of isotope tracing with lipid abundances highlights specific lipid class transformations downstream of β-catenin signaling in HCC and suggests future HCC-specific lipid metabolic targets.
- Published
- 2023
- Full Text
- View/download PDF
13. Three pathologic criteria for reproducible diagnosis of colonic sessile serrated lesion versus hyperplastic polyp.
- Author
-
Boylan KE, Kanth P, Delker D, Hazel MW, Boucher KM, Affolter K, Clayton F, Evason KJ, Jedrzkiewicz J, Pletneva M, Samowitz W, Swanson E, and Bronner MP
- Subjects
- Humans, Biopsy, Colonic Polyps pathology, Adenoma pathology, Colonic Neoplasms genetics, Colorectal Neoplasms pathology
- Abstract
Colonic SSLs are thought to predispose to ∼30% of colonic adenocarcinomas. This increased risk, compared to benign HPs, makes their distinction vitally important. However, no gold standard exists to differentiate them, and wide observer variability is reported. To better distinguish these polyps, we investigated 94 serrated polyps (53 SSLs and 41 HPs) using an easy-to-apply pathologic scoring system that combines, for the first time, three established distinguishing features: polyp morphology, location, and size. As an additional novel approach, polyp size was assessed by serrated biopsy number compared to endoscopic size. RNA expression profiling served as an additional biomarker. The considerable morphologic overlap across serrated polyps was quantitated for the first time. Interobserver variability was assessed by 8 expert gastrointestinal pathologists. By ROC analysis, polyp size by biopsy number performed best, followed by polyp location and morphology (areas under the curves [AUCs] = 85.9%, 81.2%, and 65.9%, respectively). Optimal discrimination combined all 3 features (AUC = 92.9%). For polyp size, the biopsy number proved superior to endoscopic size (AUC = 85.9% versus 55.2%, P = .001). Interobserver variability analysis yielded the highest reported Fleiss and Kappa statistics (0.879) and percent agreement (96.8%), showing great promise toward improved diagnosis. The proposed 3-criteria pathologic system, combining size by biopsy number, location, and morphology, yields an improved, easy-to-use, and highly reproducible diagnostic approach for differentiating SSLs and HPs., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
14. The KEAP1-NRF2 pathway regulates TFEB/TFE3-dependent lysosomal biogenesis.
- Author
-
Ong AJS, Bladen CE, Tigani TA, Karamalakis AP, Evason KJ, Brown KK, and Cox AG
- Subjects
- Gene Expression Regulation, Kelch-Like ECH-Associated Protein 1 genetics, Kelch-Like ECH-Associated Protein 1 metabolism, Lysosomes metabolism, Animals, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors metabolism, NF-E2-Related Factor 2 metabolism
- Abstract
The maintenance of redox and metabolic homeostasis is integral to embryonic development. Nuclear factor erythroid 2-related factor 2 (NRF2) is a stress-induced transcription factor that plays a central role in the regulation of redox balance and cellular metabolism. Under homeostatic conditions, NRF2 is repressed by Kelch-like ECH-associated protein 1 (KEAP1). Here, we demonstrate that Keap1 deficiency induces Nrf2 activation and postdevelopmental lethality. Loss of viability is preceded by severe liver abnormalities characterized by an accumulation of lysosomes. Mechanistically, we demonstrate that loss of Keap1 promotes aberrant activation of transcription factor EB (TFEB)/transcription factor binding to IGHM Enhancer 3 (TFE3)-dependent lysosomal biogenesis. Importantly, we find that NRF2-dependent regulation of lysosomal biogenesis is cell autonomous and evolutionarily conserved. These studies identify a role for the KEAP1-NRF2 pathway in the regulation of lysosomal biogenesis and suggest that maintenance of lysosomal homeostasis is required during embryonic development.
- Published
- 2023
- Full Text
- View/download PDF
15. Histologic features of allograft livers in patients treated for rejection before biopsy.
- Author
-
Leonard NB, Hale GL, Boylan KE, Ou Z, Zhang C, Kim R, Chandna S, Dong ZM, and Evason KJ
- Subjects
- Humans, Retrospective Studies, Biopsy, Inflammation pathology, Allografts, Liver pathology, Graft Rejection pathology
- Abstract
Liver biopsy is essential for management in liver transplant patients with clinical features suspicious for acute cellular rejection (ACR). As more patients are transplanted for noninfectious indications, it has become increasingly common for them to receive treatment for presumed ACR before biopsy. The effect of pretreatment on the classic histologic triad of ACR's mixed portal inflammation, endothelialitis, and bile duct damage is not well described. Here we report a retrospective study of 70 liver transplant biopsies performed on 53 patients for suspected ACR between 2018 and 2021. Thirty-seven biopsies had a clinical diagnosis of ACR after biopsy. Pretreatment with steroids, antithymocyte globulin, or other increased immunosuppression was given before biopsy in 17 of 37 cases; 20 not-pretreated cases acted as controls. A representative hematoxylin and eosin-stained slide from each biopsy was reviewed independently in a blinded fashion by 3 hepatic pathologists, graded according to the Banff system, assigned a Rejection Activity Index (RAI), and assessed for other histologic features. We found that pretreated biopsies had significantly less portal inflammation (P < .001), less endothelialitis (P < .001), lower RAI (P < .001), and less prominent eosinophils (P = .048) compared to not-pretreated biopsies. There was no significant difference for the other examined variables, including bile duct inflammation/damage (P = .32). Our findings suggest that portal inflammation and endothelialitis become less prominent with pretreatment, whereas bile duct inflammation/damage may take longer to resolve. When evaluating biopsies for suspected ACR, the finding of bile duct inflammation/damage should raise the possibility of partially treated ACR, even in the absence of endothelialitis and portal inflammation., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
16. p53-Bad* Fusion Gene Therapy Induces Apoptosis In Vitro and Reduces Zebrafish Tumor Burden in Hepatocellular Carcinoma.
- Author
-
Bowman KER, Ahne L, O'Brien L, Vander Mause ER, Lu P, Wallis B, Evason KJ, and Lim CS
- Subjects
- Animals, Zebrafish genetics, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Tumor Burden, Apoptosis genetics, Apoptosis Regulatory Proteins genetics, Genetic Therapy, Cell Line, Tumor, Carcinoma, Hepatocellular genetics, Carcinoma, Hepatocellular therapy, Carcinoma, Hepatocellular metabolism, Liver Neoplasms genetics, Liver Neoplasms therapy, Liver Neoplasms metabolism
- Abstract
With few curative treatments and a global yearly death rate of over 800,000, hepatocellular carcinoma (HCC) desperately needs new therapies. Although wild-type p53 gene therapy has been shown to be safe in HCC patients, it has not shown enough efficacy to merit approval. This work aims to show how p53 can be re-engineered through fusion to the pro-apoptotic BH3 protein Bcl-2 antagonist of cell death (Bad) to improve anti-HCC activity and potentially lead to a novel HCC therapeutic, p53-Bad*. p53-Bad* is a fusion of p53 and Bad, with two mutations, S112A and S136A. We determined mitochondrial localization of p53-Bad* in liver cancer cell lines with varying p53 mutation statuses via fluorescence microscopy. We defined the apoptotic activity of p53-Bad* in four liver cancer cell lines using flow cytometry. To determine the effects of p53-Bad* in vivo, we generated and analyzed transgenic zebrafish expressing hepatocyte-specific p53-Bad*. p53-Bad* localized to the mitochondria regardless of the p53 mutation status and demonstrated superior apoptotic activity over WT p53 in early, middle, and late apoptosis assays. Tumor burden in zebrafish HCC was reduced by p53-Bad* as measured by the liver-to-body mass ratio and histopathology. p53-Bad* induced significant apoptosis in zebrafish HCC as measured by TUNEL staining but did not induce apoptosis in non-HCC fish. p53-Bad* can induce apoptosis in a panel of liver cancer cell lines with varying p53 mutation statuses and induce apoptosis/reduce HCC tumor burden in vivo in zebrafish. p53-Bad* warrants further investigation as a potential new HCC therapeutic.
- Published
- 2023
- Full Text
- View/download PDF
17. A missense mutation in the proprotein convertase gene furinb causes hepatic cystogenesis during liver development in zebrafish.
- Author
-
Ellis JL, Evason KJ, Zhang C, Fourman MN, Liu J, Ninov N, Delous M, Vanhollebeke B, Fiddes I, Otis JP, Houvras Y, Farber SA, Xu X, Lin X, Stainier DYR, and Yin C
- Subjects
- Animals, Proprotein Convertases genetics, Mutation, Missense genetics, Tunicamycin, Actins genetics, Disease Models, Animal, Liver pathology, TOR Serine-Threonine Kinases genetics, Mammals, Zebrafish genetics, Cysts genetics
- Abstract
Hepatic cysts are fluid-filled lesions in the liver that are estimated to occur in 5% of the population. They may cause hepatomegaly and abdominal pain. Progression to secondary fibrosis, cirrhosis, or cholangiocarcinoma can lead to morbidity and mortality. Previous studies of patients and rodent models have associated hepatic cyst formation with increased proliferation and fluid secretion in cholangiocytes, which are partially due to impaired primary cilia. Congenital hepatic cysts are thought to originate from faulty bile duct development, but the underlying mechanisms are not fully understood. In a forward genetic screen, we identified a zebrafish mutant that developed hepatic cysts during larval stages. The cyst formation was not due to changes in biliary cell proliferation, bile secretion, or impairment of primary cilia. Instead, time-lapse live imaging data showed that the mutant biliary cells failed to form interconnecting bile ducts because of defects in motility and protrusive activity. Accordingly, immunostaining revealed a disorganized actin and microtubule cytoskeleton in the mutant biliary cells. By whole-genome sequencing, we determined that the cystic phenotype in the mutant was caused by a missense mutation in the furinb gene, which encodes a proprotein convertase. The mutation altered Furinb localization and caused endoplasmic reticulum (ER) stress. The cystic phenotype could be suppressed by treatment with the ER stress inhibitor 4-phenylbutyric acid and exacerbated by treatment with the ER stress inducer tunicamycin. The mutant liver also exhibited increased mammalian target of rapamycin (mTOR) signaling. Treatment with mTOR inhibitors halted cyst formation at least partially through reducing ER stress. Conclusion: Our study has established a vertebrate model for studying hepatic cystogenesis and illustrated the contribution of ER stress in the disease pathogenesis., (© 2022 The Authors. Hepatology Communications published by Wiley Periodicals LLC on behalf of American Association for the Study of Liver Diseases.)
- Published
- 2022
- Full Text
- View/download PDF
18. Intratumor Heterogeneity in Hepatocellular Carcinoma: Challenges and Opportunities.
- Author
-
Kalasekar SM, VanSant-Webb CH, and Evason KJ
- Abstract
Hepatocellular carcinoma (HCC) represents a leading cause of cancer-related death, but it remains difficult to treat. Intratumor genetic and phenotypic heterogeneity are inherent properties of breast, skin, lung, prostate, and brain tumors, and intratumor heterogeneity (ITH) helps define prognosis and therapeutic response in these cancers. Several recent studies estimate that ITH is inherent to HCC and attribute the clinical intractability of HCC to this heterogeneity. In this review, we examine the evidence for genomic, phenotypic, and tumor microenvironment ITH in HCC, with a focus on two of the top molecular drivers of HCC: β-catenin (CTNNB1) and Telomerase reverse transcriptase (TERT). We discuss the influence of ITH on HCC diagnosis, prognosis, and therapy, while highlighting the gaps in knowledge and possible future directions.
- Published
- 2021
- Full Text
- View/download PDF
19. Exploring the Interplay of Telomerase Reverse Transcriptase and β-Catenin in Hepatocellular Carcinoma.
- Author
-
Kotiyal S and Evason KJ
- Abstract
Hepatocellular carcinoma (HCC) is one of the deadliest human cancers. Activating mutations in the telomerase reverse transcriptase ( TERT) promoter ( TERTp) and CTNNB1 gene encoding β-catenin are widespread in HCC (~50% and ~30%, respectively). TERTp mutations are predicted to increase TERT transcription and telomerase activity. This review focuses on exploring the role of TERT and β-catenin in HCC and the current findings regarding their interplay. TERT can have contradictory effects on tumorigenesis via both its canonical and non-canonical functions. As a critical regulator of proliferation and differentiation in progenitor and stem cells, activated β-catenin drives HCC; however, inhibiting endogenous β-catenin can also have pro-tumor effects. Clinical studies revealed a significant concordance between TERTp and CTNNB1 mutations in HCC. In stem cells, TERT acts as a co-factor in β-catenin transcriptional complexes driving the expression of WNT/β-catenin target genes, and β-catenin can bind to the TERTp to drive its transcription. A few studies have examined potential interactions between TERT and β-catenin in HCC in vivo, and their results suggest that the coexpression of these two genes promotes hepatocarcinogenesis. Further studies are required with vertebrate models to better understand how TERT and β-catenin influence hepatocarcinogenesis.
- Published
- 2021
- Full Text
- View/download PDF
20. Deleterious Variants in ABCC12 are Detected in Idiopathic Chronic Cholestasis and Cause Intrahepatic Bile Duct Loss in Model Organisms.
- Author
-
Pham DH, Kudira R, Xu L, Valencia CA, Ellis JL, Shi T, Evason KJ, Osuji I, Matuschek N, Pfuhler L, Mullen M, Mohanty SK, Husami A, Bull LN, Zhang K, Wali S, Yin C, and Miethke A
- Subjects
- ATP-Binding Cassette Transporters metabolism, Animals, Apoptosis, Bile Ducts, Intrahepatic metabolism, Case-Control Studies, Cholestasis, Intrahepatic metabolism, Chronic Disease, Female, Gene Editing, Genetic Association Studies, Genetic Predisposition to Disease, Humans, Infant, Mice, Mice, Inbred C57BL, Phenotype, Exome Sequencing, Zebrafish, Zebrafish Proteins metabolism, ATP-Binding Cassette Transporters genetics, Bile Ducts, Intrahepatic pathology, Cholestasis, Intrahepatic genetics, Cholestasis, Intrahepatic pathology, Mutation, Zebrafish Proteins genetics
- Abstract
Background & Aims: The etiology of cholestasis remains unknown in many children. We surveyed the genome of children with chronic cholestasis for variants in genes not previously associated with liver disease and validated their biological relevance in zebrafish and murine models., Method: Whole-exome (n = 4) and candidate gene sequencing (n = 89) was completed on 93 children with cholestasis and normal serum γ-glutamyl transferase (GGT) levels without pathogenic variants in genes known to cause low GGT cholestasis such as ABCB11 or ATP8B1. CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 genome editing was used to induce frameshift pathogenic variants in the candidate gene in zebrafish and mice., Results: In a 1-year-old female patient with normal GGT cholestasis and bile duct paucity, we identified a homozygous truncating pathogenic variant (c.198delA, p.Gly67Alafs∗6) in the ABCC12 gene (NM_033226). Five additional rare ABCC12 variants, including a pathogenic one, were detected in our cohort. ABCC12 encodes multidrug resistance-associated protein 9 (MRP9) that belongs to the adenosine 5'-triphosphate-binding cassette transporter C family with unknown function and no previous implication in liver disease. Immunohistochemistry and Western blotting revealed conserved MRP9 protein expression in the bile ducts in human, mouse, and zebrafish. Zebrafish abcc12-null mutants were prone to cholangiocyte apoptosis, which caused progressive bile duct loss during the juvenile stage. MRP9-deficient mice had fewer well-formed interlobular bile ducts and higher serum alkaline phosphatase levels compared with wild-type mice. They exhibited aggravated cholangiocyte apoptosis, hyperbilirubinemia, and liver fibrosis upon cholic acid challenge., Conclusions: Our work connects MRP9 with bile duct homeostasis and cholestatic liver disease for the first time. It identifies a potential therapeutic target to attenuate bile acid-induced cholangiocyte injury., (Copyright © 2021 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
21. Immune Checkpoint Inhibitors in Combinations for Hepatocellular Carcinoma.
- Author
-
Kalasekar SM, Garrido-Laguna I, and Evason KJ
- Subjects
- Antineoplastic Agents, Immunological pharmacology, Biomarkers, Pharmacological analysis, Clinical Trials as Topic, Drug Monitoring methods, Humans, Progression-Free Survival, Antineoplastic Combined Chemotherapy Protocols pharmacology, Carcinoma, Hepatocellular drug therapy, Carcinoma, Hepatocellular immunology, Carcinoma, Hepatocellular pathology, Immune Checkpoint Inhibitors pharmacology, Liver Neoplasms drug therapy, Liver Neoplasms immunology, Liver Neoplasms pathology, Receptors, Vascular Endothelial Growth Factor antagonists & inhibitors
- Published
- 2021
- Full Text
- View/download PDF
22. Regulation of Tumor Initiation by the Mitochondrial Pyruvate Carrier.
- Author
-
Bensard CL, Wisidagama DR, Olson KA, Berg JA, Krah NM, Schell JC, Nowinski SM, Fogarty S, Bott AJ, Wei P, Dove KK, Tanner JM, Panic V, Cluntun A, Lettlova S, Earl CS, Namnath DF, Vázquez-Arreguín K, Villanueva CJ, Tantin D, Murtaugh LC, Evason KJ, Ducker GS, Thummel CS, and Rutter J
- Subjects
- Animals, Cell Transformation, Neoplastic metabolism, Drosophila, Female, Male, Mice, Mice, Inbred C57BL, Adenoma metabolism, Carcinogenesis metabolism, Colorectal Neoplasms metabolism, Mitochondria metabolism, Mitochondrial Membrane Transport Proteins metabolism, Pyruvic Acid metabolism
- Abstract
Although metabolic adaptations have been demonstrated to be essential for tumor cell proliferation, the metabolic underpinnings of tumor initiation are poorly understood. We found that the earliest stages of colorectal cancer (CRC) initiation are marked by a glycolytic metabolic signature, including downregulation of the mitochondrial pyruvate carrier (MPC), which couples glycolysis and glucose oxidation through mitochondrial pyruvate import. Genetic studies in Drosophila suggest that this downregulation is required because hyperplasia caused by loss of the Apc or Notch tumor suppressors in intestinal stem cells can be completely blocked by MPC overexpression. Moreover, in two distinct CRC mouse models, loss of Mpc1 prior to a tumorigenic stimulus doubled the frequency of adenoma formation and produced higher grade tumors. MPC loss was associated with a glycolytic metabolic phenotype and increased expression of stem cell markers. These data suggest that changes in cellular pyruvate metabolism are necessary and sufficient to promote cancer initiation., Competing Interests: Declaration of Interests The University of Utah has filed a patent related to the mitochondrial pyruvate carrier, of which J.R. and C.S.T. are listed as co-inventors. All other authors declare no competing interests., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
23. Quantifying Liver Size in Larval Zebrafish Using Brightfield Microscopy.
- Author
-
Kotiyal S, Fulbright A, O'Brien LK, and Evason KJ
- Subjects
- Animals, Animals, Genetically Modified, Fixatives, Larva anatomy & histology, Liver diagnostic imaging, Microscopy methods, Organ Size, Zebrafish genetics, Liver anatomy & histology, Zebrafish anatomy & histology
- Abstract
In several transgenic zebrafish models of hepatocellular carcinoma (HCC), hepatomegaly can be observed during early larval stages. Quantifying larval liver size in zebrafish HCC models provides a means to rapidly assess the effects of drugs and other manipulations on an oncogene-related phenotype. Here we show how to fix zebrafish larvae, dissect the tissues surrounding the liver, photograph livers using bright-field microscopy, measure liver area, and analyze results. This protocol enables rapid, precise quantification of liver size. As this method involves measuring liver area, it may underestimate differences in liver volume, and complementary methodologies are required to differentiate between changes in cell size and changes in cell number. The dissection technique described herein is an excellent tool to visualize the liver, gut, and pancreas in their natural positions for myriad downstream applications including immunofluorescence staining and in situ hybridization. The described strategy for quantifying larval liver size is applicable to many aspects of liver development and regeneration.
- Published
- 2020
- Full Text
- View/download PDF
24. Heterogeneous beta-catenin activation is sufficient to cause hepatocellular carcinoma in zebrafish.
- Author
-
Kalasekar SM, Kotiyal S, Conley C, Phan C, Young A, and Evason KJ
- Abstract
Up to 41% of hepatocellular carcinomas (HCCs) result from activating mutations in the CTNNB1 gene encoding β-catenin. HCC-associated CTNNB1 mutations stabilize the β-catenin protein, leading to nuclear and/or cytoplasmic localization of β-catenin and downstream activation of Wnt target genes. In patient HCC samples, β-catenin nuclear and cytoplasmic localization are typically patchy, even among HCC with highly active CTNNB1 mutations. The functional and clinical relevance of this heterogeneity in β-catenin activation are not well understood. To define mechanisms of β-catenin-driven HCC initiation, we generated a Cre-lox system that enabled switching on activated β-catenin in (1) a small number of hepatocytes in early development; or (2) the majority of hepatocytes in later development or adulthood. We discovered that switching on activated β-catenin in a subset of larval hepatocytes was sufficient to drive HCC initiation. To determine the role of Wnt/β-catenin signaling heterogeneity later in hepatocarcinogenesis, we performed RNA-seq analysis of zebrafish β-catenin-driven HCC. At the single-cell level, 2.9% to 15.2% of hepatocytes from zebrafish β-catenin-driven HCC expressed two or more of the Wnt target genes axin2 , mtor , glula , myca and wif1 , indicating focal activation of Wnt signaling in established tumors. Thus, heterogeneous β-catenin activation drives HCC initiation and persists throughout hepatocarcinogenesis., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2019. Published by The Company of Biologists Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
25. Epithelial Inclusions in Gallbladder May Mimic Parasite Infection.
- Author
-
Evason KJ and Swanson EA
- Subjects
- Gallbladder, Humans, Prevalence, Gallbladder Diseases, Infections, Parasitic Diseases
- Published
- 2019
- Full Text
- View/download PDF
26. Endogenous development of Cystoisospora belli in intestinal and biliary epithelium of humans.
- Author
-
Dubey JP, Evason KJ, and Walther Z
- Subjects
- Adult, Biliary Tract parasitology, Biliary Tract pathology, Biopsy, Coccidiosis parasitology, Duodenum pathology, Humans, Life Cycle Stages, Male, Merozoites growth & development, Oocysts growth & development, Young Adult, Biliary Tract cytology, Duodenum cytology, Duodenum parasitology, Epithelial Cells parasitology, Isospora growth & development
- Abstract
Cystoisospora (Isospora) belli is a coccidian parasite of humans. It can cause serious digestive disorders involving infection of intestines, biliary tract and gallbladder, especially in those with depressed immunity. It has a direct fecal-oral transmission cycle. After ingestion of sporulated oocysts, the parasite multiplies asexually and sexually within host epithelial cells, resulting in unsporulated oocysts that are excreted in feces. The details of asexual and sexual stages are not known and certain inclusions in epithelial cells in biopsy samples have been erroneously identified recently as C. belli. Here, we provide details of developmental stages of C. belli in two patients, in duodenal biopsy of one and biliary epithelium of the other. Immature and mature asexual stages (schizonts/meronts) were seen in epithelial cells. The merozoites were seen singly, in pairs and in groups in single parasitophorous vacuole (pv) in host cytoplasm. Immature and mature meronts were seen together in the same pv; up to eight nuclei were seen in meronts that retained elongated crescent shape; round multinucleated schizonts, seen in other coccidians, were not found. Meronts were up to 25 µm long and contained up to ten merozoites that were 8-11 µm long. The merozoites and meronts contained PAS-positive granules. Microgamonts (male) contained up to 30 nuclei that were arranged at the periphery and had condensed chromatin; 1-3 PAS-positive, eosinophilic, residual bodies were left when microgametes were formed. The microgametes were 4 µm long and PAS-negative. All stages of macrogamonts, including oocysts were PAS-positive. The detailed description of the life cycle stages of C. belli reported here should facilitate in histopathologic diagnosis of this parasite.
- Published
- 2019
- Full Text
- View/download PDF
27. Estrogen Activation of G-Protein-Coupled Estrogen Receptor 1 Regulates Phosphoinositide 3-Kinase and mTOR Signaling to Promote Liver Growth in Zebrafish and Proliferation of Human Hepatocytes.
- Author
-
Chaturantabut S, Shwartz A, Evason KJ, Cox AG, Labella K, Schepers AG, Yang S, Acuña M, Houvras Y, Mancio-Silva L, Romano S, Gorelick DA, Cohen DE, Zon LI, Bhatia SN, North TE, and Goessling W
- Subjects
- 9,10-Dimethyl-1,2-benzanthracene, Animals, Carcinogenesis drug effects, Carcinoma, Hepatocellular pathology, Cell Proliferation drug effects, Female, Gene Expression drug effects, Hepatocytes, Humans, Liver metabolism, Liver Cirrhosis metabolism, Liver Neoplasms pathology, Liver Regeneration, Male, Organ Size drug effects, Phosphatidylinositol 3-Kinase metabolism, Receptors, G-Protein-Coupled genetics, Sex Factors, Signal Transduction, TOR Serine-Threonine Kinases genetics, TOR Serine-Threonine Kinases metabolism, Tumor Burden drug effects, Zebrafish, Zebrafish Proteins genetics, Carcinoma, Hepatocellular metabolism, Estradiol pharmacology, Estrogens pharmacology, Liver growth & development, Liver Neoplasms metabolism, Receptors, Estrogen metabolism, Receptors, G-Protein-Coupled metabolism, Zebrafish Proteins metabolism
- Abstract
Background & Aims: Patients with cirrhosis are at high risk for hepatocellular carcinoma (HCC) and often have increased serum levels of estrogen. It is not clear how estrogen promotes hepatic growth. We investigated the effects of estrogen on hepatocyte proliferation during zebrafish development, liver regeneration, and carcinogenesis. We also studied human hepatocytes and liver tissues., Methods: Zebrafish were exposed to selective modifiers of estrogen signaling at larval and adult stages. Liver growth was assessed by gene expression, fluorescent imaging, and histologic analyses. We monitored liver regeneration after hepatocyte ablation and HCC development after administration of chemical carcinogens (dimethylbenzanthrazene). Proliferation of human hepatocytes was measured in a coculture system. We measured levels of G-protein-coupled estrogen receptor (GPER1) in HCC and nontumor liver tissues from 68 patients by immunohistochemistry., Results: Exposure to 17β-estradiol (E2) increased proliferation of hepatocytes and liver volume and mass in larval and adult zebrafish. Chemical genetic and epistasis experiments showed that GPER1 mediates the effects of E2 via the phosphoinositide 3-kinase-protein kinase B-mechanistic target of rapamycin pathway: gper1-knockout and mtor-knockout zebrafish did not increase liver growth in response to E2. HCC samples from patients had increased levels of GPER1 compared with nontumor tissue samples; estrogen promoted proliferation of human primary hepatocytes. Estrogen accelerated hepatocarcinogenesis specifically in male zebrafish. Chemical inhibition or genetic loss of GPER1 significantly reduced tumor development in the zebrafish., Conclusions: In an analysis of zebrafish and human liver cells and tissues, we found GPER1 to be a hepatic estrogen sensor that regulates liver growth during development, regeneration, and tumorigenesis. Inhibitors of GPER1 might be developed for liver cancer prevention or treatment., Transcript Profiling: The accession number in the Gene Expression Omnibus is GSE92544., (Copyright © 2019 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
28. Anti-inflammatory microRNA-146a protects mice from diet-induced metabolic disease.
- Author
-
Runtsch MC, Nelson MC, Lee SH, Voth W, Alexander M, Hu R, Wallace J, Petersen C, Panic V, Villanueva CJ, Evason KJ, Bauer KM, Mosbruger T, Boudina S, Bronner M, Round JL, Drummond MJ, and O'Connell RM
- Subjects
- Animals, Blood Glucose metabolism, Diet, High-Fat adverse effects, Disease Models, Animal, Female, Gene Expression, Humans, Hyperglycemia genetics, Hyperglycemia metabolism, Hyperglycemia prevention & control, Inflammation genetics, Inflammation metabolism, Insulin blood, Intra-Abdominal Fat metabolism, Intra-Abdominal Fat pathology, Macrophages metabolism, Male, Metabolic Diseases genetics, Metabolic Diseases metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, MicroRNAs antagonists & inhibitors, NF-kappa B metabolism, Obesity genetics, Obesity metabolism, Obesity prevention & control, Proto-Oncogene Proteins c-akt genetics, Sirolimus pharmacology, TOR Serine-Threonine Kinases antagonists & inhibitors, TOR Serine-Threonine Kinases genetics, Weight Gain drug effects, Weight Gain genetics, Inflammation prevention & control, Metabolic Diseases prevention & control, MicroRNAs genetics, MicroRNAs metabolism
- Abstract
Identifying regulatory mechanisms that influence inflammation in metabolic tissues is critical for developing novel metabolic disease treatments. Here, we investigated the role of microRNA-146a (miR-146a) during diet-induced obesity in mice. miR-146a is reduced in obese and type 2 diabetic patients and our results reveal that miR-146a-/- mice fed a high-fat diet (HFD) have exaggerated weight gain, increased adiposity, hepatosteatosis, and dysregulated blood glucose levels compared to wild-type controls. Pro-inflammatory genes and NF-κB activation increase in miR-146a-/- mice, indicating a role for this miRNA in regulating inflammatory pathways. RNA-sequencing of adipose tissue macrophages demonstrated a role for miR-146a in regulating both inflammation and cellular metabolism, including the mTOR pathway, during obesity. Further, we demonstrate that miR-146a regulates inflammation, cellular respiration and glycolysis in macrophages through a mechanism involving its direct target Traf6. Finally, we found that administration of rapamycin, an inhibitor of mTOR, was able to rescue the obesity phenotype in miR-146a-/- mice. Altogether, our study provides evidence that miR-146a represses inflammation and diet-induced obesity and regulates metabolic processes at the cellular and organismal levels, demonstrating how the combination of diet and miRNA genetics influences obesity and diabetic phenotypes., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
29. Epithelial Inclusions in Gallbladder Specimens Mimic Parasite Infection: Histologic and Molecular Examination of Reported Cystoisospora belli Infection in Gallbladders of Immunocompetent Patients.
- Author
-
Swanson EA, March JK, Clayton F, Couturier MR, Arcega R, Smith R, and Evason KJ
- Subjects
- Adult, Aged, DNA, Protozoan genetics, Databases, Factual, Diagnosis, Differential, Epithelial Cells immunology, Epithelial Cells parasitology, Female, Gallbladder immunology, Gallbladder parasitology, Gallbladder Diseases immunology, Gallbladder Diseases pathology, Host-Pathogen Interactions, Humans, Inclusion Bodies immunology, Inclusion Bodies parasitology, Isospora genetics, Isospora immunology, Isosporiasis immunology, Isosporiasis pathology, Male, Middle Aged, Polymerase Chain Reaction, Predictive Value of Tests, Retrospective Studies, Staining and Labeling methods, Epithelial Cells pathology, Gallbladder pathology, Gallbladder Diseases parasitology, Immunocompetence, Inclusion Bodies pathology, Isospora isolation & purification, Isosporiasis parasitology
- Abstract
Recent publications have described epithelial cytoplasmic vacuoles and inclusions incidentally noted within gallbladder epithelium and concluded that they represent coccidian parasite infection, in particular, Cystoisospora belli. We identified 8 gallbladder specimens from our institution in the past 3 years in which this diagnosis was suggested or in which similar epithelial alterations were prominent. Molecular analysis was performed on the 8 gallbladder specimens and on 3 positive control specimens: small bowel biopsies from acquired immunodeficiency syndrome patients with diarrhea. Polymerase chain reaction using primers designed to amplify an internal transcribed spacer (ITS2) in the C. belli ribosomal gene cluster was performed on the DNA samples. All 8 gallbladder specimens were negative for amplification, while a product consistent with C. belli was amplified from all 3 positive controls. Histologically, the gallbladder cytoplasmic inclusions stained diffusely positive for Grocott-Gomori's methenamine silver and Periodic acid-Schiff with diastase. In contrast, sections from a positive control small bowel biopsy demonstrated organisms that were negative for Grocott-Gomori's methenamine silver and showed a distinct capsular and punctate internal staining on Periodic acid-Schiff with diastase in various parasite forms. Together, the lack of molecular evidence of C. belli and the distinct morphologic and special staining patterns in these gallbladders compared with positive control small bowel suggest that these epithelial changes do not represent true C. belli infection. Our results suggest that gallbladders of immunocompetent patients may occasionally show epithelial changes that can morphologically mimic C. belli infection. Pathologists should be aware of this histologic variant to minimize unnecessary treatment, testing, and patient anxiety.
- Published
- 2018
- Full Text
- View/download PDF
30. MYC-driven inhibition of the glutamate-cysteine ligase promotes glutathione depletion in liver cancer.
- Author
-
Anderton B, Camarda R, Balakrishnan S, Balakrishnan A, Kohnz RA, Lim L, Evason KJ, Momcilovic O, Kruttwig K, Huang Q, Xu G, Nomura DK, and Goga A
- Subjects
- Animals, Cell Line, Tumor, Cluster Analysis, Disease Models, Animal, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Glutamate-Cysteine Ligase genetics, Glutamate-Cysteine Ligase metabolism, Glutamine metabolism, Humans, Liver Neoplasms genetics, Metabolic Networks and Pathways genetics, Metabolome, Metabolomics methods, Mice, Mice, Transgenic, MicroRNAs genetics, Oxidative Stress, Proto-Oncogene Proteins c-myc genetics, RNA Interference, Glutamate-Cysteine Ligase antagonists & inhibitors, Glutathione metabolism, Liver Neoplasms metabolism, Proto-Oncogene Proteins c-myc metabolism
- Abstract
How MYC reprograms metabolism in primary tumors remains poorly understood. Using integrated gene expression and metabolite profiling, we identify six pathways that are coordinately deregulated in primary MYC-driven liver tumors: glutathione metabolism; glycine, serine, and threonine metabolism; aminoacyl-tRNA biosynthesis; cysteine and methionine metabolism; ABC transporters; and mineral absorption. We then focus our attention on glutathione (GSH) and glutathione disulfide (GSSG), as they are markedly decreased in MYC-driven tumors. We find that fewer glutamine-derived carbons are incorporated into GSH in tumor tissue relative to non-tumor tissue. Expression of GCLC, the rate-limiting enzyme of GSH synthesis, is attenuated by the MYC-induced microRNA miR-18a. Inhibition of miR-18a in vivo leads to increased GCLC protein expression and GSH abundance in tumor tissue. Finally, MYC-driven liver tumors exhibit increased sensitivity to acute oxidative stress. In summary, MYC-dependent attenuation of GCLC by miR-18a contributes to GSH depletion in vivo , and low GSH corresponds with increased sensitivity to oxidative stress in tumors. Our results identify new metabolic pathways deregulated in primary MYC tumors and implicate a role for MYC in regulating a major antioxidant pathway downstream of glutamine., (© 2017 The Authors.)
- Published
- 2017
- Full Text
- View/download PDF
31. Selenoprotein H is an essential regulator of redox homeostasis that cooperates with p53 in development and tumorigenesis.
- Author
-
Cox AG, Tsomides A, Kim AJ, Saunders D, Hwang KL, Evason KJ, Heidel J, Brown KK, Yuan M, Lien EC, Lee BC, Nissim S, Dickinson B, Chhangawala S, Chang CJ, Asara JM, Houvras Y, Gladyshev VN, and Goessling W
- Subjects
- Animals, DNA Damage genetics, DNA-Binding Proteins metabolism, Female, Gastrointestinal Neoplasms pathology, Gene Expression Regulation, Neoplastic, Homeostasis, Humans, Male, Oxidation-Reduction, Oxidative Stress genetics, Selenium metabolism, Selenoproteins metabolism, Transcriptome genetics, Zebrafish genetics, Carcinogenesis genetics, DNA-Binding Proteins genetics, Gastrointestinal Neoplasms genetics, Selenoproteins genetics, Tumor Suppressor Protein p53 genetics
- Abstract
Selenium, an essential micronutrient known for its cancer prevention properties, is incorporated into a class of selenocysteine-containing proteins (selenoproteins). Selenoprotein H (SepH) is a recently identified nucleolar oxidoreductase whose function is not well understood. Here we report that seph is an essential gene regulating organ development in zebrafish. Metabolite profiling by targeted LC-MS/MS demonstrated that SepH deficiency impairs redox balance by reducing the levels of ascorbate and methionine, while increasing methionine sulfoxide. Transcriptome analysis revealed that SepH deficiency induces an inflammatory response and activates the p53 pathway. Consequently, loss of seph renders larvae susceptible to oxidative stress and DNA damage. Finally, we demonstrate that seph interacts with p53 deficiency in adulthood to accelerate gastrointestinal tumor development. Overall, our findings establish that seph regulates redox homeostasis and suppresses DNA damage. We hypothesize that SepH deficiency may contribute to the increased cancer risk observed in cohorts with low selenium levels., Competing Interests: The authors declare no conflict of interest.
- Published
- 2016
- Full Text
- View/download PDF
32. Monocytes promote liver carcinogenesis in an oncogene-specific manner.
- Author
-
Juric V, Ruffell B, Evason KJ, Hu J, Che L, Wang L, Chen X, and Bishop JM
- Subjects
- Animals, Antigens, Ly analysis, Female, Genes, ras, Mice, Neutrophil Infiltration, Proto-Oncogene Proteins c-akt genetics, Receptors, Chemokine analysis, Genes, myc physiology, Liver Neoplasms, Experimental etiology, Monocytes physiology
- Abstract
Background & Aims: The leukocyte composition of tumors is heterogeneous, as is the involvement of each leukocyte subset in promoting or restraining tumorigenesis. This heterogeneity reflects the tissue of origin, tumor stage, and the functional state of leukocyte activation, but its biological roots remain poorly understood. Since tumorigenesis is driven by various genetic events, we assessed the role of driver genes in shaping the profiles and the roles of leukocytes in tumorigenesis., Methods: Mouse liver tumors were induced by hepatic overexpression of either MYC or the combination of myristoylated AKT and NRAS(V12) oncogenes via hydrodynamic transfection. A comparative, flow cytometry- and histology-based immunophenotyping of liver-infiltrating leukocytes was performed at various stages of liver tumorigenesis. The roles of the most abundant leukocyte subsets in tumorigenesis were addressed by immunodepletion. The contribution of liver injury was assessed by comparing the injury-inducing hydrodynamic transfection model to a model in which MYC is an inducible transgene., Results: Myristoylated AKT and NRAS(V12) promoted a marked recruitment of CD11b(+)Ly6G(hi)Ly6C(int) neutrophils and CD11b(+)Ly6G(-)Ly6C(hi) monocytes to the liver, but their immunodepletion did not alter tumorigenesis. In contrast, despite minimal invasion by monocytes/neutrophils during MYC-driven tumorigenesis, immunodepletion of these cells reduced MYC tumor burden and extended survival. MYC-driven tumor initiation was augmented specifically by Ly6C+ monocytes and their ability to promote liver injury., Conclusions: Our results demonstrate that leukocyte profiles do not necessarily predict their involvement in tumorigenesis, the functional role of leukocytes can be shaped by oncogenes, and that monocyte-dependent tissue injury selectively cooperates with MYC during tumorigenesis., (Copyright © 2015 European Association for the Study of the Liver. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
33. Identification of Chemical Inhibitors of β-Catenin-Driven Liver Tumorigenesis in Zebrafish.
- Author
-
Evason KJ, Francisco MT, Juric V, Balakrishnan S, Lopez Pazmino Mdel P, Gordan JD, Kakar S, Spitsbergen J, Goga A, and Stainier DY
- Subjects
- Animals, Animals, Genetically Modified, Carcinoma, Hepatocellular mortality, Carcinoma, Hepatocellular pathology, Cell Line, Tumor, Cell Transformation, Neoplastic drug effects, Disease Models, Animal, Gene Expression Regulation, Neoplastic, Hepatocytes metabolism, Hepatocytes pathology, Humans, JNK Mitogen-Activated Protein Kinases antagonists & inhibitors, JNK Mitogen-Activated Protein Kinases metabolism, Liver pathology, Liver Neoplasms mortality, Liver Neoplasms pathology, MAP Kinase Signaling System drug effects, Mesothelin, Mice, Selective Serotonin Reuptake Inhibitors therapeutic use, Xenopus laevis, Zebrafish, beta Catenin genetics, Amitriptyline therapeutic use, Antidepressive Agents, Tricyclic therapeutic use, Carcinoma, Hepatocellular drug therapy, Liver Neoplasms drug therapy, beta Catenin metabolism
- Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal human cancers. The search for targeted treatments has been hampered by the lack of relevant animal models for the genetically diverse subsets of HCC, including the 20-40% of HCCs that are defined by activating mutations in the gene encoding β-catenin. To address this chemotherapeutic challenge, we created and characterized transgenic zebrafish expressing hepatocyte-specific activated β-catenin. By 2 months post fertilization (mpf), 33% of transgenic zebrafish developed HCC in their livers, and 78% and 80% of transgenic zebrafish showed HCC at 6 and 12 mpf, respectively. As expected for a malignant process, transgenic zebrafish showed significantly decreased mean adult survival compared to non-transgenic control siblings. Using this novel transgenic model, we screened for druggable pathways that mediate β-catenin-induced liver growth and identified two c-Jun N-terminal kinase (JNK) inhibitors and two antidepressants (one tricyclic antidepressant, amitriptyline, and one selective serotonin reuptake inhibitor) that suppressed this phenotype. We further found that activated β-catenin was associated with JNK pathway hyperactivation in zebrafish and in human HCC. In zebrafish larvae, JNK inhibition decreased liver size specifically in the presence of activated β-catenin. The β-catenin-specific growth-inhibitory effect of targeting JNK was conserved in human liver cancer cells. Our other class of hits, antidepressants, has been used in patient treatment for decades, raising the exciting possibility that these drugs could potentially be repurposed for cancer treatment. In support of this proposal, we found that amitriptyline decreased tumor burden in a mouse HCC model. Our studies implicate JNK inhibitors and antidepressants as potential therapeutics for β-catenin-induced liver tumors.
- Published
- 2015
- Full Text
- View/download PDF
34. CDK1 inhibition targets the p53-NOXA-MCL1 axis, selectively kills embryonic stem cells, and prevents teratoma formation.
- Author
-
Huskey NE, Guo T, Evason KJ, Momcilovic O, Pardo D, Creasman KJ, Judson RL, Blelloch R, Oakes SA, Hebrok M, and Goga A
- Subjects
- Animals, Apoptosis genetics, CDC2 Protein Kinase genetics, Cell Differentiation, Cell Line, Cyclin A genetics, Cyclin B1 genetics, Cyclin B2 genetics, DNA Damage drug effects, Drug Resistance drug effects, Drug Resistance genetics, Embryonic Stem Cells cytology, Embryonic Stem Cells drug effects, Embryonic Stem Cells pathology, Gene Expression Regulation, Gene Knockout Techniques, Humans, Mice, Myeloid Cell Leukemia Sequence 1 Protein genetics, Octamer Transcription Factor-3 genetics, Octamer Transcription Factor-3 metabolism, Protein Kinase Inhibitors pharmacology, Proto-Oncogene Proteins c-bcl-2 genetics, RNA Interference, Teratoma pathology, Tumor Suppressor Protein p53 genetics, CDC2 Protein Kinase antagonists & inhibitors, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Embryonic Stem Cells metabolism, Myeloid Cell Leukemia Sequence 1 Protein metabolism, Proto-Oncogene Proteins c-bcl-2 metabolism, Signal Transduction drug effects, Teratoma etiology, Tumor Suppressor Protein p53 metabolism
- Abstract
Embryonic stem cells (ESCs) have adopted an accelerated cell-cycle program with shortened gap phases and precocious expression of cell-cycle regulatory proteins, including cyclins and cyclin-dependent kinases (CDKs). We examined the effect of CDK inhibition on the pathways regulating proliferation and survival of ESCs. We found that inhibiting cyclin-dependent kinase 1 (CDK1) leads to activation of the DNA damage response, nuclear p53 stabilization, activation of a subset of p53 target genes including NOXA, and negative regulation of the anti-apoptotic protein MCL1 in human and mouse ESCs, but not differentiated cells. We demonstrate that MCL1 is highly expressed in ESCs and loss of MCL1 leads to ESC death. Finally, we show that clinically relevant CDK1 inhibitors prevent formation of ESC-derived tumors and induce necrosis in established ESC-derived tumors. Our data demonstrate that ES cells are uniquely sensitive to CDK1 inhibition via a p53/NOXA/MCL1 pathway., (Copyright © 2015 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
35. Well-differentiated hepatocellular neoplasm of uncertain malignant potential: proposal for a new diagnostic category--reply.
- Author
-
Kakar S, Evason KJ, and Ferrell LD
- Subjects
- Female, Humans, Male, Adenoma, Liver Cell genetics, Carcinoma, Hepatocellular genetics, Liver Neoplasms genetics, beta Catenin genetics
- Published
- 2014
- Full Text
- View/download PDF
36. The perivascular niche regulates breast tumour dormancy.
- Author
-
Ghajar CM, Peinado H, Mori H, Matei IR, Evason KJ, Brazier H, Almeida D, Koller A, Hajjar KA, Stainier DY, Chen EI, Lyden D, and Bissell MJ
- Subjects
- Animals, Bone Marrow Neoplasms blood supply, Bone Marrow Neoplasms metabolism, Brain Neoplasms blood supply, Brain Neoplasms metabolism, Breast Neoplasms blood supply, Breast Neoplasms metabolism, Cell Adhesion Molecules metabolism, Endothelium, Vascular metabolism, Female, Fluorescent Antibody Technique, Humans, Lung Neoplasms blood supply, Lung Neoplasms metabolism, Mice, Neoplasm, Residual blood supply, Neoplasm, Residual metabolism, Pericytes metabolism, Stem Cell Niche physiology, Thrombospondin 1 metabolism, Transforming Growth Factor beta metabolism, Tumor Cells, Cultured, Tumor Microenvironment, Zebrafish growth & development, Zebrafish metabolism, Bone Marrow Neoplasms secondary, Brain Neoplasms secondary, Breast Neoplasms pathology, Endothelium, Vascular pathology, Lung Neoplasms secondary, Neoplasm, Residual pathology, Neovascularization, Pathologic, Pericytes pathology
- Abstract
In a significant fraction of breast cancer patients, distant metastases emerge after years or even decades of latency. How disseminated tumour cells (DTCs) are kept dormant, and what wakes them up, are fundamental problems in tumour biology. To address these questions, we used metastasis assays in mice and showed that dormant DTCs reside on microvasculature of lung, bone marrow and brain. We then engineered organotypic microvascular niches to determine whether endothelial cells directly influence breast cancer cell (BCC) growth. These models demonstrated that endothelial-derived thrombospondin-1 induces sustained BCC quiescence. This suppressive cue was lost in sprouting neovasculature; time-lapse analysis showed that sprouting vessels not only permit, but accelerate BCC outgrowth. We confirmed this surprising result in dormancy models and in zebrafish, and identified active TGF-β1 and periostin as tumour-promoting factors derived from endothelial tip cells. Our work reveals that stable microvasculature constitutes a dormant niche, whereas sprouting neovasculature sparks micrometastatic outgrowth.
- Published
- 2013
- Full Text
- View/download PDF
37. Hepatic stellate cells in liver development, regeneration, and cancer.
- Author
-
Yin C, Evason KJ, Asahina K, and Stainier DY
- Subjects
- Animals, Carcinoma, Hepatocellular metabolism, Cell Differentiation, Endothelial Cells cytology, Fibrosis metabolism, Humans, Liver embryology, Liver pathology, Liver physiopathology, Liver Diseases metabolism, Liver Failure, Acute metabolism, Liver Neoplasms pathology, Mice, Microscopy, Phase-Contrast methods, Hepatic Stellate Cells cytology, Liver physiology, Liver Neoplasms metabolism, Liver Regeneration
- Abstract
Hepatic stellate cells are liver-specific mesenchymal cells that play vital roles in liver physiology and fibrogenesis. They are located in the space of Disse and maintain close interactions with sinusoidal endothelial cells and hepatic epithelial cells. It is becoming increasingly clear that hepatic stellate cells have a profound impact on the differentiation, proliferation, and morphogenesis of other hepatic cell types during liver development and regeneration. In this Review, we summarize and evaluate the recent advances in our understanding of the formation and characteristics of hepatic stellate cells, as well as their function in liver development, regeneration, and cancer. We also discuss how improved knowledge of these processes offers new perspectives for the treatment of patients with liver diseases.
- Published
- 2013
- Full Text
- View/download PDF
38. Atypical hepatocellular adenoma-like neoplasms with β-catenin activation show cytogenetic alterations similar to well-differentiated hepatocellular carcinomas.
- Author
-
Evason KJ, Grenert JP, Ferrell LD, and Kakar S
- Subjects
- Adenoma, Liver Cell pathology, Adult, Aged, Carcinoma, Hepatocellular pathology, Chromosome Aberrations, Comparative Genomic Hybridization, Female, Humans, In Situ Hybridization, Fluorescence, Liver Neoplasms pathology, Male, Middle Aged, Adenoma, Liver Cell genetics, Carcinoma, Hepatocellular genetics, Liver Neoplasms genetics, beta Catenin genetics
- Abstract
The distinction of hepatocellular adenoma from well-differentiated hepatocellular carcinoma (HCC) arising in noncirrhotic liver can be challenging, particularly when tumors histologically resembling hepatocellular adenoma occur in unusual clinical settings such as in a man or an older woman or show focal atypical morphologic features. In this study, we examine the morphologic, immunohistochemical, and cytogenetic features of hepatocellular adenoma-like neoplasms occurring in men, women 50 years or older or younger than 15 years, and/or those with focal atypia (small cell change, pseudogland formation, and/or nuclear atypia), designated atypical hepatocellular neoplasms, where the distinction of hepatocellular adenoma versus HCC could not be clearly established. Immunohistochemistry was performed for β-catenin, glutamine synthetase, and serum amyloid A in 31 hepatocellular adenomas, 20 well-differentiated HCCs, and 40 atypical hepatocellular neoplasms. Chromosomal gains/losses had previously been determined in 37 cases using comparative genomic hybridization or fluorescence in situ hybridization. β-Catenin activation was observed in 35% of atypical hepatocellular neoplasms compared with 10% of typical hepatocellular adenomas (P < .05) and 55% of well-differentiated HCCs (P = .14). Cytogenetic changes typically observed in HCC were present in all atypical hepatocellular neoplasms with β-catenin activation. β-Catenin activation in atypical hepatocellular neoplasms was also associated with atypical morphologic features. Follow-up data were limited, but adverse outcome was observed in 2 atypical hepatocellular neoplasms with β-catenin activation (1 recurrence, 1 metastasis); transition to areas of HCC was observed in 1 case. The similarity in morphologic and cytogenetic features of β-catenin-activated hepatocellular adenoma-like tumors and HCC suggests that the former tumors represent an extremely well-differentiated variant of HCC., Competing Interests: Disclosure/Conflict of interest: The authors have no conflicts of interest to declare., (Published by Elsevier Inc.)
- Published
- 2013
- Full Text
- View/download PDF
39. The basic helix-loop-helix transcription factor, heart and neural crest derivatives expressed transcript 2, marks hepatic stellate cells in zebrafish: analysis of stellate cell entry into the developing liver.
- Author
-
Yin C, Evason KJ, Maher JJ, and Stainier DY
- Subjects
- Animals, Antineoplastic Agents pharmacology, Basic Helix-Loop-Helix Transcription Factors genetics, Basic Helix-Loop-Helix Transcription Factors metabolism, Benzoates pharmacology, Cell Count, Cell Differentiation, Cell Proliferation drug effects, Ethanol pharmacology, Hepatic Stellate Cells drug effects, Hepatic Stellate Cells metabolism, Liver growth & development, Myocardium metabolism, Neural Crest metabolism, RNA, Messenger metabolism, Receptors, Retinoic Acid agonists, Retinoic Acid Receptor alpha, Signal Transduction, Tetrahydronaphthalenes pharmacology, Zebrafish, Zebrafish Proteins genetics, Cell Communication, Cell Movement, Endothelial Cells cytology, Hepatic Stellate Cells cytology, Liver cytology, Vascular Endothelial Growth Factor A antagonists & inhibitors
- Abstract
Unlabelled: Hepatic stellate cells (HSCs) are liver-specific mesenchymal cells that play vital roles in liver development and injury. Our knowledge of HSC biology is limited by the paucity of in vivo data. HSCs and sinusoidal endothelial cells (SECs) reside in close proximity, and interactions between these two cell types are potentially critical for their development and function. Here, we introduce a transgenic zebrafish line, Tg(hand2:EGFP), that labels HSCs. We find that zebrafish HSCs share many similarities with their mammalian counterparts, including morphology, location, lipid storage, gene-expression profile, and increased proliferation and matrix production, in response to an acute hepatic insult. Using the Tg(hand2:EGFP) line, we conducted time-course analyses during development to reveal that HSCs invade the liver after SECs do. However, HSCs still enter the liver in mutants that lack most endothelial cells, including SECs, indicating that SECs are not required for HSC differentiation or their entry into the liver. In the absence of SECs, HSCs become abnormally associated with hepatic biliary cells, suggesting that SECs influence HSC localization during liver development. We analyzed factors that regulate HSC development and show that inhibition of vascular endothelial growth factor signaling significantly reduces the number of HSCs that enter the liver. We also performed a pilot chemical screen and identified two compounds that affect HSC numbers during development., Conclusion: Our work provides the first comprehensive description of HSC development in zebrafish and reveals the requirement of SECs in HSC localization. The Tg(hand2:EGFP) line represents a unique tool for in vivo analysis and molecular dissection of HSC behavior., (Copyright © 2012 American Association for the Study of Liver Diseases.)
- 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.