101
|
Klammt J, Neumann D, Gevers EF, Andrew SF, Schwartz ID, Rockstroh D, Colombo R, Sanchez MA, Vokurkova D, Kowalczyk J, Metherell LA, Rosenfeld RG, Pfäffle R, Dattani MT, Dauber A, Hwa V. Dominant-negative STAT5B mutations cause growth hormone insensitivity with short stature and mild immune dysregulation. Nat Commun 2018; 9:2105. [PMID: 29844444 PMCID: PMC5974024 DOI: 10.1038/s41467-018-04521-0] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 05/07/2018] [Indexed: 12/24/2022] Open
Abstract
Growth hormone (GH) insensitivity syndrome (GHIS) is a rare clinical condition in which production of insulin-like growth factor 1 is blunted and, consequently, postnatal growth impaired. Autosomal-recessive mutations in signal transducer and activator of transcription (STAT5B), the key signal transducer for GH, cause severe GHIS with additional characteristics of immune and, often fatal, pulmonary complications. Here we report dominant-negative, inactivating STAT5B germline mutations in patients with growth failure, eczema, and elevated IgE but without severe immune and pulmonary problems. These STAT5B missense mutants are robustly tyrosine phosphorylated upon stimulation, but are unable to nuclear localize, or fail to bind canonical STAT5B DNA response elements. Importantly, each variant retains the ability to dimerize with wild-type STAT5B, disrupting the normal transcriptional functions of wild-type STAT5B. We conclude that these STAT5B variants exert dominant-negative effects through distinct pathomechanisms, manifesting in milder clinical GHIS with general sparing of the immune system. Severe growth hormone insensitivity syndrome (GHIS) with immunodeficiency is caused by autosomal recessive mutations in STAT5B. Here the authors report heterozygous STAT5B mutations with dominant-negative effects, causing mild GHIS without immune defects.
Collapse
Affiliation(s)
- Jürgen Klammt
- Department of Women's and Child Health, University Hospital Leipzig, Liebigstrasse 20a, 04103, Leipzig, Germany
| | - David Neumann
- Department of Pediatrics, Faculty of Medicine, University Hospital Hradec Kralove, Charles University, Prague, 500 05, Hradec Kralove, Czech Republic
| | - Evelien F Gevers
- Department of Pediatric Endocrinology, Royal London Children's Hospital, Barts Health NHS Trust, Whitechapel Road, London, E1 1 BB, UK.,Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, First Floor North, John Vane Building, Charterhouse Square, London, EC1M 6BQ, UK
| | - Shayne F Andrew
- Division of Endocrinology, 240 Albert Sabin Way, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - I David Schwartz
- Mercy Kids Pediatric Endocrinology & Diabetes, Mercy Children's Hospital and Mercy Clinic, 1965 S. Fremont, Suite 260, Springfield, MO, 65804, USA
| | - Denise Rockstroh
- Department of Women's and Child Health, University Hospital Leipzig, Liebigstrasse 20a, 04103, Leipzig, Germany
| | - Roberto Colombo
- Institute of Clinical Biochemistry, Faculty of Medicine, Catholic University and IRCCS Policlinico Agostino Gemelli, Largo Francesco Vito 1, I-00168, Rome, Italy.,Center for the Study of Rare Hereditary Diseases, Niguarda Ca' Granda Metropolitan Hospital, Milan, Italy
| | - Marco A Sanchez
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
| | - Doris Vokurkova
- Department of Clinical Immunology and Allergology, Faculty of Medicine, University Hospital Hradec Kralove, Charles University, Prague, 500 05, Hradec Kralove, Czech Republic
| | - Julia Kowalczyk
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, First Floor North, John Vane Building, Charterhouse Square, London, EC1M 6BQ, UK
| | - Louise A Metherell
- Centre for Endocrinology, William Harvey Research Institute, Queen Mary University of London, First Floor North, John Vane Building, Charterhouse Square, London, EC1M 6BQ, UK
| | - Ron G Rosenfeld
- Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Roland Pfäffle
- Department of Women's and Child Health, University Hospital Leipzig, Liebigstrasse 20a, 04103, Leipzig, Germany
| | - Mehul T Dattani
- Section of Genetics and Epigenetics in Health and Disease, Genetics and Genomic Medicine Programme, University College London, Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Andrew Dauber
- Division of Endocrinology, 240 Albert Sabin Way, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA
| | - Vivian Hwa
- Division of Endocrinology, 240 Albert Sabin Way, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, 45229, USA.
| |
Collapse
|
102
|
Liu CZ, Luo Y, Limbu SM, Chen LQ, Du ZY. IGF-1 induces SOCS-2 but not SOCS-1 and SOCS-3 transcription in juvenile Nile tilapia ( Oreochromis niloticus). ACTA ACUST UNITED AC 2018; 221:jeb.179291. [PMID: 29650756 DOI: 10.1242/jeb.179291] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 04/08/2018] [Indexed: 12/11/2022]
Abstract
Insulin-like growth factor-1 (IGF-1) plays a crucial role in regulating growth in vertebrates whereas suppressors of cytokine signaling (SOCS) act as feedback inhibitors of the GH/IGF-1 axis. Although SOCS-2 binds the IGF-1 receptor and inhibits IGF-1-induced STAT3 activation, presently there is no clear evidence as to whether IGF-1 could induce SOCS gene expression. The current study aimed to determine whether IGF-1 could induce the transcription of SOCS in juvenile Nile tilapia (Oreochromis niloticus). We show that there is a common positive relationship between the mRNA expression of IGF-I and SOCS-2 under different nutritional statuses and stimulants, but not the mRNA expression of SOCS-1 and SOCS-3 Furthermore, rhIGF-1 treatment and transcriptional activity assay confirmed the hypothesis that IGF-1 could induce SOCS-2 expression, whereas it had no effect or even decreased the expression of SOCS-1 and SOCS-3 Overall, we obtained evidence that the transcription of SOCS-2, but not SOCS-1 or SOCS-3, could be induced by IGF signaling, suggesting that SOCS-2 serves as a feedback suppressor of the IGF-1 axis in juvenile Nile tilapia.
Collapse
Affiliation(s)
- Cai-Zhi Liu
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai 200241, People's Republic of China
| | - Yuan Luo
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai 200241, People's Republic of China
| | - Samwel Mchele Limbu
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai 200241, People's Republic of China.,Department of Aquatic Sciences and Fisheries Technology, P. O. Box 35064, University of Dar es Salaam, Dar es Salaam, Tanzania
| | - Li-Qiao Chen
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai 200241, People's Republic of China
| | - Zhen-Yu Du
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai 200241, People's Republic of China
| |
Collapse
|
103
|
Caslin HL, Kiwanuka KN, Haque TT, Taruselli MT, MacKnight HP, Paranjape A, Ryan JJ. Controlling Mast Cell Activation and Homeostasis: Work Influenced by Bill Paul That Continues Today. Front Immunol 2018; 9:868. [PMID: 29755466 PMCID: PMC5932183 DOI: 10.3389/fimmu.2018.00868] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/09/2018] [Indexed: 01/13/2023] Open
Abstract
Mast cells are tissue resident, innate immune cells with heterogenous phenotypes tuned by cytokines and other microenvironmental stimuli. Playing a protective role in parasitic, bacterial, and viral infections, mast cells are also known for their role in the pathogenesis of allergy, asthma, and autoimmune diseases. Here, we review factors controlling mast cell activation, with a focus on receptor signaling and potential therapies for allergic disease. Specifically, we will discuss our work with FcεRI and FγR signaling, IL-4, IL-10, and TGF-β1 treatment, and Stat5. We conclude with potential therapeutics for allergic disease. Much of these efforts have been influenced by the work of Bill Paul. With many mechanistic targets for mast cell activation and different classes of therapeutics being studied, there is reason to be hopeful for continued clinical progress in this area.
Collapse
Affiliation(s)
- Heather L Caslin
- Department of Biology, Virginia Commonwealth University, Richmond, VA, United States
| | - Kasalina N Kiwanuka
- Department of Biology, Virginia Commonwealth University, Richmond, VA, United States
| | - Tamara T Haque
- Department of Biology, Virginia Commonwealth University, Richmond, VA, United States
| | - Marcela T Taruselli
- Department of Biology, Virginia Commonwealth University, Richmond, VA, United States
| | - H Patrick MacKnight
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA, United States
| | - Anuya Paranjape
- Department of Biology, Virginia Commonwealth University, Richmond, VA, United States
| | - John J Ryan
- Department of Biology, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
104
|
Hasan S, Naqvi AR, Rizvi A. Transcriptional Regulation of Emergency Granulopoiesis in Leukemia. Front Immunol 2018; 9:481. [PMID: 29593731 PMCID: PMC5858521 DOI: 10.3389/fimmu.2018.00481] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 02/23/2018] [Indexed: 12/16/2022] Open
Abstract
Neutropenic conditions are prevalent in leukemia patients and are often associated with increased susceptibility to infections. In fact, emergency granulopoiesis (EG), a process regulating neutrophil homeostasis in inflammatory conditions and infections, may occur improperly in leukemic conditions, leading to reduced neutrophil counts. Unfortunately, the mechanisms central to dysfunctional EG remain understudied in both leukemia patients and leukemic mouse models. However, despite no direct studies on EG response in leukemia are reported, recently certain transcription factors (TFs) have been found to function at the crossroads of leukemia and EG. In this review, we present an update on TFs that can potentially govern the fate of EG in leukemia. Transcriptional control of Fanconi DNA repair pathway genes is also highlighted, as well as the newly discovered role of Fanconi proteins in innate immune response and EG. Identifying the TFs regulating EG in leukemia and dissecting their underlying mechanisms may facilitate the discovery of therapeutic drugs for the treatment of neutropenia.
Collapse
Affiliation(s)
- Shirin Hasan
- Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Afsar R Naqvi
- Department of Periodontics, University of Illinois at Chicago, Chicago, IL, United States
| | - Asim Rizvi
- Department of Biochemistry, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
105
|
Kusama K, Bai R, Imakawa K. Regulation of human trophoblast cell syncytialization by transcription factors STAT5B and NR4A3. J Cell Biochem 2018; 119:4918-4927. [PMID: 29377304 DOI: 10.1002/jcb.26721] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 01/24/2018] [Indexed: 11/08/2022]
Abstract
In human trophoblast cells, cyclic AMP or its inducer forskolin (FSK) activates two downstream signaling molecules, protein kinase A (PKA) and exchange protein directly activated by cAMP (EPAC), both of which induce syncytialization, cell fusion, and the production of human chorionic gonadotropin (hCG) and progesterone. However, a transcription factor other than GCM1 and molecular mechanisms associated with these events have not been well characterized. To identify novel transcription factors involved in syncytialization of cAMP-stimulated human choriocarcinoma BeWo cells, the microarray analysis was performed with RNAs extracted from PKA- or EPAC-selective cAMP analog-stimulated BeWo cells, from which two up-regulated transcription factors, STAT5 and NR4A3, were found. The knockdown of STAT5B decreased FSK-induced cell fusion and the expression of syncytialization markers, CGB, syncytin1, syncytin2, GCM1, and OVOL1, but NR4A3 knockdown increased FSK-induced cell fusion and the expression of CGB and syncytin2. These findings indicated that cAMP-PKA up-regulated STAT5B, followed by increase in syncytin2 expression through GCM1 and OVOL1, resulting in cell fusion and hCG production, while cAMP-PKA-up-regulated NR4A3 could decrease syncytin2 expression, and suggested that both positive and negative effects of STAT5B and NR4A3, respectively, are required to control the degree of syncytialization in human trophoblast cells.
Collapse
Affiliation(s)
- Kazuya Kusama
- Graduate School of Agricultural and Life Science, The University of Tokyo, Animal Resource Science Center, Kasama, Ibaraki, Japan
| | - Rulan Bai
- Graduate School of Agricultural and Life Science, The University of Tokyo, Animal Resource Science Center, Kasama, Ibaraki, Japan
| | - Kazuhiko Imakawa
- Graduate School of Agricultural and Life Science, The University of Tokyo, Animal Resource Science Center, Kasama, Ibaraki, Japan
| |
Collapse
|
106
|
Tong J, Thompson I, Zhao X, Lacasse P. Effect of 17β-estradiol on milk production, hormone secretion, and mammary gland gene expression in dairy cows. J Dairy Sci 2018; 101:2588-2601. [DOI: 10.3168/jds.2017-13353] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 11/02/2017] [Indexed: 11/19/2022]
|
107
|
Xiao Q, Wu J, Wang WJ, Chen S, Zheng Y, Yu X, Meeth K, Sahraei M, Bothwell ALM, Chen L, Bosenberg M, Chen J, Sexl V, Sun L, Li L, Tang W, Wu D. DKK2 imparts tumor immunity evasion through β-catenin-independent suppression of cytotoxic immune-cell activation. Nat Med 2018; 24:262-270. [PMID: 29431745 PMCID: PMC5840007 DOI: 10.1038/nm.4496] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 01/12/2018] [Indexed: 12/11/2022]
Abstract
Immunotherapy offers new options for cancer treatment, but efficacy varies across cancer types. Colorectal cancers (CRCs) are largely refractory to immune-checkpoint blockade, which suggests the presence of yet uncharacterized immune-suppressive mechanisms. Here we report that the loss of adenomatosis polyposis coli (APC) in intestinal tumor cells or of the tumor suppressor PTEN in melanoma cells upregulates the expression of Dickkopf-related protein 2 (DKK2), which, together with its receptor LRP5, provides an unconventional mechanism for tumor immune evasion. DKK2 secreted by tumor cells acts on cytotoxic lymphocytes, inhibiting STAT5 signaling by impeding STAT5 nuclear localization via LRP5, but independently of LRP6 and the Wnt-β-catenin pathway. Genetic or antibody-mediated ablation of DKK2 activates natural killer (NK) cells and CD8+ T cells in tumors, impedes tumor progression, and enhances the effects of PD-1 blockade. Thus, we have identified a previously unknown tumor immune-suppressive mechanism and immunotherapeutic targets particularly relevant for CRCs and a subset of melanomas.
Collapse
Affiliation(s)
- Qian Xiao
- Vascular Biology and Therapeutic Program and Department of Pharmacology, Yale School of Medicine, New Haven, CT 06520
| | - Jibo Wu
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of sciences, Shanghai 200031, China
| | - Wei-Jia Wang
- Vascular Biology and Therapeutic Program and Department of Pharmacology, Yale School of Medicine, New Haven, CT 06520
| | - Shiyang Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Yingxia Zheng
- Vascular Biology and Therapeutic Program and Department of Pharmacology, Yale School of Medicine, New Haven, CT 06520
| | - Xiaoqing Yu
- Biostatistics Department, Yale University, New Haven, CT 06520
| | - Katrina Meeth
- Departments of Dermatology and Pathology, Yale School of Medicine, New Haven, CT 06520
| | - Mahnaz Sahraei
- Vascular Biology and Therapeutic Program and Department of Pharmacology, Yale School of Medicine, New Haven, CT 06520
| | - Alfred L. M. Bothwell
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520
| | - Lieping Chen
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520
| | - Marcus Bosenberg
- Departments of Dermatology and Pathology, Yale School of Medicine, New Haven, CT 06520
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06520
| | - Jianfeng Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences; University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, Department for Biomedical Sciences, University of Veterinary Medicine Vienna, Vienna, Austria
| | | | - Lin Li
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of sciences, Shanghai 200031, China
| | - Wenwen Tang
- Vascular Biology and Therapeutic Program and Department of Pharmacology, Yale School of Medicine, New Haven, CT 06520
| | - Dianqing Wu
- Vascular Biology and Therapeutic Program and Department of Pharmacology, Yale School of Medicine, New Haven, CT 06520
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520
| |
Collapse
|
108
|
Brain STAT5 signaling modulates learning and memory formation. Brain Struct Funct 2018; 223:2229-2241. [PMID: 29460051 DOI: 10.1007/s00429-018-1627-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 02/12/2018] [Indexed: 01/02/2023]
Abstract
The signal transducer and activator of transcription 5 (STAT5) is a transcription factor recruited by numerous cytokines. STAT5 is important for several physiological functions, including body and tissue growth, mammary gland development, immune system and lipid metabolism. However, the role of STAT5 signaling for brain functions is still poorly investigated, especially regarding cognitive aspects. Therefore, the objective of the present study was to investigate whether brain STAT5 signaling modulates learning and memory formation. For this purpose, brain-specific STAT5 knockout (STAT5 KO) mice were studied in well-established memory tests. Initially, we confirmed a robust reduction in STAT5a and STAT5b mRNA levels in different brain structures of STAT5 KO mice. STAT5 KO mice showed no significant alterations in metabolism, growth, somatotropic axis and spontaneous locomotor activity. In contrast, brain-specific STAT5 ablation impaired learning and memory formation in the novel object recognition, Barnes maze and contextual fear conditioning tests. To unravel possible mechanisms that might underlie the memory deficits of STAT5 KO mice, we assessed neurogenesis in the hippocampus, but no significant differences were observed between groups. On the other hand, reduced insulin-like growth factor-1 (IGF-1) mRNA expression was found in the hippocampus and hypothalamus of STAT5 KO mice. These findings collectively indicate that brain STAT5 signaling is required to attain normal learning and memory. Therefore, STAT5 is an important downstream cellular mechanism shared by several cytokines to regulate cognitive functions.
Collapse
|
109
|
Leehy KA, Truong TH, Mauro LJ, Lange CA. Progesterone receptors (PR) mediate STAT actions: PR and prolactin receptor signaling crosstalk in breast cancer models. J Steroid Biochem Mol Biol 2018; 176:88-93. [PMID: 28442393 PMCID: PMC5653461 DOI: 10.1016/j.jsbmb.2017.04.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/28/2017] [Accepted: 04/20/2017] [Indexed: 12/17/2022]
Abstract
Estrogen is the major mitogenic stimulus of mammary gland development during puberty wherein ER signaling acts to induce abundant PR expression. PR signaling, in contrast, is the primary driver of mammary epithelial cell proliferation in adulthood. The high circulating levels of progesterone during pregnancy signal through PR, inducing expression of the prolactin receptor (PRLR). Cooperation between PR and prolactin (PRL) signaling, via regulation of downstream components in the PRL signaling pathway including JAKs and STATs, facilitates the alveolar morphogenesis observed during pregnancy. Indeed, these pathways are fully integrated via activation of shared signaling pathways (i.e. JAKs, MAPKs) as well as by the convergence of PRs and STATs at target genes relevant to both mammary gland biology and breast cancer progression (i.e. proliferation, stem cell outgrowth, tissue cell type heterogeneity). Thus, rather than a single mediator such as ER, transcription factor cascades (ER>PR>STATs) are responsible for rapid proliferative and developmental programming in the normal mammary gland. It is not surprising that these same mediators typify uncontrolled proliferation in a majority of breast cancers, where ER and PR are most often co-expressed and may cooperate to drive malignant tumor progression. This review will primarily focus on the integration of PR and PRL signaling in breast cancer models and the importance of this cross-talk in cancer progression in the context of mammographic density. Components of these PR/PRL signaling pathways could offer alternative drug targets and logical complements to anti-ER or anti-estrogen-based endocrine therapies.
Collapse
Affiliation(s)
- Katherine A Leehy
- Departments of Medicine and Pharmacology, University of Minnesota Masonic Cancer Center, Minneapolis, MN, 55455, United States
| | - Thu H Truong
- Departments of Medicine and Pharmacology, University of Minnesota Masonic Cancer Center, Minneapolis, MN, 55455, United States
| | - Laura J Mauro
- Department of Animal Sciences, University of Minnesota Masonic Cancer Center, Minneapolis, MN, 55455, United States
| | - Carol A Lange
- Departments of Medicine and Pharmacology, University of Minnesota Masonic Cancer Center, Minneapolis, MN, 55455, United States.
| |
Collapse
|
110
|
Reichenstein M, Rauner G, Kfir S, Kisliouk T, Barash I. Luminal STAT5 mediates H2AX promoter activity in distinct population of basal mammary epithelial cells. Oncotarget 2018; 7:41781-41797. [PMID: 27260000 PMCID: PMC5173096 DOI: 10.18632/oncotarget.9718] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 05/20/2016] [Indexed: 01/08/2023] Open
Abstract
Deregulated STAT5 activity in the mammary gland causes parity-dependent tumorigenesis. Epithelial cell cultures transfected with constitutively active STAT5 express higher levels of the histone H2AX than their non-transfected counterparts. Higher H2AX expression may be involved in tumorigenesis. Here, we aimed to link high STAT5 activity to H2AX–GFP expression by looking for distinct types of mammary cells that express these proteins. In vitro and in transgenic mice, only 0.2 and 0.02%, respectively, of the cells expressed the H2AX–GFP hybrid gene. Its expression correlated with that of the endogenous H2AX gene, suggesting that detectable H2AX–GFP expression marks high levels of H2AX transcript. Methylation of the H2AX promoter characterized non-GFP-expressing H2AX–GFP cells and was inversely correlated with promoter activity. Administration of 5-azacytidine increased H2AX promoter activity in an activated STAT5-dependent manner. In transgenic mice, H2AX–GFP expression peaked at pregnancy. The number of H2AX–GFP-expressing cells and GFP expression decreased in a Stat5a-null background and increased in mice expressing the hyperactivated STAT5. Importantly, H2AX–GFP activity was allocated to basal mammary cells lacking stem-cell properties, whereas STAT5 hyperactivity was detected in the adjacent luminal cells. Knockdown of RANKL by siRNA suggested its involvement in signaling between the two layers. These results suggest paracrine activation of H2AX via promoter demethylation in specific populations of basal mammary cells that is induced by a signal from neighboring luminal cells with hyper STAT5 activity. This pathway provides an alternative route for the luminally confined STAT5 to affect basal mammary cell activity.
Collapse
Affiliation(s)
| | - Gat Rauner
- Institute of Animal Science, ARO, The Volcani Center, Bet-Dagan, Israel.,The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Shenhav Kfir
- Institute of Animal Science, ARO, The Volcani Center, Bet-Dagan, Israel.,The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Tatiana Kisliouk
- Institute of Animal Science, ARO, The Volcani Center, Bet-Dagan, Israel
| | - Itamar Barash
- Institute of Animal Science, ARO, The Volcani Center, Bet-Dagan, Israel
| |
Collapse
|
111
|
Liao Y, Du W. An Rb family-independent E2F3 transcription factor variant impairs STAT5 signaling and mammary gland remodeling during pregnancy in mice. J Biol Chem 2018; 293:3156-3167. [PMID: 29330306 DOI: 10.1074/jbc.ra117.000583] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 01/08/2018] [Indexed: 01/02/2023] Open
Abstract
E2F transcription factors are regulated by binding to the retinoblastoma (Rb) tumor suppressor family of proteins. Previously, we reported an E2FLQ mutation that disrupts the binding with Rb proteins without affecting the transcriptional activity of E2F. We also showed that mouse embryonic fibroblasts with an E2F3LQ mutation exhibit increased E2F activity and more rapid cell proliferation. In this report, we analyzed E2F3LQ mice to further characterize the in vivo consequences of Rb family-independent E2F3 activity. We found that homozygous E2F3LQ mice were viable and had no obvious developmental defects or tumor growth. Our results also indicated that E2F3LQ cells largely retain normal control of cell proliferation in vivo However, female E2F3LQ mice had partial nursing defects. Examination of the E2F3LQ mammary glands revealed increased caveolin-1 (CAV1) expression, reduced prolactin receptor/Stat5 signaling, and impaired pregnancy-induced cell proliferation and differentiation. Of note, ChIP experiments disclosed that E2F3 binds the CAV1 promoter. Furthermore, E2F3 overexpression induced CAV1 expression, and CRISPR/CAS9-mediated E2F3 knockout reduced CAV1 levels and also increased prolactin receptor-induced Stat5 signaling in mammary epithelial cells. Our results suggest that the Rb family-independent E2F3 LQ variant inhibits pregnancy-induced mammary gland cell proliferation and differentiation by up-regulating CAV1 expression and inhibiting Stat5 signaling.
Collapse
Affiliation(s)
- Yang Liao
- From the Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois 60637
| | - Wei Du
- From the Ben May Department for Cancer Research, University of Chicago, Chicago, Illinois 60637
| |
Collapse
|
112
|
|
113
|
|
114
|
Rotwein P. Diversification of the insulin-like growth factor 1 gene in mammals. PLoS One 2017; 12:e0189642. [PMID: 29240807 PMCID: PMC5730178 DOI: 10.1371/journal.pone.0189642] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 11/29/2017] [Indexed: 12/12/2022] Open
Abstract
Insulin-like growth factor 1 (IGF1), a small, secreted peptide growth factor, is involved in a variety of physiological and patho-physiological processes, including somatic growth, tissue repair, and metabolism of carbohydrates, proteins, and lipids. IGF1 gene expression appears to be controlled by several different signaling cascades in the few species in which it has been evaluated, with growth hormone playing a major role by activating a pathway involving the Stat5b transcription factor. Here, genes encoding IGF1 have been evaluated in 25 different mammalian species representing 15 different orders and ranging over ~180 million years of evolutionary diversification. Parts of the IGF1 gene have been fairly well conserved. Like rat Igf1 and human IGF1, 21 of 23 other genes are composed of 6 exons and 5 introns, and all 23 also contain recognizable tandem promoters, each with a unique leader exon. Exon and intron lengths are similar in most species, and DNA sequence conservation is moderately high in orthologous exons and proximal promoter regions. In contrast, putative growth hormone-activated Stat5b-binding enhancers found in analogous locations in rodent Igf1 and in human IGF1 loci, have undergone substantial variation in other mammals, and a processed retro-transposed IGF1 pseudogene is found in the sloth locus, but not in other mammalian genomes. Taken together, the fairly high level of organizational and nucleotide sequence similarity in the IGF1 gene among these 25 species supports the contention that some common regulatory pathways had existed prior to the beginning of mammalian speciation.
Collapse
Affiliation(s)
- Peter Rotwein
- Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas, United States of America
- * E-mail:
| |
Collapse
|
115
|
Wilkinson DS, Ghosh D, Nickle RA, Moorman CD, Mannie MD. Partial CD25 Antagonism Enables Dominance of Antigen-Inducible CD25 high FOXP3 + Regulatory T Cells As a Basis for a Regulatory T Cell-Based Adoptive Immunotherapy. Front Immunol 2017; 8:1782. [PMID: 29312311 PMCID: PMC5735073 DOI: 10.3389/fimmu.2017.01782] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 11/29/2017] [Indexed: 02/02/2023] Open
Abstract
FOXP3+ regulatory T cells (Tregs) represent a promising platform for effective adoptive immunotherapy of chronic inflammatory disease, including autoimmune diseases such as multiple sclerosis. Successful Treg immunotherapy however requires new technologies to enable long-term expansion of stable, antigen-specific FOXP3+ Tregs in cell culture. Antigen-specific activation of naïve T cells in the presence of TGF-β elicits the initial differentiation of the FOXP3+ lineage, but these Treg lines lack phenotypic stability and rapidly transition to a conventional T cell (Tcon) phenotype during in vitro propagation. Because Tregs and Tcons differentially express CD25, we hypothesized that anti-CD25 monoclonal antibodies (mAbs) would only partially block IL-2 signaling in CD25high FOXP3+ Tregs while completely blocking IL-2 responses of CD25low-intermediate Tcons to enable preferential outgrowth of Tregs during in vitro propagation. Indeed, murine TGF-β-induced MOG-specific Treg lines from 2D2 transgenic mice that were maintained in IL-2 with the anti-CD25 PC61 mAb rapidly acquired and indefinitely maintained a FOXP3high phenotype during long-term in vitro propagation (>90% FOXP3+ Tregs), whereas parallel cultures lacking PC61 rapidly lost FOXP3. These results pertained to TGF-β-inducible "iTregs" because Tregs from 2D2-FIG Rag1-/- mice, which lack thymic or natural Tregs, were stabilized by continuous culture in IL-2 and PC61. MOG-specific and polyclonal Tregs upregulated the Treg-associated markers Neuropilin-1 (NRP1) and Helios (IKZF2). Just as PC61 stabilized FOXP3+ Tregs during expansion in IL-2, TGF-β fully stabilized FOXP3+ Tregs during cellular activation in the presence of dendritic cells and antigen/mitogen. Adoptive transfer of blastogenic CD25high FOXP3+ Tregs from MOG35-55-specific 2D2 TCR transgenic mice suppressed experimental autoimmune encephalomyelitis in pretreatment and therapeutic protocols. In conclusion, low IL-2 concentrations coupled with high PC61 concentrations constrained IL-2 signaling to a low-intensity range that enabled dominant stable outgrowth of suppressive CD25high FOXP3+ Tregs. The ability to indefinitely expand stable Treg lines will provide insight into FOXP3+ Treg physiology and will be foundational for Treg-based immunotherapy.
Collapse
Affiliation(s)
- Daniel S Wilkinson
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Debjani Ghosh
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Rebecca A Nickle
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Cody D Moorman
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Mark D Mannie
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| |
Collapse
|
116
|
Richard AJ, Hang H, Stephens JM. Pyruvate dehydrogenase complex (PDC) subunits moonlight as interaction partners of phosphorylated STAT5 in adipocytes and adipose tissue. J Biol Chem 2017; 292:19733-19742. [PMID: 28982698 PMCID: PMC5712614 DOI: 10.1074/jbc.m117.811794] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/03/2017] [Indexed: 12/19/2022] Open
Abstract
STAT5 proteins play a role in adipocyte development and function, but their specific functions are largely unknown. To this end, we used an unbiased MS-based approach to identify novel STAT5-interacting proteins. We observed that STAT5A bound the E1β and E2 subunits of the pyruvate dehydrogenase complex (PDC). Whereas STAT5A typically localizes to the cytosol or nucleus, PDC normally resides within the mitochondrial matrix where it converts pyruvate to acetyl-CoA. We employed affinity purification and immunoblotting to validate the interaction between STAT5A and PDC subunits in murine and human cultured adipocytes, as well as in adipose tissue. We found that multiple PDC subunits interact with hormone-activated STAT5A in a dose- and time-dependent manner that coincides with tyrosine phosphorylation of STAT5. Using subcellular fractionation and immunofluorescence microscopy, we observed that PDC-E2 is present within the adipocyte nucleus where it associates with STAT5A. Because STAT5A is a transcription factor, we used chromatin immunoprecipitation (ChIP) to assess PDC's ability to interact with STAT5 DNA-binding sites. These analyses revealed that PDC-E2 is bound to a STAT5-binding site in the promoter of the STAT5 target gene cytokine-inducible SH2-containing protein (cish). We have demonstrated a compelling interaction between STAT5A and PDC subunits in adipocytes under physiological conditions. There is previous evidence that PDC localizes to cancer cell nuclei where it plays a role in histone acetylation. On the basis of our ChIP data and these previous findings, we hypothesize that PDC may modulate STAT5's ability to regulate gene expression by controlling histone or STAT5 acetylation.
Collapse
Affiliation(s)
- Allison J Richard
- From the Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808 and
| | - Hardy Hang
- From the Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808 and
| | - Jacqueline M Stephens
- From the Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808 and
- the Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana 70803
| |
Collapse
|
117
|
|
118
|
Gillinder KR, Tuckey H, Bell CC, Magor GW, Huang S, Ilsley MD, Perkins AC. Direct targets of pSTAT5 signalling in erythropoiesis. PLoS One 2017; 12:e0180922. [PMID: 28732065 PMCID: PMC5521770 DOI: 10.1371/journal.pone.0180922] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 06/25/2017] [Indexed: 12/29/2022] Open
Abstract
Erythropoietin (EPO) acts through the dimeric erythropoietin receptor to stimulate proliferation, survival, differentiation and enucleation of erythroid progenitor cells. We undertook two complimentary approaches to find EPO-dependent pSTAT5 target genes in murine erythroid cells: RNA-seq of newly transcribed (4sU-labelled) RNA, and ChIP-seq for pSTAT5 30 minutes after EPO stimulation. We found 302 pSTAT5-occupied sites: ~15% of these reside in promoters while the rest reside within intronic enhancers or intergenic regions, some >100kb from the nearest TSS. The majority of pSTAT5 peaks contain a central palindromic GAS element, TTCYXRGAA. There was significant enrichment for GATA motifs and CACCC-box motifs within the neighbourhood of pSTAT5-bound peaks, and GATA1 and/or KLF1 co-occupancy at many sites. Using 4sU-RNA-seq we determined the EPO-induced transcriptome and validated differentially expressed genes using dynamic CAGE data and qRT-PCR. We identified known direct pSTAT5 target genes such as Bcl2l1, Pim1 and Cish, and many new targets likely to be involved in driving erythroid cell differentiation including those involved in mRNA splicing (Rbm25), epigenetic regulation (Suv420h2), and EpoR turnover (Clint1/EpsinR). Some of these new EpoR-JAK2-pSTAT5 target genes could be used as biomarkers for monitoring disease activity in polycythaemia vera, and for monitoring responses to JAK inhibitors.
Collapse
Affiliation(s)
- Kevin R. Gillinder
- Cancer Genomics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Queensland, Australia
| | - Hugh Tuckey
- Cancer Genomics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Queensland, Australia
- Faculty of Medicine and Biomedical Sciences, University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Charles C. Bell
- Cancer Genomics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Queensland, Australia
| | - Graham W. Magor
- Cancer Genomics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Queensland, Australia
| | - Stephen Huang
- Cancer Genomics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Queensland, Australia
- Faculty of Medicine and Biomedical Sciences, University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Melissa D. Ilsley
- Cancer Genomics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Queensland, Australia
- Faculty of Medicine and Biomedical Sciences, University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Andrew C. Perkins
- Cancer Genomics Group, Mater Research Institute - University of Queensland, Translational Research Institute, Woolloongabba, Brisbane, Queensland, Australia
- Faculty of Medicine and Biomedical Sciences, University of Queensland, St. Lucia, Brisbane, Queensland, Australia
- Princess Alexandra Hospital, Brisbane, Queensland, Australia
- * E-mail:
| |
Collapse
|
119
|
Sivasubramaniyam T, Schroer SA, Li A, Luk CT, Shi SY, Besla R, Dodington DW, Metherel AH, Kitson AP, Brunt JJ, Lopes J, Wagner KU, Bazinet RP, Bendeck MP, Robbins CS, Woo M. Hepatic JAK2 protects against atherosclerosis through circulating IGF-1. JCI Insight 2017; 2:93735. [PMID: 28724798 DOI: 10.1172/jci.insight.93735] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 06/06/2017] [Indexed: 01/12/2023] Open
Abstract
Atherosclerosis is considered both a metabolic and inflammatory disease; however, the specific tissue and signaling molecules that instigate and propagate this disease remain unclear. The liver is a central site of inflammation and lipid metabolism that is critical for atherosclerosis, and JAK2 is a key mediator of inflammation and, more recently, of hepatic lipid metabolism. However, precise effects of hepatic Jak2 on atherosclerosis remain unknown. We show here that hepatic Jak2 deficiency in atherosclerosis-prone mouse models exhibited accelerated atherosclerosis with increased plaque macrophages and decreased plaque smooth muscle cell content. JAK2's essential role in growth hormone signalling in liver that resulted in reduced IGF-1 with hepatic Jak2 deficiency played a causal role in exacerbating atherosclerosis. As such, restoring IGF-1 either pharmacologically or genetically attenuated atherosclerotic burden. Together, our data show hepatic Jak2 to play a protective role in atherogenesis through actions mediated by circulating IGF-1 and, to our knowledge, provide a novel liver-centric mechanism in atheroprotection.
Collapse
Affiliation(s)
- Tharini Sivasubramaniyam
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science
| | - Stephanie A Schroer
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Angela Li
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Immunology
| | - Cynthia T Luk
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science
| | - Sally Yu Shi
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science
| | - Rickvinder Besla
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology
| | - David W Dodington
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Adam H Metherel
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Alex P Kitson
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Jara J Brunt
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science
| | - Joshua Lopes
- Department of Laboratory Medicine and Pathobiology
| | - Kay-Uwe Wagner
- Eppley Institute for Research in Cancer and Allied Diseases and the Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Richard P Bazinet
- Department of Nutritional Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Michelle P Bendeck
- Department of Laboratory Medicine and Pathobiology.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Clinton S Robbins
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Immunology.,Department of Laboratory Medicine and Pathobiology
| | - Minna Woo
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Medical Science.,Department of Immunology.,Division of Endocrinology and Metabolism, Department of Medicine, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
120
|
Silveira MA, Furigo IC, Zampieri TT, Bohlen TM, de Paula DG, Franci CR, Donato J, Frazao R. STAT5 signaling in kisspeptin cells regulates the timing of puberty. Mol Cell Endocrinol 2017; 448:55-65. [PMID: 28344041 DOI: 10.1016/j.mce.2017.03.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 03/03/2017] [Accepted: 03/22/2017] [Indexed: 12/26/2022]
Abstract
Previous studies have shown that kisspeptin neurons are important mediators of prolactin's effects on reproduction. However, the cellular mechanisms recruited by prolactin to affect kisspeptin neurons remain unknown. Using whole-cell patch-clamp recordings of brain slices from kisspeptin reporter mice, we observed that 20% of kisspeptin neurons in the anteroventral periventricular nucleus was indirectly depolarized by prolactin via an unknown population of prolactin responsive neurons. This effect required the phosphatidylinositol 3-kinase signaling pathway. No effects on the activity of arcuate kisspeptin neurons were observed, despite a high percentage (70%) of arcuate neurons expressing prolactin-induced STAT5 phosphorylation. To determine whether STAT5 expression in kisspeptin cells regulates reproduction, mice carrying Stat5a/b inactivation specifically in kisspeptin cells were generated. These mutants exhibited an early onset of estrous cyclicity, indicating that STAT5 transcription factors exert an inhibitory effect on the timing of puberty.
Collapse
Affiliation(s)
- Marina Augusto Silveira
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Isadora C Furigo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Thais T Zampieri
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Tabata M Bohlen
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Daniella G de Paula
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Celso Rodrigues Franci
- Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Renata Frazao
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
121
|
Murtaza M, Khan G, Aftab MF, Afridi SK, Ghaffar S, Ahmed A, Hafizur RM, Waraich RS. Cucurbitacin E reduces obesity and related metabolic dysfunction in mice by targeting JAK-STAT5 signaling pathway. PLoS One 2017; 12:e0178910. [PMID: 28598969 PMCID: PMC5466318 DOI: 10.1371/journal.pone.0178910] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 05/19/2017] [Indexed: 01/14/2023] Open
Abstract
Several members of cucurbitaceae family have been reported to regulate growth of cancer by interfering with STAT3 signaling. In the present study, we investigated the unique role and molecular mechanism of cucurbitacins (Cucs) in reducing symptoms of metabolic syndrome in mice. Cucurbitacin E (CuE) was found to reduce adipogenesis in murine adipocytes. CuE treatment diminished hypertrophy of adipocytes, visceral obesity and lipogenesis gene expression in diet induced mice model of metabolic syndrome (MetS). CuE also ameliorated adipose tissue dysfunction by reducing hyperleptinemia and TNF-alpha levels and enhancing hypoadiponectinemia. Results show that CuE mediated these effects by attenuating Jenus kinase- Signal transducer and activator of transcription 5 (JAK- STAT5) signaling in visceral fat tissue. As a result, CuE treatment also reduced PPAR gamma expression. Glucose uptake enhanced in adipocytes after stimulation with CuE and insulin resistance diminished in mice treated with CuE, as reflected by reduced glucose intolerance and glucose stimulated insulin secretion. CuE restored insulin sensitivity indirectly by inhibiting JAK phosphorylation and improving AMPK activity. Consequently, insulin signaling was up-regulated in mice muscle. As CuE positively regulated adipose tissue function and suppressed visceral obesity, dyslipedemia, hyperglycemia and insulin resistance in mice model of MetS, we suggest that CuE can be used as novel approach to treat metabolic diseases.
Collapse
Affiliation(s)
- Munazza Murtaza
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Gulnaz Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Meha Fatima Aftab
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Shabbir Khan Afridi
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Safina Ghaffar
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Ayaz Ahmed
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Rahman M Hafizur
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Rizwana Sanaullah Waraich
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| |
Collapse
|
122
|
Rosenfeld RG, Hwa V. Biology of the somatotroph axis (after the pituitary). ANNALES D'ENDOCRINOLOGIE 2017; 78:80-82. [PMID: 28495324 DOI: 10.1016/j.ando.2017.04.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Normal growth requires that pituitary-secreted growth hormone (GH) bind to its specific receptor and activate a complex signaling cascade, leaving to production of insulin-like growth factor-I (IGF-I), which, in turn, activates its own receptor (IGF1R). The GH receptor (GHR) is preformed as a dimer and is transported in a nonligand bound state to the cell surface. Binding of GH to the GHR dimer, results in a conformational change of the dimer, activation of the intracellular Janus Kinase 2 (JAK2) and phosphorylation of signal transducer and activator of transcription (STAT) 5B. Phosphorylated STAT5B dimers are then translocated to the nucleus, where they transcriptionally activate multiple genes, including those for IGF-I, IGF binding protein-3 and the acid-labile subunit (ALS).
Collapse
Affiliation(s)
- Ron G Rosenfeld
- Oregon Health & Science University, Portland, Oregon, United States.
| | - Vivian Hwa
- Cincinnati Center for Growth Disorders, Cincinnati, Ohio, United States; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States
| |
Collapse
|
123
|
Pedersen MH, Svart MV, Lebeck J, Bidlingmaier M, Stødkilde-Jørgensen H, Pedersen SB, Møller N, Jessen N, Jørgensen JOL. Substrate Metabolism and Insulin Sensitivity During Fasting in Obese Human Subjects: Impact of GH Blockade. J Clin Endocrinol Metab 2017; 102:1340-1349. [PMID: 28324055 DOI: 10.1210/jc.2016-3835] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 01/13/2017] [Indexed: 12/23/2022]
Abstract
CONTEXT Insulin resistance and metabolic inflexibility are features of obesity and are amplified by fasting. Growth hormone (GH) secretion increases during fasting and GH causes insulin resistance. OBJECTIVE To study the metabolic effects of GH blockade during fasting in obese subjects. SUBJECTS AND METHODS Nine obese males were studied thrice in a randomized design: (1) after an overnight fast (control), (2) after 72 hour fasting (fasting), and (3) after 72 hour fasting with GH blockade (pegvisomant) [fasting plus GH antagonist (GHA)]. Each study day consisted of a 4-hour basal period followed by a 2-hour hyperinsulinemic, euglycemic clamp combined with indirect calorimetry, assessment of glucose and palmitate turnover, and muscle and fat biopsies. RESULTS GH levels increased with fasting (P < 0.01), and the fasting-induced reduction of serum insulin-like growth factor I was enhanced by GHA (P < 0.05). Fasting increased lipolysis and lipid oxidation independent of GHA, but fasting plus GHA caused a more pronounced suppression of lipid intermediates in response to hyperinsulinemic, euglycemic clamp. Fasting-induced insulin resistance was abrogated by GHA (P < 0.01) primarily due to reduced endogenous glucose production (P = 0.003). Fasting plus GHA also caused elevated glycerol levels and reduced levels of counterregulatory hormones. Fasting significantly reduced the expression of antilipolytic signals in adipose tissue independent of GHA. CONCLUSIONS Suppression of GH activity during fasting in obese subjects reverses insulin resistance and amplifies insulin-stimulated suppression of lipid intermediates, indicating that GH is an important regulator of substrate metabolism, insulin sensitivity, and metabolic flexibility also in obese subjects.
Collapse
Affiliation(s)
- Morten Høgild Pedersen
- Medical Research Laboratory, Department of Endocrinology and Internal Medicine
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Mads Vandsted Svart
- Medical Research Laboratory, Department of Endocrinology and Internal Medicine
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | | | - Martin Bidlingmaier
- Endocrine Research Unit, Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians-Universität, 80336 Munich, Germany
| | - Hans Stødkilde-Jørgensen
- The MR Research Center, and
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Steen Bønløkke Pedersen
- Medical Research Laboratory, Department of Endocrinology and Internal Medicine
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Niels Møller
- Medical Research Laboratory, Department of Endocrinology and Internal Medicine
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - Niels Jessen
- Department of Clinical Pharmacology, Aarhus University Hospital 8000 Aarhus, Denmark
| | - Jens O L Jørgensen
- Medical Research Laboratory, Department of Endocrinology and Internal Medicine
- Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
124
|
Mota de Sá P, Richard AJ, Hang H, Stephens JM. Transcriptional Regulation of Adipogenesis. Compr Physiol 2017; 7:635-674. [PMID: 28333384 DOI: 10.1002/cphy.c160022] [Citation(s) in RCA: 256] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Adipocytes are the defining cell type of adipose tissue. Once considered a passive participant in energy storage, adipose tissue is now recognized as a dynamic organ that contributes to several important physiological processes, such as lipid metabolism, systemic energy homeostasis, and whole-body insulin sensitivity. Therefore, understanding the mechanisms involved in its development and function is of great importance. Adipocyte differentiation is a highly orchestrated process which can vary between different fat depots as well as between the sexes. While hormones, miRNAs, cytoskeletal proteins, and many other effectors can modulate adipocyte development, the best understood regulators of adipogenesis are the transcription factors that inhibit or promote this process. Ectopic expression and knockdown approaches in cultured cells have been widely used to understand the contribution of transcription factors to adipocyte development, providing a basis for more sophisticated in vivo strategies to examine adipogenesis. To date, over two dozen transcription factors have been shown to play important roles in adipocyte development. These transcription factors belong to several families with many different DNA-binding domains. While peroxisome proliferator-activated receptor gamma (PPARγ) is undoubtedly the most important transcriptional modulator of adipocyte development in all types of adipose tissue, members of the CCAAT/enhancer-binding protein, Krüppel-like transcription factor, signal transducer and activator of transcription, GATA, early B cell factor, and interferon-regulatory factor families also regulate adipogenesis. The importance of PPARγ activity is underscored by several covalent modifications that modulate its activity and its ability to modulate adipocyte development. This review will primarily focus on the transcriptional control of adipogenesis in white fat cells and on the mechanisms involved in this fine-tuned developmental process. © 2017 American Physiological Society. Compr Physiol 7:635-674, 2017.
Collapse
Affiliation(s)
- Paula Mota de Sá
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Allison J Richard
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Hardy Hang
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Jacqueline M Stephens
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| |
Collapse
|
125
|
Hall MD, Murray CA, Valdez MJ, Perantoni AO. Mesoderm-specific Stat3 deletion affects expression of Sox9 yielding Sox9-dependent phenotypes. PLoS Genet 2017; 13:e1006610. [PMID: 28166224 PMCID: PMC5319801 DOI: 10.1371/journal.pgen.1006610] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 02/21/2017] [Accepted: 01/30/2017] [Indexed: 01/14/2023] Open
Abstract
To date, mutations within the coding region and translocations around the SOX9 gene both constitute the majority of genetic lesions underpinning human campomelic dysplasia (CD). While pathological coding-region mutations typically result in a non-functional SOX9 protein, little is known about what mechanism(s) controls normal SOX9 expression, and subsequently, which signaling pathways may be interrupted by alterations occurring around the SOX9 gene. Here, we report the identification of Stat3 as a key modulator of Sox9 expression in nascent cartilage and developing chondrocytes. Stat3 expression is predominant in tissues of mesodermal origin, and its conditional ablation using mesoderm-specific TCre, in vivo, causes dwarfism and skeletal defects characteristic of CD. Specifically, Stat3 loss results in the expansion of growth plate hypertrophic chondrocytes and deregulation of normal endochondral ossification in all bones examined. Conditional deletion of Stat3 with a Sox9Cre driver produces palate and tracheal irregularities similar to those described in Sox9+/- mice. Furthermore, mesodermal deletion of Stat3 causes global embryonic down regulation of Sox9 expression and function in vivo. Mechanistic experiments ex vivo suggest Stat3 can directly activate the expression of Sox9 by binding to its proximal promoter following activation. These findings illuminate a novel role for Stat3 in chondrocytes during skeletal development through modulation of a critical factor, Sox9. Importantly, they further provide the first evidence for the modulation of a gene product other than Sox9 itself which is capable of modeling pathological aspects of CD and underscore a potentially valuable therapeutic target for patients with the disorder.
Collapse
Affiliation(s)
- Michael D. Hall
- The Cancer and Developmental Biology Laboratory, National Cancer Institute-Frederick, Frederick, Maryland, United States of America
| | - Caroline A. Murray
- The Cancer and Developmental Biology Laboratory, National Cancer Institute-Frederick, Frederick, Maryland, United States of America
| | - Michael J. Valdez
- The Cancer and Developmental Biology Laboratory, National Cancer Institute-Frederick, Frederick, Maryland, United States of America
| | - Alan O. Perantoni
- The Cancer and Developmental Biology Laboratory, National Cancer Institute-Frederick, Frederick, Maryland, United States of America
| |
Collapse
|
126
|
Hirai M, Arita Y, McGlade CJ, Lee KF, Chen J, Evans SM. Adaptor proteins NUMB and NUMBL promote cell cycle withdrawal by targeting ERBB2 for degradation. J Clin Invest 2017; 127:569-582. [PMID: 28067668 PMCID: PMC5272190 DOI: 10.1172/jci91081] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 11/10/2016] [Indexed: 12/20/2022] Open
Abstract
Failure of trabecular myocytes to undergo appropriate cell cycle withdrawal leads to ventricular noncompaction and heart failure. Signaling of growth factor receptor ERBB2 is critical for myocyte proliferation and trabeculation. However, the mechanisms underlying appropriate downregulation of trabecular ERBB2 signaling are little understood. Here, we have found that the endocytic adaptor proteins NUMB and NUMBL were required for downregulation of ERBB2 signaling in maturing trabeculae. Loss of NUMB and NUMBL resulted in a partial block of late endosome formation, resulting in sustained ERBB2 signaling and STAT5 activation. Unexpectedly, activated STAT5 overrode Hippo-mediated inhibition and drove YAP1 to the nucleus. Consequent aberrant cardiomyocyte proliferation resulted in ventricular noncompaction that was markedly rescued by heterozygous loss of function of either ERBB2 or YAP1. Further investigations revealed that NUMB and NUMBL interacted with small GTPase Rab7 to transition ERBB2 from early to late endosome for degradation. Our studies provide insight into mechanisms by which NUMB and NUMBL promote cardiomyocyte cell cycle withdrawal and highlight previously unsuspected connections between pathways that are important for cardiomyocyte cell cycle reentry, with relevance to ventricular noncompaction cardiomyopathy and regenerative medicine.
Collapse
Affiliation(s)
- Maretoshi Hirai
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD, La Jolla, California, USA
| | - Yoh Arita
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD, La Jolla, California, USA
| | - C. Jane McGlade
- Program in Cell Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kuo-Fen Lee
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute, La Jolla, California, USA
| | | | - Sylvia M. Evans
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD, La Jolla, California, USA
- Department of Medicine and
- Department of Pharmacology, UCSD, La Jolla, California, USA
| |
Collapse
|
127
|
Abstract
STAT5 plays a critical role in the development and function of many cell types. Here, we review the role of STAT5 in the development of T lymphocytes in the thymus and its subsequent role in the differentiation of distinct CD4 + helper and regulatory T-cell subsets.
Collapse
Affiliation(s)
- David L. Owen
- Center for Immunology, Masonic Cancer Center, and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Michael A. Farrar
- Center for Immunology, Masonic Cancer Center, and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA
| |
Collapse
|
128
|
Furigo IC, Ramos-Lobo AM, Frazão R, Donato J. Brain STAT5 signaling and behavioral control. Mol Cell Endocrinol 2016; 438:70-76. [PMID: 27118133 DOI: 10.1016/j.mce.2016.04.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 04/19/2016] [Accepted: 04/22/2016] [Indexed: 02/06/2023]
Abstract
Several growth factors and cytokines recruit the signal transducer and activator of transcription 5 (STAT5) signaling pathway to control cell proliferation, differentiation and apoptosis. Nonetheless, the importance of this transcription factor for brain functions is still poorly understood. Because some STAT5-inducing hormones, such as prolactin and leptin, act in the brain to regulate the expression of motivated behaviors, this signaling pathway is likely involved in behavioral modulation. Therefore, the objective of the present review was to summarize and discuss the available data regarding the possible role of central STAT5 signaling in the regulation of brain functions, especially on behavioral control. We discussed studies that investigated the importance of STAT5 signaling in the regulation of maternal and feeding behaviors. Additionally, we highlighted other behaviors that could be potentially affected by STAT5 signaling. This knowledge may help to understand how motivated behaviors are regulated at the cellular level.
Collapse
Affiliation(s)
- Isadora C Furigo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-000, Brazil
| | - Angela M Ramos-Lobo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-000, Brazil
| | - Renata Frazão
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-900, Brazil
| | - J Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
129
|
Abstract
Adipose tissue plays a central role in regulating whole-body energy and glucose homeostasis through its subtle functions at both organ and systemic levels. On one hand, adipose tissue stores energy in the form of lipid and controls the lipid mobilization and distribution in the body. On the other hand, adipose tissue acts as an endocrine organ and produces numerous bioactive factors such as adipokines that communicate with other organs and modulate a range of metabolic pathways. Moreover, brown and beige adipose tissue burn lipid by dissipating energy in the form of heat to maintain euthermia, and have been considered as a new way to counteract obesity. Therefore, adipose tissue dysfunction plays a prominent role in the development of obesity and its related disorders such as insulin resistance, cardiovascular disease, diabetes, depression and cancer. In this review, we will summarize the recent findings of adipose tissue in the control of metabolism, focusing on its endocrine and thermogenic function.
Collapse
Affiliation(s)
- Liping Luo
- Department of Metabolism and EndocrinologyMetabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Meilian Liu
- Department of Metabolism and EndocrinologyMetabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital,
Central South University, Changsha, Hunan, China
- Department of Biochemistry and Molecular BiologyUniversity of New Mexico Health Sciences Center,
Albuquerque, New Mexico, USA
| |
Collapse
|
130
|
Baddela VS, Onteru SK, Singh D. A syntenic locus on buffalo chromosome 20: novel genomic hotspot for miRNAs involved in follicular-luteal transition. Funct Integr Genomics 2016; 17:321-334. [PMID: 27866284 DOI: 10.1007/s10142-016-0535-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 11/02/2016] [Accepted: 11/04/2016] [Indexed: 12/17/2022]
Abstract
The developmental reorganization of ovarian follicular granulosa cells (GC) during follicular maturation, ovulation, and luteinization require a well-controlled regulation of dynamic gene expression profiles. Recently, microRNAs (miRNAs) were found to be key players of ovarian follicular dynamics. The current study aimed to understand the miRNA regulatory role in follicular-luteal transition by characterizing the miRNA profile through miRNA-seq at different follicular (small, medium, and large) and luteal (early, mid, and late) stages in Indian water buffaloes, mono-ovulatory animals like humans. A total of 517 miRNAs were identified in follicular granulosa cells (GC) and corpus luteum (CL) together. Among them, 2 unique and 40 novel miRNAs were in GC; 15 unique and 45 novel miRNAs were in CL. Among the remaining 415 annotated common miRNAs between GC and CL, 43 have showed significant (p < 0.05) differential expression between GC and CL. Particularly, 39 and 4 miRNAs showed higher expression in CL and GC, respectively, with respect to each other. Genome mapping analysis revealed that 71.7% of differential miRNAs having higher expression in CL compared to GC, and 93% of the unique miRNAs in CL were mapped to a short chromosomal region of 0.7 Mb (67.4 to 68.1 Mb) on chromosome 21 of cows which is syntenic to the buffalo chromosome 20. Clustering of all these miRNAs at this locus suggests it as a chromosomal hotspot for miRNAs involved in follicular-luteal transition, especially for CL physiological functions.
Collapse
Affiliation(s)
- Vijay Simha Baddela
- Molecular Endocrinology, Functional Genomics & Systems Biology Laboratory, Animal Biochemistry Division, ICAR-National Dairy Research Institute (Deemed University), Karnal, 132001, Haryana, India
| | - Suneel Kumar Onteru
- Molecular Endocrinology, Functional Genomics & Systems Biology Laboratory, Animal Biochemistry Division, ICAR-National Dairy Research Institute (Deemed University), Karnal, 132001, Haryana, India
| | - Dheer Singh
- Molecular Endocrinology, Functional Genomics & Systems Biology Laboratory, Animal Biochemistry Division, ICAR-National Dairy Research Institute (Deemed University), Karnal, 132001, Haryana, India.
| |
Collapse
|
131
|
Donato J, Frazão R. Interactions between prolactin and kisspeptin to control reproduction. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2016; 60:587-595. [PMID: 27901187 PMCID: PMC10522168 DOI: 10.1590/2359-3997000000230] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 09/26/2016] [Indexed: 11/21/2022]
Abstract
Prolactin is best known for its effects of stimulating mammary gland development and lactogenesis. However, prolactin is a pleiotropic hormone that is able to affect several physiological functions, including fertility. Prolactin receptors (PRLRs) are widely expressed in several tissues, including several brain regions and reproductive tract organs. Upon activation, PRLRs may exert prolactin's functions through several signaling pathways, although the recruitment of the signal transducer and activator of transcription 5 causes most of the known effects of prolactin. Pathological hyperprolactinemia is mainly due to the presence of a prolactinoma or pharmacological effects induced by drugs that interact with the dopamine system. Notably, hyperprolactinemia is a frequent cause of reproductive dysfunction and may lead to infertility in males and females. Recently, several studies have indicated that prolactin may modulate the reproductive axis by acting on specific populations of hypothalamic neurons that express the Kiss1 gene. The Kiss1 gene encodes neuropeptides known as kisspeptins, which are powerful activators of gonadotropin-releasing hormone neurons. In the present review, we will summarize the current knowledge about prolactin's actions on reproduction. Among other aspects, we will discuss whether the interaction between prolactin and the Kiss1-expressing neurons can affect reproduction and how kisspeptins may become a novel therapeutic approach to treat prolactin-induced infertility.
Collapse
Affiliation(s)
- Jose Donato
- Departamento de Fisiologia e BiofísicaInstituto de Ciências BiomédicasUniversidade de São PauloSão PauloSPBrasilDepartamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo (USP), São Paulo, SP, Brasil
| | - Renata Frazão
- Departamento de AnatomiaInstituto de Ciências BiomédicasUSPSão PauloSPBrasilDepartamento de Anatomia, Instituto de Ciências Biomédicas, USP, São Paulo, SP, Brasil
| |
Collapse
|
132
|
Inhibition of interleukin-3- and interferon- α-induced JAK/STAT signaling by the synthetic α-X-2′,3,4,4′-tetramethoxychalcones α-Br-TMC and α-CF3-TMC. Biol Chem 2016; 397:1187-1204. [DOI: 10.1515/hsz-2016-0148] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 06/21/2016] [Indexed: 12/18/2022]
Abstract
Abstract
The JAK/STAT pathway is an essential mediator of cytokine signaling, often upregulated in human diseases and therefore recognized as a relevant therapeutic target. We previously identified the synthetic chalcone α-bromo-2′,3,4,4′-tetramethoxychalcone (α-Br-TMC) as a novel JAK2/STAT5 inhibitor. We also found that treatment with α-Br-TMC resulted in a downward shift of STAT5 proteins in SDS-PAGE, suggesting a post-translational modification that might affect STAT5 function. In the present study, we show that a single cysteine within STAT5 is responsible for the α-Br-TMC-induced protein shift, and that this modification does not alter STAT5 transcriptional activity. We also compared the inhibitory activity of α-Br-TMC to that of another synthetic chalcone, α-trifluoromethyl-2′,3,4,4′-tetramethoxychalcone (α-CF3-TMC). We found that, like α-Br-TMC, α-CF3-TMC inhibits JAK2 and STAT5 phosphorylation in response to interleukin-3, however without altering STAT5 mobility in SDS-PAGE. Moreover, we demonstrate that both α-Br-TMC and α-CF3-TMC inhibit interferon-α-induced activation of STAT1 and STAT2, by inhibiting their phosphorylation and the expression of downstream interferon-stimulated genes. Together with the previous finding that α-Br-TMC and α-CF3-TMC inhibit the response to inflammation by inducing Nrf2 and blocking NF-κB activities, our data suggest that synthetic chalcones might be useful as anti-inflammatory, anti-cancer and immunomodulatory agents in the treatment of human diseases.
Collapse
|
133
|
Lin Z, Jiang J, Liu XS. Ursolic acid-mediated apoptosis of K562 cells involves Stat5/Akt pathway inhibition through the induction of Gfi-1. Sci Rep 2016; 6:33358. [PMID: 27634378 PMCID: PMC5025887 DOI: 10.1038/srep33358] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 08/25/2016] [Indexed: 02/05/2023] Open
Abstract
Ursolic acid (UA) is a promising natural compound for cancer prevention and therapy. We previously reported that UA induced apoptosis in CML-derived K562 cells. Here we show that the apoptotic process is accompanied by down-regulation of Bcl-xL and Mcl-1 expression and dephosphorylation of Bad. These events are associated with Stat5 inhibition, which is partially mediated through elevated expression of transcriptional repressor Gfi-1. Gfi-1 knockdown using siRNA abrogates the ability of UA to decrease Stat5b expression and attenuates apoptosis induction by UA. We also demonstrate that UA suppresses the Akt kinase activity by inhibiting Akt1/2 expression, which correlates with Stat5 inhibition. Stat5 activity inhibited by a chemical inhibitor or siRNA, Akt1/2 mRNA expression is suppressed. Moreover, we show that UA exerts growth-inhibition in Imatinib-resistant K562/G01. UA has synergistic effects when used in combination with Imatinib in both K562 and K562/G01. Altogether, the data provide evidence that UA's pro-apoptotic effect in K562 cells is associated with the Gfi-1/Stat5/Akt pathway. The findings indicate that UA could potentially be a useful agent in the treatment of CML.
Collapse
Affiliation(s)
- Ze Lin
- Department of Biochemistry, Shantou University Medical College, No. 22 Xinlin Road, Jinping District, Shantou, 510451, China
| | - Jikai Jiang
- Department of Biochemistry, Shantou University Medical College, No. 22 Xinlin Road, Jinping District, Shantou, 510451, China
| | - Xiao-Shan Liu
- Department of Biochemistry, Guangzhou Medical University, Xinzao Town, Panyu District, Guangzhou 511436, China
| |
Collapse
|
134
|
Gergics P, Christian HC, Choo MS, Ajmal A, Camper SA. Gene Expression in Mouse Thyrotrope Adenoma: Transcription Elongation Factor Stimulates Proliferation. Endocrinology 2016; 157:3631-46. [PMID: 27580811 PMCID: PMC5007889 DOI: 10.1210/en.2016-1183] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Thyrotrope hyperplasia and hypertrophy are common responses to primary hypothyroidism. To understand the genetic regulation of these processes, we studied gene expression changes in the pituitaries of Cga(-/-) mice, which are deficient in the common α-subunit of TSH, LH, and FSH. These mice have thyrotrope hypertrophy and hyperplasia and develop thyrotrope adenoma. We report that cell proliferation is increased, but the expression of most stem cell markers is unchanged. The α-subunit is required for secretion of the glycoprotein hormone β-subunits, and mutants exhibit elevated expression of many genes involved in the unfolded protein response, consistent with dilation and stress of the endoplasmic reticulum. Mutants have elevated expression of transcription factors that are important in thyrotrope function, such as Gata2 and Islet 1, and those that stimulate proliferation, including Nupr1, E2f1, and Etv5. We characterized the expression and function of a novel, overexpressed gene, transcription elongation factor A (SII)-like 5 (Tceal5). Stable expression of Tceal5 in a pituitary progenitor cell line is sufficient to increase cell proliferation. Thus, Tceal5 may act as a proto-oncogene. This study provides a rich resource for comparing pituitary transcriptomes and an analysis of gene expression networks.
Collapse
Affiliation(s)
- Peter Gergics
- Department of Human Genetics (P.G., M.S.C., S.A.C.), University of Michigan, Ann Arbor, Michigan 48109; Department of Physiology, Anatomy and Genetics (H.C.C.), University of Oxford, Oxford OX3 0RZ, United Kingdom; and Department of Internal Medicine, Metabolism, Endocrinology and Diabetes (A.A.), University of Michigan, Ann Arbor, Michigan 48105
| | - Helen C Christian
- Department of Human Genetics (P.G., M.S.C., S.A.C.), University of Michigan, Ann Arbor, Michigan 48109; Department of Physiology, Anatomy and Genetics (H.C.C.), University of Oxford, Oxford OX3 0RZ, United Kingdom; and Department of Internal Medicine, Metabolism, Endocrinology and Diabetes (A.A.), University of Michigan, Ann Arbor, Michigan 48105
| | - Monica S Choo
- Department of Human Genetics (P.G., M.S.C., S.A.C.), University of Michigan, Ann Arbor, Michigan 48109; Department of Physiology, Anatomy and Genetics (H.C.C.), University of Oxford, Oxford OX3 0RZ, United Kingdom; and Department of Internal Medicine, Metabolism, Endocrinology and Diabetes (A.A.), University of Michigan, Ann Arbor, Michigan 48105
| | - Adnan Ajmal
- Department of Human Genetics (P.G., M.S.C., S.A.C.), University of Michigan, Ann Arbor, Michigan 48109; Department of Physiology, Anatomy and Genetics (H.C.C.), University of Oxford, Oxford OX3 0RZ, United Kingdom; and Department of Internal Medicine, Metabolism, Endocrinology and Diabetes (A.A.), University of Michigan, Ann Arbor, Michigan 48105
| | - Sally A Camper
- Department of Human Genetics (P.G., M.S.C., S.A.C.), University of Michigan, Ann Arbor, Michigan 48109; Department of Physiology, Anatomy and Genetics (H.C.C.), University of Oxford, Oxford OX3 0RZ, United Kingdom; and Department of Internal Medicine, Metabolism, Endocrinology and Diabetes (A.A.), University of Michigan, Ann Arbor, Michigan 48105
| |
Collapse
|
135
|
Futawaka K, Tagami T, Fukuda Y, Koyama R, Nushida A, Nezu S, Imamoto M, Kasahara M, Moriyama K. Growth hormone regulates the expression of UCP2 in myocytes. Growth Horm IGF Res 2016; 29:57-62. [PMID: 27150070 DOI: 10.1016/j.ghir.2016.04.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Revised: 03/03/2016] [Accepted: 04/10/2016] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To determine if and how growth hormone (GH) signaling is involved in energy metabolism. DESIGN We used human embryonic kidney TSA201 cells, human H-EMC-SS chondrosarcoma cells, rat L6 skeletal muscle cells, and murine C2C12 skeletal muscle myoblasts to investigate GH-induced expression of uncoupling protein2 (UCP2) to the GHR/JAK/STAT5 pathway by a combination of a reporter assay, electrophoretic mobility shift assay (EMSA), real-time quantitative PCR, Western blotting. RESULTS We demonstrated that the regulation energy metabolism, which was hypothesized to be directly acted on by GH, involves UCP2 via activated STAT5B, a signal transducer downstream of GH. We also showed that the sequence at the -586 'TTCnGA' may function as a novel putative consensus sequence of STAT5s. CONCLUSION The results suggest that GH regulates energy metabolism directly in myocytes and that UCP2 participates in the signal transduction pathway that functions downstream of the GHR/JAK/STAT.
Collapse
Affiliation(s)
- Kumi Futawaka
- Medicine and Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan.
| | - Tetsuya Tagami
- Clinical Research Institute for Endocrine and Metabolic Diseases, National Hospital Organization Kyoto Medical Center, Kyoto 612-8555, Japan
| | - Yuki Fukuda
- Medicine and Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Rie Koyama
- Medicine and Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Ayaka Nushida
- Medicine and Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Syoko Nezu
- Medicine and Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan
| | - Miyuki Imamoto
- Clinical Research Institute for Endocrine and Metabolic Diseases, National Hospital Organization Kyoto Medical Center, Kyoto 612-8555, Japan
| | - Masato Kasahara
- Clinical Research Institute for Endocrine and Metabolic Diseases, National Hospital Organization Kyoto Medical Center, Kyoto 612-8555, Japan
| | - Kenji Moriyama
- Medicine and Clinical Science, Faculty of Pharmaceutical Sciences, Mukogawa Women's University, Hyogo 663-8179, Japan; Clinical Research Institute for Endocrine and Metabolic Diseases, National Hospital Organization Kyoto Medical Center, Kyoto 612-8555, Japan; Department of Nephrology and Blood Purification, Institute of Biomedical Research and Innovation, Kobe Medical Frontier Center, Kobe 650-0047, Japan
| |
Collapse
|
136
|
Han W, Sfondouris ME, Jones FE. Direct coupling of the HER4 intracellular domain (4ICD) and STAT5A signaling is required to induce mammary epithelial cell differentiation. Biochem Biophys Rep 2016; 7:323-327. [PMID: 28955922 PMCID: PMC5613636 DOI: 10.1016/j.bbrep.2016.07.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 06/17/2016] [Accepted: 07/18/2016] [Indexed: 11/29/2022] Open
Abstract
The HER4 receptor tyrosine kinase and STAT5A cooperate to promote mammary luminal progenitor cell maturation and mammary epithelial cell differentiation. Coupled HER4 and STAT5A signaling is mediated, in part, through association of the HER4 intracellular domain (4ICD) with STAT5A at STAT5A target gene promoters where 4ICD functions as a STAT5A transcriptional coactivator. Despite an essential role for coupled 4ICD and STAT5A signaling in mammary gland development, the mechanistic basis of 4ICD and STAT5A cooperative signaling remains unexplored. Here we show for the first time that 4ICD and STAT5A directly interact through STAT5A recruitment and binding to HER4/4ICD residue Y984. Accordingly, altering the 4ICD Y984 to phenylalanine results in a dramatic reduction of STAT5A and 4ICD-Y984F interacting complexes coimmunoprecipitated with HER4 or STAT5A specific antibodies. We further show that disrupting the 4ICD and STAT5A interaction has an important physiological impact on mammary epithelial cell differentiation. HC11 mammary epithelial cells with stable expression of 4ICD undergo differentiation with significantly increased expression of the STAT5A target genes and differentiation markers β-casein and WAP. In contrast, HC11 cells stably expressing 4ICD-Y984F failed to undergo differentiation with basal expression levels of β-casein and WAP. Differentiation in this cell system was induced in the absence of exogenous prolactin indicating that 4ICD activity is sufficient to induce mammary epithelial cell differentiation. Finally, we show that suppression of STAT5A expression abolishes the ability of 4ICD to induce HC11 differentiation and activate β-casein or WAP expression. Taken together our results demonstrate for the first time that direct coupling of 4ICD and STAT5A is both necessary and sufficient to drive mammary epithelial differentiation. In conclusion, our findings that 4ICD and STAT5A directly interact to form a physiologically important transcriptional activation complex, provide a mechanistic basis for the in vivo observations that HER4/4ICD and STAT5A cooperate to promote mammary gland progenitor cell maturation and initiate lactation at parturition. HER4/4ICD tyrosine 984 mediates a direct interaction with STAT5A. 4ICD expression with an intact Y984 is sufficient to induce mammary differentiation. Mammary differentiation is abolished by disrupting the 4ICD and STAT5A interaction. STAT5 expression is required for 4ICD-induced mammary differentiation.
Collapse
Key Words
- 4ICD, HER4 intracellular domain
- ATCC, American type culture collection
- EGF, epidermal growth factor
- EGFP, enhanced green fluorescent protein
- EGFR, epidermal growth factor family
- EGFR-family
- ERα, estrogen receptor alpha
- FBS, fetal bovine serum
- Gene expression
- HEK, human embryonic kidney
- HER4/ERBB4
- HRGα, heregulin alpha
- HRGα1, heregulin beta 1
- Mammary epithelial differentiation
- NLS, nuclear localization signal
- PCR, polymerase chain reaction
- PI3K, phosphoinositide 3-kinase
- RIP, regulated intramembrane cleavage
- RT, reverse transcription
- RTK, receptor tyrosine kinase
- SH2, src homology 2
- STAT5A
- STAT5A, signal transducer and activator of transcription 5A
- TACE, tumor necrosis factor-α-converting enzyme
- Transactivation
- WAP, whey acidic protein
- YAP, yes-associated protein
Collapse
Affiliation(s)
- Wen Han
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Mary E Sfondouris
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Frank E Jones
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| |
Collapse
|
137
|
Sachs Z, Been RA, DeCoursin KJ, Nguyen HT, Mohd Hassan NA, Noble-Orcutt KE, Eckfeldt CE, Pomeroy EJ, Diaz-Flores E, Geurts JL, Diers MD, Hasz DE, Morgan KJ, MacMillan ML, Shannon KM, Largaespada DA, Wiesner SM. Stat5 is critical for the development and maintenance of myeloproliferative neoplasm initiated by Nf1 deficiency. Haematologica 2016; 101:1190-1199. [PMID: 27418650 DOI: 10.3324/haematol.2015.136002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 06/15/2016] [Indexed: 11/09/2022] Open
Abstract
Juvenile myelomonocytic leukemia is a rare myeloproliferative neoplasm characterized by hyperactive RAS signaling. Neurofibromin1 (encoded by the NF1 gene) is a negative regulator of RAS activation. Patients with neurofibromatosis type 1 harbor loss-of-function mutations in NF1 and have a 200- to 500-fold increased risk of juvenile myelomonocytic leukemia. Leukemia cells from patients with juvenile myelomonocytic leukemia display hypersensitivity to certain cytokines, such as granulocyte-macrophage colony-stimulating factor. The granulocyte-macrophage colony-stimulating factor receptor utilizes pre-associated JAK2 to initiate signals after ligand binding. JAK2 subsequently activates STAT5, among other downstream effectors. Although STAT5 is gaining recognition as an important mediator of growth factor signaling in myeloid leukemias, the contribution of STAT5 to the development of hyperactive RAS-initiated myeloproliferative disease has not been well described. In this study, we investigated the consequence of STAT5 attenuation via genetic and pharmacological approaches in Nf1-deficient murine models of juvenile myelomonocytic leukemia. We found that homozygous Stat5 deficiency extended the lifespan of Nf1-deficient mice and eliminated the development of myeloproliferative neoplasm associated with Nf1 gene loss. Likewise, we found that JAK inhibition with ruxolitinib attenuated myeloproliferative neoplasm in Nf1-deficient mice. Finally, we found that primary cells from a patient with KRAS-mutant juvenile myelomonocytic leukemia displayed reduced colony formation in response to JAK2 inhibition. Our findings establish a central role for STAT5 activation in the pathogenesis of juvenile myelomonocytic leukemia and suggest that targeting this pathway may be of clinical utility in these patients.
Collapse
Affiliation(s)
- Zohar Sachs
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Raha A Been
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA College of Veterinary Medicine and Department of Comparative and Molecular Biosciences, University of Minnesota, St. Paul, MN, USA
| | | | - Hanh T Nguyen
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | | | - Klara E Noble-Orcutt
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Craig E Eckfeldt
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Emily J Pomeroy
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Ernesto Diaz-Flores
- Department of Pediatrics, University of California, San Francisco, CA, USA Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Jennifer L Geurts
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Miechaleen D Diers
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Diane E Hasz
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Kelly J Morgan
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
| | - Margaret L MacMillan
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA Blood and Marrow Transplantation Program, University of Minnesota, Minneapolis, MN, USA
| | - Kevin M Shannon
- Department of Pediatrics, University of California, San Francisco, CA, USA Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - David A Largaespada
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA Blood and Marrow Transplantation Program, University of Minnesota, Minneapolis, MN, USA
| | - Stephen M Wiesner
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA Center for Allied Health Programs, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
138
|
Shen S, Wang Y, Wang C, Wu YN, Xing Y. SURVIV for survival analysis of mRNA isoform variation. Nat Commun 2016; 7:11548. [PMID: 27279334 PMCID: PMC4906168 DOI: 10.1038/ncomms11548] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Accepted: 04/07/2016] [Indexed: 01/07/2023] Open
Abstract
The rapid accumulation of clinical RNA-seq data sets has provided the opportunity to associate mRNA isoform variations to clinical outcomes. Here we report a statistical method SURVIV (Survival analysis of mRNA Isoform Variation), designed for identifying mRNA isoform variation associated with patient survival time. A unique feature and major strength of SURVIV is that it models the measurement uncertainty of mRNA isoform ratio in RNA-seq data. Simulation studies suggest that SURVIV outperforms the conventional Cox regression survival analysis, especially for data sets with modest sequencing depth. We applied SURVIV to TCGA RNA-seq data of invasive ductal carcinoma as well as five additional cancer types. Alternative splicing-based survival predictors consistently outperform gene expression-based survival predictors, and the integration of clinical, gene expression and alternative splicing profiles leads to the best survival prediction. We anticipate that SURVIV will have broad utilities for analysing diverse types of mRNA isoform variation in large-scale clinical RNA-seq projects.
Collapse
Affiliation(s)
- Shihao Shen
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Yuanyuan Wang
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Chengyang Wang
- Bioinformatics Interdepartmental Graduate Program, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Ying Nian Wu
- Department of Statistics, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Yi Xing
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, California 90095, USA
| |
Collapse
|
139
|
Hwa V. STAT5B deficiency: Impacts on human growth and immunity. Growth Horm IGF Res 2016; 28:16-20. [PMID: 26703237 PMCID: PMC4846566 DOI: 10.1016/j.ghir.2015.12.006] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 11/24/2015] [Accepted: 12/06/2015] [Indexed: 02/06/2023]
Abstract
Growth hormone (GH) promotes postnatal human growth primarily by regulating insulin-like growth factor (IGF)-I production through activation of the GH receptor (GHR)-signal transducer and activator of transcription (STAT)-5B signaling cascade. The critical importance of STAT5B in human IGF-I production was confirmed with the identification of the first homozygous, autosomal recessive, STAT5B mutation in a young female patient who phenotypically resembled patients with classical growth hormone insensitivity (GHI) syndrome (Laron syndrome) due to mutations in the GHR gene, presenting with severe postnatal growth failure and marked IGF-I deficiency. Of note, the closely related STAT5A, which shares >95% amino acid identity with STAT5B, could not compensate for loss of functional STAT5B. To date, 7 homozygous, inactivating, STAT5B mutations in 10 patients have been reported. STAT5B deficient patients, unlike patients deficient in GHR, can also present with a novel, potentially fatal, primary immunodeficiency, which can manifest as chronic pulmonary disease. STAT5B deficiency may be underestimated in endocrine, immunology and pulmonary clinics.
Collapse
Affiliation(s)
- Vivian Hwa
- Cincinnati Center for Growth Disorders, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, United States.
| |
Collapse
|
140
|
Marzec M, Hawkes CP, Eletto D, Boyle S, Rosenfeld R, Hwa V, Wit JM, van Duyvenvoorde HA, Oostdijk W, Losekoot M, Pedersen O, Yeap BB, Flicker L, Barzilai N, Atzmon G, Grimberg A, Argon Y. A Human Variant of Glucose-Regulated Protein 94 That Inefficiently Supports IGF Production. Endocrinology 2016; 157:1914-28. [PMID: 26982636 PMCID: PMC4870884 DOI: 10.1210/en.2015-2058] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 03/10/2016] [Indexed: 02/08/2023]
Abstract
IGFs are critical for normal intrauterine and childhood growth and sustaining health throughout life. We showed previously that the production of IGF-1 and IGF-2 requires interaction with the chaperone glucose-regulated protein 94 (GRP94) and that the amount of secreted IGFs is proportional to the GRP94 activity. Therefore, we tested the hypothesis that functional polymorphisms of human GRP94 affect IGF production and thereby human health. We describe a hypomorphic variant of human GRP94, P300L, whose heterozygous carriers have 9% lower circulating IGF-1 concentration. P300L was found first in a child with primary IGF deficiency and was later shown to be a noncommon single-nucleotide polymorphism with frequencies of 1%-4% in various populations. When tested in the grp94(-/-) cell-based complementation assay, P300L supported only approximately 58% of IGF secretion relative to wild-type GRP94. Furthermore, recombinant P300L showed impaired nucleotide binding activity. These in vitro data strongly support a causal relationship between the GRP94 variant and the decreased concentration of circulating IGF-1, as observed in human carriers of P300L. Thus, mutations in GRP94 that affect its IGF chaperone activity represent a novel causal genetic mechanism that limits IGF biosynthesis, quite a distinct mechanism from the known genes in the GH/IGF signaling network.
Collapse
Affiliation(s)
- Michal Marzec
- Department of Pathology and Laboratory Medicine (M.M., D.E., S.B., Y.A.), The Children's Hospital of Philadelphia and The University of Pennsylvania, Philadelphia,; Division of Endocrinology and Diabetes (C.P.H., A.G.), The Children's Hospital of Philadelphia, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Pennsylvania 19104; National Children's Research Centre (C.P.H.), Dublin 12, Ireland; STAT5, LLC (R.R.), Los Altos, California 94022; Department of Pediatrics (R.R., V.H.), Oregon Health and Science University, Portland, Oregon 97239; Departments of Pediatrics (J.-M.W., H.A.v.D., W.O.), Endocrinology and Metabolic Diseases (H.A.v.D.), and Clinical Genetics (H.A.v.D., M.L.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands; Faculty of Health and Medical Sciences (O.P.), University of Copenhagen, DK-2400 Copenhagen, Denmark; School of Medicine and Pharmacology (B.B.Y.), Western Australia Centre for Health and Ageing (L.F.), Centre for Medical Research (L.F.), and School of Medicine and Pharmacology (L.F.), University of Western Australia, Perth, Western Australia 6872, Australia; Department of Endocrinology and Diabetes (B.B.Y.), Fiona Stanley Hospital, Perth, Western Australia 6150, Australia; Department of Human Biology (G.A.), Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel; and Departments of Medicine and Genetics (N.B., G.A.), Albert Einstein College of Medicine, Bronx, New York 10461
| | - Colin P Hawkes
- Department of Pathology and Laboratory Medicine (M.M., D.E., S.B., Y.A.), The Children's Hospital of Philadelphia and The University of Pennsylvania, Philadelphia,; Division of Endocrinology and Diabetes (C.P.H., A.G.), The Children's Hospital of Philadelphia, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Pennsylvania 19104; National Children's Research Centre (C.P.H.), Dublin 12, Ireland; STAT5, LLC (R.R.), Los Altos, California 94022; Department of Pediatrics (R.R., V.H.), Oregon Health and Science University, Portland, Oregon 97239; Departments of Pediatrics (J.-M.W., H.A.v.D., W.O.), Endocrinology and Metabolic Diseases (H.A.v.D.), and Clinical Genetics (H.A.v.D., M.L.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands; Faculty of Health and Medical Sciences (O.P.), University of Copenhagen, DK-2400 Copenhagen, Denmark; School of Medicine and Pharmacology (B.B.Y.), Western Australia Centre for Health and Ageing (L.F.), Centre for Medical Research (L.F.), and School of Medicine and Pharmacology (L.F.), University of Western Australia, Perth, Western Australia 6872, Australia; Department of Endocrinology and Diabetes (B.B.Y.), Fiona Stanley Hospital, Perth, Western Australia 6150, Australia; Department of Human Biology (G.A.), Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel; and Departments of Medicine and Genetics (N.B., G.A.), Albert Einstein College of Medicine, Bronx, New York 10461
| | - Davide Eletto
- Department of Pathology and Laboratory Medicine (M.M., D.E., S.B., Y.A.), The Children's Hospital of Philadelphia and The University of Pennsylvania, Philadelphia,; Division of Endocrinology and Diabetes (C.P.H., A.G.), The Children's Hospital of Philadelphia, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Pennsylvania 19104; National Children's Research Centre (C.P.H.), Dublin 12, Ireland; STAT5, LLC (R.R.), Los Altos, California 94022; Department of Pediatrics (R.R., V.H.), Oregon Health and Science University, Portland, Oregon 97239; Departments of Pediatrics (J.-M.W., H.A.v.D., W.O.), Endocrinology and Metabolic Diseases (H.A.v.D.), and Clinical Genetics (H.A.v.D., M.L.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands; Faculty of Health and Medical Sciences (O.P.), University of Copenhagen, DK-2400 Copenhagen, Denmark; School of Medicine and Pharmacology (B.B.Y.), Western Australia Centre for Health and Ageing (L.F.), Centre for Medical Research (L.F.), and School of Medicine and Pharmacology (L.F.), University of Western Australia, Perth, Western Australia 6872, Australia; Department of Endocrinology and Diabetes (B.B.Y.), Fiona Stanley Hospital, Perth, Western Australia 6150, Australia; Department of Human Biology (G.A.), Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel; and Departments of Medicine and Genetics (N.B., G.A.), Albert Einstein College of Medicine, Bronx, New York 10461
| | - Sarah Boyle
- Department of Pathology and Laboratory Medicine (M.M., D.E., S.B., Y.A.), The Children's Hospital of Philadelphia and The University of Pennsylvania, Philadelphia,; Division of Endocrinology and Diabetes (C.P.H., A.G.), The Children's Hospital of Philadelphia, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Pennsylvania 19104; National Children's Research Centre (C.P.H.), Dublin 12, Ireland; STAT5, LLC (R.R.), Los Altos, California 94022; Department of Pediatrics (R.R., V.H.), Oregon Health and Science University, Portland, Oregon 97239; Departments of Pediatrics (J.-M.W., H.A.v.D., W.O.), Endocrinology and Metabolic Diseases (H.A.v.D.), and Clinical Genetics (H.A.v.D., M.L.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands; Faculty of Health and Medical Sciences (O.P.), University of Copenhagen, DK-2400 Copenhagen, Denmark; School of Medicine and Pharmacology (B.B.Y.), Western Australia Centre for Health and Ageing (L.F.), Centre for Medical Research (L.F.), and School of Medicine and Pharmacology (L.F.), University of Western Australia, Perth, Western Australia 6872, Australia; Department of Endocrinology and Diabetes (B.B.Y.), Fiona Stanley Hospital, Perth, Western Australia 6150, Australia; Department of Human Biology (G.A.), Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel; and Departments of Medicine and Genetics (N.B., G.A.), Albert Einstein College of Medicine, Bronx, New York 10461
| | - Ron Rosenfeld
- Department of Pathology and Laboratory Medicine (M.M., D.E., S.B., Y.A.), The Children's Hospital of Philadelphia and The University of Pennsylvania, Philadelphia,; Division of Endocrinology and Diabetes (C.P.H., A.G.), The Children's Hospital of Philadelphia, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Pennsylvania 19104; National Children's Research Centre (C.P.H.), Dublin 12, Ireland; STAT5, LLC (R.R.), Los Altos, California 94022; Department of Pediatrics (R.R., V.H.), Oregon Health and Science University, Portland, Oregon 97239; Departments of Pediatrics (J.-M.W., H.A.v.D., W.O.), Endocrinology and Metabolic Diseases (H.A.v.D.), and Clinical Genetics (H.A.v.D., M.L.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands; Faculty of Health and Medical Sciences (O.P.), University of Copenhagen, DK-2400 Copenhagen, Denmark; School of Medicine and Pharmacology (B.B.Y.), Western Australia Centre for Health and Ageing (L.F.), Centre for Medical Research (L.F.), and School of Medicine and Pharmacology (L.F.), University of Western Australia, Perth, Western Australia 6872, Australia; Department of Endocrinology and Diabetes (B.B.Y.), Fiona Stanley Hospital, Perth, Western Australia 6150, Australia; Department of Human Biology (G.A.), Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel; and Departments of Medicine and Genetics (N.B., G.A.), Albert Einstein College of Medicine, Bronx, New York 10461
| | - Vivian Hwa
- Department of Pathology and Laboratory Medicine (M.M., D.E., S.B., Y.A.), The Children's Hospital of Philadelphia and The University of Pennsylvania, Philadelphia,; Division of Endocrinology and Diabetes (C.P.H., A.G.), The Children's Hospital of Philadelphia, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Pennsylvania 19104; National Children's Research Centre (C.P.H.), Dublin 12, Ireland; STAT5, LLC (R.R.), Los Altos, California 94022; Department of Pediatrics (R.R., V.H.), Oregon Health and Science University, Portland, Oregon 97239; Departments of Pediatrics (J.-M.W., H.A.v.D., W.O.), Endocrinology and Metabolic Diseases (H.A.v.D.), and Clinical Genetics (H.A.v.D., M.L.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands; Faculty of Health and Medical Sciences (O.P.), University of Copenhagen, DK-2400 Copenhagen, Denmark; School of Medicine and Pharmacology (B.B.Y.), Western Australia Centre for Health and Ageing (L.F.), Centre for Medical Research (L.F.), and School of Medicine and Pharmacology (L.F.), University of Western Australia, Perth, Western Australia 6872, Australia; Department of Endocrinology and Diabetes (B.B.Y.), Fiona Stanley Hospital, Perth, Western Australia 6150, Australia; Department of Human Biology (G.A.), Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel; and Departments of Medicine and Genetics (N.B., G.A.), Albert Einstein College of Medicine, Bronx, New York 10461
| | - Jan M Wit
- Department of Pathology and Laboratory Medicine (M.M., D.E., S.B., Y.A.), The Children's Hospital of Philadelphia and The University of Pennsylvania, Philadelphia,; Division of Endocrinology and Diabetes (C.P.H., A.G.), The Children's Hospital of Philadelphia, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Pennsylvania 19104; National Children's Research Centre (C.P.H.), Dublin 12, Ireland; STAT5, LLC (R.R.), Los Altos, California 94022; Department of Pediatrics (R.R., V.H.), Oregon Health and Science University, Portland, Oregon 97239; Departments of Pediatrics (J.-M.W., H.A.v.D., W.O.), Endocrinology and Metabolic Diseases (H.A.v.D.), and Clinical Genetics (H.A.v.D., M.L.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands; Faculty of Health and Medical Sciences (O.P.), University of Copenhagen, DK-2400 Copenhagen, Denmark; School of Medicine and Pharmacology (B.B.Y.), Western Australia Centre for Health and Ageing (L.F.), Centre for Medical Research (L.F.), and School of Medicine and Pharmacology (L.F.), University of Western Australia, Perth, Western Australia 6872, Australia; Department of Endocrinology and Diabetes (B.B.Y.), Fiona Stanley Hospital, Perth, Western Australia 6150, Australia; Department of Human Biology (G.A.), Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel; and Departments of Medicine and Genetics (N.B., G.A.), Albert Einstein College of Medicine, Bronx, New York 10461
| | - Hermine A van Duyvenvoorde
- Department of Pathology and Laboratory Medicine (M.M., D.E., S.B., Y.A.), The Children's Hospital of Philadelphia and The University of Pennsylvania, Philadelphia,; Division of Endocrinology and Diabetes (C.P.H., A.G.), The Children's Hospital of Philadelphia, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Pennsylvania 19104; National Children's Research Centre (C.P.H.), Dublin 12, Ireland; STAT5, LLC (R.R.), Los Altos, California 94022; Department of Pediatrics (R.R., V.H.), Oregon Health and Science University, Portland, Oregon 97239; Departments of Pediatrics (J.-M.W., H.A.v.D., W.O.), Endocrinology and Metabolic Diseases (H.A.v.D.), and Clinical Genetics (H.A.v.D., M.L.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands; Faculty of Health and Medical Sciences (O.P.), University of Copenhagen, DK-2400 Copenhagen, Denmark; School of Medicine and Pharmacology (B.B.Y.), Western Australia Centre for Health and Ageing (L.F.), Centre for Medical Research (L.F.), and School of Medicine and Pharmacology (L.F.), University of Western Australia, Perth, Western Australia 6872, Australia; Department of Endocrinology and Diabetes (B.B.Y.), Fiona Stanley Hospital, Perth, Western Australia 6150, Australia; Department of Human Biology (G.A.), Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel; and Departments of Medicine and Genetics (N.B., G.A.), Albert Einstein College of Medicine, Bronx, New York 10461
| | - Wilma Oostdijk
- Department of Pathology and Laboratory Medicine (M.M., D.E., S.B., Y.A.), The Children's Hospital of Philadelphia and The University of Pennsylvania, Philadelphia,; Division of Endocrinology and Diabetes (C.P.H., A.G.), The Children's Hospital of Philadelphia, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Pennsylvania 19104; National Children's Research Centre (C.P.H.), Dublin 12, Ireland; STAT5, LLC (R.R.), Los Altos, California 94022; Department of Pediatrics (R.R., V.H.), Oregon Health and Science University, Portland, Oregon 97239; Departments of Pediatrics (J.-M.W., H.A.v.D., W.O.), Endocrinology and Metabolic Diseases (H.A.v.D.), and Clinical Genetics (H.A.v.D., M.L.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands; Faculty of Health and Medical Sciences (O.P.), University of Copenhagen, DK-2400 Copenhagen, Denmark; School of Medicine and Pharmacology (B.B.Y.), Western Australia Centre for Health and Ageing (L.F.), Centre for Medical Research (L.F.), and School of Medicine and Pharmacology (L.F.), University of Western Australia, Perth, Western Australia 6872, Australia; Department of Endocrinology and Diabetes (B.B.Y.), Fiona Stanley Hospital, Perth, Western Australia 6150, Australia; Department of Human Biology (G.A.), Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel; and Departments of Medicine and Genetics (N.B., G.A.), Albert Einstein College of Medicine, Bronx, New York 10461
| | - Monique Losekoot
- Department of Pathology and Laboratory Medicine (M.M., D.E., S.B., Y.A.), The Children's Hospital of Philadelphia and The University of Pennsylvania, Philadelphia,; Division of Endocrinology and Diabetes (C.P.H., A.G.), The Children's Hospital of Philadelphia, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Pennsylvania 19104; National Children's Research Centre (C.P.H.), Dublin 12, Ireland; STAT5, LLC (R.R.), Los Altos, California 94022; Department of Pediatrics (R.R., V.H.), Oregon Health and Science University, Portland, Oregon 97239; Departments of Pediatrics (J.-M.W., H.A.v.D., W.O.), Endocrinology and Metabolic Diseases (H.A.v.D.), and Clinical Genetics (H.A.v.D., M.L.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands; Faculty of Health and Medical Sciences (O.P.), University of Copenhagen, DK-2400 Copenhagen, Denmark; School of Medicine and Pharmacology (B.B.Y.), Western Australia Centre for Health and Ageing (L.F.), Centre for Medical Research (L.F.), and School of Medicine and Pharmacology (L.F.), University of Western Australia, Perth, Western Australia 6872, Australia; Department of Endocrinology and Diabetes (B.B.Y.), Fiona Stanley Hospital, Perth, Western Australia 6150, Australia; Department of Human Biology (G.A.), Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel; and Departments of Medicine and Genetics (N.B., G.A.), Albert Einstein College of Medicine, Bronx, New York 10461
| | - Oluf Pedersen
- Department of Pathology and Laboratory Medicine (M.M., D.E., S.B., Y.A.), The Children's Hospital of Philadelphia and The University of Pennsylvania, Philadelphia,; Division of Endocrinology and Diabetes (C.P.H., A.G.), The Children's Hospital of Philadelphia, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Pennsylvania 19104; National Children's Research Centre (C.P.H.), Dublin 12, Ireland; STAT5, LLC (R.R.), Los Altos, California 94022; Department of Pediatrics (R.R., V.H.), Oregon Health and Science University, Portland, Oregon 97239; Departments of Pediatrics (J.-M.W., H.A.v.D., W.O.), Endocrinology and Metabolic Diseases (H.A.v.D.), and Clinical Genetics (H.A.v.D., M.L.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands; Faculty of Health and Medical Sciences (O.P.), University of Copenhagen, DK-2400 Copenhagen, Denmark; School of Medicine and Pharmacology (B.B.Y.), Western Australia Centre for Health and Ageing (L.F.), Centre for Medical Research (L.F.), and School of Medicine and Pharmacology (L.F.), University of Western Australia, Perth, Western Australia 6872, Australia; Department of Endocrinology and Diabetes (B.B.Y.), Fiona Stanley Hospital, Perth, Western Australia 6150, Australia; Department of Human Biology (G.A.), Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel; and Departments of Medicine and Genetics (N.B., G.A.), Albert Einstein College of Medicine, Bronx, New York 10461
| | - Bu Beng Yeap
- Department of Pathology and Laboratory Medicine (M.M., D.E., S.B., Y.A.), The Children's Hospital of Philadelphia and The University of Pennsylvania, Philadelphia,; Division of Endocrinology and Diabetes (C.P.H., A.G.), The Children's Hospital of Philadelphia, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Pennsylvania 19104; National Children's Research Centre (C.P.H.), Dublin 12, Ireland; STAT5, LLC (R.R.), Los Altos, California 94022; Department of Pediatrics (R.R., V.H.), Oregon Health and Science University, Portland, Oregon 97239; Departments of Pediatrics (J.-M.W., H.A.v.D., W.O.), Endocrinology and Metabolic Diseases (H.A.v.D.), and Clinical Genetics (H.A.v.D., M.L.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands; Faculty of Health and Medical Sciences (O.P.), University of Copenhagen, DK-2400 Copenhagen, Denmark; School of Medicine and Pharmacology (B.B.Y.), Western Australia Centre for Health and Ageing (L.F.), Centre for Medical Research (L.F.), and School of Medicine and Pharmacology (L.F.), University of Western Australia, Perth, Western Australia 6872, Australia; Department of Endocrinology and Diabetes (B.B.Y.), Fiona Stanley Hospital, Perth, Western Australia 6150, Australia; Department of Human Biology (G.A.), Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel; and Departments of Medicine and Genetics (N.B., G.A.), Albert Einstein College of Medicine, Bronx, New York 10461
| | - Leon Flicker
- Department of Pathology and Laboratory Medicine (M.M., D.E., S.B., Y.A.), The Children's Hospital of Philadelphia and The University of Pennsylvania, Philadelphia,; Division of Endocrinology and Diabetes (C.P.H., A.G.), The Children's Hospital of Philadelphia, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Pennsylvania 19104; National Children's Research Centre (C.P.H.), Dublin 12, Ireland; STAT5, LLC (R.R.), Los Altos, California 94022; Department of Pediatrics (R.R., V.H.), Oregon Health and Science University, Portland, Oregon 97239; Departments of Pediatrics (J.-M.W., H.A.v.D., W.O.), Endocrinology and Metabolic Diseases (H.A.v.D.), and Clinical Genetics (H.A.v.D., M.L.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands; Faculty of Health and Medical Sciences (O.P.), University of Copenhagen, DK-2400 Copenhagen, Denmark; School of Medicine and Pharmacology (B.B.Y.), Western Australia Centre for Health and Ageing (L.F.), Centre for Medical Research (L.F.), and School of Medicine and Pharmacology (L.F.), University of Western Australia, Perth, Western Australia 6872, Australia; Department of Endocrinology and Diabetes (B.B.Y.), Fiona Stanley Hospital, Perth, Western Australia 6150, Australia; Department of Human Biology (G.A.), Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel; and Departments of Medicine and Genetics (N.B., G.A.), Albert Einstein College of Medicine, Bronx, New York 10461
| | - Nir Barzilai
- Department of Pathology and Laboratory Medicine (M.M., D.E., S.B., Y.A.), The Children's Hospital of Philadelphia and The University of Pennsylvania, Philadelphia,; Division of Endocrinology and Diabetes (C.P.H., A.G.), The Children's Hospital of Philadelphia, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Pennsylvania 19104; National Children's Research Centre (C.P.H.), Dublin 12, Ireland; STAT5, LLC (R.R.), Los Altos, California 94022; Department of Pediatrics (R.R., V.H.), Oregon Health and Science University, Portland, Oregon 97239; Departments of Pediatrics (J.-M.W., H.A.v.D., W.O.), Endocrinology and Metabolic Diseases (H.A.v.D.), and Clinical Genetics (H.A.v.D., M.L.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands; Faculty of Health and Medical Sciences (O.P.), University of Copenhagen, DK-2400 Copenhagen, Denmark; School of Medicine and Pharmacology (B.B.Y.), Western Australia Centre for Health and Ageing (L.F.), Centre for Medical Research (L.F.), and School of Medicine and Pharmacology (L.F.), University of Western Australia, Perth, Western Australia 6872, Australia; Department of Endocrinology and Diabetes (B.B.Y.), Fiona Stanley Hospital, Perth, Western Australia 6150, Australia; Department of Human Biology (G.A.), Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel; and Departments of Medicine and Genetics (N.B., G.A.), Albert Einstein College of Medicine, Bronx, New York 10461
| | - Gil Atzmon
- Department of Pathology and Laboratory Medicine (M.M., D.E., S.B., Y.A.), The Children's Hospital of Philadelphia and The University of Pennsylvania, Philadelphia,; Division of Endocrinology and Diabetes (C.P.H., A.G.), The Children's Hospital of Philadelphia, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Pennsylvania 19104; National Children's Research Centre (C.P.H.), Dublin 12, Ireland; STAT5, LLC (R.R.), Los Altos, California 94022; Department of Pediatrics (R.R., V.H.), Oregon Health and Science University, Portland, Oregon 97239; Departments of Pediatrics (J.-M.W., H.A.v.D., W.O.), Endocrinology and Metabolic Diseases (H.A.v.D.), and Clinical Genetics (H.A.v.D., M.L.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands; Faculty of Health and Medical Sciences (O.P.), University of Copenhagen, DK-2400 Copenhagen, Denmark; School of Medicine and Pharmacology (B.B.Y.), Western Australia Centre for Health and Ageing (L.F.), Centre for Medical Research (L.F.), and School of Medicine and Pharmacology (L.F.), University of Western Australia, Perth, Western Australia 6872, Australia; Department of Endocrinology and Diabetes (B.B.Y.), Fiona Stanley Hospital, Perth, Western Australia 6150, Australia; Department of Human Biology (G.A.), Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel; and Departments of Medicine and Genetics (N.B., G.A.), Albert Einstein College of Medicine, Bronx, New York 10461
| | - Adda Grimberg
- Department of Pathology and Laboratory Medicine (M.M., D.E., S.B., Y.A.), The Children's Hospital of Philadelphia and The University of Pennsylvania, Philadelphia,; Division of Endocrinology and Diabetes (C.P.H., A.G.), The Children's Hospital of Philadelphia, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Pennsylvania 19104; National Children's Research Centre (C.P.H.), Dublin 12, Ireland; STAT5, LLC (R.R.), Los Altos, California 94022; Department of Pediatrics (R.R., V.H.), Oregon Health and Science University, Portland, Oregon 97239; Departments of Pediatrics (J.-M.W., H.A.v.D., W.O.), Endocrinology and Metabolic Diseases (H.A.v.D.), and Clinical Genetics (H.A.v.D., M.L.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands; Faculty of Health and Medical Sciences (O.P.), University of Copenhagen, DK-2400 Copenhagen, Denmark; School of Medicine and Pharmacology (B.B.Y.), Western Australia Centre for Health and Ageing (L.F.), Centre for Medical Research (L.F.), and School of Medicine and Pharmacology (L.F.), University of Western Australia, Perth, Western Australia 6872, Australia; Department of Endocrinology and Diabetes (B.B.Y.), Fiona Stanley Hospital, Perth, Western Australia 6150, Australia; Department of Human Biology (G.A.), Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel; and Departments of Medicine and Genetics (N.B., G.A.), Albert Einstein College of Medicine, Bronx, New York 10461
| | - Yair Argon
- Department of Pathology and Laboratory Medicine (M.M., D.E., S.B., Y.A.), The Children's Hospital of Philadelphia and The University of Pennsylvania, Philadelphia,; Division of Endocrinology and Diabetes (C.P.H., A.G.), The Children's Hospital of Philadelphia, and Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Pennsylvania 19104; National Children's Research Centre (C.P.H.), Dublin 12, Ireland; STAT5, LLC (R.R.), Los Altos, California 94022; Department of Pediatrics (R.R., V.H.), Oregon Health and Science University, Portland, Oregon 97239; Departments of Pediatrics (J.-M.W., H.A.v.D., W.O.), Endocrinology and Metabolic Diseases (H.A.v.D.), and Clinical Genetics (H.A.v.D., M.L.), Leiden University Medical Center, 2300 RC Leiden, The Netherlands; Faculty of Health and Medical Sciences (O.P.), University of Copenhagen, DK-2400 Copenhagen, Denmark; School of Medicine and Pharmacology (B.B.Y.), Western Australia Centre for Health and Ageing (L.F.), Centre for Medical Research (L.F.), and School of Medicine and Pharmacology (L.F.), University of Western Australia, Perth, Western Australia 6872, Australia; Department of Endocrinology and Diabetes (B.B.Y.), Fiona Stanley Hospital, Perth, Western Australia 6150, Australia; Department of Human Biology (G.A.), Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel; and Departments of Medicine and Genetics (N.B., G.A.), Albert Einstein College of Medicine, Bronx, New York 10461
| |
Collapse
|
141
|
Distribution of growth hormone-responsive cells in the mouse brain. Brain Struct Funct 2016; 222:341-363. [PMID: 27072946 DOI: 10.1007/s00429-016-1221-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 03/30/2016] [Indexed: 12/31/2022]
Abstract
Growth hormone (GH) exerts important biological effects primarily related to growth and metabolism. However, the role of GH signaling in the brain is still elusive. To better understand GH functions in the brain, we mapped the distribution of GH-responsive cells and identified the receptors involved in GH central effects. For this purpose, mice received an acute intraperitoneal challenge with specific ligands of the GH receptor (mouse GH), prolactin receptor (prolactin) or both receptors (human GH), and their brains were subsequently processed immunohistochemically to detect the phosphorylated form of STAT5 (pSTAT5). GH induced pSTAT5 immunoreactivity in neurons, but not in astroglial cells of numerous brain regions, including the cerebral cortex, nucleus accumbens, hippocampus, septum and amygdala. The most prominent populations of GH-responsive neurons were located in hypothalamic areas, including several preoptic divisions, and the supraoptic, paraventricular, suprachiasmatic, periventricular, arcuate, ventromedial, dorsomedial, tuberal, posterior and ventral premammillary nuclei. Interestingly, many brainstem structures also exhibited GH-responsive cells. Experiments combining immunohistochemistry for pSTAT5 and in situ hybridization for GH and prolactin receptors revealed that human GH induced pSTAT5 in most, but not all, brain regions through both prolactin and GH receptors. Additionally, males and females exhibited a similar number of GH-responsive cells in forebrain structures known to be sexually dimorphic. In summary, we found GH-responsive cells primarily distributed in brain regions implicated in neurovegetative, emotional/motivational and cognitive functions. Our findings deepen the understanding of GH signaling in the brain and suggest that central GH signaling is likely more ample and complex than formerly recognized.
Collapse
|
142
|
Villarino A, Laurence A, Robinson GW, Bonelli M, Dema B, Afzali B, Shih HY, Sun HW, Brooks SR, Hennighausen L, Kanno Y, O'Shea JJ. Signal transducer and activator of transcription 5 (STAT5) paralog dose governs T cell effector and regulatory functions. eLife 2016; 5. [PMID: 26999798 PMCID: PMC4856466 DOI: 10.7554/elife.08384] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 03/18/2016] [Indexed: 12/13/2022] Open
Abstract
The transcription factor STAT5 is fundamental to the mammalian immune system. However, the relationship between its two paralogs, STAT5A and STAT5B, and the extent to which they are functionally distinct, remain uncertain. Using mouse models of paralog deficiency, we demonstrate that they are not equivalent for CD4+ 'helper' T cells, the principal orchestrators of adaptive immunity. Instead, we find that STAT5B is dominant for both effector and regulatory (Treg) responses and, therefore, uniquely necessary for immunological tolerance. Comparative analysis of genomic distribution and transcriptomic output confirm that STAT5B has fargreater impact but, surprisingly, the data point towards asymmetric expression (i.e. paralog dose), rather than distinct functional properties, as the key distinguishing feature. Thus, we propose a quantitative model of STAT5 paralog activity whereby relative abundance imposes functional specificity (or dominance) in the face of widespread structural homology. DOI:http://dx.doi.org/10.7554/eLife.08384.001 The immune system in mammals is one of the most complex networks in the animal kingdom. One way that its many components communicate is via proteins called cytokines, which are released by cells and detected by receptors on the surface of other cells. This leads to the activation of signals inside the responding cells that alter the activity of genes and, ultimately, direct how they behave. STAT5 is a signal protein that is activated when certain cytokines bind to receptors on the cell surface. Consequently, it is an attractive target for drug therapies that seek to alter immune responses and there is keen interest in understanding how it works. It is an unusual protein in that there are two versions – termed STAT5A and STAT5B – that are produced by two separate genes. Together, STAT5A and STAT5B are fundamental to the immune system but there is considerable debate about whether they perform the same job or have distinct roles. Villarino et al. used a combination of genetic and genomic approaches to investigate how both versions of STAT5 work in mice. The experiments show that STAT5B plays a much bigger role in immune cells than STAT5A. Unexpectedly, the experiments indicate that the disparity is not due to differences in protein activity, but is caused by differences in the amount of these proteins in cells. Villarino et al.’s findings resolve longstanding questions about the relationship between STAT5A and STAT5B within the immune system. A logical next step is to find the molecular mechanisms responsible for causing different amounts of STAT5A and STAT5B to be produced in immune cells. Future work will also compare the roles of STAT5A and STAT5B in non-immune cells and explore whether it might be possible to develop therapies that specifically target one version and not the other. DOI:http://dx.doi.org/10.7554/eLife.08384.002
Collapse
Affiliation(s)
- Alejandro Villarino
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, United States
| | - Arian Laurence
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, United States
| | - Gertraud W Robinson
- Laboratory of Genetics and Physiology, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, United States
| | - Michael Bonelli
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, United States
| | - Barbara Dema
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, United States
| | - Behdad Afzali
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, United States
| | - Han-Yu Shih
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, United States
| | - Hong-Wei Sun
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, United States
| | - Stephen R Brooks
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, United States
| | - Lothar Hennighausen
- Laboratory of Genetics and Physiology, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, United States
| | - Yuka Kanno
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, United States
| | - John J O'Shea
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis, Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, United States
| |
Collapse
|
143
|
Park HJ, Li J, Hannah R, Biddie S, Leal-Cervantes AI, Kirschner K, Flores Santa Cruz D, Sexl V, Göttgens B, Green AR. Cytokine-induced megakaryocytic differentiation is regulated by genome-wide loss of a uSTAT transcriptional program. EMBO J 2016; 35:580-94. [PMID: 26702099 PMCID: PMC4801948 DOI: 10.15252/embj.201592383] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 11/27/2015] [Accepted: 12/01/2015] [Indexed: 11/29/2022] Open
Abstract
Metazoan development is regulated by transcriptional networks, which must respond to extracellular cues including cytokines. The JAK/STAT pathway is a highly conserved regulatory module, activated by many cytokines, in which tyrosine-phosphorylated STATs (pSTATs) function as transcription factors. However, the mechanisms by which STAT activation modulates lineage-affiliated transcriptional programs are unclear. We demonstrate that in the absence of thrombopoietin (TPO), tyrosine-unphosphorylated STAT5 (uSTAT5) is present in the nucleus where it colocalizes with CTCF and represses a megakaryocytic transcriptional program. TPO-mediated phosphorylation of STAT5 triggers its genome-wide relocation to STAT consensus sites with two distinct transcriptional consequences, loss of a uSTAT5 program that restrains megakaryocytic differentiation and activation of a canonical pSTAT5-driven program which includes regulators of apoptosis and proliferation. Transcriptional repression by uSTAT5 reflects restricted access of the megakaryocytic transcription factor ERG to target genes. These results identify a previously unrecognized mechanism of cytokine-mediated differentiation.
Collapse
Affiliation(s)
- Hyun Jung Park
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Stem Cell Institute, Cambridge, UK Department of Haematology, University of Cambridge, Cambridge, UK
| | - Juan Li
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Stem Cell Institute, Cambridge, UK Department of Haematology, University of Cambridge, Cambridge, UK
| | - Rebecca Hannah
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Stem Cell Institute, Cambridge, UK Department of Haematology, University of Cambridge, Cambridge, UK
| | - Simon Biddie
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Stem Cell Institute, Cambridge, UK Department of Haematology, University of Cambridge, Cambridge, UK
| | - Ana I Leal-Cervantes
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Stem Cell Institute, Cambridge, UK Department of Haematology, University of Cambridge, Cambridge, UK
| | - Kristina Kirschner
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Stem Cell Institute, Cambridge, UK Department of Haematology, University of Cambridge, Cambridge, UK
| | - David Flores Santa Cruz
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Stem Cell Institute, Cambridge, UK Department of Haematology, University of Cambridge, Cambridge, UK
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, Veterinary University Vienna, Vienna, Austria
| | - Berthold Göttgens
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Stem Cell Institute, Cambridge, UK Department of Haematology, University of Cambridge, Cambridge, UK
| | - Anthony R Green
- Cambridge Institute for Medical Research, Wellcome Trust/MRC Stem Cell Institute, Cambridge, UK Department of Haematology, University of Cambridge, Cambridge, UK Department of Haematology, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
144
|
Chemical and Hormonal Effects on STAT5b-Dependent Sexual Dimorphism of the Liver Transcriptome. PLoS One 2016; 11:e0150284. [PMID: 26959237 PMCID: PMC4784907 DOI: 10.1371/journal.pone.0150284] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 02/11/2016] [Indexed: 12/21/2022] Open
Abstract
The growth hormone (GH)-activated transcription factor signal transducer and activator of transcription 5b (STAT5b) is a key regulator of sexually dimorphic gene expression in the liver. Suppression of hepatic STAT5b signaling is associated with lipid metabolic dysfunction leading to steatosis and liver cancer. In the companion publication, a STAT5b biomarker gene set was identified and used in a rank-based test to predict both increases and decreases in liver STAT5b activation status/function with high (≥ 97%) accuracy. Here, this computational approach was used to identify chemicals and hormones that activate (masculinize) or suppress (feminize) STAT5b function in a large, annotated mouse liver and primary hepatocyte gene expression compendium. Exposure to dihydrotestosterone and thyroid hormone caused liver masculinization, whereas glucocorticoids, fibroblast growth factor 15, and angiotensin II caused liver feminization. In mouse models of diabetes and obesity, liver feminization was consistently observed and was at least partially reversed by leptin or resveratrol exposure. Chemical-induced feminization of male mouse liver gene expression profiles was a relatively frequent phenomenon: of 156 gene expression biosets from chemically-treated male mice, 29% showed feminization of liver STAT5b function, while <1% showed masculinization. Most (93%) of the biosets that exhibited feminization of male liver were also associated with activation of one or more xenobiotic-responsive receptors, most commonly constitutive activated receptor (CAR) or peroxisome proliferator-activated receptor alpha (PPARα). Feminization was consistently associated with increased expression of peroxisome proliferator-activated receptor gamma (Pparg) but not other lipogenic transcription factors linked to steatosis. GH-activated STAT5b signaling in mouse liver is thus commonly altered by diverse chemicals, and provides a linkage between chemical exposure and dysregulated gene expression associated with adverse effects on the liver.
Collapse
|
145
|
Bathige SDNK, Umasuthan N, Park HC, Lee J. An invertebrate signal transducer and activator of transcription 5 (STAT5) ortholog from the disk abalone, Haliotis discus discus: Genomic structure, early developmental expression, and immune responses to bacterial and viral stresses. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 56:46-56. [PMID: 26616564 DOI: 10.1016/j.dci.2015.11.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 11/19/2015] [Accepted: 11/19/2015] [Indexed: 06/05/2023]
Abstract
Signal transducer and activator of transcription (STAT) family members are key signaling molecules that transduce cellular responses from the cell membrane to the nucleus upon Janus kinase (JAK) activation. Although seven STAT members have been reported in mammals, very limited information on STAT genes in molluscans is available. In this study, we identified and characterized a STAT paralog that is homologous to STAT5 from the disk abalone, Haliotis discus discus, and designated as AbSTAT5. Comparison of the deduced amino acid sequence for AbSTAT5 (790 amino acids) with other counterparts revealed conserved residues important for functions and typical domain regions, including the N-terminal domain, coiled-coil domain, DNA-binding domain, linker domain, and Src homology 2 (SH2) domains as mammalian counterparts. Analysis of STAT phylogeny revealed that AbSTAT5 was clustered with the molluscan subgroup in STAT5 clade with distinct evolution. According to the genomic structure of AbSTAT5, the coding sequence was distributed into 20 exons with 19 introns. Immunologically essential transcription factor-binding sites, such as GATA-1, HNF, SP1, C/EBP, Oct-1, AP1, c-Jun, and Sox-2, were predicted at the 5'-proximal region of AbSTAT5. Expression of AbSTAT5 mRNA was detected in different stages of embryonic development and observed at considerably higher levels in the morula and late veliger stages. Tissue-specific expressional studies revealed that the highest level of AbSTAT5 transcripts was detected in hemocytes, followed by gill tissues. Temporal expressions of AbSTAT5 were analyzed upon live bacterial (Vibrio parahemolyticus and Listeria monocytogenes), viral (viral hemorrhagic septicemia virus), and pathogen-associated molecular pattern (lipopolysaccharides and Poly I:C) stimulations, and significant elevations indicated immune modulation. These results suggest that AbSTAT5 may be involved in maintaining innate immune responses from developmental to adult stages in the disk abalone. Further, this study provides a basis for structural and functional exploration of STAT members in the invertebrate JAK/STAT signaling pathway.
Collapse
Affiliation(s)
- S D N K Bathige
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju Self-Governing Province 690-756, Republic of Korea; Fish Vaccine Research Center, Jeju National University, Jeju Special Self-Governing Province 690-756, Republic of Korea
| | - Navaneethaiyer Umasuthan
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju Self-Governing Province 690-756, Republic of Korea; Fish Vaccine Research Center, Jeju National University, Jeju Special Self-Governing Province 690-756, Republic of Korea
| | - Hae-Chul Park
- Graduate School of Medicine, Korea University, Ansan, Gyeonggido 425-707, Republic of Korea.
| | - Jehee Lee
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju Self-Governing Province 690-756, Republic of Korea; Fish Vaccine Research Center, Jeju National University, Jeju Special Self-Governing Province 690-756, Republic of Korea.
| |
Collapse
|
146
|
Artesunate suppresses tumor growth and induces apoptosis through the modulation of multiple oncogenic cascades in a chronic myeloid leukemia xenograft mouse model. Oncotarget 2016; 6:4020-35. [PMID: 25738364 PMCID: PMC4414170 DOI: 10.18632/oncotarget.3004] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 12/21/2014] [Indexed: 12/16/2022] Open
Abstract
Artesunate (ART), a semi-synthetic derivative of artemisinin, is one of the most commonly used anti-malarial drugs. Also, ART possesses anticancer potential albeit through incompletely understood molecular mechanism(s). Here, the effect of ART on various protein kinases, associated gene products, cellular response, and apoptosis was investigated. The in vivo effect of ART on the growth of human CML xenograft tumors in athymic nu/nu mice was also examined. In our preliminary experiments, we first observed that phosphorylation of p38, ERK, CREB, Chk-2, STAT5, and RSK proteins were suppressed upon ART exposure. Interestingly, ART induced the expression of SOCS-1 protein and depletion of SOCS-1 using siRNA abrogated the STAT5 inhibitory effect of the drug. Also various dephosphorylations caused by ART led to the suppression of various survival gene products and induced apoptosis through caspase-3 activation. Moreover, ART also substantially potentiated the apoptosis induced by chemotherapeutic agents. Finally, when administered intraperitoneally, ART inhibited p38, ERK, STAT5, and CREB activation in tumor tissues and the growth of human CML xenograft tumors in mice without exhibiting any significant adverse effects. Overall, our results suggest that ART exerts its anti-proliferative and pro-apoptotic effects through suppression of multiple signaling cascades in CML both in vitro and in vivo.
Collapse
|
147
|
Waite C, Mejia R, Ascoli M. Gq/11-Dependent Changes in the Murine Ovarian Transcriptome at the End of Gestation. Biol Reprod 2016; 94:62. [PMID: 26843449 PMCID: PMC4829089 DOI: 10.1095/biolreprod.115.136952] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 02/01/2016] [Indexed: 11/26/2022] Open
Abstract
Parturition in rodents is highly dependent on the engagement of the luteal prostaglandin F2 alpha receptor, which, through activation of the Gq/11 family of G proteins, increases the expression of Akr1c18, leading to an increase in progesterone catabolism. To further understand the involvement of Gq/11 on luteolysis and parturition, we used microarray analysis to compare the ovarian transcriptome of mice with a granulosa/luteal cell-specific deletion of Galphaq/11 with their control littermates on Day 18 of pregnancy, when mice from both genotypes are pregnant, and on Day 22, when mice with a granulosa/luteal cell-specific deletion of Galphaq/11 are still pregnant but their control littermates are 1–2 days postpartum. Ovarian genes up-regulated at the end of gestation in a Galphaq/11 -dependent fashion include genes involved in focal adhesion and extracellular matrix interactions. Genes down-regulated at the end of gestation in a Galphaq/11-dependent manner include Serpina6 (which encodes corticosteroid-binding globulin); Enpp2 (which encodes autotaxin, the enzyme responsible for the synthesis of lysophosphatidic acid); genes involved in protein processing and export; reproductive genes, such as Lhcgr; the three genes needed to convert progesterone to estradiol (Cyp17a1, Hsd17b7, and Cyp19a1); and Inha. Activation of ovarian Gq/11 by engagement of the prostaglandin F2 alpha receptor on Day 18 of pregnancy recapitulated the regulation of many but not all of these genes. Thus, although the ovarian transcriptome at the end of gestation is highly dependent on the activation of Gq/11, not all of these changes are dependent on the actions of prostaglandin F2 alpha.
Collapse
Affiliation(s)
- Courtney Waite
- Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Rachel Mejia
- Department of Obstetrics and Gynecology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Mario Ascoli
- Department of Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa
| |
Collapse
|
148
|
White UA, Maier J, Zhao P, Richard AJ, Stephens JM. The modulation of adiponectin by STAT5-activating hormones. Am J Physiol Endocrinol Metab 2016; 310:E129-36. [PMID: 26601851 PMCID: PMC4719028 DOI: 10.1152/ajpendo.00068.2015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 11/23/2015] [Indexed: 11/22/2022]
Abstract
Adiponectin is a hormone secreted from adipocytes that plays an important role in insulin sensitivity and protects against metabolic syndrome. Growth hormone (GH) and prolactin (PRL) are potent STAT5 activators that regulate the expression of several genes in adipocytes. Studies have shown that the secretion of adiponectin from adipose tissue is decreased by treatment with PRL and GH. In this study, we demonstrate that 3T3-L1 adipocytes treated with GH or PRL exhibit a reduction in adiponectin protein levels. Furthermore, we identified three putative STAT5 binding sites in the murine adiponectin promoter and show that only one of these, located at -3,809, binds nuclear protein in a GH- or PRL-dependent manner. Mutation of the STAT5 binding site reduced PRL-dependent protein binding, and supershift analysis revealed that STAT5A and -5B, but not STAT1 and -3, bind to this site in response to PRL. Chromatin immunoprecipitation (IP) analysis demonstrated that only STAT5A, and not STAT1 and -3, bind to the murine adiponectin promoter in a GH-dependent manner in vivo. Adiponectin promoter/reporter constructs were responsive to GH, and chromatin IP analysis reveals that STAT5 binds the adiponectin promoter in vivo following GH stimulation. Overall, these data strongly suggest that STAT5 activators regulate adiponectin transcription through the binding of STAT5 to the -3,809 site that leads to decreased adiponectin expression and secretion. These mechanistic observations are highly consistent with studies in mice and humans that have high GH or PRL levels that are accompanied by lower circulating levels of adiponectin.
Collapse
Affiliation(s)
- Ursula A White
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana; and
| | - Joel Maier
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana
| | - Peng Zhao
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana; and Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana
| | - Allison J Richard
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana; and
| | - Jacqueline M Stephens
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana; and Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana
| |
Collapse
|
149
|
Cordero A, Pellegrini P, Sanz-Moreno A, Trinidad EM, Serra-Musach J, Deshpande C, Dougall WC, Pujana MA, González-Suárez E. Rankl Impairs Lactogenic Differentiation Through Inhibition of the Prolactin/Stat5 Pathway at Midgestation. Stem Cells 2016; 34:1027-39. [PMID: 26695351 DOI: 10.1002/stem.2271] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 10/06/2015] [Accepted: 11/04/2015] [Indexed: 11/12/2022]
Abstract
Prolactin and progesterone both orchestrate the proliferation and differentiation of the mammary gland during gestation. Differentiation of milk secreting alveoli depends on the presence of prolactin receptor, the downstream Jak2-Stat5 pathway and the transcription factor Elf5. A strict regulation of Rank signaling is essential for the differentiation of the mammary gland and in particular for alveolar commitment. Impaired alveologenesis and lactation failure are observed in both, knockout and Rank overexpressing mice; however, the underlying molecular mechanism responsible for these phenotypes remains largely unknown. Using genome-wide expression analyses and functional studies, we show here that Rankl (RL) exposure leads to impaired secretory differentiation of alveolar cells not only in MMTV-RANK but also in wild-type (WT) mammary acini. Conversely, pharmacological blockage of Rank signaling at midgestation in WT mice leads to precocious and exacerbated lactogenesis. Mechanistically, RL negatively regulates Stat5 phosphorylation and Elf5 expression at the onset of lactogenesis. Continuous RL exposure leads to the expansion of basal and bipotent cells in WT and MMTV-RANK acini. Overall, we demonstrate that enhanced Rank signaling impairs secretory differentiation during pregnancy by inhibition of the prolactin/p-Stat5 pathway.
Collapse
Affiliation(s)
- Alex Cordero
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, Barcelona, Spain
| | - Pasquale Pellegrini
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, Barcelona, Spain
| | - Adrián Sanz-Moreno
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, Barcelona, Spain
| | - Eva M Trinidad
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, Barcelona, Spain
| | | | - Chetan Deshpande
- Department of Molecular Sciences and Computational Biology, Amgen Inc., Thousand Oaks, California, USA
| | | | | | - Eva González-Suárez
- Cancer Epigenetics and Biology Program, Bellvitge Biomedical Research Institute, Barcelona, Spain
| |
Collapse
|
150
|
Abstract
Signal transducers and activators of transcription 5 (STAT5a and STAT5b) are highly homologous proteins that are encoded by 2 separate genes and are activated by Janus-activated kinases (JAK) downstream of cytokine receptors. STAT5 proteins are activated by a wide variety of hematopoietic and nonhematopoietic cytokines and growth factors, all of which use the JAK-STAT signalling pathway as their main mode of signal transduction. STAT5 proteins critically regulate vital cellular functions such as proliferation, differentiation, and survival. The physiological importance of STAT5 proteins is underscored by the plethora of primary human tumors that have aberrant constitutive activation of these proteins, which significantly contributes to tumor cell survival and malignant progression of disease. STAT5 plays an important role in the maintenance of normal immune function and homeostasis, both of which are regulated by specific members of IL-2 family of cytokines, which share a common gamma chain (γ(c)) in their receptor complex. STAT5 critically mediates the biological actions of members of the γ(c) family of cytokines in the immune system. Essentially, STAT5 plays a critical role in the function and development of Tregs, and consistently activated STAT5 is associated with a suppression in antitumor immunity and an increase in proliferation, invasion, and survival of tumor cells. Thus, therapeutic targeting of STAT5 is promising in cancer.
Collapse
Affiliation(s)
- Aradhana Rani
- Department of Biomedical Sciences, University of Westminster , London, United Kingdom
| | - John J Murphy
- Department of Biomedical Sciences, University of Westminster , London, United Kingdom
| |
Collapse
|