1
|
Sarver DC, Garcia-Diaz J, Saqib M, Riddle RC, Wong GW. Tmem263 deletion disrupts the GH/IGF-1 axis and causes dwarfism and impairs skeletal acquisition. eLife 2024; 12:RP90949. [PMID: 38241182 PMCID: PMC10945605 DOI: 10.7554/elife.90949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2024] Open
Abstract
Genome-wide association studies (GWAS) have identified a large number of candidate genes believed to affect longitudinal bone growth and bone mass. One of these candidate genes, TMEM263, encodes a poorly characterized plasma membrane protein. Single nucleotide polymorphisms in TMEM263 are associated with bone mineral density in humans and mutations are associated with dwarfism in chicken and severe skeletal dysplasia in at least one human fetus. Whether this genotype-phenotype relationship is causal, however, remains unclear. Here, we determine whether and how TMEM263 is required for postnatal growth. Deletion of the Tmem263 gene in mice causes severe postnatal growth failure, proportional dwarfism, and impaired skeletal acquisition. Mice lacking Tmem263 show no differences in body weight within the first 2 weeks of postnatal life. However, by P21 there is a dramatic growth deficit due to a disrupted growth hormone (GH)/insulin-like growth factor 1 (IGF-1) axis, which is critical for longitudinal bone growth. Tmem263-null mice have low circulating IGF-1 levels and pronounced reductions in bone mass and growth plate length. The low serum IGF-1 in Tmem263-null mice is associated with reduced hepatic GH receptor (GHR) expression and GH-induced JAK2/STAT5 signaling. A deficit in GH signaling dramatically alters GH-regulated genes and feminizes the liver transcriptome of Tmem263-null male mice, with their expression profile resembling wild-type female, hypophysectomized male, and Stat5b-null male mice. Collectively, our data validates the causal role for Tmem263 in regulating postnatal growth and raises the possibility that rare mutations or variants of TMEM263 may potentially cause GH insensitivity and impair linear growth.
Collapse
Affiliation(s)
- Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Jean Garcia-Diaz
- Department of Orthopaedic Surgery, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Orthopaedics, University of Maryland School of MedicineBaltimoreUnited States
- Cell and Molecular Medicine graduate program, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Muzna Saqib
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Ryan C Riddle
- Department of Orthopaedic Surgery, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Orthopaedics, University of Maryland School of MedicineBaltimoreUnited States
- Research and Development Service, Baltimore Veterans Administration Medical CenterBaltimoreUnited States
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of MedicineBaltimoreUnited States
| |
Collapse
|
2
|
Sarver DC, Garcia-Diaz J, Saqib M, Riddle RC, Wong GW. Tmem263 deletion disrupts the GH/IGF-1 axis and causes dwarfism and impairs skeletal acquisition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.02.551694. [PMID: 37577461 PMCID: PMC10418210 DOI: 10.1101/2023.08.02.551694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Genome-wide association studies (GWAS) have identified a large number of candidate genes believed to affect longitudinal bone growth and bone mass. One of these candidate genes, TMEM263, encodes a poorly characterized plasma membrane protein. Single nucleotide polymorphisms in TMEM263 are associated with bone mineral density in humans and mutations are associated with dwarfism in chicken and severe skeletal dysplasia in at least one human fetus. Whether this genotype-phenotype relationship is causal, however, remains unclear. Here, we determine whether and how TMEM263 is required for postnatal growth. Deletion of the Tmem263 gene in mice causes severe postnatal growth failure, proportional dwarfism, and impaired skeletal acquisition. Mice lacking Tmem263 show no differences in body weight within the first two weeks of postnatal life. However, by P21 there is a dramatic growth deficit due to a disrupted GH/IGF-1 axis, which is critical for longitudinal bone growth. Tmem263-null mice have low circulating IGF-1 levels and pronounced reductions in bone mass and growth plate length. The low serum IGF-1 in Tmem263-null mice is associated with reduced hepatic GH receptor (GHR) expression and GH-induced JAK2/STAT5 signaling. A deficit in GH signaling dramatically alters GH-regulated genes and feminizes the liver transcriptome of Tmem263-null male mice, with their expression profile resembling a wild-type female, hypophysectomized male, and Stat5b-null male mice. Collectively, our data validates the causal role for Tmem263 in regulating postnatal growth and raises the possibility that rare mutations or variants of TMEM263 may potentially cause GH insensitivity and impair linear growth.
Collapse
Affiliation(s)
- Dylan C Sarver
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jean Garcia-Diaz
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Cell and Molecular Medicine graduate program, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Muzna Saqib
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ryan C Riddle
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Research and Development Service, Baltimore Veterans Administration Medical Center, Baltimore, Maryland, USA
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
3
|
Rizzolo D, Kong B, Piekos S, Chen L, Zhong X, Lu J, Shi J, Zhu HJ, Yang Q, Li A, Li L, Wang H, Siemiątkowska A, Park C, Kagan L, Guo GL. Effects of Overexpression of Fibroblast Growth Factor 15/19 on Hepatic Drug Metabolizing Enzymes. Drug Metab Dispos 2022; 50:468-477. [PMID: 34965924 PMCID: PMC11022908 DOI: 10.1124/dmd.121.000416] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 12/20/2021] [Indexed: 11/22/2022] Open
Abstract
Fibroblast growth factors 15 (FGF15) and 19 (FGF19) are endocrine growth factors that play an important role in maintaining bile acid homeostasis. FGF15/19-based therapies are currently being tested in clinical trials for the treatment of nonalcoholic steatohepatitis and cholestatic liver diseases. To determine the physiologic impact of long-term elevations of FGF15/19, a transgenic mouse model with overexpression of Fgf15 (Fgf15 Tg) was used in the current study. The RNA sequencing (RNA-seq) analysis revealed elevations of the expression of several genes encoding phase I drug metabolizing enzymes (DMEs), including Cyp2b10 and Cyp3a11, in Fgf15 Tg mice. We found that the induction of several Cyp2b isoforms resulted in increased function of CYP2B in microsomal metabolism and pharmacokinetics studies. Because the CYP2B family is known to be induced by constitutive androstane receptor (CAR), to determine the role of CAR in the observed inductions, we crossed Fgf15 Tg mice with CAR knockout mice and found that CAR played a minor role in the observed alterations in DME expression. Interestingly, we found that the overexpression of Fgf15 in male mice resulted in a phenotypical switch from the male hepatic expression pattern of DMEs to that of female mice. Differences in secretion of growth hormone (GH) between male and female mice are known to drive sexually dimorphic, STAT5b-dependent expression patterns of hepatic genes. We found that male Fgf15 Tg mice presented with many features similar to GH deficiency, including lowered body length and weight, Igf-1 and Igfals expression, and STAT5 signaling. SIGNIFICANCE STATEMENT: The overexpression of Fgf15 in mice causes an alteration in DMEs at the mRNA, protein, and functional levels, which is not entirely due to CAR activation but associated with lower GH signaling.
Collapse
Affiliation(s)
- Daniel Rizzolo
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (D.R., B.K., G.L.G.), Department of Pharmaceutical Sciences, Ernest Mario School of Pharmacy (A.S., C.P., L.K.), Center of Excellence for Pharmaceutical Translational Research and Education (A.S., C.P., L.K.), and Environmental and Occupational Health Sciences Institute (EOHSI) (D.R., G.L.G.), Rutgers University, Piscataway, New Jersey; Rutgers Center for Lipid Research, Rutgers University-New Brunswick, New Brunswick, New Jersey (D.R., G.L.G.); VA New Jersey Health Care System, Veterans Administration Medical Center, East Orange, New Jersey (G.L.G.); Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (S.P., L.C., X.Z.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (J.L.); Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.S., H.-J.Z.); In Vitro ADMET Laboratories, LLC, Columbia, Maryland (Q.Y., A.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland (L.L., H.W.); and Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Poznań, Poland (A.S.)
| | - Bo Kong
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (D.R., B.K., G.L.G.), Department of Pharmaceutical Sciences, Ernest Mario School of Pharmacy (A.S., C.P., L.K.), Center of Excellence for Pharmaceutical Translational Research and Education (A.S., C.P., L.K.), and Environmental and Occupational Health Sciences Institute (EOHSI) (D.R., G.L.G.), Rutgers University, Piscataway, New Jersey; Rutgers Center for Lipid Research, Rutgers University-New Brunswick, New Brunswick, New Jersey (D.R., G.L.G.); VA New Jersey Health Care System, Veterans Administration Medical Center, East Orange, New Jersey (G.L.G.); Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (S.P., L.C., X.Z.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (J.L.); Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.S., H.-J.Z.); In Vitro ADMET Laboratories, LLC, Columbia, Maryland (Q.Y., A.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland (L.L., H.W.); and Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Poznań, Poland (A.S.)
| | - Stephanie Piekos
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (D.R., B.K., G.L.G.), Department of Pharmaceutical Sciences, Ernest Mario School of Pharmacy (A.S., C.P., L.K.), Center of Excellence for Pharmaceutical Translational Research and Education (A.S., C.P., L.K.), and Environmental and Occupational Health Sciences Institute (EOHSI) (D.R., G.L.G.), Rutgers University, Piscataway, New Jersey; Rutgers Center for Lipid Research, Rutgers University-New Brunswick, New Brunswick, New Jersey (D.R., G.L.G.); VA New Jersey Health Care System, Veterans Administration Medical Center, East Orange, New Jersey (G.L.G.); Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (S.P., L.C., X.Z.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (J.L.); Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.S., H.-J.Z.); In Vitro ADMET Laboratories, LLC, Columbia, Maryland (Q.Y., A.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland (L.L., H.W.); and Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Poznań, Poland (A.S.)
| | - Liming Chen
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (D.R., B.K., G.L.G.), Department of Pharmaceutical Sciences, Ernest Mario School of Pharmacy (A.S., C.P., L.K.), Center of Excellence for Pharmaceutical Translational Research and Education (A.S., C.P., L.K.), and Environmental and Occupational Health Sciences Institute (EOHSI) (D.R., G.L.G.), Rutgers University, Piscataway, New Jersey; Rutgers Center for Lipid Research, Rutgers University-New Brunswick, New Brunswick, New Jersey (D.R., G.L.G.); VA New Jersey Health Care System, Veterans Administration Medical Center, East Orange, New Jersey (G.L.G.); Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (S.P., L.C., X.Z.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (J.L.); Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.S., H.-J.Z.); In Vitro ADMET Laboratories, LLC, Columbia, Maryland (Q.Y., A.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland (L.L., H.W.); and Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Poznań, Poland (A.S.)
| | - Xiaobo Zhong
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (D.R., B.K., G.L.G.), Department of Pharmaceutical Sciences, Ernest Mario School of Pharmacy (A.S., C.P., L.K.), Center of Excellence for Pharmaceutical Translational Research and Education (A.S., C.P., L.K.), and Environmental and Occupational Health Sciences Institute (EOHSI) (D.R., G.L.G.), Rutgers University, Piscataway, New Jersey; Rutgers Center for Lipid Research, Rutgers University-New Brunswick, New Brunswick, New Jersey (D.R., G.L.G.); VA New Jersey Health Care System, Veterans Administration Medical Center, East Orange, New Jersey (G.L.G.); Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (S.P., L.C., X.Z.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (J.L.); Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.S., H.-J.Z.); In Vitro ADMET Laboratories, LLC, Columbia, Maryland (Q.Y., A.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland (L.L., H.W.); and Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Poznań, Poland (A.S.)
| | - Jie Lu
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (D.R., B.K., G.L.G.), Department of Pharmaceutical Sciences, Ernest Mario School of Pharmacy (A.S., C.P., L.K.), Center of Excellence for Pharmaceutical Translational Research and Education (A.S., C.P., L.K.), and Environmental and Occupational Health Sciences Institute (EOHSI) (D.R., G.L.G.), Rutgers University, Piscataway, New Jersey; Rutgers Center for Lipid Research, Rutgers University-New Brunswick, New Brunswick, New Jersey (D.R., G.L.G.); VA New Jersey Health Care System, Veterans Administration Medical Center, East Orange, New Jersey (G.L.G.); Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (S.P., L.C., X.Z.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (J.L.); Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.S., H.-J.Z.); In Vitro ADMET Laboratories, LLC, Columbia, Maryland (Q.Y., A.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland (L.L., H.W.); and Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Poznań, Poland (A.S.)
| | - Jian Shi
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (D.R., B.K., G.L.G.), Department of Pharmaceutical Sciences, Ernest Mario School of Pharmacy (A.S., C.P., L.K.), Center of Excellence for Pharmaceutical Translational Research and Education (A.S., C.P., L.K.), and Environmental and Occupational Health Sciences Institute (EOHSI) (D.R., G.L.G.), Rutgers University, Piscataway, New Jersey; Rutgers Center for Lipid Research, Rutgers University-New Brunswick, New Brunswick, New Jersey (D.R., G.L.G.); VA New Jersey Health Care System, Veterans Administration Medical Center, East Orange, New Jersey (G.L.G.); Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (S.P., L.C., X.Z.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (J.L.); Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.S., H.-J.Z.); In Vitro ADMET Laboratories, LLC, Columbia, Maryland (Q.Y., A.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland (L.L., H.W.); and Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Poznań, Poland (A.S.)
| | - Hao-Jie Zhu
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (D.R., B.K., G.L.G.), Department of Pharmaceutical Sciences, Ernest Mario School of Pharmacy (A.S., C.P., L.K.), Center of Excellence for Pharmaceutical Translational Research and Education (A.S., C.P., L.K.), and Environmental and Occupational Health Sciences Institute (EOHSI) (D.R., G.L.G.), Rutgers University, Piscataway, New Jersey; Rutgers Center for Lipid Research, Rutgers University-New Brunswick, New Brunswick, New Jersey (D.R., G.L.G.); VA New Jersey Health Care System, Veterans Administration Medical Center, East Orange, New Jersey (G.L.G.); Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (S.P., L.C., X.Z.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (J.L.); Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.S., H.-J.Z.); In Vitro ADMET Laboratories, LLC, Columbia, Maryland (Q.Y., A.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland (L.L., H.W.); and Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Poznań, Poland (A.S.)
| | - Qian Yang
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (D.R., B.K., G.L.G.), Department of Pharmaceutical Sciences, Ernest Mario School of Pharmacy (A.S., C.P., L.K.), Center of Excellence for Pharmaceutical Translational Research and Education (A.S., C.P., L.K.), and Environmental and Occupational Health Sciences Institute (EOHSI) (D.R., G.L.G.), Rutgers University, Piscataway, New Jersey; Rutgers Center for Lipid Research, Rutgers University-New Brunswick, New Brunswick, New Jersey (D.R., G.L.G.); VA New Jersey Health Care System, Veterans Administration Medical Center, East Orange, New Jersey (G.L.G.); Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (S.P., L.C., X.Z.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (J.L.); Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.S., H.-J.Z.); In Vitro ADMET Laboratories, LLC, Columbia, Maryland (Q.Y., A.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland (L.L., H.W.); and Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Poznań, Poland (A.S.)
| | - Albert Li
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (D.R., B.K., G.L.G.), Department of Pharmaceutical Sciences, Ernest Mario School of Pharmacy (A.S., C.P., L.K.), Center of Excellence for Pharmaceutical Translational Research and Education (A.S., C.P., L.K.), and Environmental and Occupational Health Sciences Institute (EOHSI) (D.R., G.L.G.), Rutgers University, Piscataway, New Jersey; Rutgers Center for Lipid Research, Rutgers University-New Brunswick, New Brunswick, New Jersey (D.R., G.L.G.); VA New Jersey Health Care System, Veterans Administration Medical Center, East Orange, New Jersey (G.L.G.); Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (S.P., L.C., X.Z.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (J.L.); Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.S., H.-J.Z.); In Vitro ADMET Laboratories, LLC, Columbia, Maryland (Q.Y., A.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland (L.L., H.W.); and Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Poznań, Poland (A.S.)
| | - Linhao Li
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (D.R., B.K., G.L.G.), Department of Pharmaceutical Sciences, Ernest Mario School of Pharmacy (A.S., C.P., L.K.), Center of Excellence for Pharmaceutical Translational Research and Education (A.S., C.P., L.K.), and Environmental and Occupational Health Sciences Institute (EOHSI) (D.R., G.L.G.), Rutgers University, Piscataway, New Jersey; Rutgers Center for Lipid Research, Rutgers University-New Brunswick, New Brunswick, New Jersey (D.R., G.L.G.); VA New Jersey Health Care System, Veterans Administration Medical Center, East Orange, New Jersey (G.L.G.); Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (S.P., L.C., X.Z.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (J.L.); Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.S., H.-J.Z.); In Vitro ADMET Laboratories, LLC, Columbia, Maryland (Q.Y., A.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland (L.L., H.W.); and Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Poznań, Poland (A.S.)
| | - Hongbing Wang
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (D.R., B.K., G.L.G.), Department of Pharmaceutical Sciences, Ernest Mario School of Pharmacy (A.S., C.P., L.K.), Center of Excellence for Pharmaceutical Translational Research and Education (A.S., C.P., L.K.), and Environmental and Occupational Health Sciences Institute (EOHSI) (D.R., G.L.G.), Rutgers University, Piscataway, New Jersey; Rutgers Center for Lipid Research, Rutgers University-New Brunswick, New Brunswick, New Jersey (D.R., G.L.G.); VA New Jersey Health Care System, Veterans Administration Medical Center, East Orange, New Jersey (G.L.G.); Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (S.P., L.C., X.Z.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (J.L.); Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.S., H.-J.Z.); In Vitro ADMET Laboratories, LLC, Columbia, Maryland (Q.Y., A.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland (L.L., H.W.); and Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Poznań, Poland (A.S.)
| | - Anna Siemiątkowska
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (D.R., B.K., G.L.G.), Department of Pharmaceutical Sciences, Ernest Mario School of Pharmacy (A.S., C.P., L.K.), Center of Excellence for Pharmaceutical Translational Research and Education (A.S., C.P., L.K.), and Environmental and Occupational Health Sciences Institute (EOHSI) (D.R., G.L.G.), Rutgers University, Piscataway, New Jersey; Rutgers Center for Lipid Research, Rutgers University-New Brunswick, New Brunswick, New Jersey (D.R., G.L.G.); VA New Jersey Health Care System, Veterans Administration Medical Center, East Orange, New Jersey (G.L.G.); Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (S.P., L.C., X.Z.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (J.L.); Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.S., H.-J.Z.); In Vitro ADMET Laboratories, LLC, Columbia, Maryland (Q.Y., A.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland (L.L., H.W.); and Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Poznań, Poland (A.S.)
| | - Celine Park
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (D.R., B.K., G.L.G.), Department of Pharmaceutical Sciences, Ernest Mario School of Pharmacy (A.S., C.P., L.K.), Center of Excellence for Pharmaceutical Translational Research and Education (A.S., C.P., L.K.), and Environmental and Occupational Health Sciences Institute (EOHSI) (D.R., G.L.G.), Rutgers University, Piscataway, New Jersey; Rutgers Center for Lipid Research, Rutgers University-New Brunswick, New Brunswick, New Jersey (D.R., G.L.G.); VA New Jersey Health Care System, Veterans Administration Medical Center, East Orange, New Jersey (G.L.G.); Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (S.P., L.C., X.Z.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (J.L.); Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.S., H.-J.Z.); In Vitro ADMET Laboratories, LLC, Columbia, Maryland (Q.Y., A.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland (L.L., H.W.); and Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Poznań, Poland (A.S.)
| | - Leonid Kagan
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (D.R., B.K., G.L.G.), Department of Pharmaceutical Sciences, Ernest Mario School of Pharmacy (A.S., C.P., L.K.), Center of Excellence for Pharmaceutical Translational Research and Education (A.S., C.P., L.K.), and Environmental and Occupational Health Sciences Institute (EOHSI) (D.R., G.L.G.), Rutgers University, Piscataway, New Jersey; Rutgers Center for Lipid Research, Rutgers University-New Brunswick, New Brunswick, New Jersey (D.R., G.L.G.); VA New Jersey Health Care System, Veterans Administration Medical Center, East Orange, New Jersey (G.L.G.); Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (S.P., L.C., X.Z.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (J.L.); Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.S., H.-J.Z.); In Vitro ADMET Laboratories, LLC, Columbia, Maryland (Q.Y., A.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland (L.L., H.W.); and Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Poznań, Poland (A.S.)
| | - Grace L Guo
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy (D.R., B.K., G.L.G.), Department of Pharmaceutical Sciences, Ernest Mario School of Pharmacy (A.S., C.P., L.K.), Center of Excellence for Pharmaceutical Translational Research and Education (A.S., C.P., L.K.), and Environmental and Occupational Health Sciences Institute (EOHSI) (D.R., G.L.G.), Rutgers University, Piscataway, New Jersey; Rutgers Center for Lipid Research, Rutgers University-New Brunswick, New Brunswick, New Jersey (D.R., G.L.G.); VA New Jersey Health Care System, Veterans Administration Medical Center, East Orange, New Jersey (G.L.G.); Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut (S.P., L.C., X.Z.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania (J.L.); Department of Clinical Pharmacy, College of Pharmacy, University of Michigan, Ann Arbor, Michigan (J.S., H.-J.Z.); In Vitro ADMET Laboratories, LLC, Columbia, Maryland (Q.Y., A.L.); Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland (L.L., H.W.); and Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Poznań, Poland (A.S.)
| |
Collapse
|
4
|
Lefebvre P, Staels B. Hepatic sexual dimorphism - implications for non-alcoholic fatty liver disease. Nat Rev Endocrinol 2021; 17:662-670. [PMID: 34417588 DOI: 10.1038/s41574-021-00538-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/06/2021] [Indexed: 12/14/2022]
Abstract
The liver is often thought of as a single functional unit, but both its structural and functional architecture make it highly multivalent and adaptable. In any given physiological situation, the liver can maintain metabolic homeostasis, conduct appropriate inflammatory responses, carry out endobiotic and xenobiotic transformation and synthesis reactions, as well as store and release multiple bioactive molecules. Moreover, the liver is a very resilient organ. This resilience means that chronic liver diseases can go unnoticed for decades, yet culminate in life-threatening clinical complications once the adaptive capacity of the liver is overwhelmed. Non-alcoholic fatty liver disease (NAFLD) predisposes individuals to cirrhosis and increases liver-related and cardiovascular disease-related mortality. This Review discusses the accumulating evidence of sexual dimorphism in NAFLD, which is currently rarely considered in preclinical and clinical studies. Increased awareness of the mechanistic causes of hepatic sexual dimorphism could lead to improved understanding of the biological processes that are dysregulated in NAFLD, to the identification of relevant therapeutic targets and to improved risk stratification of patients with NAFLD undergoing therapeutic intervention.
Collapse
Affiliation(s)
- Philippe Lefebvre
- Université Lille, INSERM, CHU Lille, Institut Pasteur de Lille, Lille, France.
| | - Bart Staels
- Université Lille, INSERM, CHU Lille, Institut Pasteur de Lille, Lille, France
| |
Collapse
|
5
|
Expression and Polymorphism of TSLP/TSLP Receptors as Potential Diagnostic Markers of Colorectal Cancer Progression. Genes (Basel) 2021; 12:genes12091386. [PMID: 34573368 PMCID: PMC8469613 DOI: 10.3390/genes12091386] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/25/2021] [Accepted: 08/29/2021] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common malignancy and the fourth leading cause of cancer-related mortality worldwide. Inflammation is considered as a critical driver for CRC development and growth. We investigated the association between polymorphisms/expression levels of thymic stromal lymphopoietin (TSLP) /TSLP receptors and CRC risk in Saudi population. DNA samples were isolated from blood samples from 220 participants. Case subjects were 112 patients diagnosed with CRC, while control subjects were 108 healthy individuals, who were not diagnosed with any type of malignancy. We selected two single nucleotide polymorphisms (SNPs) located in the thymic stromal lymphopoietin gene (rs10043985 and rs2289276), three SNPs in TSLP receptor gene (TSLPR; rs36139698, rs36177645, and rs36133495), and two other SNPs in interleukin-7 receptor gene (IL-7R; rs12516866 and rs1053496), and designated these SNPs for a case-control genotyping study. The gene expression was analyzed using quantitative RT-PCR and immunohistochemistry assays array on 20 matching colorectal cancer/normal tissues. mRNA expressions and protein levels of TSLP, TSLPR-α subunit, and IL-7R-α subunit showed a 4-fold increase in colon cancer tissues when compared to normal colon tissues. Furthermore, two SNPs (rs10043985 of TSLP and rs1053496 of IL-7R) showed statistically significant correlations with CRC susceptibility. Interestingly, only rs10043985 showed a statistically significant association (p < 0.0001) in the genotypic and phenotypic levels with CRC for all clinical parameters (age, gender, and tumor location) tested. However, IL-7R rs1053496 genotyping results presented a significant correlation (p < 0.05) in male CRC patients and in individuals under 57 years of age. TSLP rs2289276, IL-7R rs12516866, and all TSLPR variants did not display any significant genotypic or phenotypic correlations in all tested clinical parameters. This study identified that TSLP rs10043985 and IL-7R rs1053496 SNPs, and the expression levels of TSLP and TSLPR-α subunit, can be used as markers for CRC development and treatment. However, additional investigations are required on larger group of patients from diverse ethnicities to confirm the genetic association of these variants to CRC.
Collapse
|
6
|
O’Brien MH, Pitot HC, Chung SH, Lambert PF, Drinkwater NR, Bilger A. Estrogen Receptor-α Suppresses Liver Carcinogenesis and Establishes Sex-Specific Gene Expression. Cancers (Basel) 2021; 13:2355. [PMID: 34068249 PMCID: PMC8153146 DOI: 10.3390/cancers13102355] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 05/10/2021] [Indexed: 02/06/2023] Open
Abstract
Estrogen protects females from hepatocellular carcinoma (HCC). To determine whether this protection is mediated by classic estrogen receptors, we tested HCC susceptibility in estrogen receptor-deficient mice. In contrast to a previous study, we found that diethylnitrosamine induces hepatocarcinogenesis to a significantly greater extent when females lack Esr1, which encodes Estrogen Receptor-α. Relative to wild-type littermates, Esr1 knockout females developed 9-fold more tumors. Deficiency of Esr2, which encodes Estrogen Receptor-β, did not affect liver carcinogenesis in females. Using microarrays and QPCR to examine estrogen receptor effects on hepatic gene expression patterns, we found that germline Esr1 deficiency resulted in the masculinization of gene expression in the female liver. Six of the most dysregulated genes have previously been implicated in HCC. In contrast, Esr1 deletion specifically in hepatocytes of Esr1 conditional null female mice (in which Cre was expressed from the albumin promoter) resulted in the maintenance of female-specific liver gene expression. Wild-type adult females lacking ovarian estrogen due to ovariectomy, which is known to make females susceptible to HCC, also maintained female-specific expression in the liver of females. These studies indicate that Esr1 mediates liver cancer risk, and its control of sex-specific liver gene expression involves cells other than hepatocytes.
Collapse
Affiliation(s)
- Mara H. O’Brien
- Department of Craniofacial Sciences, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030, USA;
| | - Henry C. Pitot
- McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin—Madison, 1111 Highland Ave, Madison, WI 53705, USA; (H.C.P.); (P.F.L.); (N.R.D.)
| | - Sang-Hyuk Chung
- Center for Nuclear Receptors and Cell Signaling, Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA;
| | - Paul F. Lambert
- McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin—Madison, 1111 Highland Ave, Madison, WI 53705, USA; (H.C.P.); (P.F.L.); (N.R.D.)
| | - Norman R. Drinkwater
- McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin—Madison, 1111 Highland Ave, Madison, WI 53705, USA; (H.C.P.); (P.F.L.); (N.R.D.)
| | - Andrea Bilger
- McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin—Madison, 1111 Highland Ave, Madison, WI 53705, USA; (H.C.P.); (P.F.L.); (N.R.D.)
| |
Collapse
|
7
|
Sayad A, Taheri M, Azari I, Oskoei VK, Ghafouri-Fard S. PIAS genes as disease markers in bipolar disorder. J Cell Biochem 2019; 120:12937-12942. [PMID: 30861611 DOI: 10.1002/jcb.28564] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/16/2018] [Accepted: 01/10/2019] [Indexed: 11/11/2022]
Abstract
The protein inhibitors of activated STAT (PIAS) are involved in regulation of many transcription factors and signaling pathways that contribute to the pathogenesis of bipolar disease (BD). In the current study, we evaluated the expression of four PIAS genes (PIAS1-4) in peripheral blood of BD patients and healthy subjects to explore their contribution in the pathogenesis of BD and their suitability as peripheral biomarkers for this disorder. All PIAS genes were significantly upregulated in total BD patients compared with total controls. The sex-based analysis confirmed upregulation of PIAS1-4 genes in male BD patients compared with male controls (P < 0.001). However, PIAS1 was significantly downregulated in female patients compared with female controls (P = 0.02). Expression levels of other PIAS genes were not significantly different between female patients and female controls. There were no significant correlations between expression levels of PIAS genes and any of the clinical data of study participants after adjustment of the effects of the sex. On the basis of the area under the curve (AUC) values in receiver operating characteristic curves, PIAS4 had the best performance in the differentiation of disease status between study participants (AUC = 0.81). PIAS3 and PIAS4 genes had the best sensitivity and specificity values, respectively. Combination of expression levels of four genes resulted in the improvement of diagnostic power (AUC = 0.82). The current data implies the role of PIAS genes in the pathogenesis of BD and denotes their suitability as peripheral markers for this disorder.
Collapse
Affiliation(s)
- Arezou Sayad
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Iman Azari
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Kholghi Oskoei
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
STAT5 expression correlates with recurrence and survival in melanoma patients treated with interferon-α. Melanoma Res 2019; 28:204-210. [PMID: 29485532 DOI: 10.1097/cmr.0000000000000435] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Interferons (IFN) have a direct growth-inhibiting effect on tumor cells through Janus kinase-dependent activation of the transcription factor signal transducer and activator of transcription (STAT1). In vitro, signaling through STAT5 has been demonstrated to counteract this effect and lead to IFN resistance of melanoma cell lines. In 32 patients treated with IFN-α in an adjuvant setting, we investigated paraffin-embedded tumor tissue from primary melanomas and melanoma metastases for expression of STAT3 and STAT5, by immunohistochemistry, and for expression of phosphorylated signaling transduction activating transcription factor (pSTAT)3 and pSTAT5, by immunofluorescence. Tumor cell expression levels of these proteins were correlated with patient characteristics and clinical outcomes. The patient cohort consisted of 12 (37.5%) patients at AJCC stage I/II (primary melanoma) and 20 (62.5%) at stage III/IV (metastatic melanoma). Recurrence was observed for 25 (78.1%) either during or after IFN-α therapy. χ Correlation of staining intensities with clinical data revealed association of pSTAT3 and STAT5 expression with sex (P=0.003 and 0.016, respectively) and of STAT3 with tumor stage (P=0.019). Recurrence of melanoma was found to be associated with high STAT5 expression (P=0.017). Multivariable regression analysis revealed STAT5 expression as an independent factor for predicting progression-free survival (P<0.0001) and overall survival (P=0.022). In summary, high expression of STAT5 correlated with melanoma recurrence and survival of patients treated with IFN-α in the adjuvant setting. Recently, it has been suggested that mutations of Janus kinases are involved in resistance to immune checkpoint blocker treatments implying a possible role of STAT5 for immune checkpoint resistance.
Collapse
|
9
|
Zhang Y, Lickteig AJ, Csanaky IL, Klaassen CD. Activation of PPARα decreases bile acids in livers of female mice while maintaining bile flow and biliary bile acid excretion. Toxicol Appl Pharmacol 2017; 338:112-123. [PMID: 29175453 DOI: 10.1016/j.taap.2017.11.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 11/16/2017] [Accepted: 11/19/2017] [Indexed: 01/20/2023]
Abstract
Fibrates are hypolipidemic drugs that act as activators of peroxisome proliferator-activated receptor α (PPARα). In both humans and rodents, females were reported to be less responsive to fibrates than males. Previous studies on fibrates and PPARα usually involved male mice, but little has been done in females. The present study aimed to provide the first comprehensive analysis of the effects of clofibrate (CLOF) and PPARα on bile acid (BA) homeostasis in female mice. Study in WT male mice showed that a 4-day CLOF treatment increased liver weight, bile flow, and biliary BA excretion, but decreased total BAs in both serum and liver. In contrast, WT female mice were less susceptible to these CLOF-mediated responses observed in males. In WT female mice, CLOF decreased total BAs in the liver, but had little effect on the mRNAs of hepatic BA-related genes. Next, a comparative analysis between WT and PPARα-null female mice showed that lack of PPARα in female mice decreased total BAs in serum, but had little effect on total BAs in liver or bile. However, lack of PPARα in female mice increased mRNAs of BA synthetic enzymes (Cyp7a1, Cyp8b1, Cyp27a1, and Cyp7b1) and transporters (Ntcp, Oatp1a1, Oatp1b2, and Mrp3). Furthermore, the increase of Cyp7a1 in PPARα-null female mice was associated with an increase in liver Fxr-Shp-Lrh-1 signaling. In conclusion, female mice are resistant to CLOF-mediated effects on BA metabolism observed in males, which could be attributed to PPARα-mediated suppression in females on genes involved in BA synthesis and transport.
Collapse
Affiliation(s)
- Youcai Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, PR China.
| | - Andrew J Lickteig
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160,USA
| | - Iván L Csanaky
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Division of Gastroenterology, Children's Mercy Hospital & Clinics, Kansas City, MO 64108; USA; Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Curtis D Klaassen
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160,USA.
| |
Collapse
|
10
|
STAT5 Activation in the Dermal Papilla Is Important for Hair Follicle Growth Phase Induction. J Invest Dermatol 2016; 136:1781-1791. [DOI: 10.1016/j.jid.2016.04.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 03/31/2016] [Accepted: 04/08/2016] [Indexed: 01/06/2023]
|
11
|
Impaired phosphorylation of JAK2-STAT5b signaling in fibroblasts from uremic children. Pediatr Nephrol 2016; 31:965-74. [PMID: 26747624 DOI: 10.1007/s00467-015-3289-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 11/20/2015] [Accepted: 12/01/2015] [Indexed: 12/28/2022]
Abstract
BACKGROUND Chronic kidney disease (CKD) in children is characterized by severe growth failure. The growth hormone/insulin-like growth factor-1 (GH/IGF-1) axis in uremic animals shows a post-receptor impaired phosphorylation of Janus kinase 2/signal transducer and activator of transcription (JAK-STAT) proteins. The objective of our study was to characterize the intracellular phosphorylation of JAK-STAT signaling in fibroblasts from children with CKD on chronic peritoneal dialysis (PD). METHODS Serum GH-binding protein (GHBP), IGF-1 and IGFBP3 were measured in 15 prepubertal CKD stage-5 children on PD. Cytoplasmic JAK2, cytoplasmic/nuclear STAT5b and nuclear IGFBP3, acid-labile subunit (ALS) and IGF-1 mRNA expression were quantified in fibroblasts obtained from skin biopsies before and after stimulation with 200 ng/ml recombinant human growth hormone (rhGH). Phosphorylation activity at both the cytoplasmic and nuclear level was expressed as the ratio phosphorylated (p)/total (t) abundance of the product (p/t) at 30 and 60 min. Fifteen healthy children were recruited as the control group. Values were expressed in arbitrary units (AU) and normalized for comparison. Significance was defined as p < 0.05. RESULTS Thirty minutes after rhGH stimulus, the cytoplasmic (p/t) JAK2 ratio was significantly lower in patients than in controls [median and interquartile range (IQR): 7.4 (4.56) vs. 20.5 (50.06) AU]. At 60 min after rhGH stimulation, median JAK2 phosphorylation activity was still significantly lower in the patients [7.14 (IQR 3.8) vs. 10.2 (IQR 29.8) AU; p < 0.05]. The increase in the cytoplasmic (p/t) STAT5b/β-actin ratio was lower at both measurement points in the patients compared to the controls, without reaching statistical significance between groups. Median IGFBP3 mRNA abundance was significantly decreased in fibroblasts from uremic patients 24 h after rhGH stimulation compared to the healthy controls [1.27 (IQR 0.83) vs. 2.37 (IQR 0.80) AU]. Median ALS and IGF-1 mRNA expression changed in response to rhGH stimuli at 24 and 48 h. CONCLUSION In this study, children with CKD undergoing PD therapy showed an impaired phosphorylation of JAK2/STAT5b signaling in fibroblasts after GH stimulation, as well as impaired IGFBP3 mRNA abundance. Both impairments may be partially responsible for the observed resistance to the growth-promoting actions of GH in chronic kidney failure.
Collapse
|
12
|
Schmölz L, Birringer M, Lorkowski S, Wallert M. Complexity of vitamin E metabolism. World J Biol Chem 2016; 7:14-43. [PMID: 26981194 PMCID: PMC4768118 DOI: 10.4331/wjbc.v7.i1.14] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 11/25/2015] [Accepted: 01/19/2016] [Indexed: 02/05/2023] Open
Abstract
Bioavailability of vitamin E is influenced by several factors, most are highlighted in this review. While gender, age and genetic constitution influence vitamin E bioavailability but cannot be modified, life-style and intake of vitamin E can be. Numerous factors must be taken into account however, i.e., when vitamin E is orally administrated, the food matrix may contain competing nutrients. The complex metabolic processes comprise intestinal absorption, vascular transport, hepatic sorting by intracellular binding proteins, such as the significant α-tocopherol-transfer protein, and hepatic metabolism. The coordinated changes involved in the hepatic metabolism of vitamin E provide an effective physiological pathway to protect tissues against the excessive accumulation of, in particular, non-α-tocopherol forms. Metabolism of vitamin E begins with one cycle of CYP4F2/CYP3A4-dependent ω-hydroxylation followed by five cycles of subsequent β-oxidation, and forms the water-soluble end-product carboxyethylhydroxychroman. All known hepatic metabolites can be conjugated and are excreted, depending on the length of their side-chain, either via urine or feces. The physiological handling of vitamin E underlies kinetics which vary between the different vitamin E forms. Here, saturation of the side-chain and also substitution of the chromanol ring system are important. Most of the metabolic reactions and processes that are involved with vitamin E are also shared by other fat soluble vitamins. Influencing interactions with other nutrients such as vitamin K or pharmaceuticals are also covered by this review. All these processes modulate the formation of vitamin E metabolites and their concentrations in tissues and body fluids. Differences in metabolism might be responsible for the discrepancies that have been observed in studies performed in vivo and in vitro using vitamin E as a supplement or nutrient. To evaluate individual vitamin E status, the analytical procedures used for detecting and quantifying vitamin E and its metabolites are crucial. The latest methods in analytics are presented.
Collapse
|
13
|
Creasy KT, Jiang J, Ren H, Peterson ML, Spear BT. Zinc Fingers and Homeoboxes 2 (Zhx2) Regulates Sexually Dimorphic Cyp Gene Expression in the Adult Mouse Liver. Gene Expr 2016; 17:7-17. [PMID: 27197076 PMCID: PMC5518317 DOI: 10.3727/105221616x691712] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The mammalian cytochrome P450 (Cyp) gene family encodes a large number of structurally related enzymes that catalyze a variety of metabolic and detoxification reactions. The liver is the primary site of Cyp expression in terms of expression levels and number of expressed genes, consistent with this organ's essential role in metabolism of endogenous and xenobiotic compounds. Many Cyp genes exhibit sexually dimorphic expression. For example, Cyp2a4 is expressed significantly higher in the adult liver of female mice compared to male mice. An exception to this pattern is seen in BALB/cJ mice, where male hepatic Cyp2a4 mRNA levels are substantially elevated compared to male mice of other strains. The Zinc fingers and homeoboxes 2 (Zhx2) protein governs the silencing of several genes in the postnatal liver, including α-fetoprotein, H19, and glypican 3. Zhx2 also regulates numerous hepatic genes that govern lipid homeostasis. We previously showed that the Zhx2 gene is mutated in BALB/cJ mice, which led us to consider whether elevated male hepatic Cyp2a4 levels in this strain are due to this Zhx2 mutation. Using mice with a conditional Zhx2 deletion, we show here that the absence of Zhx2 in hepatocytes results in increased Cyp2a4 expression in adult male liver. We extend this finding to show that additional Cyp genes are disregulated in the absence of Zhx2. We also show that mRNA levels of Cyp2a4 and several other female-biased Cyp genes are increased, and male-biased Cyp4a12 is decreased in mouse liver tumors. These data indicate that Zhx2 is a novel regulator of sex-biased Cyp gene expression in the normal and diseased liver.
Collapse
Affiliation(s)
- Kate Townsend Creasy
- *Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Jieyun Jiang
- †Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, USA
| | - Hui Ren
- †Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, USA
| | - Martha L. Peterson
- †Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, USA
- ‡Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| | - Brett T. Spear
- †Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, USA
- ‡Markey Cancer Center, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
14
|
Veldhuis JD, Bowers CY. Factors other than sex steroids modulate GHRH and GHRP-2 efficacies in men: evaluation using a GnRH agonist/testosterone clamp. J Clin Endocrinol Metab 2009; 94:2544-50. [PMID: 19351731 PMCID: PMC2708950 DOI: 10.1210/jc.2008-2767] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Sex steroids are prominent regulators of pulsatile GH secretion. HYPOTHESIS An experimentally controlled sex-steroid milieu will permit detection of nonsteroidal factors that determine GH secretion. SUBJECTS Eleven young (age, 24 +/- 0.99 yr) and 11 older (64 +/- 2.4 yr) men participated in the study. LOCATION The study was conducted at a tertiary medical center. METHODS The study consisted of GnRH-agonist down-regulation of the gonadal axis followed by fixed-dose testosterone (T) replacement (leuprolide/T clamp) and consecutive infusion of l-arginine and GHRH or GH-releasing peptide-2 (GHRP-2) to quantify peptide-secretagogue efficacies. OUTCOMES The experimental leuprolide/T clamp yielded statistically age-comparable total, bioavailable, and free T and estradiol (E(2)) concentrations. In this controlled milieu, sequential l-arginine/GHRH infusion stimulated 1.4-fold more (P = 0.021) and l-arginine/GHRP-2 1.3-fold more (P = 0.045) GH release in young than older men. Abdominal visceral fat (AVF) correlated negatively with both GHRH (P = 0.0006; R(2) = 0.39) and GHRP-2 (R(2) = 0.29) efficacy, whereas IGF-I positively predicted the same endpoints (R(2) = 0.25 to 0.30). In multivariate analysis, AVF emerged as a dominant negative determinant of GHRH efficacy (P = 0.002; R(2) = 0.41) and IGF-I as a primary positive determinant of GHRP-2 efficacy (P = 0.007; R(2) = 0.31). CONCLUSION During fixed T/E(2) availability, AVF contributes 41% of the GH-response variability to maximal GHRH drive, whereas IGF-I accounts for 31% of that for GHRP-2. Accordingly, a statistically equalized sex-steroid milieu permits dissection of age-independent and T/E(2)-independent modulators of GHRH and GHRP efficacy in men.
Collapse
Affiliation(s)
- Johannes D Veldhuis
- Department of Medicine, Endocrine Research Unit, Mayo School of Graduate Medical Education, Clinical Translational Science Center, Mayo Clinic, Rochester, Minnesota 55905, USA.
| | | |
Collapse
|
15
|
Veldhuis JD, Keenan DM, Bailey JN, Adeniji AM, Miles JM, Bowers CY. Novel relationships of age, visceral adiposity, insulin-like growth factor (IGF)-I and IGF binding protein concentrations to growth hormone (GH) releasing-hormone and GH releasing-peptide efficacies in men during experimental hypogonadal clamp. J Clin Endocrinol Metab 2009; 94:2137-43. [PMID: 19351723 PMCID: PMC2690428 DOI: 10.1210/jc.2009-0136] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Sex steroids influence GH secretion in complex ways. HYPOTHESIS Analyses in a low sex-steroid milieu will help unveil the effects of age and other nonsteroidal regulators on GH secretion. CONTEXT The study was conducted in a tertiary medical center. SUBJECTS The study group included 13 healthy young men and 12 healthy older men. METHODS We used GnRH agonist-induced down-regulation of testosterone and estradiol secretion, followed by consecutive infusion of l-arginine and GHRH or GHRP-2, to test secretagogue efficacies. OUTCOMES We measured basal and pulsatile GH secretion. RESULTS During experimental testosterone/estradiol deprivation, older (57 +/- 1.7 yr) men maintained: 1) 6.8-fold less pulsatile GH secretion (P < 0.001); and 2) 2-fold lower maximal GH responses to GHRH (P = 0.0065) and GHRP-2 (P = 0.022) than young (23 +/- 1.1 yr old) individuals. Stepwise forward-selection regression analyses identified: 1) abdominal visceral fat as a dominant negative predictor of both GHRH (R(2) = 0.49; P = 0.001) and GHRP-2 (R(2) = 0.38; P = 0.005) efficacies; and 2) fasting IGF-I concentration as a major positive correlate of GHRH (R(2) = 0.52; P < 0.001) and GHRP-2 (R(2) = 0.31; P = 0.018) efficacies. Unstimulated pulsatile GH secretion was jointly correlated with IGF-I and IGFBP-3 (P = 0.039). CONCLUSION Measures of body composition (abdominal visceral fat) and pulsatile GH action (IGF-I) explain up to one half of interindividual variability in the efficacies of GHRH and GHRP-2 in sex steroid-depleted men. Accordingly, normative ranges for maximal single peptide-stimulated GH secretion in short-term hypogonadal states should incorporate the influence of these determinants as well as age.
Collapse
Affiliation(s)
- Johannes D Veldhuis
- Endocrine Research Unit, Mayo School of Graduate Medical Education, Clinical Translational Science Center, Mayo Clinic, Rochester, Minnesota 55905, USA.
| | | | | | | | | | | |
Collapse
|
16
|
Brunet-Dunand SE, Vouyovitch C, Araneda S, Pandey V, Vidal LJP, Print C, Mertani HC, Lobie PE, Perry JK. Autocrine human growth hormone promotes tumor angiogenesis in mammary carcinoma. Endocrinology 2009; 150:1341-52. [PMID: 18974274 DOI: 10.1210/en.2008-0608] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Accumulating literature implicates pathological angiogenesis and lymphangiogenesis as playing key roles in tumor progression. Autocrine human growth hormone (hGH) is a wild-type orthotopically expressed oncogene for the human mammary epithelial cell. Herein we demonstrate that autocrine hGH expression in the human mammary carcinoma cell line MCF-7 stimulated the survival, proliferation, migration, and invasion of a human microvascular endothelial cell line (HMEC-1). Autocrine/paracrine hGH secreted from mammary carcinoma cells also promoted HMEC-1 in vitro tube formation as a consequence of increased vascular endothelial growth factor-A (VEGF-A) expression. Semiquantitative RT-PCR analysis demonstrated that HMEC-1 cells express both hGH and the hGH receptor (hGHR). Functional antagonism of HMEC-1-derived hGH reduced HMEC-1 survival, proliferation, migration/invasion, and tube formation in vitro. Autocrine/paracrine hGH secreted by mammary carcinoma cells increased tumor blood and lymphatic microvessel density in a xenograft model of human mammary carcinoma. Autocrine hGH is therefore a potential master regulator of tumor neovascularization, coordinating two critical processes in mammary neoplastic progression, angiogenesis and lymphangiogenesis. Consideration of hGH antagonism to inhibit angiogenic processes in mammary carcinoma is therefore warranted.
Collapse
|
17
|
Oldham JM, Osepchook CC, Jeanplong F, Falconer SJ, Matthews KG, Conaglen JV, Gerrard DF, Smith HK, Wilkins RJ, Bass JJ, McMahon CD. The decrease in mature myostatin protein in male skeletal muscle is developmentally regulated by growth hormone. J Physiol 2008; 587:669-77. [PMID: 19047209 DOI: 10.1113/jphysiol.2008.161521] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Myostatin inhibits myogenesis and there is reduced abundance of the mature protein in skeletal muscles of adult male compared with female mice. This reduction probably occurs after translation, which suggests that it is a regulated mechanism to reduce the availability of myostatin in males. Reduced myostatin may, thereby, contribute to the development of sexually dimorphic growth of skeletal muscle. Our first objective was to determine if the decrease in mature myostatin protein occurs before the linear growth phase to aid growth, or afterwards to maintain the mass of adult muscle. Mice were killed from 2 to 32 weeks and the gastrocnemius muscle was excised. Myostatin mRNA increased from 2 to 32 weeks and was higher in males than females (P < 0.001). In contrast, mature protein decreased in males after 6 weeks (P < 0.001). Our second objective was to determine if growth hormone (GH) induces the decrease in mature myostatin protein. GH increased myostatin mRNA and decreased the abundance of mature protein in hypophysectomised mice (P < 0.05). Our final objective was to determine if the decrease in mature protein occurs in skeletal muscles of male Stat5b(-/-) mice (Stat5b mediates the actions of GH). As expected, mature myostatin protein was not reduced in Stat5b(-/-) males compared with females. However, myostatin mRNA remained higher in males than females irrespective of genotype. These data suggest that: (1) the decrease in mature myostatin protein is developmentally regulated, (2) GH acting via Stat5b regulates the abundance of mature myostatin and (3) GH acts via a non-Stat5b pathway to regulate myostatin mRNA.
Collapse
Affiliation(s)
- Jenny M Oldham
- Growth Physiology Group, AgResearch Ltd., Hamilton, New Zealand
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Wong JH, Dukes J, Levy RE, Sos B, Mason SE, Fong TS, Weiss EJ. Sex differences in thrombosis in mice are mediated by sex-specific growth hormone secretion patterns. J Clin Invest 2008; 118:2969-78. [PMID: 18618017 DOI: 10.1172/jci34957] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Accepted: 05/27/2008] [Indexed: 11/17/2022] Open
Abstract
Sex differences in thrombosis are well described, but their underlying mechanism(s) are not completely understood. Coagulation proteins are synthesized in the liver, and liver gene expression is sex specific and depends on sex differences in growth hormone (GH) secretion--males secrete GH in a pulsatile fashion, while females secrete GH continuously. Accordingly, we tested the hypothesis that sex-specific GH secretion patterns cause sex differences in thrombosis. Male mice were more susceptible to thrombosis than females in the thromboplastin-induced pulmonary embolism model and showed shorter clotting times ex vivo. GH-deficient little (lit) mice were protected from thrombosis, and pulsatile GH given to lit mice restored the male clotting phenotype. Moreover, pulsatile GH administration resulted in a male clotting phenotype in control female mice, while continuous GH caused a female clotting phenotype in control male mice. Expression of the coagulation inhibitors Proc, Serpinc1, Serpind1, and Serpina5 were strongly modulated by sex-specific GH patterns, and GH modulated resistance to activated protein C. These results reveal what we believe to be a novel mechanism whereby sex-specific GH patterns mediate sex differences in thrombosis through coordinated changes in the expression of coagulation inhibitor genes in the liver.
Collapse
Affiliation(s)
- Joshua H Wong
- Cardiovascular Research Institute, Department of Medicine, Division of Cardiology, UCSF, San Francisco, California 94143-0124, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Perry JK, Mohankumar KM, Emerald BS, Mertani HC, Lobie PE. The contribution of growth hormone to mammary neoplasia. J Mammary Gland Biol Neoplasia 2008; 13:131-45. [PMID: 18253708 PMCID: PMC2665193 DOI: 10.1007/s10911-008-9070-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2007] [Accepted: 01/02/2008] [Indexed: 12/13/2022] Open
Abstract
While the effects of growth hormone (GH) on longitudinal growth are well established, the observation that GH contributes to neoplastic progression is more recent. Accumulating literature implicates GH-mediated signal transduction in the development and progression of a wide range malignancies including breast cancer. Recently autocrine human GH been demonstrated to be an orthotopically expressed oncogene for the human mammary gland. This review will highlight recent evidence linking GH and mammary carcinoma and discuss GH-antagonism as a potential therapeutic approach for treatment of breast cancer.
Collapse
Affiliation(s)
- Jo K Perry
- The Liggins Institute and the National Research Centre for Growth and Development
University of Auckland2-6 Park Avenue, Grafton, Private Bag 92019, Auckland 1023,NZ
| | - Kumarasamypet M Mohankumar
- The Liggins Institute and the National Research Centre for Growth and Development
University of Auckland2-6 Park Avenue, Grafton, Private Bag 92019, Auckland 1023,NZ
| | - B Starling Emerald
- The Liggins Institute and the National Research Centre for Growth and Development
University of Auckland2-6 Park Avenue, Grafton, Private Bag 92019, Auckland 1023,NZ
| | - Hichem C Mertani
- PICM, Physiologie intégrative, cellulaire et moléculaire
CNRS : UMR5123Université Claude Bernard - Lyon IBât. R. Dubois
43, Bvd du 11 Novembre 1918
69622 VILLEURBANNE CEDEX,FR
| | - Peter E Lobie
- The Liggins Institute and the National Research Centre for Growth and Development
University of Auckland2-6 Park Avenue, Grafton, Private Bag 92019, Auckland 1023,NZ
- Department of Molecular Medicine and Pathology
University of AucklandFaculty of Medical and Health Sciences, Private Bag 92019, Auckland, New Zealand,NZ
- * Correspondence should be adressed to: Peter E Lobie
| |
Collapse
|
20
|
Haus E. Chronobiology in the endocrine system. Adv Drug Deliv Rev 2007; 59:985-1014. [PMID: 17804113 DOI: 10.1016/j.addr.2007.01.001] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2006] [Accepted: 01/15/2007] [Indexed: 12/13/2022]
Abstract
Biological signaling occurs in a complex web with participation and interaction of the central nervous system, the autonomous nervous system, the endocrine glands, peripheral endocrine tissues including the intestinal tract and adipose tissue, and the immune system. All of these show an intricate time structure with rhythms and pulsatile variations in multiple frequencies. Circadian (about 24-hour) and circannual (about 1-year) rhythms are kept in step with the cyclic environmental surrounding by the timing and length of the daily light span. Rhythmicity of many endocrine variables is essential for their efficacy and, even in some instances, for the qualitative nature of their effects. Indeed, the continuous administration of certain hormones and their synthetic analogues may show substantially different effects than expected. In the design of drug-delivery systems and treatment schedules involving directly or indirectly the endocrine system, consideration of the human time organization is essential. A large amount of information on the endocrine time structure has accumulated, some of which is discussed in this review.
Collapse
Affiliation(s)
- Erhard Haus
- Department of Laboratory Medicine and Pathology, University of Minnesota, Health Partners Medical Group, Regions Hospital, 640 Jackson Street, St. Paul, Minnesota 55101, USA.
| |
Collapse
|
21
|
Han X, Osuntokun B, Benight N, Loesch K, Frank SJ, Denson LA. Signal transducer and activator of transcription 5b promotes mucosal tolerance in pediatric Crohn's disease and murine colitis. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 169:1999-2013. [PMID: 17148664 PMCID: PMC1762482 DOI: 10.2353/ajpath.2006.060186] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Growth hormone (GH) regulates anabolic metabolism via activation of the STAT5b transcription factor and reduces mucosal inflammation in colitis. Peroxisome proliferator-activated receptor (PPAR) gamma suppresses mucosal inflammation and is regulated by GH through STAT5b. We hypothesized that the GH:STAT5b axis influences susceptibility to colitis via regulation of local PPARgamma abundance. Colon biopsies from children with newly diagnosed Crohn's disease (CD) and controls were exposed to GH in short-term organ culture. Trinitrobenzene sulfonic acid (TNBS) administration was used to induce colitis in STAT5b-deficient mice and wild-type controls, with and without rosiglitazone pretreatment. GH receptor, STAT5b, PPARgamma, and nuclear factor kappaB activation and expression were determined. Epithelial cell GH receptor expression and GH-dependent STAT5b activation and PPARgamma expression were reduced in CD colon. STAT5b-deficient mice exhibited reduced basal PPARgamma nuclear abundance and developed more severe proximal colitis after TNBS administration. This was associated with a significant increase in mucosal nuclear factor kappaB activation at baseline and after TNBS administration. Rosiglitazone ameliorated colitis in wild-type mice but not STAT5b-deficient mice. GH-dependent STAT5b activation is impaired in affected CD colon and contributes to chronic mucosal inflammation via down-regulation of local PPARgamma expression. Therapeutic activation of the GH:STAT5b axis therefore represents a novel target for restoring both normal anabolic metabolism and mucosal tolerance in CD.
Collapse
Affiliation(s)
- Xiaonan Han
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, MLC 2010, 3333 Burnet Ave., Cincinnati, OH 45229-3039, USA
| | | | | | | | | | | |
Collapse
|
22
|
Perry JK, Emerald BS, Mertani HC, Lobie PE. The oncogenic potential of growth hormone. Growth Horm IGF Res 2006; 16:277-289. [PMID: 17101287 DOI: 10.1016/j.ghir.2006.09.006] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2006] [Accepted: 09/28/2006] [Indexed: 10/23/2022]
Abstract
A growing body of recent literature indicates that in addition to an essential role in growth and development, growth hormone may also play a more sinister role in oncogenic transformation and neoplastic progression. Here we review the accumulating evidence implicating growth hormone in the development and progression of cancer and describe what is known of the mechanisms utilised by this hormone in neoplastic transformation.
Collapse
Affiliation(s)
- Jo K Perry
- The Liggins Institute and the National Research Centre for Growth and Development, University of Auckland, 2-6 Park Avenue, Grafton, Private Bag 92019, Auckland 1023, New Zealand
| | | | | | | |
Collapse
|
23
|
Veldhuis JD, Roemmich JN, Richmond EJ, Bowers CY. Somatotropic and gonadotropic axes linkages in infancy, childhood, and the puberty-adult transition. Endocr Rev 2006; 27:101-40. [PMID: 16434512 DOI: 10.1210/er.2005-0006] [Citation(s) in RCA: 178] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Integrative neuroendocrine control of the gonadotropic and somatotropic axes in childhood, puberty, and young adulthood proceeds via multiple convergent and divergent pathways in the human and experimental animal. Emerging ensemble concepts are required to embody independent, parallel, and interacting mechanisms that subserve physiological adaptations and pathological disruption of reproduction and growth. Significant advances in systems biology will be needed to address these challenges.
Collapse
Affiliation(s)
- Johannes D Veldhuis
- Endocrine Research Unit, Department of Internal Medicine, Mayo Medical School, Mayo School of Graduate Medical Education, General Clinical Research Center, Mayo Clinic, Rochester, Minnesota 55905, USA.
| | | | | | | |
Collapse
|
24
|
Rowland JE, Kerr LM, White M, Noakes PG, Waters MJ. Heterozygote effects in mice with partial truncations in the growth hormone receptor cytoplasmic domain: assessment of growth parameters and phenotype. Endocrinology 2005; 146:5278-86. [PMID: 16166215 DOI: 10.1210/en.2005-0939] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The GH receptor (GHR) is essential for normal postnatal growth and development, and the molecular basis of GHR action has been studied intensively. Clinical case studies and more recently mouse models have revealed the extensive phenotype of impaired GH action. We recently reported two new mouse models, possessing cytoplasmic truncations at position 569 (plus Y539/545-F) and 391, which were created to identify functional subdomains within the cytoplasmic signaling domain. In the homozygous state, these animals show progressively impaired postnatal growth coupled with complex changes in gene expression. We describe here an extended phenotype analysis encompassing the heterozygote state to identify whether single copies of these mutant receptors bring about partial or dominant-negative phenotypes. It appears that the retention of the ubiquitin-dependent endocytosis motif in the N-terminal cytoplasmic domain permits turnover of these mutant receptors because no dominant-negative phenotype is seen. Nonetheless, we do observe partial impairment of postnatal growth in heterozygotes supporting limited haploinsufficiency. Reproductive function is impaired in these models in a progressive manner, in parallel with loss of signal transducer and activator of transcription-5 activation ability. In summary, we describe a more comprehensive phenotypic analysis of these mouse models, encompassing overall and longitudinal body growth, reproductive function, and hormonal status in both the heterozygote and homozygote state. Our results suggest that patients expressing single copies of similarly mutated GHRs would not display an obvious clinical phenotype.
Collapse
Affiliation(s)
- Jennifer E Rowland
- Institute for Molecular Bioscience, University of Queensland, St. Lucia, Australia
| | | | | | | | | |
Collapse
|
25
|
He K, Loesch K, Cowan JW, Li X, Deng L, Wang X, Jiang J, Frank SJ. Janus kinase 2 enhances the stability of the mature growth hormone receptor. Endocrinology 2005; 146:4755-65. [PMID: 16081639 DOI: 10.1210/en.2005-0514] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The abundance of surface GH receptor (GHR) is an important determinant of cellular GH sensitivity and is regulated at both transcriptional and posttranscriptional levels. In previous studies of GHR-expressing Janus kinase 2 (JAK2)-deficient human fibrosarcoma cells (gamma2A-GHR), we demonstrated that stable transfection with JAK2 resulted in increased steady-state levels of mature GHR (endoH-resistant; relative molecular mass, 115-140 kDa) relative to precursor GHR (endoH-sensitive; relative molecular mass, 100 kDa). We now examine further the effects of JAK2 on GHR trafficking by comparing gamma2A-GHR to gamma2A-GHR cells stably reconstituted with JAK2 (C14 cells). In the presence of JAK2, GHR surface expression was increased, as assessed by surface biotinylation, 125I-labeled human GH cell surface binding, and immunofluorescence microscopy assays. Although the absence of JAK2 precluded GH-stimulated signaling, GH-induced GHR disulfide linkage (a proxy for the GH-induced conformational changes in the GHR dimer) proceeded independent of JAK2 expression, indicating that the earliest steps in GH-induced GHR triggering are not prevented by the absence of JAK2. RNA interference-mediated knockdown of JAK2 in C14 cells resulted in a decreased mature to precursor ratio, supporting a primary role for JAK2 either in enhancing GHR biogenesis or dampening mature GHR degradation. To address these potential mechanisms, metabolic pulse-chase labeling experiments and experiments in which the fate of previously synthesized GHR was followed by anti-GHR immunoblotting after cycloheximide treatment (cycloheximide chase experiments) were performed. These indicated that the presence of JAK2 conferred modest enhancement (1.3- to 1.5-fold) in GHR maturation but substantially prolonged the t1/2 of the mature GHR, suggesting a predominant effect on mature GHR stability. Cycloheximide chase experiments with metalloprotease, proteasome, and lysosome inhibitors indicated that the enhanced stability of mature GHR conferred by JAK2 is not related to effects on constitutive receptor metalloproteolysis but rather is a result of reduced constitutive endosomal/lysosomal degradation of the mature GHR. These results are discussed in the context of emerging information on how JAK-family members modulate surface expression of other cytokine receptors.
Collapse
Affiliation(s)
- Kai He
- Endocrinology Section Medical Service, Veterans Affairs Medical Center, and Department of Medicine, University of Alabama at Birmingham, 1530 3rd Avenue South, BDB 861, Birmingham, Alabama 35294-0012, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
David A, Metherell LA, Clark AJL, Camacho-Hübner C, Savage MO. Diagnostic and therapeutic advances in growth hormone insensitivity. Endocrinol Metab Clin North Am 2005; 34:581-95, viii. [PMID: 16085161 DOI: 10.1016/j.ecl.2005.04.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Diagnostic and therapeutic advances in growth hormone insensitivity (GHI) have occurred principally in two areas: the molecular characterization of patients with GHI and treatment with recombinant human insulin like growth factor-I (IGF-I). This article discusses the current status of molecular diagnosis across the spectrum of the disorder. Treatment with recombinant human IGF-I in classical cases is summarized, and potential new targets for treatment are discussed together with the potential for therapy using the newly developed compound recombinant human IGF-I/IGF binding protein-3.
Collapse
Affiliation(s)
- Alessia David
- Molecular Endocrinology Centre, William Harvey Research Institute, St. Bartholomew's Hospital, London, UK
| | | | | | | | | |
Collapse
|
27
|
DiFedele LM, He J, Bonkowski EL, Han X, Held MA, Bohan A, Menon RK, Denson LA. Tumor necrosis factor alpha blockade restores growth hormone signaling in murine colitis. Gastroenterology 2005; 128:1278-91. [PMID: 15887111 DOI: 10.1053/j.gastro.2005.02.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Cytokines including tumor necrosis factor alpha (TNFalpha) may create a state of growth hormone (GH) resistance in Crohn's disease. Anabolic effects of GH are mediated via phosphorylation of the signal transducer and activator of transcription (STAT)5b transcription factor. Although GH resistance in other settings has been linked to a defect in janus kinase-STAT signaling, the molecular basis for GH resistance in colitis was not known. We hypothesized that the GH-induced phosphorylation of STAT5b would be impaired in colitis, and that TNFalpha blockade would restore GH signaling. METHODS Growth, body composition, and molecular regulators of GH signaling were determined in interleukin-10 null mice with chronic colitis and wild-type controls, +/- treatment with an anti-TNFalpha antibody. RESULTS Interleukin-10 null mice exhibited significant alterations in growth, body composition, and feed efficiency. Liver insulin-like growth factor 1 expression was reduced in colitic mice. This was associated with down-regulation of GH receptor (GHR) expression and impaired GH-dependent STAT5b activation. Down-regulation of GHR expression was associated with reduced nuclear abundance and DNA binding of the GHR gene-promoter transactivator, Sp3. TNFalpha down-regulated GHR abundance and prevented GH-induced tyrosine phosphorylation of STAT5 in rat hepatocytes in culture. TNFalpha neutralization up-regulated liver GHR abundance and restored GH activation of STAT5 and serum insulin-like growth factor 1 levels in colitic mice; this preceded improvements in weight gain and disease activity. CONCLUSIONS GH resistance in experimental colitis is caused by down-regulation of GHR expression, thereby reducing GH-dependent STAT5 activation. TNFalpha blockade restores liver GH signaling and improves anabolic metabolism in this setting.
Collapse
Affiliation(s)
- Lisa M DiFedele
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Kawana K, Kawana Y, Schust DJ. Female steroid hormones use signal transducers and activators of transcription protein-mediated pathways to modulate the expression of T-bet in epithelial cells: a mechanism for local immune regulation in the human reproductive tract. Mol Endocrinol 2005; 19:2047-59. [PMID: 15860546 DOI: 10.1210/me.2004-0489] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The transcription factor T-bet promotes the differentiation of inflammatory Th1 T helper cells. T-bet expression in lymphoid cells is regulated by cytoplasmic signaling through Janus kinase phosphorylation, nuclear signaling using signal transducers and activators of transcription (Stat) family proteins, and autocrine/paracrine feedback involving interferon (IFN)-gamma. T-bet is here shown to be present in epithelial cells of the human female reproductive tract. Regulation of T-bet expression was modulated by cytokines and the female reproductive steroids, estrogen, and progesterone. The mechanisms of T-bet regulation in epithelia differ from those in conventional immune cells. During a 15-d exposure to progesterone, T-bet levels in endometrial epithelial cells (EECs) undulated. Prior exposure to estrogen enhanced these effects. More prolonged exposure of EECs to these hormones, singly or in combination, suppressed T-bet production. Stat1 and Stat5 bound to the EEC T-bet regulatory region (TRR) at the IFN-gamma-activated sequence site, but Stat3 and Stat4 did not. Binding of Stat1 and Stat5 to the TRR were modified by progesterone in distinct ways. Estrogen suppressed the binding of Stat1 and Stat5 to the TRR. Mutation of gamma-activated sequence element reduced T-bet promoter activity, binding of Stat proteins to the TRR and regulation of the promoter by cytokines and hormones. In EECs, cytokine exposure caused phosphorylation of Janus kinase 2 and TRR-bound Stat proteins; female steroid hormones altered only phosphorylation of TRR-bound Stat5. Although there is no autocrine IFN-gamma feedback loop in reproductive tract epithelial cells, an IL-15/T-bet positive feedback loop may exist. The implications of hormonally regulated T-bet expression are discussed.
Collapse
Affiliation(s)
- Kei Kawana
- Fearing Research Laboratory, Department of Obstetrics, Gynecology, and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
29
|
Wiwi CA, Waxman DJ. Role of Hepatocyte Nuclear Factors in Transcriptional Regulation of Male-specific CYP2A2. J Biol Chem 2005; 280:3259-68. [PMID: 15539409 DOI: 10.1074/jbc.m409294200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cytochrome P450 2A2 (CYP2A2) is an adult male-specific rat liver steroid hydroxylase whose sex-dependent expression is regulated at the transcriptional level by sexually dimorphic pituitary growth hormone (GH) secretory patterns. In contrast to CYP2C11 and other male-specific, plasma GH pulse-inducible liver genes, CYP2A2 is highly expressed in hypophysectomized rat liver, despite the absence of GH stimulation. CYP2A2 promoter fragments 0.9-6.2 kb long exhibited unusually high basal promoter activity when transfected into the liver cell line HepG2. A further approximately 2.5-fold increase in activity was obtained by cotransfection of hepatocyte nuclear factor (HNF) 3gamma or HNF4alpha. CYP2A2 promoter activity was inhibited approximately 85% by transfection of HNF3beta or HNF6, both of which are more highly expressed in female than male liver and can strongly trans-activate the female-specific CYP2C12 promoter. The male GH pulse-activated transcription factor STAT5b had no effect on CYP2A2 promoter activity, either alone or in combination with HNF3gamma and HNF4alpha, consistent with the GH pulse-independence of CYP2A2 expression. By contrast, STAT5b synergistically enhanced the transcriptional activity of HNF4alpha toward two other male-specific liver target genes, Cyp2d9 and CYP8B1. Furthermore, STAT5b in combination with the HNF4alpha coactivator peroxisome proliferator-activated receptor gamma coactivator-1alpha strongly enhanced the transcriptional activity of HNF4alpha toward CYP8B1 but not toward CYP2A2. These findings support the hypothesis that sex-dependent HNFs contribute to the sexually dimorphic expression of CYP2A2 and other liver CYPs and highlight the ability of STAT5b to act in concert with HNF4alpha to regulate select male-specific liver CYP genes.
Collapse
Affiliation(s)
- Christopher A Wiwi
- Division of Cell and Molecular Biology, Department of Biology Boston University, Boston, Massachusetts 02215, USA
| | | |
Collapse
|
30
|
Zhu T, Starling-Emerald B, Zhang X, Lee KO, Gluckman PD, Mertani HC, Lobie PE. Oncogenic Transformation of Human Mammary Epithelial Cells by Autocrine Human Growth Hormone. Cancer Res 2005. [DOI: 10.1158/0008-5472.317.65.1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
The human growth hormone (hGH) gene is expressed in the normal human mammary epithelial cell and its expression increases concomitant with the acquisition of proliferative lesions. Herein we demonstrate that autocrine production of hGH in human mammary carcinoma cells dramatically enhances anchorage-independent growth in a Janus kinase 2–dependent manner. Forced expression of the hGH gene in immortalized human mammary epithelial cells increased proliferation, decreased apoptosis, altered the cellular morphology and resulted in oncogenic transformation. Autocrine hGH was therefore sufficient to support anchorage-independent growth of immortalized human mammary epithelial cells and tumor formation in vivo. Moreover, autocrine hGH disrupted normal mammary acinar architecture with luminal filling and deregulated proliferation in three-dimensional epithelial cell culture. Autocrine hGH utilized homeobox A1 to govern the transcriptional program required for autocrine hGH–stimulated oncogenic transformation of human mammary epithelial cells, including transcriptional up-regulation of c-Myc, cyclin D1, and Bcl-2. Forced expression of a single orthotopically expressed wild-type gene is therefore sufficient for oncogenic transformation of the immortalized human mammary epithelial cell.
Collapse
Affiliation(s)
- Tao Zhu
- 1Institute of Molecular and Cell Biology and
| | - B. Starling-Emerald
- 3Liggins Institute and National Research Centre for Growth and Development, University of Auckland, Auckland, New Zealand; and
| | - Xin Zhang
- 2Department of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Kok-Onn Lee
- 2Department of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Peter D. Gluckman
- 3Liggins Institute and National Research Centre for Growth and Development, University of Auckland, Auckland, New Zealand; and
| | - Hichem C. Mertani
- 4Centre National de la Recherche Scientifique UMR 5578, Physiologies Energetiques Cellulaires et Moléculaires, Université Claude Bernard, Lyon-1, France
| | - Peter E. Lobie
- 1Institute of Molecular and Cell Biology and
- 3Liggins Institute and National Research Centre for Growth and Development, University of Auckland, Auckland, New Zealand; and
| |
Collapse
|
31
|
Abstract
GH plays a pivotal role in regulating body growth and development, which is modulated by sex steroids. A close interplay between estrogen and GH leads to attainment of gender-specific body composition during puberty. The physiological basis of the interaction is not well understood. Most previous studies have focused on the effects of estrogen on GH secretion. There is also strong evidence that estrogen modulates GH action independent of secretion. Oral but not transdermal administration of estrogen impairs the metabolic action of GH in the liver, causing a fall in IGF-I production and fat oxidation. This results in a loss of lean tissue and a gain of body fat in postmenopausal women and an impairment of GH effect in hypopituitary women on GH replacement. The negative metabolic sequelae are potentially important because of the widespread use of oral estrogen and estrogen-related compounds. Estrogen affects GH action at the level of receptor expression and signaling. More recently, estrogen has been shown to inhibit Janus kinase/signal transducer and activator of transcription signaling by GH via the induction of suppressor of cytokine signaling-2, a protein inhibitor for cytokine signaling. This represents a novel paradigm of steroid regulation of cytokine receptors and is likely to have significance for a diverse range of cytokine function.
Collapse
Affiliation(s)
- Kin-Chuen Leung
- Pituitary Research Unit, Garvan Institute of Medical Research, St. Vincent's Hospital, Sydney, New South Wales 2010, Australia
| | | | | | | |
Collapse
|
32
|
Wiwi CA, Gupte M, Waxman DJ. Sexually dimorphic P450 gene expression in liver-specific hepatocyte nuclear factor 4alpha-deficient mice. Mol Endocrinol 2004; 18:1975-87. [PMID: 15155787 DOI: 10.1210/me.2004-0129] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Hepatocyte nuclear factor (HNF) 4alpha is a liver-enriched nuclear receptor that plays a critical role in regulating the expression of numerous hepatic genes, including members of the cytochrome P450 (CYP) superfamily, several of which are expressed in a sex-dependent manner. Presently, we use a liver-specific Hnf4alpha-deficient mouse model to investigate the role of HNF4alpha in regulating liver-enriched transcription factors and sexually dimorphic Cyps in liver in vivo. Real-time PCR analysis of RNA isolated from livers of wild-type and Hnf4alpha-deficient mice revealed the following: 1) HNF4alpha exerts both positive regulation (Hnfalpha, C/ebpalpha, and C/ebpbeta) and negative regulation (Hnf3alpha and the HNF4alpha coactivator Pgc-1alpha) on liver transcription factor expression; 2) a strong dependence on HNF4alpha characterizes several male-predominant Cyps (2d9 and 8b1), female-predominant Cyps (2b10, 2b13, 3a41, and 3a44) and Cyps, whose expression is sex independent (3a11, 3a25); 3) HNF4alpha confers a unique, positive regulation of two male-expressed genes (Cyp4a12 and GSTpi) and a negative regulation of several female-predominant genes (Cyp2a4, Cyp2b9, Hnf3beta, and Hnf6), both of which are manifest in male but not female mouse liver. These trends were confirmed at the protein level by Western blot analysis using antibodies raised to Cyp2a, Cyp2b, and Cyp3a family members. Thus, HNF4alpha is an essential player in the complex regulatory network of liver-enriched transcription factors and the sexually dimorphic mouse Cyp genes that they regulate. HNF4alpha is proposed to contribute to the sex specificity of liver gene expression by positively regulating a subset of male-specific Cyp genes while concomitantly inhibiting the expression of certain female-specific Cyps and liver transcription factors, by mechanisms that are operative in male, but not female, mouse liver.
Collapse
Affiliation(s)
- Christopher A Wiwi
- Department of Biology, Boston University, 5 Cummington Street, Boston, Massachusetts 02215, USA
| | | | | |
Collapse
|
33
|
Crispi S, Sanzari E, Monfregola J, De Felice N, Fimiani G, Ambrosio R, D'Urso M, Ursini MV. Characterization of the human STAT5A and STAT5B promoters: evidence of a positive and negative mechanism of transcriptional regulation. FEBS Lett 2004; 562:27-34. [PMID: 15043997 DOI: 10.1016/s0014-5793(04)00166-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2003] [Revised: 02/12/2004] [Accepted: 02/12/2004] [Indexed: 11/23/2022]
Abstract
We recently published the genomic characterization of the STAT5A and STAT5B paralogous genes that are located head to head in the 17q21 chromosome and share large regions of sequence identity. We here demonstrate by transient in vitro transfection that STAT5A and STAT5B promoters are able to direct comparable levels of transcription. The expression of basal promoters is enhanced after Sp1 up-regulation in HeLa and SL2 cells while DNA methylation associated to the recruitment of MeCP2 methyl CpG binding protein down-regulates STAT5A and B promoters by interfering with Sp1-induced transcription. In addition, cross-species sequence comparison identified a bi-directional negative cis-acting regulatory element located in the STAT5 intergenic region.
Collapse
Affiliation(s)
- Stefania Crispi
- Institute of Genetics and Biophysics Adriano Buzzati-Traverso, CNR, Via P. Castellino 111, 80131 Naples, Italy
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Sakuma T, Kitajima K, Nishiyama M, Endo Y, Miyauchi K, Jarukamjorn K, Nemoto N. Collaborated regulation of female-specific murine Cyp3a41 gene expression by growth and glucocorticoid hormones. Biochem Biophys Res Commun 2004; 314:495-500. [PMID: 14733933 DOI: 10.1016/j.bbrc.2003.12.114] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CYP3A41 is a female-specific major CYP3A in mouse livers. Adrenalectomy decreased expression of CYP3A41 as well as CYP3A11, another major CYP3A, and dexamethasone (DEX) restored the decreased expression. Hypophysectomy completely abolished CYP3A41 expression and growth hormone (GH) replacement only slightly restored the expression. Treatment with DEX alone did not induce expression of either CYP3A41 or CYP3A11 in hypophysectomized mice. However, combined treatment with GH and DEX strongly induced expression of CYP3A41 but not CYP3A11. In primary cultured mouse hepatocytes, DEX induced expression of both CYP3A41 and CYP3A11, and DEX-inducible expression of CYP3A41 was suppressed by RU486, a potent antiglucocorticoid. In contrast, RU486 by itself enhanced basal expression of CYP3A11 mRNA, while it showed no inhibitory effect on DEX-inducible expression. These observations indicate that glucocorticoids may participate in the GH-dependent control of the Cyp3a41 gene expression, probably mediated via the glucocorticoid receptor, which may be different from that of the Cyp3a11 gene expression.
Collapse
Affiliation(s)
- Tsutomu Sakuma
- Department of Toxicology, Faculty of Pharmaceutical Sciences, Toyama Medical and Pharmaceutical University, 2630 Sugitani, 930-0194 Toyama, Japan.
| | | | | | | | | | | | | |
Collapse
|
35
|
Takebe K, Oka Y, Radisky D, Tsuda H, Tochigui K, Koshida S, Kogo K, Hirai Y. Epimorphin acts to induce hair follicle anagen in C57BL/6 mice. FASEB J 2003; 17:2037-47. [PMID: 14597673 DOI: 10.1096/fj.03-0386com] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Epimorphin is a mesenchymal morphogen that has been shown to mediate epithelial-mesenchymal signaling interactions in various organs. We now show that epimorphin functions in hair follicle morphogenesis; using a novel ex vivo organ culture assay, we define a mechanism for epimorphin signaling that may provide insight into general developmental processes. We found that epimorphin was produced by follicular mesenchymal cells and bound selectively to follicular epithelial cells, and that treatment with recombinant epimorphin could stimulate procession of hair follicles from telogen (resting stage) to anagen (growing stage). Based on analyses of epimorphin proteolytic digests that suggested a smaller peptide might be able to substitute for the full-length epimorphin molecule, we determined that pep7, a 10-amino acid peptide, was capable of inducing telogen-to-anagen transition both in the culture assay and in the mouse. That pep7 showed maximal activity only when modified with specific sulfhydryl-reactive reagents suggested that a particular structural conformation of the peptide was essential for activity; molecular dynamics studies were pursued to investigate the active peptide structure. These findings define a previously unknown morphogenic process in the hair follicle that may have applications to many other organs.
Collapse
Affiliation(s)
- Kyoko Takebe
- EPM project groups, Osaka R and D Laboratories, Sumitomo Electric Industries LTD. 1, Taya-cho Sakae-ku, Yokohama 244-8588, Japan
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Wolbold R, Klein K, Burk O, Nüssler AK, Neuhaus P, Eichelbaum M, Schwab M, Zanger UM. Sex is a major determinant of CYP3A4 expression in human liver. Hepatology 2003. [DOI: 10.1002/hep.1840380424] [Citation(s) in RCA: 229] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
|
37
|
Badger TM, Ronis MJJ, Frank SJ, Chen Y, He L. Effects of chronic ethanol on hepatic and renal CYP2C11 in the male rat: interactions with the Janus-kinase 2-signal transducer and activators of transcription proteins 5b pathway. Endocrinology 2003; 144:3969-76. [PMID: 12933671 DOI: 10.1210/en.2002-0163] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Chronic alcohol intake in male rats results in: 1) demasculinization of the GH pulse pattern; 2) reduced serum testosterone concentrations; and 3) decreased expression hepatic CYP2C11. Hepatic CYP2C11 expression is regulated by the male pattern of GH through the Janus-kinase/signal transducer and activators of transcription proteins (JAK/STAT) signal transduction pathway in the male rat. Renal CYP2C11 is regulated by testosterone, not GH. The involvement of the JAK/STAT5b signal transduction pathway in renal CYP2C11 signaling has not been studied. We tested the hypothesis that ethanol reduces CYP2C11 levels by interfering with the JAK/STAT5b pathway. Using a total enteral nutrition (TEN) model to feed rats a well-balanced diet, we have studied the effects of chronic ethanol intake (21 d) on hepatic and renal JAK/STAT pathway of adult male rats (8-10/group). We found decreased hepatic and renal expression of CYP2C11 in ethanol-fed rats with concomitant decreases in STAT5b and phospho-STAT5b, decreased in vitro hepatic STAT5b binding to a CYP2C11 promoter element and no effects on hepatic GHR levels. Ethanol caused tissue specific effects in phospho-JAK2 and JAK2, with increased levels in the liver, but decreased JAK2 expression in the kidney. We conclude that ethanol suppression of CYP2C11 expression is clearly associated with reductions in STAT5b levels, but not necessarily in reductions of JAK2 levels. The mechanisms underlying ethanol-induced suppression of STAT5b is yet to be determined, as is the question of whether this is secondary to hormonal effects or a direct ethanol effect.
Collapse
Affiliation(s)
- T M Badger
- Arkansas Children's Nutrition Center and Department of Physiology/Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72202, USA.
| | | | | | | | | |
Collapse
|
38
|
Lampl M, Jeanty P. Timing is everything: a reconsideration of fetal growth velocity patterns identifies the importance of individual and sex differences. Am J Hum Biol 2003; 15:667-80. [PMID: 12953179 DOI: 10.1002/ajhb.10204] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Fetal growth has been posited to follow a "timing hypothesis" sequence in which the second trimester favors a single growth velocity peak in body length and the third trimester accommodates a single growth velocity peak in weight accrual. To our knowledge, this proposition has never been tested with high-frequency longitudinal ultrasound data from normally growing human fetuses. The present study examined whether fetal growth in leg length had its peak velocity at or about 20-26 gestational weeks and declined subsequently and whether estimated fetal weight velocity was maximal at or about 33 weeks and declined subsequently; if the greatest acquisition of leg length occurred in the second trimester and weight in the third trimester; and if birth outcomes reflected these relationships. The data in this study included approximately weekly longitudinal ultrasound data collected from 44 maternal/fetal pairs in Brussels, Belgium. Diaphyseal lengths of the femur and tibia provided information on leg growth and estimated fetal weight was assessed from the biparietal and occipital-frontal head diameters and transverse and anterior-posterior diameters of the abdomen. Growth patterns were investigated from individual growth curves derived from daily growth velocity z-scores. Paired t-tests compared individuals' trimestral increments in leg length and fetal weight. Least-squares regression models employing the robust procedure for repeated measurements were used to test for relationships between trimester, size, growth rates, and birth outcome, controlling for day of measurement, sex, maternal smoking, and gestational age at birth. The normal fetuses in this study grew by pulsatile patterns of leg and estimated weight acquisition, not a single peak and decline process. Greater incremental growth in estimated fetal weight occurred during the second trimester and leg length in the third trimester. Individual and sex effects were significant in growth velocity patterns. Girls grew with greater synchrony between leg and weight growth and were accelerated by comparison with boys, with faster leg growth predicting lower ponderal index by the second trimester. Birth outcomes were sex-specific in timing effects and predictive variables. These results support the importance of sex-specific analyses, reemphasize the common notion that girls grow faster than boys, and direct attention to cross-talk between energy resources and growth.
Collapse
Affiliation(s)
- Michelle Lampl
- Department of Anthropology, Emory University, Atlanta, Georgia 30322, USA.
| | | |
Collapse
|
39
|
Abstract
The unfolding of pubertal growth and maturation entails multisystem collaboration. Most notably, the outflow of gonadotropins and growth hormone (GH) proceeds both independently and jointly. The current update highlights this unique dependency in the human.
Collapse
|
40
|
Growth and lactogenic hormones, insulin-like growth factor and insulin. ACTA ACUST UNITED AC 2003. [DOI: 10.1016/s1567-7443(03)80045-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
41
|
|
42
|
Morales O, Faulds MH, Lindgren UJ, Haldosén LA. 1Alpha,25-dihydroxyvitamin D3 inhibits GH-induced expression of SOCS-3 and CIS and prolongs growth hormone signaling via the Janus kinase (JAK2)/signal transducers and activators of transcription (STAT5) system in osteoblast-like cells. J Biol Chem 2002; 277:34879-84. [PMID: 12107179 DOI: 10.1074/jbc.m204819200] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Growth hormone (GH) and 1alpha,25-dihydroxyvitamin D(3) (1,25-(OH)(2)D(3)) are regulators of bone growth and bone metabolism. In target cells, GH activates several signaling pathways, among them the Janus kinase (JAK)/signal transducers and activators of transcription (STAT) pathway. GH mainly activates JAK2 and STAT5a and b. The effects of 1,25-(OH)(2)D(3) are mediated via a nuclear receptor, the vitamin D receptor, which, when bound by 1,25-(OH)(2)D(3), activates the transcription of target genes. In earlier studies (Morel, G., Chavassieux, P., Barenton, B., Dubois, P. M., Meunier, P. J., and Boivin, G. (1993) Cell Tissue Res. 273, 279-286) synergistic interaction between 1,25-(OH)(2)D(3) and GH regarding expression of osteoblastic markers has been described. The UMR 106 cell line is a rat osteosarcoma cell line with osteoblast-like properties. We have recently shown (Morales, O., Lindgren, U., and Haldosen, L. A. (2000) J. Bone Miner. Res. 15, 2284-2290) that UMR 106 cells express a GH-responsive JAK2/STAT5 signaling system. These cells also express the vitamin D receptor and respond to 1,25-(OH)(2)D(3). In the present study we have investigated whether 1,25-(OH)(2)D(3) influences GH signaling via the JAK2/STAT5 pathway in UMR 106 cells. We found that 1,25-(OH)(2)D(3) prolonged GH signaling via the JAK2/STAT5 pathway. Pretreatment of cells with 1,25-(OH)(2)D(3) was also necessary in order to detect GH-induced STAT5 transcriptional response. Furthermore, the pretreatment of cells with 1,25-(OH)(2)D(3) rendered to the cells the capacity to respond to repetitive GH-stimulation. In UMR 106 cells, GH induced the expression of the JAK/STAT negative regulatory proteins SOCS-3 and CIS. Interestingly, pretreatment with 1,25-(OH)(2)D(3) inhibited GH-induced expression of these proteins. From these results we propose that 1,25-(OH)(2)D(3) has an inhibitory effect on negative regulatory pathways acting on JAK2 and/or STAT5 in UMR 106 cells and that this, in all or partly, explains the effects of 1,25-(OH)(2)D(3) on GH-signaling via the JAK/STAT pathway.
Collapse
Affiliation(s)
- Orlando Morales
- Department of Orthopedic Surgery, Karolinska Institutet, Huddinge Hospital, S-141 86 Huddinge, Sweden
| | | | | | | |
Collapse
|
43
|
Ursini MV, Gaetaniello L, Ambrosio R, Matrecano E, Apicella AJ, Salerno MC, Pignata C. Atypical X-linked SCID phenotype associated with growth hormone hyporesponsiveness. Clin Exp Immunol 2002; 129:502-9. [PMID: 12197892 PMCID: PMC1906458 DOI: 10.1046/j.1365-2249.2002.01823.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2002] [Indexed: 01/17/2023] Open
Abstract
Severe combined immunodeficiency (SCID) is a heterogeneous group of disorders characterized by defect of T- and B-cell immunity. In many cases of autosomal recessive SCID, thus far described, the molecular alteration involves genes encoding for molecules that participate in the signal transduction. We report on a patient affected by a combined immunodeficiency, characterized by severe T-cell functional impairment, in spite of a close to normal number of circulating mature type T and B cells. NK cells were absent. Associated with the immunodeficiency, this patient also showed short stature characterized by very low growth velocity, delayed bone age and absence of increase of the plasma levels of Insulin growth factor-I (IGF-I) after growth hormone (GH) in vivo stimulation indicating peripheral hyporesponsiveness to GH. Evaluation of the protein tyrosine phosphorylation events occurring following either T-cell receptor (TCR) or GH receptor (GHR) triggering revealed striking abnormalities. No molecular alteration of GHR gene was found, thus suggesting the presence of postreceptorial blockage. Mutational screening and expression analysis failed to reveal any molecular alteration of JAK2 and STAT 5 A/B genes thus ruling out the involvement of these genes in the pathogenesis of this form of SCID. Mutational analysis of IL2Rgamma chain gene revealed the presence of a L183S missense mutation, thus indicating an atypical and a more complex clinical presentation of this X-linked form of SCID. At our knowledge, this is the first report on the GH hyporesponsiveness in this disease.
Collapse
Affiliation(s)
- M V Ursini
- Department of Paediatrics Federico II University, Naples, Italy
| | | | | | | | | | | | | |
Collapse
|
44
|
Kalsotra A, Anakk S, Boehme CL, Strobel HW. Sexual dimorphism and tissue specificity in the expression of CYP4F forms in Sprague Dawley rats. Drug Metab Dispos 2002; 30:1022-8. [PMID: 12167568 DOI: 10.1124/dmd.30.9.1022] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The cytochromes P450 belong to a superfamily of enzymes involved in a diverse array of endobiotic and xenobiotic metabolic pathways. Several members of a novel family of cytochrome P450 (CYP4F), which specifically mediate leukotriene B(4) omega-hydroxylation, have now been identified in various species including rat, mouse, and human. In rats, the CYP4F family consists of four related genes, CYP4F1, CYP4F4, CYP4F5, and CYP4F6. Here we report development of fluorescent real-time quantitative polymerase chain reaction assays (TaqMan), which allow us to carry out specific quantitation of mRNA expression of all four members of this subfamily. Since no inducers for the CYP4F family are known to date, we validated these assays using clofibrate, a known suppressor of rat CYP4Fs. Additionally, Northern blot hybridization was carried out to validate these assays. Using this approach, we demonstrate quantitatively, for the first time, that each of the rat CYP4Fs is expressed in a tissue- and sex-dependent manner showing a significantly higher expression in females vis-à-vis males. Western blot analysis using a CYP4F polyclonal antibody also shows a considerably higher protein expression in female liver, kidneys, and lungs when compared with male rats. Furthermore, we observe a significant decrease in the CYP4F1, CYP4F4, and CYP4F6 message in kidneys and liver of ovariectomized rats when compared with control females. This loss of expression is partially restored by estrogen treatment in both tissues, suggesting a role of estrogen in regulating CYP4F expression.
Collapse
Affiliation(s)
- Auinash Kalsotra
- Department of Biochemistry and Molecular Biology, the University of Texas Medical School of Houston, Houston, Texas 77225, USA
| | | | | | | |
Collapse
|
45
|
Sakuma T, Endo Y, Mashino M, Kuroiwa M, Ohara A, Jarukamjorn K, Nemoto N. Regulation of the expression of two female-predominant CYP3A mRNAs (CYP3A41 and CYP3A44) in mouse liver by sex and growth hormones. Arch Biochem Biophys 2002; 404:234-42. [PMID: 12147261 DOI: 10.1016/s0003-9861(02)00329-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A second female-predominant murine CYP3A, CYP3A44, was isolated from liver and its mRNA expression was compared with that of the previously described CYP3A41. The expression of CYP3A44 was relatively constant after birth in females, whereas it gradually declined in males after 5 weeks of age. The expression of CYP3A41 increased with age in females after 3 weeks of age, whereas it gradually declined in males after 5 weeks of age. Hypophysectomy and growth hormone replacement indicated that expression of both CYP3A mRNAs in females was dependent on the feminine plasma growth hormone profile. Estradiol induced the expression of both mRNAs and the effect was dependent on the presence of the pituitary gland. These observations suggest that endocrine control of expression might be similar, but not identical, for two female-predominant CYP3A mRNAs.
Collapse
Affiliation(s)
- Tsutomu Sakuma
- Department of Toxicology, Faculty of Pharmaceutical Sciences, Toyama Medical and Pharmaceutical University, 2630 Sugitani, Toyama 930-0194, Japan.
| | | | | | | | | | | | | |
Collapse
|
46
|
Zhou YC, Davey HW, McLachlan MJ, Xie T, Waxman DJ. Elevated basal expression of liver peroxisomal beta-oxidation enzymes and CYP4A microsomal fatty acid omega-hydroxylase in STAT5b(-/-) mice: cross-talk in vivo between peroxisome proliferator-activated receptor and signal transducer and activator of transcription signaling pathways. Toxicol Appl Pharmacol 2002; 182:1-10. [PMID: 12127257 DOI: 10.1006/taap.2002.9426] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Long-term treatment of rodents with peroxisome proliferator chemicals, a group of structurally diverse nongenotoxic carcinogens, leads to liver cancer in a process dependent on the nuclear receptor peroxisome proliferator-activated receptor-alpha (PPARalpha). Previous in vitro studies have shown that growth hormone (GH) can inhibit PPARalpha-dependent gene expression by down-regulation of PPARalpha expression and by a novel inhibitory cross-talk involving the GH-activated transcription factor STAT5b. Presently, we evaluate the role of STAT5b in mediating these inhibitory actions of GH on PPAR function using a STATb-deficient mouse model. Protein levels of three PPARalpha-responsive peroxisomal beta-oxidation pathway enzymes (fatty acyl-CoA oxidase, 3-ketoacyl-CoA thiolase, and L-bifunctional enzyme) were increased up to two- to threefold in STAT5b(-/-) relative to wild-type control mouse liver, as was the basal expression of two PPARalpha-regulated cytochrome P450 4A proteins. In contrast, protein levels of two PPARalpha-unresponsive peroxisomal enzymes, catalase and urate oxidase, were not affected by the loss of STAT5b. A corresponding increase in expression of fatty acyl-CoA oxidase and L-bifunctional enzyme mRNA, as well as PPARalpha mRNA, was observed in the STAT5b-deficient mice, suggesting a transcriptional mechanism for the observed increases. Although basal liver expression of PPARalpha and its target genes was thus elevated in STAT5b(-/-) mice, the clofibrate-induced level of enzyme expression was unaffected, suggesting that the inhibitory effects of STAT5b are overcome at high concentrations of PPARalpha activators. These findings support the hypothesis that GH and potentially other endogenous activators of STAT5b help to maintain liver PPARalpha function at a low basal level and may thereby moderate PPARalpha-dependent hepatocarcinogenesis and other responses stimulated by exposure to low levels of environmental chemicals of the peroxisome proliferator class.
Collapse
MESH Headings
- Acetyl-CoA C-Acyltransferase/biosynthesis
- Acetyl-CoA C-Acyltransferase/genetics
- Acyl-CoA Oxidase
- Animals
- Blotting, Western
- Catalase/biosynthesis
- Catalase/genetics
- Cytochrome P-450 CYP4A
- Cytochrome P-450 Enzyme System/biosynthesis
- Cytochrome P-450 Enzyme System/genetics
- DNA-Binding Proteins/metabolism
- Enoyl-CoA Hydratase/biosynthesis
- Enoyl-CoA Hydratase/genetics
- Female
- Gene Expression Regulation, Enzymologic/physiology
- Liver/enzymology
- Liver/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microsomes, Liver/enzymology
- Microsomes, Liver/metabolism
- Milk Proteins
- Mixed Function Oxygenases/biosynthesis
- Mixed Function Oxygenases/genetics
- Oxidoreductases/biosynthesis
- Oxidoreductases/genetics
- RNA, Messenger/chemistry
- RNA, Messenger/genetics
- Receptor Cross-Talk/physiology
- Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors
- Receptors, Cytoplasmic and Nuclear/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- STAT5 Transcription Factor
- Trans-Activators/metabolism
- Transcription Factors/antagonists & inhibitors
- Transcription Factors/metabolism
- Urate Oxidase/biosynthesis
Collapse
Affiliation(s)
- Yuan Chun Zhou
- Department of Biology, Division of Cell and Molecular Biology, Boston University, Boston, Massachusetts 02215, USA
| | | | | | | | | |
Collapse
|
47
|
Grant A, Staffas L, Mancowitz L, Kelly VP, Manson MM, DePierre JW, Hayes JD, Ellis EM, Mancowiz L. Expression of rat aldehyde reductase AKR7A1: influence of age and sex and tissue-specific inducibility. Biochem Pharmacol 2001; 62:1511-9. [PMID: 11728387 DOI: 10.1016/s0006-2952(01)00771-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The regulation of the aldo-keto reductase AKR7A1 was examined in the livers of male and female rats during development by using Western blots, and its contribution to carbonyl metabolism was assessed by using enzyme assays. Hepatic levels of AKR7A1 are low in fetal rats and rise to a peak at around 6 weeks of age in animals of both sexes. Higher levels of the enzyme are found in adult male rat liver than in adult female rat liver. The reductase, therefore, appears to be subject to sex-specific regulation. The effect of growth hormone in mediating this difference in expression was examined by using hypophysectomized animals whose serum growth hormone levels had been feminized by continuous administration. Results demonstrate that such treatment leads to a reduction in AKR7A1 expression. AKR7A1 was found to be constitutively expressed in rat tissues such as liver, kidney, small intestine, and testis, but it was not detected in nasal mucosa, skeletal muscle, heart, adrenal gland, brain, or spleen. However, AKR7A1 was inducible by the synthetic antioxidant ethoxyquin in liver, kidney, and small intestine, but not in the other tissues examined. These results show that levels of this important detoxication enzyme vary considerably according to age and sex and that dietary antioxidants can also influence its level in several tissues.
Collapse
Affiliation(s)
- A Grant
- Department of Bioscience & Biotechnology, University of Strathclyde, 204, George Street, G1 1XW, Scotland, Glasgow, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Park SH, Waxman DJ. Inhibitory cross-talk between STAT5b and liver nuclear factor HNF3beta: impact on the regulation of growth hormone pulse-stimulated, male-specific liver cytochrome P-450 gene expression. J Biol Chem 2001; 276:43031-9. [PMID: 11562369 DOI: 10.1074/jbc.m107597200] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
STAT5b is repeatedly activated in rodent liver by the male pattern of intermittent plasma growth hormone (GH) stimulation and is required to maintain the GH pulse-regulated, male-specific pattern of liver gene expression. We presently investigate the interactions between STAT5b and hepatocyte-enriched nuclear factors (HNFs) that contribute to regulation of GH pulse-inducible, male-specific liver cytochrome P-450 (CYP) genes. STAT5 binding sites were identified in the 5'-flank of the adult male-expressed genes CYP2A2 (nucleotides -2255 to -2247), CYP4A2 (nucleotides -1872 to -1864), and CYP2C11 (nucleotides -1150 to -1142). STAT5-DNA complexes were formed by each CYP sequence with nuclear extract from GH pulse-activated male, but not female, rat liver. The CYP2C11 STAT5 site, which is flanked by HNF3 consensus sequences, conferred STAT5b-inducible reporter gene activity in GH-treated HepG2 cells. trans-Activation of the intact CYP2C11 promoter (1.8-kilobase 5'-flank) was strongly induced by the liver nuclear factors HNF1alpha and HNF3beta but, unexpectedly, was inhibited by GH-activated STAT5b. This STAT5b inhibitory effect could be reversed by HNF1alpha and reflects a functional antagonism between STAT5b and HNF3beta, as evidenced by the inhibition of HNF3beta DNA binding and transcriptional activity by STAT5b. HNF3beta, in turn, inhibited STAT5b by a novel mechanism that leads to suppression of GH-inducible STAT5b tyrosine phosphorylation, DNA binding activity, and transcriptional activity. The potential for GH-activated STAT5b to stimulate male-specific liver CYP expression can thus be modulated by HNF3beta, highlighting the complex interrelationship between STAT5b and liver transcription factors controlling expression of GH-regulated CYP genes.
Collapse
Affiliation(s)
- S H Park
- Division of Cell and Molecular Biology, Department of Biology, Boston University, Boston, Massachusetts 02215, USA
| | | |
Collapse
|
49
|
Horikawa K, Kaku H, Nakajima H, Davey HW, Hennighausen L, Iwamoto I, Yasue T, Kariyone A, Takatsu K. Essential role of Stat5 for IL-5-dependent IgH switch recombination in mouse B cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:5018-26. [PMID: 11673510 DOI: 10.4049/jimmunol.167.9.5018] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-5 stimulation of CD38-activated murine splenic B cells induces mu-gamma1 CSR at the DNA level leading to a high level of IgG1 production. Further addition of IL-4 in the system enhances IL-5-dependent mu-gamma1 CSR. Although some of the postreceptor signaling events initiated by IL-5 in activated B cells have been characterized, the involvement of Stat in IL-5 signaling has not been thoroughly evaluated. In this study, we examined the activation of Stat5 and activation-induced cytidine deaminase (AID) in CD38-activated murine splenic B cells by IL-5. The role of Stat5a and Stat5b in IL-5-induced mu-gamma1 CSR and also IgG1 and IgM production was documented, as IL-5 does not act on CD38-stimulated splenic B cells from Stat5a(-/-) and Stat5b(-/-) mice. Expression levels of CD38-induced germline gamma1 transcripts and AID in Stat5a(-/-) and Stat5b(-/-) B cells upon IL-5 stimulation were comparable to those of wild-type B cells. The impaired mu-gamma1 CSR by Stat5b(-/-) B cells, but not by Stat5a(-/-) B cells, was rescued in part by IL-4, as the addition of IL-4 to the culture of CD38- and IL-5-stimulated B cells induced mu-gamma1 CSR leading to IgG1 production. Analysis of cell division cycle number of wild-type B cells revealed that mu-gamma1 CSR was observed after five or six cell divisions. Stat5a(-/-) and Stat5b(-/-) B cells showed similar cell division cycles, but they did not undergo mu-gamma1 CSR. Our data support the notion that both Stat5a and Stat5b are essential for IL-5-dependent mu;-gamma1 CSR and Ig secretion; however, their major target may not be AID. Stat5a and Stat5b are not redundant, but rather are at least partially distinctive in their function.
Collapse
Affiliation(s)
- K Horikawa
- Department of Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Davey HW, Xie T, McLachlan MJ, Wilkins RJ, Waxman DJ, Grattan DR. STAT5b is required for GH-induced liver IGF-I gene expression. Endocrinology 2001; 142:3836-41. [PMID: 11517160 DOI: 10.1210/endo.142.9.8400] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Although the increased expression of Igf-I in liver in response to GH is well characterized, the intracellular signaling pathways that mediate this effect have not been identified. Intracellular signaling molecules belonging to the Janus kinase-signal transducer and activator of transcription 5b (JAK2-STAT5b) pathway are activated by GH and have previously been shown to be required for sexually dimorphic body growth and the expression of liver cytochrome P450 proteins known to be regulated by the gender-specific temporal patterns of pituitary GH secretion. Here, we evaluate the role of STAT5b in GH activation of Igf-I by monitoring the induction of Igf-I mRNA in livers of wild-type and Stat5b(-/-)mice stimulated with exogenous pulses of GH. GH induced the expression of liver Igf-I mRNA in hypophysectomized male wild-type, but not in hypophysectomized male Stat5b(-/-) mice, although the Stat5b(-/-) mice exhibit both normal liver GH receptor expression and strong GH induction of Cytokine-inducible SH2 protein (Cis), which is believed to contribute to the down-regulation of GH-induced liver STAT5b signaling. Thus, STAT5b plays an important and specific role in liver Igf-I gene expression.
Collapse
Affiliation(s)
- H W Davey
- AgResearch, Ruakura Research Centre, Hamilton 2001, New Zealand.
| | | | | | | | | | | |
Collapse
|