1
|
Zhu A, Yan X, Chen M, Lin Y, Li L, Wang Y, Huang J, He J, Yang M, Hua W, Chen K, Qi J, Zhou Z. Sappanone A alleviates metabolic dysfunction-associated steatohepatitis by decreasing hepatocyte lipotoxicity via targeting Mup3 in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156341. [PMID: 39733550 DOI: 10.1016/j.phymed.2024.156341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/01/2024] [Accepted: 12/19/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND AND PURPOSE Metabolic dysfunction-associated steatohepatitis (MASH) is an inflammatory lipotoxic disorder marked by hepatic steatosis, hepatocyte damage, inflammation, and varying stages of fibrosis. Sappanone A (SA), a flavonoid, exhibits anti-inflammatory and hepatoprotection activities. Nevertheless, the effects of SA on MASH remain ambiguous. We evaluated the effects of SA on hepatocyte lipotoxicity, inflammation, and fibrosis conditions in MASH mice, as well as the underlying mechanisms. METHODS A conventional murine MASH model fed a methionine-choline-deficient (MCD) diet was utilized to assess the role of SA on MASH in vivo. Drug target prediction and liver transcriptomics were employed to elucidate the potential actions of SA. AML12 cells were applied to further explore the effects and mechanisms of SA in vitro. RESULTS The in silico prediction indicated that SA could modulate inflammation, insulin resistance, lipid metabolism, and collagen catabolic process. Treating with SA dose-dependently lessened the elevated levels of serum ALT and AST in mice with diet-triggered MASH, and high-dose SA treatment exhibited a similar effect to silymarin. Additionally, SA treatment significantly reduced lipid deposition, inflammation, and fibrosis subjected to metabolic stress in a dose-dependent manner. Besides, SA mitigated palmitate-triggered lipotoxicity in hepatocytes. Liver transcriptomics further confirmed the aforementioned findings. Of note, mRNA-sequencing analysis and molecular biology experiments demonstrated that SA statistically up-regulated the hepatic expression of major urinary protein 3 (Mup3), thereby facilitating lipid transportation and inhibiting lipotoxicity. Furthermore, Mup3 knockdown in hepatocytes significantly abolished the hepatoprotection provided by SA. CONCLUSION SA alleviates MASH by decreasing lipid accumulation and lipotoxicity in hepatocytes, at least partially by targeting Mup3, and subsequently blocks MASH process. Therefore, SA could be a promising hepatoprotective agent in the context of MASH.
Collapse
Affiliation(s)
- An Zhu
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, 1 Xue Fu North Road, Fuzhou 350122, China
| | - Xueqing Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fujian Medical University, No.1, Xuefu North Road, University Town, Fuzhou, Fujian 350122, China
| | - Mengting Chen
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, 1 Xue Fu North Road, Fuzhou 350122, China
| | - Yifan Lin
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, 1 Xue Fu North Road, Fuzhou 350122, China
| | - Lanqian Li
- Department of Pathology & Diagnosis Pathological Center, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Yufei Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fujian Medical University, No.1, Xuefu North Road, University Town, Fuzhou, Fujian 350122, China
| | - Jiabin Huang
- Department of Pathology & Diagnosis Pathological Center, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Jiale He
- Department of Pathology & Diagnosis Pathological Center, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Mengchen Yang
- Department of Pathology & Diagnosis Pathological Center, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Wenxi Hua
- Department of Pathology & Diagnosis Pathological Center, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| | - Kunqi Chen
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fujian Medical University, 1 Xue Fu North Road, Fuzhou 350122, China.
| | - Jing Qi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fujian Medical University, No.1, Xuefu North Road, University Town, Fuzhou, Fujian 350122, China.
| | - Zixiong Zhou
- Department of Pathology & Diagnosis Pathological Center, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China.
| |
Collapse
|
2
|
Creasy KT, Ren H, Jiang J, Peterson ML, Spear BT. Elongation of very long chain fatty acids-3 ( Elovl3) is activated by ZHX2 and is a regulator of cell cycle progression. Am J Physiol Gastrointest Liver Physiol 2023; 325:G582-G592. [PMID: 37847682 PMCID: PMC10894669 DOI: 10.1152/ajpgi.00235.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/11/2023] [Accepted: 10/11/2023] [Indexed: 10/19/2023]
Abstract
Zinc fingers and homeoboxes 2 (Zhx2) are transcriptional regulators of liver gene expression with key functions in embryonic development as well as tissue regeneration in response to damage and disease, presumably through its control of target genes. Previous microarray data suggested that elongation of very long chain fatty acids-3 (Elovl3), a member of the ELOVL family of enzymes that synthesize very long chain fatty acids (VLCFAs), is a putative Zhx2 target gene. VLCFAs are core component of ceramides and other bioactive sphingolipids that are often dysregulated in diseases and regulate key cellular processes including proliferation. Since several previously identified Zhx2 targets become dysregulated in liver damage, we investigated the relationship between Zhx2 and Elovl3 in liver development, damage, and regeneration. Here, using mouse and cell models, we demonstrate that Zhx2 positively regulates Elovl3 expression in the liver and that male-biased hepatic Elovl3 expression is established between 4 and 8 wk of age in mice. Elovl3 is dramatically repressed in mouse models of liver regeneration, and the reduced Elovl3 levels in the regenerating liver are associated with changes in hepatic VLCFAs. Human hepatoma cell lines with forced Elovl3 expression have lower rates of cell growth; analysis of synchronized cells indicates that this reduced proliferation correlates with cells stalling in S-phase and lower mRNA levels of cell cyclins. Taken together, these data indicate that Elovl3 expression helps regulate cellular proliferation during liver development and regeneration, possibly through control of VLCFAs.NEW & NOTEWORTHY Numerous targets of the transcription factor Zhx2 are dysregulated in liver disease. We show that the elongase Elovl3 is a novel Zhx2 target. Elovl3 and Zhx2 expression change during liver regeneration, which is associated with changes in very long chain fatty acids. Forced Elovl3 expression reduces cell growth and blocks cell cycle progression. This suggests that Elovl3 may account, at least in part, for the relationship between Zhx2 and proliferation during liver development and disease.
Collapse
Affiliation(s)
- Kate Townsend Creasy
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Hui Ren
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Jieyun Jiang
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Martha L Peterson
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, United States
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| | - Brett T Spear
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, United States
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, Kentucky, United States
| |
Collapse
|
3
|
Hou J, Dong C, Chen J, Chen H, Na R, Zhou B, Hou J, Jiang DK. An intronic genetic variant of ZHX2 predicts response to pegylated interferon α therapy in HBeAg-positive chronic hepatitis B patients. Antiviral Res 2023; 220:105741. [PMID: 39492517 DOI: 10.1016/j.antiviral.2023.105741] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/21/2023] [Accepted: 10/27/2023] [Indexed: 11/05/2024]
Abstract
ZHX2 plays a crucial role in host immunity and modulates hepatitis B virus (HBV) replication. However, its correlation with immunomodulator-related treatment, i.g., pegylated interferon α(PegIFNα), for HBV remains uncertain. To explore the link between single nucleotide polymorphisms (SNPs) in ZHX2 and response to PegIFNα therapy for chronic hepatitis B (CHB) patients, we conducted a retrospective study in 945 hepatitis B e antigen (HBeAg)-positive CHB patients with at least 48 weeks of PegIFNα treatment and 24 weeks of follow-up from two phase-IV, multicenter trials (Cohort 1, n = 238; Cohort 2, n = 707). Thirty-eight tag SNPs were selected across the whole ZHX2 gene region. A polygenic score (PGS) was constructed by integrating multiple SNPs. The associations of ZHX2 SNPs and PGS with treatment response were assessed in both cohorts and their combination. Among the 38 tag SNPs, ZHX2_rs17289471 (T>C) was significantly associated with combined response (CR, i.e., HBeAg seroconversion and HBV DNA level <3.3log10IU/mL) at week 72 in both cohorts. The CR rate at week 72 increased steadily from rs17289471 TT to CT and CC genotype carriers in both Cohort 1 (P = 0.002) and Cohort 2 (P = 0.025) as well as Cohort 1 + 2 (P = 3.50 × 10-4). Moreover, a PGS integrating ZHX2_rs17289471 and five other previously identified SNPs was further significantly associated with CR rate at week 72 in Cohort 1 (P = 2.38 × 10-6), Cohort 2 (P = 1.04 × 10-7) and Cohort 1 + 2 (P = 9.37 × 10-13). Overall, ZHX2_rs17289471 and PGS have the potential to predict response to PegIFNα treatment of HBeAg-positive CHB patients.
Collapse
Affiliation(s)
- Jia Hou
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Institute of Liver Diseases, Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; The Key Laboratory of Molecular Pathology (Hepatic Diseases) of Guangxi, Department of Pathology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, 533099, China
| | - Chao Dong
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, China
| | - Jiaxuan Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Institute of Liver Diseases, Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Haitao Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Institute of Liver Diseases, Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, 528406, China
| | - Rong Na
- Division of Urology, Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Bin Zhou
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Institute of Liver Diseases, Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jinlin Hou
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Institute of Liver Diseases, Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - De-Ke Jiang
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Institute of Liver Diseases, Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China; The Key Laboratory of Molecular Pathology (Hepatic Diseases) of Guangxi, Department of Pathology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi, 533099, China.
| |
Collapse
|
4
|
Hoffmann LB, McVicar EA, Harris RV, Collar-Fernández C, Clark MB, Hannan AJ, Pang TY. Increased paternal corticosterone exposure influences offspring behaviour and expression of urinary pheromones. BMC Biol 2023; 21:186. [PMID: 37667240 PMCID: PMC10478242 DOI: 10.1186/s12915-023-01678-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 08/07/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Studies have shown that paternal stress prior to conception can influence the innate behaviours of their offspring. The evolutionary impacts of such intergenerational effects are therefore of considerable interest. Our group previously showed in a model of daily stress that glucocorticoid treatment of adult male mouse breeders prior to conception leads to increased anxiety-related behaviours in male offspring. Here, we aimed to understand the transgenerational effects of paternal stress exposure on the social behaviour of progeny and its potential influence on reproductive success. RESULTS We assessed social parameters including social reward, male attractiveness and social dominance, in the offspring (F1) and grand-offspring (F2). We report that paternal corticosterone treatment was associated with increased display of subordination towards other male mice. Those mice were unexpectedly more attractive to female mice while expressing reduced levels of the key rodent pheromone Darcin, contrary to its conventional role in driving female attraction. We investigated the epigenetic regulation of major urinary protein (Mup) expression by performing the first Oxford Nanopore direct methylation of sperm DNA in a mouse model of stress, but found no differences in Mup genes that could be attributed to corticosterone-treatment. Furthermore, no overt differences of the prefrontal cortex transcriptome were found in F1 offspring, implying that peripheral mechanisms are likely contributing to the phenotypic differences. Interestingly, no phenotypic differences were observed in the F2 grand-offspring. CONCLUSIONS Overall, our findings highlight the potential of moderate paternal stress to affect intergenerational (mal)adaptive responses, informing future studies of adaptiveness in rodents, humans and other species.
Collapse
Affiliation(s)
- Lucas B Hoffmann
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Florey Department of Neuroscience and Mental Health, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, VIC, Australia
| | - Evangeline A McVicar
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Rebekah V Harris
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Coralina Collar-Fernández
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Florey Department of Neuroscience and Mental Health, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, VIC, Australia
| | - Michael B Clark
- Centre for Stem Cell Systems, Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Anthony J Hannan
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Florey Department of Neuroscience and Mental Health, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, VIC, Australia
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia
| | - Terence Y Pang
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia.
- Florey Department of Neuroscience and Mental Health, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, VIC, Australia.
- Department of Anatomy and Physiology, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
5
|
Del Nogal Avila M, Das R, Kharlyngdoh J, Molina-Jijon E, Donoro Blazquez H, Gambut S, Crowley M, Crossman DK, Gbadegesin RA, Chugh SS, Chugh SS, Avila-Casado C, Macé C, Clement LC, Chugh SS. Cytokine storm-based mechanisms for extrapulmonary manifestations of SARS-CoV-2 infection. JCI Insight 2023; 8:e166012. [PMID: 37040185 PMCID: PMC10322692 DOI: 10.1172/jci.insight.166012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 04/05/2023] [Indexed: 04/12/2023] Open
Abstract
Viral illnesses like SARS-CoV-2 have pathologic effects on nonrespiratory organs in the absence of direct viral infection. We injected mice with cocktails of rodent equivalents of human cytokine storms resulting from SARS-CoV-2/COVID-19 or rhinovirus common cold infection. At low doses, COVID-19 cocktails induced glomerular injury and albuminuria in zinc fingers and homeoboxes 2 (Zhx2) hypomorph and Zhx2+/+ mice to mimic COVID-19-related proteinuria. Common Cold cocktail induced albuminuria selectively in Zhx2 hypomorph mice to model relapse of minimal change disease, which improved after depletion of TNF-α, soluble IL-4Rα, or IL-6. The Zhx2 hypomorph state increased cell membrane to nuclear migration of podocyte ZHX proteins in vivo (both cocktails) and lowered phosphorylated STAT6 activation (COVID-19 cocktail) in vitro. At higher doses, COVID-19 cocktails induced acute heart injury, myocarditis, pericarditis, acute liver injury, acute kidney injury, and high mortality in Zhx2+/+ mice, whereas Zhx2 hypomorph mice were relatively protected, due in part to early, asynchronous activation of STAT5 and STAT6 pathways in these organs. Dual depletion of cytokine combinations of TNF-α with IL-2, IL-13, or IL-4 in Zhx2+/+ mice reduced multiorgan injury and eliminated mortality. Using genome sequencing and CRISPR/Cas9, an insertion upstream of ZHX2 was identified as a cause of the human ZHX2 hypomorph state.
Collapse
Affiliation(s)
- Maria Del Nogal Avila
- Glomerular Disease Therapeutics Laboratory, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Ranjan Das
- Glomerular Disease Therapeutics Laboratory, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Joubert Kharlyngdoh
- Glomerular Disease Therapeutics Laboratory, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Eduardo Molina-Jijon
- Glomerular Disease Therapeutics Laboratory, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Hector Donoro Blazquez
- Glomerular Disease Therapeutics Laboratory, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Stéphanie Gambut
- Glomerular Disease Therapeutics Laboratory, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Michael Crowley
- Genomics Core Lab, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - David K. Crossman
- Genomics Core Lab, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Rasheed A. Gbadegesin
- Division of Nephrology, Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA
| | - Sunveer S. Chugh
- Glomerular Disease Therapeutics Laboratory, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Sunjeet S. Chugh
- Glomerular Disease Therapeutics Laboratory, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Carmen Avila-Casado
- Department of Anatomical Pathology, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
- Instituto Nacional de Cardiología, Mexico City, Mexico
| | - Camille Macé
- Glomerular Disease Therapeutics Laboratory, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Lionel C. Clement
- Glomerular Disease Therapeutics Laboratory, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Sumant S. Chugh
- Glomerular Disease Therapeutics Laboratory, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
6
|
Jiang J, Turpin C, Qiu G(S, Xu M, Lee E, Hinds TD, Peterson ML, Spear BT. Zinc fingers and homeoboxes 2 is required for diethylnitrosamine-induced liver tumor formation in C57BL/6 mice. Hepatol Commun 2022; 6:3550-3562. [PMID: 36194180 PMCID: PMC9701486 DOI: 10.1002/hep4.2106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/31/2022] [Accepted: 09/13/2022] [Indexed: 01/21/2023] Open
Abstract
Liver cancer, comprised primarily of hepatocellular carcinoma (HCC), is the third leading cause of cancer deaths worldwide and increasing in Western countries. We previously identified the transcription factor zinc fingers and homeoboxes 2 (Zhx2) as a regulator of hepatic gene expression, and many Zhx2 target genes are dysregulated in HCC. Here, we investigate HCC in Zhx2-deficient mice using the diethylnitrosamine (DEN)-induced liver tumor model. Our study using whole-body Zhx2 knockout (Zhx2KO ) mice revealed the complete absence of liver tumors 9 and 10 months after DEN exposure. Analysis soon after DEN treatment showed no differences in expression of the DEN bioactivating enzyme cytochrome P450 2E1 (CYP2E1) and DNA polymerase delta 2, or in the numbers of phosphorylated histone variant H2AX foci between Zhx2KO and wild-type (Zhx2wt ) mice. The absence of Zhx2, therefore, did not alter DEN bioactivation or DNA damage. Zhx2KO livers showed fewer positive foci for Ki67 staining and reduced interleukin-6 and AKT serine/threonine kinase 2 expression compared with Zhx2wt livers, suggesting that Zhx2 loss reduces liver cell proliferation and may account for reduced tumor formation. Tumors were reduced but not absent in DEN-treated liver-specific Zhx2 knockout mice, suggesting that Zhx2 acts in both hepatocytes and nonparenchymal cells to inhibit tumor formation. Analysis of data from the Cancer Genome Atlas and Clinical Proteomic Tumor Consortium indicated that ZHX2 messenger RNA and protein levels were significantly higher in patients with HCC and associated with clinical pathological parameters. Conclusion: In contrast to previous studies in human hepatoma cell lines and other HCC mouse models showing that Zhx2 acts as a tumor suppressor, our data indicate that Zhx2 acts as an oncogene in the DEN-induced HCC model and is consistent with the higher ZHX2 expression in patients with HCC.
Collapse
Affiliation(s)
- Jieyun Jiang
- Department of Microbiology, Immunology and Molecular GeneticsUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| | - Courtney Turpin
- Department of Pharmacology and Nutritional SciencesUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| | - Guofang (Shirley) Qiu
- Department of Microbiology, Immunology and Molecular GeneticsUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| | - Mei Xu
- Department of Pharmacology and Nutritional SciencesUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| | - Eun Lee
- Department of Pathology and Laboratory MedicineUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| | - Terry D. Hinds
- Department of Pharmacology and Nutritional SciencesUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
- Barnstable Brown Diabetes CenterUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
- Markey Cancer CenterUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| | - Martha L. Peterson
- Department of Microbiology, Immunology and Molecular GeneticsUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
- Markey Cancer CenterUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| | - Brett T. Spear
- Department of Microbiology, Immunology and Molecular GeneticsUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
- Department of Pharmacology and Nutritional SciencesUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
- Markey Cancer CenterUniversity of Kentucky College of MedicineLexingtonKentuckyUSA
| |
Collapse
|
7
|
Abstract
As a transcriptional factor and the negative regulator of alpha fetal protein (AFP), Zinc fingers and homeoboxes 2 (ZHX2) has a well-established role in protection against hepatocellular carcinoma (HCC). However, recent studies have suggested ZHX2 as an oncogene in clear cell renal cell carcinoma (ccRCC) and triple-negative breast cancer (TNBC). Moreover, mounting evidence has illustrated a much broader role of ZHX2 in multiple cellular processes, including cell proliferation, cell differentiation, lipid metabolism, and immunoregulation. This comprehensive review emphasizes the role of ZHX2 in health and diseases which have been more recently uncovered.
Collapse
Affiliation(s)
- Na Li
- Key Laboratory for Experimental Teratology of Ministry of Education and Dept. Immunology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, Shandong, China
| | - Zhuanchang Wu
- Key Laboratory for Experimental Teratology of Ministry of Education and Dept. Immunology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, Shandong, China
| | - Chunhong Ma
- Key Laboratory for Experimental Teratology of Ministry of Education and Dept. Immunology, School of Basic Medical Sciences, Cheeloo Medical College, Shandong University, Jinan, Shandong, China
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong University, Jinan, Shandong, China
| |
Collapse
|
8
|
Bretes E, Wróblewski J, Wyszczelska-Rokiel M, Jakubowski H. Cystathionine β-synthase gene inactivation dysregulates major urinary protein biogenesis and impairs sexual signaling in mice. FASEB J 2022; 36:e22547. [PMID: 36098436 DOI: 10.1096/fj.202200969r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/23/2022] [Accepted: 08/30/2022] [Indexed: 11/11/2022]
Abstract
Reproductive success in mice depends on sexually dimorphic major urinary proteins (Mup) that facilitate interactions between females and males. Deletion of cystathionine β-synthase (Cbs) gene, a metabolic gene important for homeostasis of one-carbon metabolism, impairs reproduction by causing female infertility in mice. Here, we examined Mup biogenesis and sexual signaling in Cbs-/- versus Cbs+/- mice. We found significantly reduced levels of total urinary Mup protein in male and female Cbs-/- versus Cbs+/- mice. SDS-PAGE/Western blot, ESI-MS, and RT-qPCR analyses of the liver, plasma, and urinary proteins identified a male-specific Mup20 in Cbs-/- , but not in Cbs+/- females. The 18 893 Da Mup20 became the most abundant in urine of Cbs-/- females and males. Effects of Cbs genotype on 18 645 Da, 18 693 Da, and 18 709 Da Mup species abundance were Mup- and sex-specific. Cbs genotype-dependent changes in hepatic Mups and Mup20 expression were similar at the protein and mRNA level. Changes in Mups, but not in Mup20, can be explained by downregulation of hepatic Zhx2 and Ghr receptors in Cbs-/- mice. Behavioral testing showed that Cbs+/- females ignored Cbs-/- male urine but were attracted to Cbs+/- male urine. Cbs+/- males ignored urine of Cbs-/- males but countermarked urine of other Cbs+/- males and were attracted to urines of Cbs-/- as well as Cbs+/- females. Cbs-/- males did not countermark urine of Cbs+/- males but were attracted to urines of Cbs+/- females. Taken together, these findings show that Cbs, a metabolic gene, interacts with the processes involved in Mup biogenesis that are essential for the maintenance of sexual dimorphism and signaling and suggest that dysregulation of these interactions impairs reproductive fitness in mice.
Collapse
Affiliation(s)
- Ewa Bretes
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, Poznań, Poland
| | - Jacek Wróblewski
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, Poznań, Poland.,Institute of Bioorganic Chemistry, Poznań, Poland
| | - Monika Wyszczelska-Rokiel
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers University, New Jersey Medical School, International Center for Public Health, Newark, New Jersey, USA
| | - Hieronim Jakubowski
- Department of Biochemistry and Biotechnology, Poznań University of Life Sciences, Poznań, Poland.,Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers University, New Jersey Medical School, International Center for Public Health, Newark, New Jersey, USA
| |
Collapse
|
9
|
Bao Y, Zhang H, Han Z, Guo Y, Yang W. Zinc Fingers and Homeobox Family in Cancer: A Double-Edged Sword. Int J Mol Sci 2022; 23:ijms231911167. [PMID: 36232466 PMCID: PMC9570228 DOI: 10.3390/ijms231911167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022] Open
Abstract
The zinc fingers and homeobox (ZHX) family includes ZHX1, ZHX2, and ZHX3, and their proteins have similar unique structures, containing two C2H2-type zinc finger motifs and four or five HOX-like homeodomains. The members of the ZHX family can form homodimers or heterodimers with each other or with a subunit of nuclear factor Y. Previous studies have suggested that ZHXs can function as positive or negative transcriptional regulators. Recent studies have further revealed their biological functions and underlying mechanisms in cancers. This review summarized the advances of ZHX-mediated functions, including tumor-suppressive and oncogenic functions in cancer formation and progression, the molecular mechanisms, and regulatory functions, such as cancer cell proliferation, migration, invasion, and metastasis. Moreover, the differential expression levels and their association with good or poor outcomes in patients with various malignancies and differential responses to chemotherapy exert opposite functions of oncogene or tumor suppressors. Therefore, the ZHXs act as a double-edged sword in cancers.
Collapse
Affiliation(s)
- Yonghua Bao
- Department of Pathology, Mudanjiang Medical University, Mudanjiang 157011, China
| | - Haifeng Zhang
- Department of Pathology, Mudanjiang Medical University, Mudanjiang 157011, China
| | - Zhixue Han
- Department of Pathology, Mudanjiang Medical University, Mudanjiang 157011, China
| | - Yongchen Guo
- Department of Immunology, Mudanjiang Medical University, Mudanjiang 157011, China
- Correspondence: (Y.G.); (W.Y.)
| | - Wancai Yang
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- Correspondence: (Y.G.); (W.Y.)
| |
Collapse
|
10
|
Beierle JA, Yao EJ, Goldstein SI, Lynch WB, Scotellaro JL, Shah AA, Sena KD, Wong AL, Linnertz CL, Averin O, Moody DE, Reilly CA, Peltz G, Emili A, Ferris MT, Bryant CD. Zhx2 Is a Candidate Gene Underlying Oxymorphone Metabolite Brain Concentration Associated with State-Dependent Oxycodone Reward. J Pharmacol Exp Ther 2022; 382:167-180. [PMID: 35688478 PMCID: PMC9341249 DOI: 10.1124/jpet.122.001217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/16/2022] [Indexed: 11/22/2022] Open
Abstract
Understanding the pharmacogenomics of opioid metabolism and behavior is vital to therapeutic success, as mutations can dramatically alter therapeutic efficacy and addiction liability. We found robust, sex-dependent BALB/c substrain differences in oxycodone behaviors and whole brain concentration of oxycodone metabolites. BALB/cJ females showed robust state-dependent oxycodone reward learning as measured via conditioned place preference when compared with the closely related BALB/cByJ substrain. Accordingly, BALB/cJ females also showed a robust increase in brain concentration of the inactive metabolite noroxycodone and the active metabolite oxymorphone compared with BALB/cByJ mice. Oxymorphone is a highly potent, full agonist at the mu opioid receptor that could enhance drug-induced interoception and state-dependent oxycodone reward learning. Quantitative trait locus (QTL) mapping in a BALB/c F2 reduced complexity cross revealed one major QTL on chromosome 15 underlying brain oxymorphone concentration that explained 32% of the female variance. BALB/cJ and BALB/cByJ differ by fewer than 10,000 variants, which can greatly facilitate candidate gene/variant identification. Hippocampal and striatal cis-expression QTL (eQTL) and exon-level eQTL analysis identified Zhx2, a candidate gene coding for a transcriptional repressor with a private BALB/cJ retroviral insertion that reduces Zhx2 expression and sex-dependent dysregulation of cytochrome P450 enzymes. Whole brain proteomics corroborated the Zhx2 eQTL and identified upregulated CYP2D11 that could increase brain oxymorphone in BALB/cJ females. To summarize, Zhx2 is a highly promising candidate gene underlying brain oxycodone metabolite levels. Future studies will validate Zhx2 and its site of action using reciprocal gene editing and tissue-specific viral manipulations in BALB/c substrains. SIGNIFICANCE STATEMENT: Our findings show that genetic variation can result in sex-specific alterations in whole brain concentration of a bioactive opioid metabolite after oxycodone administration, reinforcing the need for sex as a biological factor in pharmacogenomic studies. The cooccurrence of female-specific increased oxymorphone and state-dependent reward learning suggests that this minor yet potent and efficacious metabolite of oxycodone could increase opioid interoception and drug-cue associative learning of opioid reward, which has implications for cue-induced relapse of drug-seeking behavior and for precision pharmacogenetics.
Collapse
Affiliation(s)
- Jacob A Beierle
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Emily J Yao
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Stanley I Goldstein
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - William B Lynch
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Julia L Scotellaro
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Anyaa A Shah
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Katherine D Sena
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Alyssa L Wong
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Colton L Linnertz
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Olga Averin
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - David E Moody
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Christopher A Reilly
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Gary Peltz
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Andrew Emili
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Martin T Ferris
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| | - Camron D Bryant
- Ph.D. Program in Biomolecular Pharmacology (J.A.B., S.I.G.), Laboratory of Addiction Genetics, Department of Pharmacology and Experimental Therapeutics and Psychiatry (J.A.B., E.J.Y., W.B.L., J.L.S., A.A.S., K.D.S., A.L.W., C.D.B.), Department of Biology and Biochemistry, Center for Network Systems Biology (S.I.G., A.E.), and Graduate Program in Neuroscience (W.B.L), Boston University School of Medicine, Boston, Massachusetts; Transformative Training Program in Addiction Science (TTPAS) (J.A.B., W.B.L.) and Undergraduate Research Opportunity Program (J.L.S., K.D.S.), Boston University, Boston, Massachusetts; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina (C.L.L., M.T.F.); Department of Pharmacology and Toxicity, Center for Human Toxicology, University of Utah, Salt Lake City, Utah (O.A., D.E.M., C.A.R.); and Department of Anesthesiology, Pain, and Preoperative Medicine Stanford University School of Medicine, Stanford, California (G.P.)
| |
Collapse
|
11
|
Conner MM, Parker HV, Falcone DR, Chung G, Schaner Tooley CE. Novel regulation of the transcription factor ZHX2 by N-terminal methylation. Transcription 2022; 13:1-15. [PMID: 35613330 DOI: 10.1080/21541264.2022.2079184] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
N-terminal methylation (Nα-methylation) by the methyltransferase NRMT1 is an important post-translational modification that regulates protein-DNA interactions. Accordingly, its loss impairs functions that are reliant on such interactions, including DNA repair and transcriptional regulation. The global loss of Nα-methylation results in severe developmental and premature aging phenotypes, but given over 300 predicted substrates, it is hard to discern which physiological substrates contribute to each phenotype. One of the most striking phenotypes in NRMT1 knockout (Nrmt1-/-) mice is early liver degeneration. To identify the disrupted signaling pathways leading to this phenotype and the NRMT1 substrates involved, we performed RNA-sequencing analysis of control and Nrmt1-/- adult mouse livers. We found both a significant upregulation of transcripts in the cytochrome P450 (CYP) family and downregulation of transcripts in the major urinary protein (MUP) family. Interestingly, transcription of both families is inversely regulated by the transcription factor zinc fingers and homeoboxes 2 (ZHX2). ZHX2 contains a non-canonical NRMT1 consensus sequence, indicating that its function could be directly regulated by Nα-methylation. We confirmed misregulation of CYP and MUP mRNA and protein levels in Nrmt1-/- livers and verified NRMT1 can methylate ZHX2 in vitro. In addition, we used a mutant of ZHX2 that cannot be methylated to directly demonstrate Nα-methylation promotes ZHX2 transcription factor activity and target promoter occupancy. Finally, we show Nrmt1-/- mice also exhibit early postnatal de-repression of ZHX2 targets involved in fetal liver development. Taken together, these data implicate ZHX2 misregulation as a driving force behind the liver phenotype seen in Nrmt1-/- mice.
Collapse
Affiliation(s)
- Meghan M Conner
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Haley V Parker
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Daniela R Falcone
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Gehoon Chung
- Department of Oral Physiology and Program in Neurobiology, School of Dentistry, Seoul National University, Seoul, South Korea
| | - Christine E Schaner Tooley
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
12
|
Penn DJ, Zala SM, Luzynski KC. Regulation of Sexually Dimorphic Expression of Major Urinary Proteins. Front Physiol 2022; 13:822073. [PMID: 35431992 PMCID: PMC9008510 DOI: 10.3389/fphys.2022.822073] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/21/2022] [Indexed: 11/15/2022] Open
Abstract
Male house mice excrete large amounts of protein in their urinary scent marks, mainly composed of Major Urinary Proteins (MUPs), and these lipocalins function as pheromones and pheromone carriers. Here, we review studies on sexually dimorphic MUP expression in house mice, including the proximate mechanisms controlling MUP gene expression and their adaptive functions. Males excrete 2 to 8 times more urinary protein than females, though there is enormous variation in gene expression across loci in both sexes. MUP expression is dynamically regulated depending upon a variety of factors. Males regulate MUP expression according to social status, whereas females do not, and males regulate expression depending upon health and condition. Male-biased MUP expression is regulated by pituitary secretion of growth hormone (GH), which binds receptors in the liver, activating the JAK2-STAT5 signaling pathway, chromatin accessibility, and MUP gene transcription. Pulsatile male GH secretion is feminized by several factors, including caloric restriction, microbiota depletion, and aging, which helps explain condition-dependent MUP expression. If MUP production has sex-specific fitness optima, then this should generate sexual antagonism over allelic expression (intra-locus sexual conflict) selectively favoring sexually dimorphic expression. MUPs influence the sexual attractiveness of male urinary odor and increased urinary protein excretion is correlated with the reproductive success of males but not females. This finding could explain the selective maintenance of sexually dimorphic MUP expression. Producing MUPs entails energetic costs, but increased excretion may reduce the net energetic costs and predation risks from male scent marking as well as prolong the release of chemical signals. MUPs may also provide physiological benefits, including regulating metabolic rate and toxin removal, which may have sex-specific effects on survival. A phylogenetic analysis on the origins of male-biased MUP gene expression in Mus musculus suggests that this sexual dimorphism evolved by increasing male MUP expression rather than reducing female expression.
Collapse
Affiliation(s)
- Dustin J. Penn
- Department of Interdisciplinary Life Sciences, Konrad Lorenz Institute of Ethology, University of Veterinary Medicine Vienna, Vienna, Austria
| | | | | |
Collapse
|
13
|
Zhou Q, Guo H, Yu C, Huang XR, Liang L, Zhang P, Yu J, Zhang J, Chan TF, Ma RCW, Lan HY. Identification of Smad3-related transcriptomes in type-2 diabetic nephropathy by whole transcriptome RNA sequencing. J Cell Mol Med 2020; 25:2052-2068. [PMID: 33369170 PMCID: PMC7882931 DOI: 10.1111/jcmm.16133] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/01/2020] [Accepted: 11/04/2020] [Indexed: 12/21/2022] Open
Abstract
Smad3 deficiency prevents the development of type 2 diabetic nephropathy; however, the underlying molecular mechanisms remain unknown. In this study, we aimed to identify Smad3‐related genes involved in the pathogenesis of diabetic kidney disease. High‐throughput RNA sequencing was performed to profile the whole transcriptome in the diabetic kidney of Smad3 WT‐db/db, Smad3 KO‐db/db, Smad3+/− db/db and their littermate control db/m mice at 20 weeks. The gene ontology, pathways and alternative splicing of differentially expressed protein‐coding genes and long non‐coding RNAs related to Smad3 in diabetic kidney were analysed. Compared to Smad3 WT‐db/db mice, Smad3 KO‐db/db mice exhibited an alteration of genes associated with RNA splicing and metabolism, whereas heterozygosity deletion of Smad3 (Smad3+/− db/db mice) significantly altered genes related to cell division and cell cycle. Notably, three protein‐coding genes (Upk1b, Psca and Gdf15) and two lncRNAs (NONMMUG023520.2 and NONMMUG032975.2) were identified to be Smad3‐dependent and to be associated with the development of diabetic nephropathy. By using whole transcriptome RNA sequencing, we identified novel Smad3 transcripts related to the development of diabetic nephropathy. Thus, targeting these transcripts may represent a novel and effective therapy for diabetic nephropathy.
Collapse
Affiliation(s)
- Qin Zhou
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,National Health Commission Key Laboratory of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Honghong Guo
- State Key Laboratory for Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Chaolun Yu
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiao-Ru Huang
- Guangdong-Hong Kong Joint Laboratory for Immunological and Genetic Kidney Disease, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China.,Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Liying Liang
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Puhua Zhang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,National Health Commission Key Laboratory of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianwen Yu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,National Health Commission Key Laboratory of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jizhou Zhang
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ting-Fung Chan
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ronald C W Ma
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.,Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
14
|
Wang Z, Kong L, Tan S, Zhang Y, Song X, Wang T, Lin Q, Wu Z, Xiang P, Li C, Gao L, Liang X, Ma C. Zhx2 Accelerates Sepsis by Promoting Macrophage Glycolysis via Pfkfb3. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:2232-2241. [PMID: 32179636 DOI: 10.4049/jimmunol.1901246] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 02/12/2020] [Indexed: 01/03/2025]
Abstract
Sepsis is a life-threatening condition with limited therapeutic options, characterized as excessive systemic inflammation and multiple organ failure. Macrophages play critical roles in sepsis pathogenesis. Metabolism orchestrates homeostasis of macrophages. However, the precise mechanism of macrophage metabolism during sepsis remains poorly elucidated. In this study, we identified the key role of zinc fingers and homeoboxes (Zhx2), a ubiquitous transcription factor, in macrophage glycolysis and sepsis by enhancing 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (Pfkfb3) expression. Mice with myeloid Zhx2-specific deletion (abbreviated as MKO) showed more resistance to cecal ligation and puncture and LPS-induced sepsis, exhibiting as prolonged survival, attenuated pulmonary injury, and reduced level of proinflammatory cytokines, such as TNF-α, IL-6, and IL-1β. Interestingly, Zhx2 deletion conferred macrophage tolerance to LPS-induced glycolysis, accompanied by reduced proinflammatory cytokines and lactate. Consistently, treatment of glycolytic inhibitor 2-deoxyglucose almost completely abrogated the protection of mice from LPS-induced sepsis initiated by Zhx2 deletion in macrophages. RNA sequencing and chromatin immunoprecipitation assays confirmed that Zhx2 enhanced transcription of Pfkfb3, the glycolysis rate-limiting enzyme, via binding with Pfkfb3 promoter. Furthermore, Pfkfb3 overexpression not only rescued the reduction of macrophage glycolysis caused by Zhx2 deficiency, displaying as extracellular acidification rates and lactate production but also destroyed the resistance of mice to LPS-induced sepsis initiated by transfer of bone marrow-derived macrophages from MKO mice. These findings highlight the novel role of transcription factor Zhx2 in sepsis via regulating Pfkfb3 expression and reprogramming macrophage metabolism, which would shed new insights into the potential strategy to intervene sepsis.
Collapse
Affiliation(s)
- Zehua Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
| | - Liang Kong
- Department of Clinical Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250012, People's Republic of China
| | - Siyu Tan
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
| | - Yankun Zhang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
| | - Xiaojia Song
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
| | - Tixiao Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
| | - Qinghai Lin
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
| | - Zhuanchang Wu
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
| | - Peng Xiang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
| | - Chunyang Li
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Histology and Embryology, Shandong University School of Basic Medical Science, Jinan, Shandong 250012, People's Republic of China; and
| | - Lifen Gao
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
| | - Xiaohong Liang
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
| | - Chunhong Ma
- Key Laboratory for Experimental Teratology of Ministry of Education, Department of Immunology, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China;
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong University School of Basic Medical Sciences, Jinan, Shandong 250012, People's Republic of China
| |
Collapse
|
15
|
Pallauf K, Günther I, Chin D, Rimbach G. In Contrast to Dietary Restriction, Application of Resveratrol in Mice Does not Alter Mouse Major Urinary Protein Expression. Nutrients 2020; 12:nu12030815. [PMID: 32204477 PMCID: PMC7146287 DOI: 10.3390/nu12030815] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/16/2020] [Accepted: 03/17/2020] [Indexed: 12/20/2022] Open
Abstract
Resveratrol (RSV) supplementation in mice has been discussed as partly mimicking the beneficial effects of dietary restriction (DR). However, data on putative benefits from resveratrol application in mice and other model organisms including humans is contradictory. Mouse major urinary proteins (MUPs) are a family of proteins that are expressed in rodent liver and secreted via urine. Impacting (mating) behavior and pheromone communication, they are severely down-regulated upon DR. We carried out two studies in C57BL/6Rj mice where RSV was either supplemented via diet or injected intraperitoneally for 8 weeks. Contrary to −40% DR, RSV did not decrease total MUP protein expression or Mup (amongst others Mup3, Mup5, Mup6, Mup15, and Mup20) mRNA levels in mouse liver when compared to ad-libitum (AL)-fed controls. Since inhibitory glucocorticoid response elements can be found in Mup promoters, we also measured glucocorticoid receptor (GR) levels in nuclear hepatic extracts. Consistent with differential MUP expression, we observed more nuclear GR in DR mice than in RSV-supplemented and AL control mice with no difference between RSV and AL. These findings point to the notion that, in mice, RSV does not mimic DR in terms of differential MUP expression.
Collapse
|
16
|
Nail AN, Smith JJ, Peterson ML, Spear BT. Evolutionary Analysis of the Zinc Finger and Homeoboxes Family of Proteins Identifies Multiple Conserved Domains and a Common Early Chordate Ancestor. Genome Biol Evol 2020; 12:174-184. [PMID: 32125369 PMCID: PMC7144352 DOI: 10.1093/gbe/evaa039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2020] [Indexed: 12/26/2022] Open
Abstract
The Zinc Fingers and Homeoboxes (Zhx) proteins, Zhx1, Zhx2, and Zhx3, comprise a small family of proteins containing two amino-terminal C2–H2 zinc fingers and four or five carboxy-terminal homeodomains. These multiple homeodomains make Zhx proteins unusual because the majority of homeodomain-containing proteins contain a single homeodomain. Studies in cultured cells and mice suggest that Zhx proteins can function as positive or negative transcriptional regulators. Zhx2 regulates numerous hepatic genes, and all three Zhx proteins have been implicated in different cancers. Because Zhx proteins contain multiple predicted homeodomains, are associated with interesting physiological traits, and seem to be only present in the vertebrate lineage, we investigated the evolutionary history of this small family by comparing Zhx homologs from a wide range of chordates. This analysis indicates that the zinc finger motifs and homeodomains are highly similar among all Zhx proteins and also identifies additional Zhx-specific conserved regions, including a 13 amino acid amino-terminal motif that is nearly identical among all gnathostome Zhx proteins. We found single Zhx proteins in the sea lamprey (Petromyzon marinus) and in the nonvertebrate chordates sea squirt (Ciona intestinalis) and lancelet (Branchiostoma floridae); these Zhx proteins are most similar to gnathostome Zhx3. Based on our analyses, we propose that a duplication of the primordial Zhx gene gave rise to Zhx3 and the precursor to Zhx1 and Zhx2. A subsequent tandem duplication of this precursor generated Zhx1 and Zhx2 found in gnathostomes.
Collapse
Affiliation(s)
- Alexandra N Nail
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky
| | - Jeramiah J Smith
- Department of Biology, University of Kentucky.,Markey Cancer Center, University of Kentucky
| | - Martha L Peterson
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky.,Markey Cancer Center, University of Kentucky
| | - Brett T Spear
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky.,Markey Cancer Center, University of Kentucky
| |
Collapse
|
17
|
Host Transcription Factors in Hepatitis B Virus RNA Synthesis. Viruses 2020; 12:v12020160. [PMID: 32019103 PMCID: PMC7077322 DOI: 10.3390/v12020160] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/27/2020] [Accepted: 01/28/2020] [Indexed: 02/06/2023] Open
Abstract
The hepatitis B virus (HBV) chronically infects over 250 million people worldwide and is one of the leading causes of liver cancer and hepatocellular carcinoma. HBV persistence is due in part to the highly stable HBV minichromosome or HBV covalently closed circular DNA (cccDNA) that resides in the nucleus. As HBV replication requires the help of host transcription factors to replicate, focusing on host protein–HBV genome interactions may reveal insights into new drug targets against cccDNA. The structural details on such complexes, however, remain poorly defined. In this review, the current literature regarding host transcription factors’ interactions with HBV cccDNA is discussed.
Collapse
|
18
|
Clinkenbeard EL, Turpin C, Jiang J, Peterson ML, Spear BT. Liver size and lipid content differences between BALB/c and BALB/cJ mice on a high-fat diet are due, in part, to Zhx2. Mamm Genome 2019; 30:226-236. [PMID: 31321500 DOI: 10.1007/s00335-019-09811-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 07/09/2019] [Indexed: 11/26/2022]
Abstract
BALB/cJ mice exhibit considerable phenotypic differences with other BALB/c substrains. Some of these traits involve the liver, including persistent postnatal expression of genes that are normally expressed only in the fetal liver and reduced expression of major urinary proteins. These traits are due to a mutation that dramatically reduces expression of the gene encoding the transcription factor Zinc fingers and homeoboxes 2 (Zhx2). BALB/cJ mice also exhibit reduced serum lipid levels and resistance to atherosclerosis compared to other mouse strains when placed on a high-fat diet. This trait is also due, at least in part, to the Zhx2 mutation. Microarray analysis identified many genes affecting lipid homeostasis, including Lipoprotein lipase, that are dysregulated in BALB/cJ liver. This led us to investigate whether hepatic lipid levels would be different between BALB/cJ and BALB/c mice when placed on a normal chow or a high-fat chow diet. On the high-fat chow, BALB/cJ mice had increased weight gain, increased liver:body weight ratio, elevated hepatic lipid accumulation and markers of liver damage when compared to BALB/c mice. These traits in BALB/cJ mice were only partially reversed by a hepatocyte-specific Zhx2 transgene. These data indicate that Zhx2 reduces liver lipid levels and is hepatoprotective in mice on a high-fat diet, but the partial rescue by the Zhx2 transgene suggests a contribution by both parenchymal and non-parenchymal cells. A model to account for the cardiovascular and liver phenotype in mice with reduced Zhx2 levels is provided.
Collapse
Affiliation(s)
- Erica L Clinkenbeard
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Courtney Turpin
- Department of Pharmacology & Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - Jieyun Jiang
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - Martha L Peterson
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - Brett T Spear
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY, 40536, USA.
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, 40536, USA.
| |
Collapse
|
19
|
Morales-Prieto N, Ruiz-Laguna J, Sheehan D, Abril N. Transcriptome signatures of p,p´-DDE-induced liver damage in Mus spretus mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 238:150-167. [PMID: 29554563 DOI: 10.1016/j.envpol.2018.03.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/19/2018] [Accepted: 03/04/2018] [Indexed: 06/08/2023]
Abstract
The use of DDT (1,1,1-trichloro-2,2-bis(p-chlorophenyl) ethane) in some countries, although regulated, is contributing to an increased worldwide risk of exposure to this organochlorine pesticide or its derivative p,p'-DDE [1,1-dichloro-2,2-bis(p-chlorophenyl) ethylene]. Many studies have associated p,p'-DDE exposure to type 2 diabetes, obesity and alterations of the reproductive system, but their molecular mechanisms of toxicity remain poorly understood. We have addressed this issue by using commercial microarrays based on probes for the entire Mus musculus genome to determine the hepatic transcriptional signatures of p,p'-DDE in the phylogenetically close mouse species Mus spretus. High-stringency hybridization conditions and analysis assured reliable results, which were also verified, in part, by qRT-PCR, immunoblotting and/or enzymatic activity. Our data linked 198 deregulated genes to mitochondrial dysfunction and perturbations of central signaling pathways (kinases, lipids, and retinoic acid) leading to enhanced lipogenesis and aerobic glycolysis, inflammation, cell proliferation and testosterone catabolism and excretion. Alterations of transcript levels of genes encoding enzymes involved in testosterone catabolism and excretion would explain the relationships established between p,p´-DDE exposure and reproductive disorders, obesity and diabetes. Further studies will help to fully understand the molecular basis of p,p´-DDE molecular toxicity in liver and reproductive organs, to identify effective exposure biomarkers and perhaps to design efficient p,p'-DDE exposure counteractive strategies.
Collapse
Affiliation(s)
- Noelia Morales-Prieto
- Departamento de Bioquímica y Biología Molecular, Campus de Excelencia Internacional Agroalimentario CeiA3, Universidad de Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071, Córdoba, Spain
| | - Julia Ruiz-Laguna
- Departamento de Bioquímica y Biología Molecular, Campus de Excelencia Internacional Agroalimentario CeiA3, Universidad de Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071, Córdoba, Spain
| | - David Sheehan
- College of Arts and Science, Khalifa University of Science and Technology, PO Box 127788, Abu Dhabi, United Arab Emirates
| | - Nieves Abril
- Departamento de Bioquímica y Biología Molecular, Campus de Excelencia Internacional Agroalimentario CeiA3, Universidad de Córdoba, Campus de Rabanales, Edificio Severo Ochoa, E-14071, Córdoba, Spain.
| |
Collapse
|
20
|
Loss of liver-specific and sexually dimorphic gene expression by aryl hydrocarbon receptor activation in C57BL/6 mice. PLoS One 2017; 12:e0184842. [PMID: 28922406 PMCID: PMC5602546 DOI: 10.1371/journal.pone.0184842] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 08/31/2017] [Indexed: 01/13/2023] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a highly conserved transcription factor that mediates a broad spectrum of species-, strain-, sex-, age-, tissue-, and cell-specific responses elicited by structurally diverse ligands including 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Dose-dependent effects on liver-specific and sexually dimorphic gene expression were examined in male and female mice gavaged with TCDD every 4 days for 28 or 92 days. RNA-seq data revealed the coordinated repression of 181 genes predominately expressed in the liver including albumin (3.7-fold), α-fibrinogen (14.5-fold), and β-fibrinogen (17.4-fold) in males with corresponding AhR enrichment at 2 hr. Liver-specific genes exhibiting sexually dimorphic expression also demonstrated diminished divergence between sexes. For example, male-biased Gstp1 was repressed 3.0-fold in males and induced 4.5-fold in females, which were confirmed at the protein level. Disrupted regulation is consistent with impaired GHR-JAK2-STAT5 signaling and inhibition of female specific CUX2-mediated transcription as well as the repression of other key transcriptional regulators including Ghr, Stat5b, Bcl6, Hnf4a, Hnf6, Foxa1/2/3, and Zhx2. Attenuated liver-specific and sexually dimorphic gene expression was concurrent with the induction of fetal genes such as alpha-fetoprotein. The results suggest AhR activation causes the loss of liver-specific and sexually dimorphic gene expression producing a functionally "de-differentiated" hepatic phenotype.
Collapse
|