151
|
Bhakkiyalakshmi E, Suganya N, Sireesh D, Krishnamurthi K, Saravana Devi S, Rajaguru P, Ramkumar KM. Carvacrol induces mitochondria-mediated apoptosis in HL-60 promyelocytic and Jurkat T lymphoma cells. Eur J Pharmacol 2015; 772:92-8. [PMID: 26724845 DOI: 10.1016/j.ejphar.2015.12.046] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 12/22/2015] [Accepted: 12/23/2015] [Indexed: 01/27/2023]
Abstract
The aim of the present study was to investigate the effect of carvacrol, a phenolic monoterpenoid on the induction of apoptosis in HL-60 (Human acute promyelocytic leukemia cells) and Jurkat (human T lymphocyte cells) cells. Carvacrol showed a potent cytotoxic effect on both cells with dose-dependent increase in the level of free radical formation as measured by an oxidation sensitive fluorescent dye, 2,7-dichlorodihydrofluorescein diacetate (H2DCFDA) levels. The reduction in the level of antioxidants such as catalase (CAT) and superoxide dismutase (SOD) (P<0.05) was observed in carvacrol-treated cells. The major cytotoxic effect appears to be intervened by the induction of apoptotic cell death as assessed by annexin-V labeling assay using flow cytometry. Western blot analysis showed that Bax expression was increased, whereas Bcl-2 expression was significantly decreased in carvacrol exposed HL-60 cells and Jurkat cells. Further studies revealed that the dissipation of mitochondrial membrane potential of intact cells was accompanied by the activation of caspase-3. Our results found that the potential mechanism of cellular apoptosis induced by carvacrol is mediated by caspase-3 and is associated with the collapse of mitochondrial membrane potential, generation of free radicals, and depletion of the intracellular antioxidant pool.
Collapse
Affiliation(s)
| | | | | | - Kannan Krishnamurthi
- Environmental Health Division, CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, India
| | - Sivanesan Saravana Devi
- Environmental Health Division, CSIR-National Environmental Engineering Research Institute (NEERI), Nagpur, India
| | - Palanisamy Rajaguru
- Department of Biotechnology, Anna University - BIT Campus, Tiruchirappalli, India
| | | |
Collapse
|
152
|
Raven LA, Cocks BG, Kemper KE, Chamberlain AJ, Vander Jagt CJ, Goddard ME, Hayes BJ. Targeted imputation of sequence variants and gene expression profiling identifies twelve candidate genes associated with lactation volume, composition and calving interval in dairy cattle. Mamm Genome 2015; 27:81-97. [DOI: 10.1007/s00335-015-9613-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 10/28/2015] [Indexed: 10/22/2022]
|
153
|
Ji MR, Lee SI, Jang YJ, Jeon MH, Kim JS, Kim KW, Park JK, Yoo JG, Jeon IS, Kwon DJ, Park CK, Byun SJ. STAT5 plays a critical role in regulating the 5'-flanking region of the porcine whey acidic protein gene in transgenic mice. Mol Reprod Dev 2015; 82:957-66. [PMID: 26256125 DOI: 10.1002/mrd.22535] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 08/06/2015] [Indexed: 11/06/2022]
Abstract
The mammary gland serves as a valuable bioreactor system for the production of recombinant proteins in lactating animals. Pharmaceutical-grade recombinant protein can be harvested from the milk of transgenic animals that carry a protein of interest under the control of promoter regions genes encoding milk proteins. Whey acidic protein (WAP), for example, is predominantly expressed in the mammary gland and is regulated by lactating hormones during pregnancy. We cloned the 5'-flanking region of the porcine WAP gene (pWAP) to confirm the sequence elements in its promoter that are required for gene-expression activity. In the present study, we investigated how lactogenic hormones--including prolactin, hydrocortisone, and insulin--contribute to the transcriptional activation of the pWAP promoter region in mammalian cells, finding that these hormones activate STAT5 signaling, which in turn induce gene expression via STAT5 binding sites in its 5'-flanking region. To confirm the expression and hormonal regulation of the 5'-flanking region of pWAP in vivo, we generated transgenic mice expressing human recombinant granulocyte colony stimulating factor (hCSF2) in the mammary gland under the control of the pWAP promoter. These mice secreted hCSF2 protein in their milk at levels ranging from 242 to 1,274.8 ng/ml. Collectively, our findings show that the pWAP promoter may be useful for confining the expression of foreign proteins to the mammary gland, where they can be secreted along with milk.
Collapse
Affiliation(s)
- Mi-Ran Ji
- Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500, Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, Republic of Korea
| | - Sang In Lee
- Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500, Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, Republic of Korea.,Department of Animal Resource and Science, Dankook University, Cheonan, Choongnam, Republic of Korea
| | - Ye-Jin Jang
- Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500, Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, Republic of Korea
| | - Mi-Hyang Jeon
- Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500, Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, Republic of Korea
| | - Jeom Sun Kim
- Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500, Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, Republic of Korea
| | - Kyung-Woon Kim
- Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500, Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, Republic of Korea
| | - Jin-Ki Park
- Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500, Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, Republic of Korea
| | - Jae Gyu Yoo
- Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500, Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, Republic of Korea
| | - Ik-Soo Jeon
- Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500, Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, Republic of Korea
| | - Dae-Jin Kwon
- Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500, Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, Republic of Korea
| | - Choon-Keun Park
- College of Animal Life Sciences, Kangwon National University, Chuncheon, Republic of Korea
| | - Sung June Byun
- Animal Biotechnology Division, National Institute of Animal Science, RDA, 1500, Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, Republic of Korea
| |
Collapse
|
154
|
Miyado M, Miyado K, Katsumi M, Saito K, Nakamura A, Shihara D, Ogata T, Fukami M. Parturition failure in mice lacking Mamld1. Sci Rep 2015; 5:14705. [PMID: 26435405 PMCID: PMC4592954 DOI: 10.1038/srep14705] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 09/07/2015] [Indexed: 11/13/2022] Open
Abstract
In mice, the onset of parturition is triggered by a rapid decline in circulating progesterone. Progesterone withdrawal occurs as a result of functional luteolysis, which is characterized by an increase in the enzymatic activity of 20α-hydroxysteroid dehydrogenase (20α-HSD) in the corpus luteum and is mediated by the prostaglandin F2α (PGF2α) signaling. Here, we report that the genetic knockout (KO) of Mamld1, which encodes a putative non-DNA-binding regulator of testicular steroidogenesis, caused defective functional luteolysis and subsequent parturition failure and neonatal deaths. Progesterone receptor inhibition induced the onset of parturition in pregnant KO mice, and MAMLD1 regulated the expression of Akr1c18, the gene encoding 20α-HSD, in cultured cells. Ovaries of KO mice at late gestation were morphologically unremarkable; however, Akr1c18 expression was reduced and expression of its suppressor Stat5b was markedly increased. Several other genes including Prlr, Cyp19a1, Oxtr, and Lgals3 were also dysregulated in the KO ovaries, whereas PGF2α signaling genes remained unaffected. These results highlight the role of MAMLD1 in labour initiation. MAMLD1 likely participates in functional luteolysis by regulating Stat5b and other genes, independent of the PGF2α signaling pathway.
Collapse
Affiliation(s)
- Mami Miyado
- Department of Molecular Endocrinology, National Research Institute of Child Health and Development, Tokyo 157-8535, Japan
| | - Kenji Miyado
- Department of Reproductive Biology, National Research Institute of Child Health and Development, Tokyo 157-8535, Japan
| | - Momori Katsumi
- Department of Molecular Endocrinology, National Research Institute of Child Health and Development, Tokyo 157-8535, Japan
| | - Kazuki Saito
- Department of Molecular Endocrinology, National Research Institute of Child Health and Development, Tokyo 157-8535, Japan
| | - Akihiro Nakamura
- Department of Reproductive Biology, National Research Institute of Child Health and Development, Tokyo 157-8535, Japan
| | - Daizou Shihara
- Department of Molecular Endocrinology, National Research Institute of Child Health and Development, Tokyo 157-8535, Japan
| | - Tsutomu Ogata
- Department of Molecular Endocrinology, National Research Institute of Child Health and Development, Tokyo 157-8535, Japan.,Department of Pediatrics, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| | - Maki Fukami
- Department of Molecular Endocrinology, National Research Institute of Child Health and Development, Tokyo 157-8535, Japan
| |
Collapse
|
155
|
Gao P, Zhang Y, Liu Y, Chen J, Zong C, Yu C, Cui S, Gao W, Qin D, Sun W, Li X, Wang X. Signal transducer and activator of transcription 5B (STAT5B) modulates adipocyte differentiation via MOF. Cell Signal 2015; 27:2434-43. [PMID: 26388045 DOI: 10.1016/j.cellsig.2015.09.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/14/2015] [Accepted: 09/15/2015] [Indexed: 11/28/2022]
Abstract
The role and mechanism of signal transducer and activator of transcription 5B (STAT5B) in adipogenesis remain unclear. In this study, our data showed that Males absent on the first (MOF) protein expression was increased during 3 T3-L1 preadipocytes differentiation accompanied with STAT5B expression increasing. Over-expression STAT5B enhanced MOF promoter trans-activation in HeLa cells. Mutagenesis assay and ChIP analysis exhibited that STAT5B was able to bind MOF promoter. Knocking-down STAT5B in 3 T3-L1 preadipocytes led to decreased expression of MOF, but resulted in increased expression of peroxisome proliferator-activated receptor γ (PPARγ), CCAAT/enhancer-binding protein α (C/EBPα) and fatty acid-binding protein 4 (Fabp4), which were important factors or enzymes for adipogenesis. We also found that knocking-down MOF in 3 T3-L1 preadipocytes resulted in increased expression of PPARγ, C/EBPα and Fabp4, which was in the same trend as STAT5B knocking-down. Over-expression MOF resulted in reduced promoter trans-activation activity of C/EBPα. These results suggest that STAT5B and MOF work as negative regulators in adipogenesis, and STAT5B modulates preadipocytes differentiation partially by regulating MOF expression.
Collapse
Affiliation(s)
- Peng Gao
- The Department of Cell Biology, Shandong University School of Medicine, Jinan 250012, China.
| | - Yuchao Zhang
- The Department of Cell Biology, Shandong University School of Medicine, Jinan 250012, China; Center for Reproductive Medicine, National Research Center for Assisted reproductive Technology and Reproductive Genetics, The Key Laboratory for Reproductive Endocrinology of Ministry of Education, Shandong Provincial Key Laboratory of Reproductive Medicine, Jinan 250021, China.
| | - Yuantao Liu
- Department of Endocrinology, Qingdao Municipal Hospital, Qingdao 266071, China.
| | - Jicui Chen
- The Department of Cell Biology, Shandong University School of Medicine, Jinan 250012, China.
| | - Chen Zong
- The Department of Cell Biology, Shandong University School of Medicine, Jinan 250012, China.
| | - Cong Yu
- The Department of Cell Biology, Shandong University School of Medicine, Jinan 250012, China.
| | - Shang Cui
- The Department of Cell Biology, Shandong University School of Medicine, Jinan 250012, China.
| | - Weina Gao
- The Department of Cell Biology, Shandong University School of Medicine, Jinan 250012, China.
| | - Dandan Qin
- The Department of Cell Biology, Shandong University School of Medicine, Jinan 250012, China.
| | - Wenchuan Sun
- Department of Nephrology, the Second Hospital of Shandong University, Jinan 250033, China.
| | - Xia Li
- The Department of Cell Biology, Shandong University School of Medicine, Jinan 250012, China.
| | - Xiangdong Wang
- The Department of Cell Biology, Shandong University School of Medicine, Jinan 250012, China; Key Laboratory of Protein Sciences for Chronic Degenerative Diseases in Universities of Shandong (Shandong University), Jinan 250012, China.
| |
Collapse
|
156
|
Alzhanov D, Mukherjee A, Rotwein P. Identifying growth hormone-regulated enhancers in the Igf1 locus. Physiol Genomics 2015; 47:559-68. [PMID: 26330488 DOI: 10.1152/physiolgenomics.00062.2015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 08/27/2015] [Indexed: 11/22/2022] Open
Abstract
Growth hormone (GH) plays a central role in regulating somatic growth and in controlling multiple physiological processes in humans and other vertebrates. A key agent in many GH actions is the secreted peptide, IGF-I. As established previously, GH stimulates IGF-I gene expression via the Stat5b transcription factor, leading to production of IGF-I mRNAs and proteins. However, the precise mechanisms by which GH-activated Stat5b promotes IGF-I gene transcription have not been defined. Unlike other GH-regulated genes, there are no Stat5b sites near either of the two IGF-I gene promoters. Although dispersed GH-activated Stat5b binding elements have been mapped in rodent Igf1 gene chromatin, it is unknown how these distal sites might function as potential transcriptional enhancers. Here we have addressed mechanisms of regulation of IGF-I gene transcription by GH by generating cell lines in which the rat Igf1 chromosomal locus has been incorporated into the mouse genome. Using these cells we find that physiological levels of GH rapidly and potently activate Igf1 gene transcription while stimulating physical interactions in chromatin between inducible Stat5b-binding elements and the Igf1 promoters. We have thus developed a robust experimental platform for elucidating how dispersed transcriptional enhancers control Igf1 gene expression under different biological conditions.
Collapse
Affiliation(s)
- Damir Alzhanov
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon; and
| | - Aditi Mukherjee
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon; and
| | - Peter Rotwein
- Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas
| |
Collapse
|
157
|
Abstract
Major progress has been recently made in understanding the molecular pathogenesis of myeloproliferative neoplasms (MPN). Mutations in one of four genes-JAK2, MPL, CALR, and CSF3R-can be found in the vast majority of patients with MPN and represent driver mutations that can induce the MPN phenotype. Hyperactive JAK/STAT signaling appears to be the common denominator of MPN, even in patients with CALR mutations and the so-called "triple-negative" MPN, where the driver gene mutation is still unknown. Mutations in epigenetic regulators, transcription factors, and signaling components modify the course of the disease and can contribute to disease initiation and/or progression. The central role of JAK2 in MPN allowed development of small molecular inhibitors that are in clinical use and are active in almost all patients with MPN. Advances in understanding the mechanism of JAK2 activation open new perspectives of developing the next generation of inhibitors that will be selective for the mutated forms of JAK2.
Collapse
|
158
|
Chen JQ, Mori H, Cardiff RD, Trott JF, Hovey RC, Hubbard NE, Engelberg JA, Tepper CG, Willis BJ, Khan IH, Ravindran RK, Chan SR, Schreiber RD, Borowsky AD. Abnormal Mammary Development in 129:STAT1-Null Mice is Stroma-Dependent. PLoS One 2015; 10:e0129895. [PMID: 26075897 PMCID: PMC4468083 DOI: 10.1371/journal.pone.0129895] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Accepted: 05/14/2015] [Indexed: 11/18/2022] Open
Abstract
Female 129:Stat1-null mice (129S6/SvEvTac-Stat1tm1Rds homozygous) uniquely develop estrogen-receptor (ER)-positive mammary tumors. Herein we report that the mammary glands (MG) of these mice have altered growth and development with abnormal terminal end buds alongside defective branching morphogenesis and ductal elongation. We also find that the 129:Stat1-null mammary fat pad (MFP) fails to sustain the growth of 129S6/SvEv wild-type and Stat1-null epithelium. These abnormalities are partially reversed by elevated serum progesterone and prolactin whereas transplantation of wild-type bone marrow into 129:Stat1-null mice does not reverse the MG developmental defects. Medium conditioned by 129:Stat1-null epithelium-cleared MFP does not stimulate epithelial proliferation, whereas it is stimulated by medium conditioned by epithelium-cleared MFP from either wild-type or 129:Stat1-null females having elevated progesterone and prolactin. Microarrays and multiplexed cytokine assays reveal that the MG of 129:Stat1-null mice has lower levels of growth factors that have been implicated in normal MG growth and development. Transplanted 129:Stat1-null tumors and their isolated cells also grow slower in 129:Stat1-null MG compared to wild-type recipient MG. These studies demonstrate that growth of normal and neoplastic 129:Stat1-null epithelium is dependent on the hormonal milieu and on factors from the mammary stroma such as cytokines. While the individual or combined effects of these factors remains to be resolved, our data supports the role of STAT1 in maintaining a tumor-suppressive MG microenvironment.
Collapse
Affiliation(s)
- Jane Q. Chen
- Center for Comparative Medicine, University of California, Davis, California, United States of America
| | - Hidetoshi Mori
- Center for Comparative Medicine, University of California, Davis, California, United States of America
| | - Robert D. Cardiff
- Center for Comparative Medicine, University of California, Davis, California, United States of America
| | - Josephine F. Trott
- Department of Animal Science, University of California, Davis, California, United States of America
| | - Russell C. Hovey
- Department of Animal Science, University of California, Davis, California, United States of America
| | - Neil E. Hubbard
- Center for Comparative Medicine, University of California, Davis, California, United States of America
| | - Jesse A. Engelberg
- Center for Comparative Medicine, University of California, Davis, California, United States of America
| | - Clifford G. Tepper
- Division of Basic Sciences, Cancer Center and Department of Biochemistry and Molecular Medicine, University of California, Davis, School of Medicine, Sacramento, California, United States of America
| | - Brandon J. Willis
- Mouse Biology Program, University of California, Davis, California, United States of America
| | - Imran H. Khan
- Center for Comparative Medicine, University of California, Davis, California, United States of America
| | - Resmi K. Ravindran
- Center for Comparative Medicine, University of California, Davis, California, United States of America
| | - Szeman R. Chan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Robert D. Schreiber
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Alexander D. Borowsky
- Center for Comparative Medicine, University of California, Davis, California, United States of America
- Department of Pathology and Laboratory Medicine, University of California, Davis, School of Medicine, Sacramento, California, United States of America
- * E-mail:
| |
Collapse
|
159
|
Boulanger CA, Rosenfield SM, George AL, Smith GH. Hormone signaling requirements for the conversion of non-mammary mouse cells to mammary cell fate(s) in vivo. J Mammary Gland Biol Neoplasia 2015; 20:93-101. [PMID: 26362796 PMCID: PMC4595519 DOI: 10.1007/s10911-015-9343-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 09/03/2015] [Indexed: 12/25/2022] Open
Abstract
Mammotropic hormones and growth factors play a very important role in mammary growth and differentiation. Here, hormones including Estrogen, Progesterone, Prolactin, their cognate receptors, and the growth factor Amphiregulin, are tested with respect to their roles in signaling non-mammary cells from the mouse to redirect to mammary epithelial cell fate(s). This was done in the context of glandular regeneration in pubertal athymic female mice. Our previous studies demonstrated that mammary stem cell niches are recapitulated during gland regeneration in vivo. During this process, cells of exogenous origin cooperate with mammary epithelial cells to form mammary stem cell niches and thus respond to normal developmental signals. In all cases tested with the possible exception of estrogen receptor alpha (ER-α), hormone signaling is dispensable for non-mammary cells to undertake mammary epithelial cell fate(s), proliferate, and contribute progeny to chimeric mammary outgrowths. Importantly, redirected non-mammary cell progeny, regardless of their source, have the ability to self-renew and contribute offspring to secondary mammary outgrowths derived from transplanted chimeric mammary fragments; thus suggesting that some of these cells are capable of mammary stem cell/progenitor functions.
Collapse
Affiliation(s)
- Corinne A Boulanger
- Mammary Stem Cell Biology Section, BRL, CCR, NCI, Bldg. 37 Rm. 1122A, Bethesda, MD, 20892, USA
| | - Sonia M Rosenfield
- Mammary Stem Cell Biology Section, BRL, CCR, NCI, Bldg. 37 Rm. 1122A, Bethesda, MD, 20892, USA
| | - Andrea L George
- Mammary Stem Cell Biology Section, BRL, CCR, NCI, Bldg. 37 Rm. 1122A, Bethesda, MD, 20892, USA
| | - Gilbert H Smith
- Mammary Stem Cell Biology Section, BRL, CCR, NCI, Bldg. 37 Rm. 1122A, Bethesda, MD, 20892, USA.
| |
Collapse
|
160
|
Gorvin CM. The prolactin receptor: Diverse and emerging roles in pathophysiology. JOURNAL OF CLINICAL AND TRANSLATIONAL ENDOCRINOLOGY 2015; 2:85-91. [PMID: 29204371 PMCID: PMC5685068 DOI: 10.1016/j.jcte.2015.05.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 05/10/2015] [Indexed: 12/21/2022]
Abstract
Investigations over two decades have revised understanding of the prolactin hormone. Long thought to be merely a lactogenic hormone, its list of functions has been extended to include: reproduction, islet differentiation, adipocyte control and immune modulation. Prolactin functions by binding cell-surface expressed prolactin receptor, initiating signaling cascades, primarily utilizing Janus kinase-signal transducer and activator of transcription (JAK-STAT). Pathway disruption has been implicated in tumorigenesis, reproductive abnormalities, and diabetes. Prolactin can also be secreted from extrapituitary sources adding complexity to understanding of its physiological functions. This review aims to describe how prolactin exerts its pathophysiological roles by endocrine and autocrine means.
Collapse
Affiliation(s)
- Caroline M Gorvin
- Academic Endocrine Unit, University of Oxford, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, Oxford, OX3 7LJ, UK
| |
Collapse
|
161
|
Buonfiglio DC, Ramos-Lobo AM, Silveira MA, Furigo IC, Hennighausen L, Frazão R, Donato J. Neuronal STAT5 signaling is required for maintaining lactation but not for postpartum maternal behaviors in mice. Horm Behav 2015; 71:60-8. [PMID: 25896118 PMCID: PMC6282758 DOI: 10.1016/j.yhbeh.2015.04.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 04/04/2015] [Accepted: 04/10/2015] [Indexed: 12/17/2022]
Abstract
Prolactin and placental lactogens control mammary development and lactation as well as play an important role in maternal behaviors. However, the molecular mechanisms in the brain responsible for this regulation remain largely unknown. Therefore, the present study investigated whether Signal Transducer and Activator of Transcription 5 (STAT5) signaling in the brain, the key transcriptional factor recruited by prolactin receptor and other hormones, is required for postpartum maternal behavior, maintenance of lactation and offspring growth. Neuronal ablation of STAT5 impaired the control of prolactin secretion and reduced the hypothalamic expression of suppressors of cytokine signaling (i.e., SOCS3 and CISH). In addition, neuronal STAT5 deletion attenuated the hyperphagia commonly observed during lactation by decreasing the hypothalamic expression of orexigenic neurotransmitters such as the neuropeptide Y and agouti-related protein. The lower food intake of lactating neuron-specific STAT5 knockout females resulted in reduced milk production and offspring growth. Unexpectedly, postpartum maternal behavior expression was not impaired in neuron-specific STAT5 knockout females. On the contrary, the latency to retrieve and group the pups into the nest was reduced in mutant dams. Finally, we demonstrated that approximately 30% of recorded neurons in the medial preoptic area were acutely depolarized by prolactin suggesting that fast STAT5-independent signaling pathways may be involved in the regulation of maternal behaviors. Overall, our results revealed important information about the molecular mechanisms recruited by hormones to orchestrate the activation of neural circuitries engaged in the induction of maternal care.
Collapse
Affiliation(s)
- Daniella C Buonfiglio
- 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
| | - Marina A Silveira
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-900, Brazil
| | - Isadora C Furigo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil
| | - Lothar Hennighausen
- Laboratory of Genetics and Physiology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Renata Frazão
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-900, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil.
| |
Collapse
|
162
|
Ortutay Z, Oksanen A, Aittomäki S, Ortutay C, Pesu M. Proprotein convertase FURIN regulates T cell receptor-induced transactivation. J Leukoc Biol 2015; 98:73-83. [PMID: 25926688 DOI: 10.1189/jlb.2a0514-257rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 04/08/2015] [Indexed: 12/23/2022] Open
Abstract
Antigen emergence rapidly stimulates T cells, which leads to changes in cytokine production, cell proliferation, and differentiation. Some of the key molecules involved in these events, such as TGF-β1 and NOTCH1, are synthesized initially as inactive precursors and are proteolytically activated during T cell activation. PCSKs regulate proprotein maturation by catalyzing the proteolytic cleavage of their substrates. The prototype PCSK FURIN is induced upon TCR activation, and its expression in T cells is critical for the maintenance of peripheral immune tolerance. In this study, we tested the hypothesis that FURIN regulates T cell activation. Our data demonstrate that IL-2 is increased initially in FURIN-deficient mouse CD4(+) T cells, but the TCR-induced IL-2 mRNA expression is not sustained in the absence of FURIN. Accordingly, the inhibition of FURIN in human Jurkat T cell lines also results in a decrease in IL-2 production, whereas the overexpression of WT FURIN is associated with elevated IL-2 levels. In Jurkat cells, FURIN is dispensable for immediate TCR signaling steps, such as ERK, ZAP70, or LAT phosphorylation. However, with the use of gene reporter assays, we demonstrate that FURIN regulates the AP-1, NFAT, and NF-κB transcription factors. Finally, by performing a transcription factor-binding site enrichment analysis on FURIN-dependent transcriptomes, we identify the FURIN-regulated transcription factors in mouse CD4(+) T cell subsets. Collectively, our work confirms the hypothesis that the TCR-regulated protease FURIN plays an important role in T cell activation and that it can specifically modulate TCR-activated transactivation.
Collapse
Affiliation(s)
- Zsuzsanna Ortutay
- *Immunoregulation, BioMediTech, University of Tampere, Finland; HiDucator Oy, Kangasala, Finland; and Department of Dermatology and Fimlab Laboratories, Tampere University Hospital, Pirkanmaa Hospital District, Tampere, Finland
| | - Anna Oksanen
- *Immunoregulation, BioMediTech, University of Tampere, Finland; HiDucator Oy, Kangasala, Finland; and Department of Dermatology and Fimlab Laboratories, Tampere University Hospital, Pirkanmaa Hospital District, Tampere, Finland
| | - Saara Aittomäki
- *Immunoregulation, BioMediTech, University of Tampere, Finland; HiDucator Oy, Kangasala, Finland; and Department of Dermatology and Fimlab Laboratories, Tampere University Hospital, Pirkanmaa Hospital District, Tampere, Finland
| | - Csaba Ortutay
- *Immunoregulation, BioMediTech, University of Tampere, Finland; HiDucator Oy, Kangasala, Finland; and Department of Dermatology and Fimlab Laboratories, Tampere University Hospital, Pirkanmaa Hospital District, Tampere, Finland
| | - Marko Pesu
- *Immunoregulation, BioMediTech, University of Tampere, Finland; HiDucator Oy, Kangasala, Finland; and Department of Dermatology and Fimlab Laboratories, Tampere University Hospital, Pirkanmaa Hospital District, Tampere, Finland
| |
Collapse
|
163
|
Keogh K, Waters SM, Kelly AK, Wylie ARG, Kenny DA. Effect of feed restriction and subsequent re-alimentation on hormones and genes of the somatotropic axis in cattle. Physiol Genomics 2015; 47:264-73. [PMID: 25921585 DOI: 10.1152/physiolgenomics.00134.2014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 04/25/2015] [Indexed: 11/22/2022] Open
Abstract
The objective of this study was to characterize the effect of feed restriction and compensatory growth during re-alimentation on the functionality of the somatotropic axis. We blocked 60 bulls into one of two groups: 1) restricted feed allowance for 125 days (period 1) (RES, n = 30) followed by ad libitum feeding for 55 days (period 2) or 2) ad libitum access to feed throughout (ADLIB, n = 30). A growth hormone releasing hormone (GHRH) challenge was performed during each period. At the end of each period, 15 animals from each treatment were slaughtered and hepatic tissue collected. Hepatic expression of 13 genes of the somatotropic axis was measured by qRT-PCR. RES displayed a lower growth rate during period 1 (0.6 vs. 1.9 kg/day; P < 0.001), subsequently gaining more than ADLIB animals during period 2 (2.5 vs. 1.4 kg/day; P < 0.001). Growth hormone response to GHRH was not different between treatments at either time-point (P > 0.05); however, resultant plasma IGF-1 was lower in period 1 and greater in period 2 in RES animals (P < 0.05). Expression of IGFBP2 was higher (P < 0.01) and IGF1 (P < 0.001) and GHRIA (P < 0.05) lower in RES compared with ADLIB during period 1, with no difference evident in period 2 (P > 0.05). Collectively, the results of this study are consistent with uncoupling of the somatotropic axis following feed restriction. However, there is no evidence from this study that the somatotropic axis per se is a significant contributor to compensatory growth.
Collapse
Affiliation(s)
- Kate Keogh
- Animal and Bioscience Research Department, Animal and Grassland Research and Innovation Centre, Teagasc, Dunsany, County Meath, Ireland; University College Dublin School of Agriculture and Food Science, Belfield, Dublin, Ireland; and
| | - Sinéad M Waters
- Animal and Bioscience Research Department, Animal and Grassland Research and Innovation Centre, Teagasc, Dunsany, County Meath, Ireland
| | - Alan K Kelly
- University College Dublin School of Agriculture and Food Science, Belfield, Dublin, Ireland; and
| | | | - David A Kenny
- Animal and Bioscience Research Department, Animal and Grassland Research and Innovation Centre, Teagasc, Dunsany, County Meath, Ireland;
| |
Collapse
|
164
|
Otero DC, Fares-Frederickson NJ, Xiao M, Baker DP, David M. IFN-β Selectively Inhibits IL-2 Production through CREM-Mediated Chromatin Remodeling. THE JOURNAL OF IMMUNOLOGY 2015; 194:5120-8. [PMID: 25888642 DOI: 10.4049/jimmunol.1403181] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 03/24/2015] [Indexed: 01/14/2023]
Abstract
IFN-β is widely used in the treatment of multiple sclerosis, yet the mechanism facilitating its efficacy remains unclear. IL-2 production by activated T cells, including those mediating autoimmunity, and subsequent autocrine stimulation is vital for T cell expansion and function. In this study, we demonstrate that in mouse and human T cells, IFN-β specifically inhibits the production of IL-2 upon TCR engagement without affecting other cytokines or activation markers. Rather than disrupting TCR signaling, IFN-β alters histone modifications in the IL-2 promoter to retain the locus in an inaccessible configuration. This in turn is mediated through the upregulation of the transcriptional suppressor CREM by IFN-β and consequent recruitment of histone deacetylases to the IL-2 promoter. In accordance, ablation of CREM expression or inhibition of histone deacetylases activity eliminates the suppressive effects of IFN-β on IL-2 production. Collectively, these findings provide a molecular basis by which IFN-β limits T cell responses.
Collapse
Affiliation(s)
- Dennis C Otero
- Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093
| | | | - Menghong Xiao
- Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093
| | | | - Michael David
- Division of Biological Sciences, University of California San Diego, La Jolla, CA 92093; UC San Diego Moores Cancer Center, University of California San Diego, La Jolla, CA 92093
| |
Collapse
|
165
|
Cuesta-Domínguez Á, León-Rico D, Álvarez L, Díez B, Bodega-Mayor I, Baños R, Martín-Rey MÁ, Santos-Roncero M, Gaspar ML, Martín-Acosta P, Almarza E, Bueren JA, Río P, Fernández-Ruiz E. BCR-JAK2 drives a myeloproliferative neoplasm in transplanted mice. J Pathol 2015; 236:219-28. [DOI: 10.1002/path.4513] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 01/28/2015] [Accepted: 02/05/2015] [Indexed: 01/13/2023]
Affiliation(s)
- Álvaro Cuesta-Domínguez
- Molecular Biology Unit; Instituto de Investigación Sanitaria Princesa (IIS-P, UAM), Hospital Universitario de La Princesa; Madrid Spain
| | - Diego León-Rico
- Division of Haematopoietic Innovative Therapies; CIEMAT/CIBERER; Madrid Spain
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD, UAM); Madrid Spain
| | - Lara Álvarez
- Division of Haematopoietic Innovative Therapies; CIEMAT/CIBERER; Madrid Spain
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD, UAM); Madrid Spain
| | - Begoña Díez
- Division of Haematopoietic Innovative Therapies; CIEMAT/CIBERER; Madrid Spain
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD, UAM); Madrid Spain
| | - Irene Bodega-Mayor
- Molecular Biology Unit; Instituto de Investigación Sanitaria Princesa (IIS-P, UAM), Hospital Universitario de La Princesa; Madrid Spain
| | - Rocío Baños
- Division of Haematopoietic Innovative Therapies; CIEMAT/CIBERER; Madrid Spain
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD, UAM); Madrid Spain
| | - Miguel Ángel Martín-Rey
- Division of Haematopoietic Innovative Therapies; CIEMAT/CIBERER; Madrid Spain
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD, UAM); Madrid Spain
| | - Matilde Santos-Roncero
- Molecular Biology Unit; Instituto de Investigación Sanitaria Princesa (IIS-P, UAM), Hospital Universitario de La Princesa; Madrid Spain
| | - María Luisa Gaspar
- Centro Nacional de Microbiología; Instituto de Salud Carlos III (ISCIII); Majadahonda Spain
| | - Paloma Martín-Acosta
- Servicio de Anatomía Patológica; Hospital Universitario Puerta de Hierro; Majadahonda Spain
| | - Elena Almarza
- Division of Haematopoietic Innovative Therapies; CIEMAT/CIBERER; Madrid Spain
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD, UAM); Madrid Spain
| | - Juan A. Bueren
- Division of Haematopoietic Innovative Therapies; CIEMAT/CIBERER; Madrid Spain
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD, UAM); Madrid Spain
| | - Paula Río
- Division of Haematopoietic Innovative Therapies; CIEMAT/CIBERER; Madrid Spain
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD, UAM); Madrid Spain
| | - Elena Fernández-Ruiz
- Molecular Biology Unit; Instituto de Investigación Sanitaria Princesa (IIS-P, UAM), Hospital Universitario de La Princesa; Madrid Spain
| |
Collapse
|
166
|
Kim WS, Zhu Y, Deng Q, Chin CJ, He CB, Grieco AJ, Dravid GG, Parekh C, Hollis RP, Lane TF, Bouhassira EE, Kohn DB, Crooks GM. Erythropoiesis from human embryonic stem cells through erythropoietin-independent AKT signaling. Stem Cells 2015; 32:1503-14. [PMID: 24677652 DOI: 10.1002/stem.1677] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 12/16/2013] [Accepted: 01/11/2014] [Indexed: 12/25/2022]
Abstract
Unlimited self renewal capacity and differentiation potential make human pluripotent stem cells (PSC) a promising source for the ex vivo manufacture of red blood cells (RBCs) for safe transfusion. Current methods to induce erythropoiesis from PSC suffer from low yields of RBCs, most of which are immature and contain embryonic and fetal rather than adult hemoglobins. We have previously shown that homodimerization of the intracellular component of MPL (ic-MPL) induces erythropoiesis from human cord blood progenitors. The goal of this study was to investigate the potential of ic-MPL dimerization to induce erythropoiesis from human embryonic stem cells (hESCs) and to identify the signaling pathways activated by this strategy. We present here the evidence that ic-MPL dimerization induces erythropoietin (EPO)-independent erythroid differentiation from hESC by inducing the generation of erythroid progenitors and by promoting more efficient erythroid maturation with increased RBC enucleation as well as increased gamma:epsilon globin ratio and production of beta-globin protein. ic-MPL dimerization is significantly more potent than EPO in inducing erythropoiesis, and its effect is additive to EPO. Signaling studies show that dimerization of ic-MPL, unlike stimulation of the wild type MPL receptor, activates AKT in the absence of JAK2/STAT5 signaling. AKT activation upregulates GATA-1 and FOXO3 transcriptional pathways with resulting inhibition of apoptosis, modulation of cell cycle, and enhanced maturation of erythroid cells. These findings open up potential new targets for the generation of therapeutically relevant RBC products from hPSC.
Collapse
Affiliation(s)
- William S Kim
- Department of Pathology and Laboratory Medicine, University of California Los Angeles (UCLA),, Los Angeles, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Sun X, Elangovan VR, Mapes B, Camp SM, Sammani S, Saadat L, Ceco E, Ma SF, Flores C, MacDougall MS, Quijada H, Liu B, Kempf CL, Wang T, Chiang ET, Garcia JGN. The NAMPT promoter is regulated by mechanical stress, signal transducer and activator of transcription 5, and acute respiratory distress syndrome-associated genetic variants. Am J Respir Cell Mol Biol 2015; 51:660-7. [PMID: 24821571 DOI: 10.1165/rcmb.2014-0117oc] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Increased nicotinamide phosphoribosyltransferase (NAMPT) transcription is mechanistically linked to ventilator-induced inflammatory lung injury (VILI), with VILI severity attenuated by reduced NAMPT bioavailability. The molecular mechanisms of NAMPT promoter regulation in response to excessive mechanical stress remain poorly understood. The objective of this study was to define the contribution of specific transcription factors, acute respiratory distress syndrome (ARDS)-associated single nucleotide polymorphisms (SNPs), and promoter demethylation to NAMPT transcriptional regulation in response to mechanical stress. In vivo NAMPT protein expression levels were examined in mice exposed to high tidal volume mechanical ventilation. In vitro NAMPT expression levels were examined in human pulmonary artery endothelial cells exposed to 5 or 18% cyclic stretch (CS), with NAMPT promoter activity assessed using NAMPT promoter luciferase reporter constructs with a series of nested deletions. In vitro NAMPT transcriptional regulation was further characterized by measuring luciferase activity, DNA demethylation, and chromatin immunoprecipitation. VILI-challenged mice exhibited significantly increased NAMPT expression in bronchoalveolar lavage leukocytes and in lung endothelium. A mechanical stress-inducible region (MSIR) was identified in the NAMPT promoter from -2,428 to -2,128 bp. This MSIR regulates NAMPT promoter activity, mRNA expression, and signal transducer and activator of transcription 5 (STAT5) binding, which is significantly increased by 18% CS. In addition, NAMPT promoter activity was increased by pharmacologic promoter demethylation and inhibited by STAT5 silencing. ARDS-associated NAMPT promoter SNPs rs59744560 (-948G/T) and rs7789066 (-2,422A/G) each significantly elevated NAMPT promoter activity in response to 18% CS in a STAT5-dependent manner. Our results show that NAMPT is a key novel ARDS therapeutic target and candidate gene with genetic/epigenetic transcriptional regulation in response to excessive mechanical stress.
Collapse
|
168
|
Brocato J, Hernandez M, Laulicht F, Sun H, Shamy M, Alghamdi MA, Khoder MI, Kluz T, Chen LC, Costa M. In Vivo Exposures to Particulate Matter Collected from Saudi Arabia or Nickel Chloride Display Similar Dysregulation of Metabolic Syndrome Genes. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2015; 78:1421-36. [PMID: 26692068 PMCID: PMC4709028 DOI: 10.1080/15287394.2015.1095689] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Particulate matter (PM) exposures have been linked to mortality, low birth weights, hospital admissions, and diseases associated with metabolic syndrome, including diabetes mellitus, cardiovascular disease, and obesity. In a previous in vitro and in vivo study, data demonstrated that PM(10μm) collected from Jeddah, Saudi Arabia (PMSA), altered expression of genes involved in lipid and cholesterol metabolism, as well as many other genes associated with metabolic disorders. PMSA contains a relatively high concentration of nickel (Ni), known to be linked to several metabolic disorders. In order to evaluate whether Ni and PM exposures induce similar gene expression profiles, mice were exposed to 100 μg/50 μl PM(SA) (PM-100), 50 μg/50 μl nickel chloride (Ni-50), or 100 μg/50 μl nickel chloride (Ni-100) twice per week for 4 wk and hepatic gene expression changes were determined. Ultimately, 55 of the same genes were altered in all 3 exposures. However, where the two Ni groups differed markedly was in the regulation (up or down) of these genes. Ni-100 and PM-100 groups displayed similar regulations, whereby 104 of the 107 genes were similarly modulated. Many of the 107 genes are involved in metabolic syndrome and include ALDH4A1, BCO2, CYP1A, CYP2U, TOP2A. In addition, the top affected pathways, such as fatty acid α-oxidation, and lipid and carbohydrate metabolism, are involved in metabolic diseases. Most notably, the top diseased outcome affected by these changes in gene expression was cardiovascular disease. Given these data, it appears that Ni and PM(SA) exposures display similar gene expression profiles, modulating the expression of genes involved in metabolic disorders.
Collapse
Affiliation(s)
- Jason Brocato
- Department of Environmental Medicine, NYU School of Medicine, NY, NY, 10016 USA
| | - Michelle Hernandez
- Department of Environmental Medicine, NYU School of Medicine, NY, NY, 10016 USA
| | - Freda Laulicht
- Department of Environmental Medicine, NYU School of Medicine, NY, NY, 10016 USA
| | - Hong Sun
- Department of Environmental Medicine, NYU School of Medicine, NY, NY, 10016 USA
| | - Magdy Shamy
- Department of Environmental Sciences, Faculty of Meteorology, Environmental and Arid Land Agriculture, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mansour A. Alghamdi
- Department of Environmental Sciences, Faculty of Meteorology, Environmental and Arid Land Agriculture, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mamdouh I. Khoder
- Department of Environmental Sciences, Faculty of Meteorology, Environmental and Arid Land Agriculture, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Excellence in Environmental Studies, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Thomas Kluz
- Department of Environmental Medicine, NYU School of Medicine, NY, NY, 10016 USA
| | - Lung-Chi Chen
- Department of Environmental Medicine, NYU School of Medicine, NY, NY, 10016 USA
| | - Max Costa
- Department of Environmental Medicine, NYU School of Medicine, NY, NY, 10016 USA
| |
Collapse
|
169
|
Nivarthi H, Prchal-Murphy M, Swoboda A, Hager M, Schlederer M, Kenner L, Tuckermann J, Sexl V, Moriggl R, Ermakova O. Stat5 gene dosage in T cells modulates CD8+ T-cell homeostasis and attenuates contact hypersensitivity response in mice. Allergy 2015; 70:67-79. [PMID: 25333229 DOI: 10.1111/all.12535] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2014] [Indexed: 12/26/2022]
Abstract
BACKGROUND Contact hypersensitivity assay (CHS) faithfully models human allergies. The Stat5 transcription factors are essential for both lymphocyte development and acute immune responses. Although consequences of Stat5 ablation and transgenic overexpression for the lymphocyte development and functions have been extensively studied, the role of Stat5 gene dosage in contact allergies has not been addressed. OBJECTIVE We investigated the effect of Stat5 gene dosage modulation in contact allergies using CHS in mice. METHODS Transgenic animals heterozygous for the germline Stat5 null allele were subjected to CHS. To dissect cell type sensitive to Stat5 gene dosage, animals with Stat5 haplo-insufficiency in T cells, where one Stat5 allele was removed by Lck-Cre-mediated deletion (Stat5(ΔT/+)), were tested by CHS. Frequency of T cells, B cells, and monocytes were analyzed in Stat5(ΔT/+) and wild-type animals by flow cytometry. Proliferation of Stat5(ΔT/+) CD8(+) T cells was studied in vitro by stimulation with IL-4 and IL-2 cytokines, and changes in the expression of Stat5 target genes were assayed by quantitative real-time PCR assay. RESULT Haplo-insufficiency of Stat5 in T cells leads to the reduction in CD8(+) T cells in all lymphoid organs and attenuates CHS response. Stat5(ΔT/+) CD8(+) T cells failed to fully activate Stat5-dependent expression of cell cycle/survival target genes, such as Bcl2 and Pim1, and to proliferate efficiently in response to IL-2 and IL-4 cytokine. CONCLUSION Our data identify Stat5 as a dose-dependent regulator of CD8(+) T-cell functions in contact allergies and suggest that modulation of Stat5 dosage could be used to target contact allergies in humans.
Collapse
Affiliation(s)
- H. Nivarthi
- Ludwig Boltzmann Institute for Cancer Research; Vienna Austria
| | - M. Prchal-Murphy
- Institute of Pharmacology and Toxicology; University of Veterinary Medicine Vienna; Vienna Austria
| | - A. Swoboda
- Ludwig Boltzmann Institute for Cancer Research; Vienna Austria
| | - M. Hager
- Ludwig Boltzmann Institute for Cancer Research; Vienna Austria
| | - M. Schlederer
- Ludwig Boltzmann Institute for Cancer Research; Vienna Austria
| | - L. Kenner
- Ludwig Boltzmann Institute for Cancer Research; Vienna Austria
| | - J. Tuckermann
- Institute of General Zoology and Endocrinology; University of Ulm; Ulm Germany
| | - V. Sexl
- Institute of Pharmacology and Toxicology; University of Veterinary Medicine Vienna; Vienna Austria
| | - R. Moriggl
- Ludwig Boltzmann Institute for Cancer Research; Vienna Austria
| | - O. Ermakova
- Mouse Biology Unit; European Molecular Biology Laboratory; Monterotondo Italy
| |
Collapse
|
170
|
Abstract
Natural killer (NK) cells are innate lymphocytes that survey the environment and protect the host from infected and cancerous cells. As their name implies, NK cells represent an early line of defense during pathogen invasion by directly killing infected cells and secreting inflammatory cytokines. Although the function of NK cells was first described more than four decades ago, the development of this cytotoxic lineage is not well understood. In recent years, we have begun to identify specific transcription factors that control each stage of development and maturation, from ontogeny of the NK cell progenitor to the effector functions of activated NK cells in peripheral organs. This chapter highlights the transcription factors that are unique to NK cells, or shared between NK cells and other hematopoietic cell lineages, but govern the biology of this cytolytic lymphocyte.
Collapse
Affiliation(s)
- Joseph C Sun
- Memorial Sloan Kettering Cancer Center, Department of Immunology and Microbial Pathogenesis, Weill Cornell Medical College, 408 East 69th Street, ZRC-1402, New York, NY, 10065, USA.
| |
Collapse
|
171
|
Mishra A, Sullivan L, Caligiuri MA. Molecular pathways: interleukin-15 signaling in health and in cancer. Clin Cancer Res 2014; 20:2044-50. [PMID: 24737791 DOI: 10.1158/1078-0432.ccr-12-3603] [Citation(s) in RCA: 171] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Interleukin-15 (IL-15) is a proinflammatory cytokine involved in the development, survival, proliferation, and activation of multiple lymphocyte lineages utilizing a variety of signaling pathways. IL-15 utilizes three distinct receptor chains in at least two different combinations to signal and exert its effects on the immune system. The binding of IL-15 to its receptor complex activates an "immune-enhancing" signaling cascade in natural killer cells and subsets of T cells, as well as the induction of a number of proto-oncogenes. Additional studies have explored the role of IL-15 in the development and progression of cancer, notably leukemia of large granular lymphocytes, cutaneous T-cell lymphoma, and multiple myeloma. This review provides an overview of the molecular events in the IL-15 signaling pathway and the aberrancies in its regulation that are associated with chronic inflammation and cancer. We briefly explore the potential therapeutic opportunities that have arisen as a result of these studies to further the treatment of cancer. These involve both targeting the disruption of IL-15 signaling as well as IL-15-mediated enhancement of innate and antigen-specific immunity.
Collapse
Affiliation(s)
- Anjali Mishra
- Authors' Affiliation: The Divisions of Dermatology and Hematology, Department of Internal Medicine, The Comprehensive Cancer Center, The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, Ohio
| | | | | |
Collapse
|
172
|
Mejia R, Waite C, Ascoli M. Activation of Gq/11 in the mouse corpus luteum is required for parturition. Mol Endocrinol 2014; 29:238-46. [PMID: 25495873 DOI: 10.1210/me.2014-1324] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Mice with a deletion of Gα(q/11) in granulosa cells were previously shown to be subfertile. They also have a reduced ovulatory response due to a deficiency in the ability of the activated LH receptor to fully induce the granulosa cell progesterone receptor. Because this conditional deletion of Gα(q/11) will interfere with the actions of any G protein-coupled receptor that activates G(q/11) in granulosa or luteal cells, we sought to determine whether the actions of other hormones that contribute to fertility were also impaired. We focused our attention on prostaglandin F2 (PGF2)α, because this hormone is known to activate phospholipase C (a prominent Gα(q/11) effector) in luteal cells and because the action of PGF2α on luteal cells is the first step in the murine parturition pathway. Our data show that the conditional deletion of Gα(q/11) from granulosa cells prevents the ability of PGF2α to induce Akr1c18 in luteal cells. Akr1c18 codes for 20α-hydroxysteroid dehydrogenase, an enzyme that inactivates progesterone. The PGF2α-mediated induction of this enzyme towards the end of pregnancy increases the inactivation of progesterone and precipitates parturition in mice. Thus, the conditional deletion of Gαq/11 from granulosa/luteal cells prevents the progesterone withdrawal that occurs at the end of pregnancy and impairs parturition. This novel molecular defect contributes to the subfertile phenotype of the mice with a deletion of Gα(q/11) from granulosa cells.
Collapse
Affiliation(s)
- Rachel Mejia
- Department of Obstetrics and Gynecology (R.M., M.A.), Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242; and Department of Pharmacology (C.W., M.A.), Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242
| | | | | |
Collapse
|
173
|
Schnöder TM, Arreba-Tutusaus P, Griehl I, Bullinger L, Buschbeck M, Lane SW, Döhner K, Plass C, Lipka DB, Heidel FH, Fischer T. Epo-induced erythroid maturation is dependent on Plcγ1 signaling. Cell Death Differ 2014; 22:974-85. [PMID: 25394487 DOI: 10.1038/cdd.2014.186] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 09/18/2014] [Accepted: 10/06/2014] [Indexed: 12/22/2022] Open
Abstract
Erythropoiesis is a tightly regulated process. Development of red blood cells occurs through differentiation of hematopoietic stem cells (HSCs) into more committed progenitors and finally into erythrocytes. Binding of erythropoietin (Epo) to its receptor (EpoR) is required for erythropoiesis as it promotes survival and late maturation of erythroid progenitors. In vivo and in vitro studies have highlighted the requirement of EpoR signaling through Janus kinase 2 (Jak2) tyrosine kinase and Stat5a/b as a central pathway. Here, we demonstrate that phospholipase C gamma 1 (Plcγ1) is activated downstream of EpoR-Jak2 independently of Stat5. Plcγ1-deficient pro-erythroblasts and erythroid progenitors exhibited strong impairment in differentiation and colony-forming potential. In vivo, suppression of Plcγ1 in immunophenotypically defined HSCs (Lin(-)Sca1(+)KIT(+)CD48(-)CD150(+)) severely reduced erythroid development. To identify Plcγ1 effector molecules involved in regulation of erythroid differentiation, we assessed changes occurring at the global transcriptional and DNA methylation level after inactivation of Plcγ1. The top common downstream effector was H2afy2, which encodes for the histone variant macroH2A2 (mH2A2). Inactivation of mH2A2 expression recapitulated the effects of Plcγ1 depletion on erythroid maturation. Taken together, our findings identify Plcγ1 and its downstream target mH2A2, as a 'non-canonical' Epo signaling pathway essential for erythroid differentiation.
Collapse
Affiliation(s)
- T M Schnöder
- Department of Hematology and Oncology, Center for Internal Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| | - P Arreba-Tutusaus
- Department of Hematology and Oncology, Center for Internal Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| | - I Griehl
- Department of Hematology and Oncology, Center for Internal Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| | - L Bullinger
- Internal Medicine III, Department of Hematology/Oncology, University Hospital Ulm, Ulm, Germany
| | - M Buschbeck
- Institute of Predictive and Personalized Medicine of Cancer (IMPPC), Campus Can Ruti, Badalona, Spain
| | - S W Lane
- Division of Immunology, QIMR Berghofer Medical Research Institute, Herston, Brisbane, Australia
| | - K Döhner
- Internal Medicine III, Department of Hematology/Oncology, University Hospital Ulm, Ulm, Germany
| | - C Plass
- Division of Epigenomics and Cancer Risk Factors (C010), German Cancer Research Center, Heidelberg, Germany
| | - D B Lipka
- 1] Department of Hematology and Oncology, Center for Internal Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany [2] Division of Epigenomics and Cancer Risk Factors (C010), German Cancer Research Center, Heidelberg, Germany
| | - F H Heidel
- Department of Hematology and Oncology, Center for Internal Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| | - T Fischer
- Department of Hematology and Oncology, Center for Internal Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| |
Collapse
|
174
|
Boehm ME, Adlung L, Schilling M, Roth S, Klingmüller U, Lehmann WD. Identification of Isoform-Specific Dynamics in Phosphorylation-Dependent STAT5 Dimerization by Quantitative Mass Spectrometry and Mathematical Modeling. J Proteome Res 2014; 13:5685-94. [DOI: 10.1021/pr5006923] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
| | | | | | - Susanne Roth
- Systems
Bioinformatics, Netherlands Institute for Systems Biology, VU University, De Boelelaan 1085, 1081 HV Amsterdam, Netherlands
| | | | | |
Collapse
|
175
|
Signal transducer and activator of transcription 5a inhibited by pimozide may regulate survival of goat mammary gland epithelial cells by regulating parathyroid hormone-related protein. Gene 2014; 551:279-89. [DOI: 10.1016/j.gene.2014.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 08/17/2014] [Accepted: 09/02/2014] [Indexed: 12/22/2022]
|
176
|
Varco-Merth B, Rotwein P. Differential effects of STAT proteins on growth hormone-mediated IGF-I gene expression. Am J Physiol Endocrinol Metab 2014; 307:E847-55. [PMID: 25205818 PMCID: PMC4216947 DOI: 10.1152/ajpendo.00324.2014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Growth hormone (GH) plays a key role regulating somatic growth and in controlling metabolism and other physiological processes in humans and other animal species. GH acts by binding to the extracellular part of its transmembrane receptor, leading to induction of multiple intracellular signal transduction pathways that culminate in changes in gene and protein expression. A key agent in GH-stimulated growth is the latent transcription factor signal transducer and activator of transcription (STAT) 5B, one of four STAT proteins induced by the GH receptor in cultured cells and in vivo. As shown by genetic and biochemical studies, GH-activated STAT5B promotes transcription of the gene encoding the critical growth peptide, insulin-like growth factor-I (IGF-I), and natural null mutations of STAT5B in humans lead to growth failure accompanied by diminished IGF-I expression. Here we have examined the possibility that other GH-activated STATs can enhance IGF-I gene transcription, and thus potentially contribute to GH-regulated somatic growth. We find that human STAT5A is nearly identical to STAT5B in its biochemical and functional responses to GH but that STAT1 and STAT3 show a weaker profile of in vitro binding to STAT DNA elements from the IGF-I gene than STAT5B, and are less potent inducers of gene transcription through these elements. Taken together, our results offer a molecular explanation for why STAT5B is a key in vivo mediator of GH-activated IGF-I gene transcription and thus of GH-regulated somatic growth.
Collapse
Affiliation(s)
- Ben Varco-Merth
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon
| | - Peter Rotwein
- Department of Biochemistry and Molecular Biology, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
177
|
Gressot LV, Doucette TA, Yang Y, Fuller GN, Heimberger AB, Bögler O, Rao A, Latha K, Rao G. Signal transducer and activator of transcription 5b drives malignant progression in a PDGFB-dependent proneural glioma model by suppressing apoptosis. Int J Cancer 2014; 136:2047-54. [PMID: 25302990 DOI: 10.1002/ijc.29264] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 09/08/2014] [Accepted: 09/15/2014] [Indexed: 12/16/2022]
Abstract
Signal transducer and activator of transcription 5b (STAT5b) is likely the relevant STAT5 isoform with respect to the process of malignant progression in gliomas. STAT5b is a latent cytoplasmic protein involved in cell signaling through the modulation of growth factors, apoptosis, and angiogenesis. Previous in vitro studies have shown increased STAT5b expression in glioblastomas relative to low-grade tumors and normal brain. We recently demonstrated that phosphorylated STAT5b associates with delta epidermal growth factor receptor in the nucleus and subsequently binds the promoters of downstream effector molecules, including aurora kinase A. Analysis of TCGA dataset reveals that STAT5b is predominantly expressed in proneural (PN) gliomas relative to mesenchymal and neural gliomas. Here, we modeled ectopic expression of STAT5b in vivo using a platelet-derived growth factor subunit B (PDGFB)-dependent mouse model of PN glioma to determine its effect on tumor formation and progression. We showed that coexpression of STAT5b and PDGFB in mice yielded a significantly higher rate of high-grade gliomas than PDGFB expression alone. We also observed shorter survival in the combined expression set. High-grade tumors from the STAT5b + PDGFB expression set were found to have a lower rate of apoptosis than those from PDGFB alone. Furthermore, we showed that increased expression of STAT5b + PDGFB led to increased expression of downstream STAT5b targets, including Bcl-xL, cyclin D1 and aurora kinase A in high-grade tumors when compared to tumors derived from PDGFB alone. Our findings show that STAT5b promotes the malignant transformation of gliomas, particularly the PN subtype, and is a potential therapeutic target.
Collapse
Affiliation(s)
- Loyola V Gressot
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | | | | | | | | | | |
Collapse
|
178
|
Uejyo T, Kuki C, Oyama S, Kumura H, Kobayashi K. Early down-regulation of milk production after weaning by pup removal and prior to involution in mouse mammary glands. Cell Tissue Res 2014; 359:643-653. [DOI: 10.1007/s00441-014-2013-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 09/18/2014] [Indexed: 01/20/2023]
|
179
|
Dobie R, MacRae VE, Huesa C, van't Hof R, Ahmed SF, Farquharson C. Direct stimulation of bone mass by increased GH signalling in the osteoblasts of Socs2-/- mice. J Endocrinol 2014; 223:93-106. [PMID: 25074853 PMCID: PMC4166176 DOI: 10.1530/joe-14-0292] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The suppressor of cytokine signalling (Socs2(-/-))-knockout mouse is characterised by an overgrowth phenotype due to enhanced GH signalling. The objective of this study was to define the Socs2(-/-) bone phenotype and determine whether GH promotes bone mass via IGF1-dependent mechanisms. Despite no elevation in systemic IGF1 levels, increased body weight in 4-week-old Socs2(-/-) mice following GH treatment was associated with increased cortical bone area (Ct.Ar) (P<0.01). Furthermore, detailed bone analysis of male and female juvenile and adult Socs2(-/-) mice revealed an altered cortical and trabecular phenotype consistent with the known anabolic effects of GH. Indeed, male Socs2(-/-) mice had increased Ct.Ar (P<0.05) and thickness associated with increased strength. Despite this, there was no elevation in hepatic Igf1 expression, suggesting that the anabolic bone phenotype was the result of increased local GH action. Mechanistic studies showed that in osteoblasts and bone of Socs2(-/-) mice, STAT5 phosphorylation was significantly increased in response to GH. Conversely, overexpression of SOCS2 decreased GH-induced STAT5 signalling. Although an increase in Igf1 expression was observed in Socs2(-/-) osteoblasts following GH, it was not evident in vivo. Igf1 expression levels were not elevated in response to GH in 4-week-old mice and no alterations in expression was observed in bone samples of 6-week-old Socs2(-/-) mice. These studies emphasise the critical role of SOCS2 in controlling the local GH anabolic bone effects. We provide compelling evidence implicating SOCS2 in the regulation of GH osteoblast signalling and ultimately bone accrual, which maybe via mechanisms that are independent of IGF1 production in vivo.
Collapse
Affiliation(s)
- R Dobie
- Division of Developmental BiologyThe Roslin Institute and R(D)SVS, The University of Edinburgh, Easter Bush, Midlothian, Edinburgh EH25 9RG, Scotland, UKInstitute of Ageing and Chronic DiseaseUniversity of Liverpool, Daulby Street, Liverpool L69 3GA, UKDevelopmental Endocrinology Research GroupSchool of Medicine, University of Glasgow, Yorkhill, Glasgow G3 8SJ, Scotland, UK
| | - V E MacRae
- Division of Developmental BiologyThe Roslin Institute and R(D)SVS, The University of Edinburgh, Easter Bush, Midlothian, Edinburgh EH25 9RG, Scotland, UKInstitute of Ageing and Chronic DiseaseUniversity of Liverpool, Daulby Street, Liverpool L69 3GA, UKDevelopmental Endocrinology Research GroupSchool of Medicine, University of Glasgow, Yorkhill, Glasgow G3 8SJ, Scotland, UK
| | - C Huesa
- Division of Developmental BiologyThe Roslin Institute and R(D)SVS, The University of Edinburgh, Easter Bush, Midlothian, Edinburgh EH25 9RG, Scotland, UKInstitute of Ageing and Chronic DiseaseUniversity of Liverpool, Daulby Street, Liverpool L69 3GA, UKDevelopmental Endocrinology Research GroupSchool of Medicine, University of Glasgow, Yorkhill, Glasgow G3 8SJ, Scotland, UK
| | - R van't Hof
- Division of Developmental BiologyThe Roslin Institute and R(D)SVS, The University of Edinburgh, Easter Bush, Midlothian, Edinburgh EH25 9RG, Scotland, UKInstitute of Ageing and Chronic DiseaseUniversity of Liverpool, Daulby Street, Liverpool L69 3GA, UKDevelopmental Endocrinology Research GroupSchool of Medicine, University of Glasgow, Yorkhill, Glasgow G3 8SJ, Scotland, UK
| | - S F Ahmed
- Division of Developmental BiologyThe Roslin Institute and R(D)SVS, The University of Edinburgh, Easter Bush, Midlothian, Edinburgh EH25 9RG, Scotland, UKInstitute of Ageing and Chronic DiseaseUniversity of Liverpool, Daulby Street, Liverpool L69 3GA, UKDevelopmental Endocrinology Research GroupSchool of Medicine, University of Glasgow, Yorkhill, Glasgow G3 8SJ, Scotland, UK
| | - C Farquharson
- Division of Developmental BiologyThe Roslin Institute and R(D)SVS, The University of Edinburgh, Easter Bush, Midlothian, Edinburgh EH25 9RG, Scotland, UKInstitute of Ageing and Chronic DiseaseUniversity of Liverpool, Daulby Street, Liverpool L69 3GA, UKDevelopmental Endocrinology Research GroupSchool of Medicine, University of Glasgow, Yorkhill, Glasgow G3 8SJ, Scotland, UK
| |
Collapse
|
180
|
Beclin-1 deficiency in the murine ovary results in the reduction of progesterone production to promote preterm labor. Proc Natl Acad Sci U S A 2014; 111:E4194-203. [PMID: 25246579 DOI: 10.1073/pnas.1409323111] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Autophagy is an important cellular process that serves as a companion pathway to the ubiquitin-proteasome system to degrade long-lived proteins and organelles to maintain cell homeostasis. Although initially characterized in yeast, autophagy is being realized as an important regulator of development and disease in mammals. Beclin1 (Becn1) is a putative tumor suppressor gene that has been shown to undergo a loss of heterozygosity in 40-75% of human breast, ovarian, and prostate cancers. Because Becn1 is a key regulator of autophagy, we sought to investigate its role in female reproduction by using a conditional knockout approach in mice. We find that pregnant females lacking Becn1 in the ovarian granulosa cell population have a defect in progesterone production and a subsequent preterm labor phenotype. Luteal cells in this model exhibit defective autophagy and a failure to accumulate lipid droplets needed for steroidogenesis. Collectively, we show that Becn1 provides essential functions in the ovary that are essential for mammalian reproduction.
Collapse
|
181
|
Shih HY, Sciumè G, Poholek AC, Vahedi G, Hirahara K, Villarino AV, Bonelli M, Bosselut R, Kanno Y, Muljo SA, O'Shea JJ. Transcriptional and epigenetic networks of helper T and innate lymphoid cells. Immunol Rev 2014; 261:23-49. [PMID: 25123275 PMCID: PMC4321863 DOI: 10.1111/imr.12208] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The discovery of the specification of CD4(+) helper T cells to discrete effector 'lineages' represented a watershed event in conceptualizing mechanisms of host defense and immunoregulation. However, our appreciation for the actual complexity of helper T-cell subsets continues unabated. Just as the Sami language of Scandinavia has 1000 different words for reindeer, immunologists recognize the range of fates available for a CD4(+) T cell is numerous and may be underestimated. Added to the crowded scene for helper T-cell subsets is the continuously growing family of innate lymphoid cells (ILCs), endowed with common effector responses and the previously defined 'master regulators' for CD4(+) helper T-cell subsets are also shared by ILC subsets. Within the context of this extraordinary complexity are concomitant advances in the understanding of transcriptomes and epigenomes. So what do terms like 'lineage commitment' and helper T-cell 'specification' mean in the early 21st century? How do we put all of this together in a coherent conceptual framework? It would be arrogant to assume that we have a sophisticated enough understanding to seriously answer these questions. Instead, we review the current status of the flexibility of helper T-cell responses in relation to their genetic regulatory networks and epigenetic landscapes. Recent data have provided major surprises as to what master regulators can or cannot do, how they interact with other transcription factors and impact global genome-wide changes, and how all these factors come together to influence helper cell function.
Collapse
Affiliation(s)
- Han-Yu Shih
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis, and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
182
|
Kermath BA, Riha PD, Woller MJ, Wolfe A, Gore AC. Hypothalamic molecular changes underlying natural reproductive senescence in the female rat. Endocrinology 2014; 155:3597-609. [PMID: 24914937 PMCID: PMC4138577 DOI: 10.1210/en.2014-1017] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The role of the hypothalamus in female reproductive senescence is unclear. Here we identified novel molecular neuroendocrine changes during the natural progression from regular reproductive cycles to acyclicity in middle-aged female rats, comparable with the perimenopausal progression in women. Expression of 48 neuroendocrine genes was quantified within three hypothalamic regions: the anteroventral periventricular nucleus, the site of steroid positive feedback onto GnRH neurons; the arcuate nucleus (ARC), the site of negative feedback and pulsatile GnRH release; and the median eminence (ME), the site of GnRH secretion. Surprisingly, the majority of changes occurred in the ARC and ME, with few effects in anteroventral periventricular nucleus. The overall pattern was increased mRNA levels with chronological age and decreases with reproductive cycle status in middle-aged rats. Affected genes included transcription factors (Stat5b, Arnt, Ahr), sex steroid hormone receptors (Esr1, Esr2, Pgr, Ar), steroidogenic enzymes (Sts, Hsd17b8), growth factors (Igf1, Tgfa), and neuropeptides (Kiss1, Tac2, Gnrh1). Bionetwork analysis revealed region-specific correlations between genes and hormones. Immunohistochemical analyses of kisspeptin and estrogen receptor-α in the ARC demonstrated age-related decreases in kisspeptin cell numbers as well as kisspeptin-estrogen receptor-α dual-labeled cells. Taken together, these results identify unexpectedly strong roles for the ME and ARC during reproductive decline and highlight fundamental differences between middle-aged rats with regular cycles and all other groups. Our data provide evidence of decreased excitatory stimulation and altered hormone feedback with aging and suggest novel neuroendocrine pathways that warrant future study. Furthermore, these changes may impact other neuroendocrine systems that undergo functional declines with age.
Collapse
Affiliation(s)
- Bailey A Kermath
- Institute for Neuroscience (B.A.K., A.C.G.), Division of Pharmacology and Toxicology (P.D.R., A.C.G.), and Institute for Cell and Molecular Biology (A.C.G.), The University of Texas at Austin, Austin, Texas 78712; Department of Biology (M.J.W.), University of Wisconsin-Whitewater, Whitewater, Wisconsin 53190; and Department of Pediatrics (A.W.), Johns Hopkins University School of Medicine, Baltimore Maryland 21287
| | | | | | | | | |
Collapse
|
183
|
Association between STAT5 polymorphisms and glioblastoma risk in Han Chinese population. Pathol Res Pract 2014; 210:582-5. [DOI: 10.1016/j.prp.2014.04.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 04/14/2014] [Accepted: 04/30/2014] [Indexed: 11/23/2022]
|
184
|
Lin G, LaPensee CR, Qin ZS, Schwartz J. Reciprocal occupancy of BCL6 and STAT5 on Growth Hormone target genes: contrasting transcriptional outcomes and promoter-specific roles of p300 and HDAC3. Mol Cell Endocrinol 2014; 395:19-31. [PMID: 25088465 PMCID: PMC4176921 DOI: 10.1016/j.mce.2014.07.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 06/30/2014] [Accepted: 07/28/2014] [Indexed: 12/30/2022]
Abstract
Expression of the Growth Hormone (GH)-stimulated gene Socs2 (Suppressor of Cytokine Signaling 2) is mediated by the transcription activator STAT5 (Signal Transducer and Activator of Transcription 5) and the transcription repressor BCL6 (B-Cell Lymphoma 6). ChIP-Sequencing identified Cish (Cytokine-Inducible SH2-containing protein) and Bcl6 as having similar patterns of reciprocal occupancy by BCL6 and STAT5 in response to GH, though GH stimulates Cish and inhibits Bcl6 expression. The co-activator p300 occupied Socs2, Cish and Bcl6 promoters, and enhanced STAT5-mediated activation of Socs2 and Cish. In contrast, on Bcl6, p300 functioned as a repressor and inhibited in conjunction with STAT5 or BCL6. The co-repressor HDAC3 (Histone deacetylase 3) inhibited the Socs2, Cish and Bcl6 promoters in the presence of STAT5. Thus transcriptional outcomes on GH-regulated genes occupied by BCL6 and STAT5 are determined in a promoter-specific fashion by co-regulatory proteins which mediate the distinction between activating and repressive transcription factors.
Collapse
Affiliation(s)
- Grace Lin
- Cellular & Molecular Biology Program, University of Michigan, Ann Arbor, MI, USA; Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Christopher R LaPensee
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Zhaohui S Qin
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, USA
| | - Jessica Schwartz
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
185
|
Hatakeyama S. Ubiquitin-mediated regulation of JAK-STAT signaling in embryonic stem cells. JAKSTAT 2014; 1:168-75. [PMID: 24058766 PMCID: PMC3670240 DOI: 10.4161/jkst.21560] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 07/20/2012] [Accepted: 07/20/2012] [Indexed: 12/12/2022] Open
Abstract
LIF activates several intracellular signaling pathways including JAK-STAT, PI3K/AKT and MAPK pathways. LIF is an important cytokine for maintenance of pluripotency and self-renewal of mouse ES cells. The JAK-STAT signal plays a key role in maintenance of the pluripotency of ESCs. Recent evidence shows that several post-translational modifications regulate activation or inhibition of intracellular signal transductions. The JAK-STAT signal is also modulated by several modifications including phosphorylation, acetylation and ubiquitination. In this review, we discuss regulation of the LIF-mediated-JAK-STAT signaling pathway that contributes to self-renewal of pluripotent ESCs.
Collapse
Affiliation(s)
- Shigetsugu Hatakeyama
- Department of Biochemistry; Hokkaido University Graduate School of Medicine; Sapporo, Hokkaido Japan
| |
Collapse
|
186
|
Huang H, Li Y, Qi X. Cytokine signaling in the differentiation of innate effector cells. JAKSTAT 2014; 2:e23531. [PMID: 24058796 PMCID: PMC3670272 DOI: 10.4161/jkst.23531] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 01/04/2013] [Accepted: 01/07/2013] [Indexed: 12/24/2022] Open
Abstract
Innate effector cells, including innate effector cells of myeloid and lymphoid lineages, are crucial components of various types of immune responses. Bone marrow progenitors differentiate into many subsets of innate effector cells after receiving instructional signals often provided by cytokines. Signal transducer and activator of transcription (STATs) have been shown to be essential in the differentiation of various types of innate effector cells. In this review, we focus specifically on the differentiation of innate effector cells, particularly the role of cytokine signaling in the differentiation of innate effector cells.
Collapse
Affiliation(s)
- Hua Huang
- Division of Allergy and Immunology; Department of Medicine; National Jewish Health; Denver, CO USA ; Integrated Department of Immunology; University of Colorado School of Medicine; Denver, CO USA
| | | | | |
Collapse
|
187
|
Zhao P, Stephens JM. Identification of STAT target genes in adipocytes. JAKSTAT 2014; 2:e23092. [PMID: 24058802 PMCID: PMC3710315 DOI: 10.4161/jkst.23092] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 12/02/2012] [Accepted: 12/03/2012] [Indexed: 02/08/2023] Open
Abstract
Adipocytes play important roles in lipid storage, energy homeostasis and whole body insulin sensitivity. Studies in the last two decades have identified the hormones and cytokines that activate specific STATs in adipocytes in vitro and in vivo. Five of the seven STAT family members are expressed in adipocyte (STATs 1, 3, 5A, 5B and 6). Many transcription factors, including STATs, have been shown to play an important role in adipose tissue development and function. This review will summarize the importance of adipocytes, indicate the cytokines and hormones that utilize the JAK-STAT signaling pathway in fat cells and focus on the identification of STAT target genes in mature adipocytes. To date, specific target genes have been identified for STATs, 1, 5A and 5B, but not for STATs 3 and 6.
Collapse
Affiliation(s)
- Peng Zhao
- Department of Biological Sciences; Louisiana State University; Baton Rouge, LA USA ; Adipocyte Biology Lab; Pennington Biomedical Research Center; Baton Rouge, LA USA
| | | |
Collapse
|
188
|
Hughes K, Watson CJ. The spectrum of STAT functions in mammary gland development. JAKSTAT 2014; 1:151-8. [PMID: 24058764 PMCID: PMC3670238 DOI: 10.4161/jkst.19691] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 02/08/2012] [Accepted: 02/13/2012] [Indexed: 12/21/2022] Open
Abstract
The signal transducer and activator of transcription (STAT) family of transcription factors have a spectrum of functions in mammary gland development. In some cases these roles parallel those of STATs in other organ systems, while in other instances the function of individual STATs in the mammary gland is specific to this tissue. In the immune system, STAT6 is associated with differentiation of T helper cells, while in the mammary gland, it has a fundamental role in the commitment of luminal epithelial cells to the alveolar lineage. STAT5A is required for the production of luminal progenitor cells from mammary stem cells and is essential for the differentiation of milk producing alveolar cells during pregnancy. By contrast, the initiation of regression following weaning heralds a dramatic and specific activation of STAT3, reflecting its pivotal role in the regulation of cell death and tissue remodeling during mammary involution. Although it has been demonstrated that STAT1 is regulated during a mammary developmental cycle, it is not yet determined whether it has a specific, non-redundant function. Thus, the mammary gland constitutes an unusual example of an adult organ in which different STATs are sequentially activated to orchestrate the processes of functional differentiation, cell death and tissue remodeling.
Collapse
|
189
|
Aupperlee MD, Zhao Y, Tan YS, Leipprandt JR, Bennett J, Haslam SZ, Schwartz RC. Epidermal growth factor receptor (EGFR) signaling is a key mediator of hormone-induced leukocyte infiltration in the pubertal female mammary gland. Endocrinology 2014; 155:2301-13. [PMID: 24693965 PMCID: PMC4020926 DOI: 10.1210/en.2013-1933] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
It is well documented that macrophages and eosinophils play important roles in normal murine pubertal mammary gland development. Although it is accepted that estrogen (E) and progesterone (P) are key players in mammary gland development, the roles these hormones might play in regulating the actions of leukocytes in that process is an understudied area. We show here that P and E, respectively, induce unique, but overlapping, sets of proinflammatory and angiogenic cytokines and chemokines, in the pubertal female BALB/c mammary gland, as well as induce infiltration of macrophages and eosinophils to the mammary periepithelium. This extends earlier studies showing P induction of proinflammatory products in pubertal and adult mammary epithelial organoids and P-induced in vivo infiltration of leukocytes to the adult mammary periepithelium. Importantly, epidermal growth factor receptor-signaling, which is likely mediated by amphiregulin (Areg), a downstream mediator of E and P, is both necessary and sufficient for both E- and P-induced recruitment of macrophages and eosinophils to the pubertal mammary periepithelium. We further show that receptor activator of nuclear factor κB ligand (RANKL), although not sufficient of itself to cause macrophage and eosinophil recruitment, contributes to an optimal response to P. The potency of Areg is highlighted by the fact that it is sufficient to induce macrophage and eosinophil recruitment at levels equivalent to that induced by either E or P. Our finding of a dominant role for Areg in hormonally induced leukocyte recruitment to the pubertal mammary gland parallels its dominance in regulating ductal outgrowth and its role in P-induced proliferation in the pubertal gland.
Collapse
Affiliation(s)
- Mark D Aupperlee
- Breast Cancer and the Environment Research Program, Departments of Physiology (M.D.A., Y.Z., Y.S.T., J.R.L., J.B., S.Z.H.) and Microbiology and Molecular Genetics (R.C.S.), Michigan State University, East Lansing, Michigan 48824
| | | | | | | | | | | | | |
Collapse
|
190
|
Oberley CC, Bilger A, Drinkwater NR. Genetic background determines if Stat5b suppresses or enhances murine hepatocarcinogenesis. Mol Carcinog 2014; 54:959-70. [PMID: 24838184 DOI: 10.1002/mc.22165] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 03/26/2014] [Accepted: 03/31/2014] [Indexed: 12/23/2022]
Abstract
Murine hepatocarcinogenesis requires growth hormone (GH). To determine if the GH-responsive transcription factor STAT5b (signal transducer and activator of transcription 5b) is also required, we compared the hepatic gene expression profiles of global Stat5b null mice to cancer-resistant mice mutant in the GH pathway-GH-deficient little and androgen receptor-null Tfm males. We found a high degree of overlap among Tfm, little, and Stat5b null males. The liver cancer susceptibility of global Stat5b null mice was assessed on three distinct genetic backgrounds: BALB/cJ (BALB), C57BL/6J (B6), and C3H/HeJ (C3H). The effect of Stat5b on hepatocarcinogenesis depended on the genetic background. B6 Stat5b null congenic males and females developed 2.4 times as many tumors as wild-type (WT) controls (P < 0.002) and the tumors were larger (P < 0.003). In BALB/c congenics, loss of STAT5b had no effect on either sex. C3H Stat5b null congenic males and females were resistant to liver cancer, developing 2.7- and 6-fold fewer tumors, respectively (P < 0.02, 0.01). These results provide the first example of a single gene behaving as both oncogene and tumor suppressor in a given tissue, depending only on the endogenous modifier alleles carried by different genetic backgrounds.
Collapse
Affiliation(s)
- Christopher C Oberley
- McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Andrea Bilger
- McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Norman R Drinkwater
- McArdle Laboratory for Cancer Research, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
191
|
Schaller-Schönitz M, Barzan D, Williamson AJK, Griffiths JR, Dallmann I, Battmer K, Ganser A, Whetton AD, Scherr M, Eder M. BCR-ABL affects STAT5A and STAT5B differentially. PLoS One 2014; 9:e97243. [PMID: 24836440 PMCID: PMC4023949 DOI: 10.1371/journal.pone.0097243] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 04/16/2014] [Indexed: 11/21/2022] Open
Abstract
Signal transducers and activators of transcription (STATs) are latent cytoplasmic transcription factors linking extracellular signals to target gene transcription. Hematopoietic cells express two highly conserved STAT5-isoforms (STAT5A/STAT5B), and STAT5 is directly activated by JAK2 downstream of several cytokine receptors and the oncogenic BCR-ABL tyrosine kinase. Using an IL-3-dependent cell line with inducible BCR-ABL-expression we compared STAT5-activation by IL-3 and BCR-ABL in a STAT5-isoform specific manner. RNAi targeting of STAT5B strongly inhibits BCR-ABL-dependent cell proliferation, and STAT5B but not STAT5A is essential for BCL-XL-expression in the presence of BCR-ABL. Although BCR-ABL induces STAT5-tyrosine phosphorylation independent of JAK2-kinase activity, BCR-ABL is less efficient in inducing active STAT5A:STAT5B-heterodimerization than IL-3, leaving constitutive STAT5A and STAT5B-homodimerization unaffected. In comparison to IL-3, nuclear accumulation of a STAT5A-eGFP fusion protein is reduced by BCR-ABL, and BCR-ABL tyrosine kinase activity induces STAT5A-eGFP translocation to the cell membrane and co-localization with the IL-3 receptor. Furthermore, BCR-ABL-dependent phosphorylation of Y682 in STAT5A was detected by mass-spectrometry. Finally, RNAi targeting STAT5B but not STAT5A sensitizes human BCR-ABL-positive cell lines to imatinib-treatment. These data demonstrate differences between IL-3 and BCR-ABL-mediated STAT5-activation and isoform-specific effects, indicating therapeutic options for isoform-specific STAT5-inhibition in BCR-ABL-positive leukemia.
Collapse
Affiliation(s)
- Michael Schaller-Schönitz
- Hannover Medical School, Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover, Germany
| | - David Barzan
- Hannover Medical School, Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover, Germany
| | - Andrew J. K. Williamson
- Faculty of Medical and Human Sciences, Manchester Academic Health Science Centre, University of Manchester, Wolfson Molecular Imaging Centre, Manchester, United Kingdom
| | - John R. Griffiths
- Faculty of Medical and Human Sciences, Manchester Academic Health Science Centre, University of Manchester, Wolfson Molecular Imaging Centre, Manchester, United Kingdom
| | - Iris Dallmann
- Hannover Medical School, Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover, Germany
| | - Karin Battmer
- Hannover Medical School, Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover, Germany
| | - Arnold Ganser
- Hannover Medical School, Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover, Germany
| | - Anthony D. Whetton
- Faculty of Medical and Human Sciences, Manchester Academic Health Science Centre, University of Manchester, Wolfson Molecular Imaging Centre, Manchester, United Kingdom
| | - Michaela Scherr
- Hannover Medical School, Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover, Germany
| | - Matthias Eder
- Hannover Medical School, Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover, Germany
- * E-mail:
| |
Collapse
|
192
|
Lewis RS, Noor SM, Fraser FW, Sertori R, Liongue C, Ward AC. Regulation of embryonic hematopoiesis by a cytokine-inducible SH2 domain homolog in zebrafish. THE JOURNAL OF IMMUNOLOGY 2014; 192:5739-48. [PMID: 24835394 DOI: 10.4049/jimmunol.1301376] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cytokine-inducible SH2 domain-containing protein (CISH), a member of the suppressor of cytokine signaling family of negative feedback regulators, is induced by cytokines that activate STAT5 and can inhibit STAT5 signaling in vitro. However, demonstration of a definitive in vivo role for CISH during development has remained elusive. This study employed expression analysis and morpholino-mediated knockdown in zebrafish in concert with bioinformatics and biochemical approaches to investigate CISH function. Two zebrafish CISH paralogs were identified, cish.a and cish.b, with high overall conservation (43-46% identity) with their mammalian counterparts. The cish.a gene was maternally derived, with transcripts present throughout embryogenesis, and increasing at 4-5 d after fertilization, whereas cish.b expression commenced at 8 h after fertilization. Expression of cish.a was regulated by the JAK2/STAT5 pathway via conserved tetrameric STAT5 binding sites (TTCN3GAA) in its promoter. Injection of morpholinos targeting cish.a, but not cish.b or control morpholinos, resulted in enhanced embryonic erythropoiesis, myelopoiesis, and lymphopoiesis, including a 2- 3-fold increase in erythrocytic markers. This occurred concomitantly with increased activation of STAT5. This study indicates that CISH functions as a conserved in vivo target and regulator of STAT5 in the control of embryonic hematopoiesis.
Collapse
Affiliation(s)
- Rowena S Lewis
- School of Life and Environmental Sciences, Deakin University, Burwood, Victoria 3125, Australia; Walter and Eliza Hall Institute for Medical Research, Parkville, Victoria 3050, Australia
| | - Suzita M Noor
- School of Medicine, Deakin University, Geelong, Victoria 3217, Australia; Strategic Research Centre in Molecular and Medical Research, Deakin University, Geelong, Victoria 3217, Australia; and Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Fiona W Fraser
- School of Medicine, Deakin University, Geelong, Victoria 3217, Australia; Strategic Research Centre in Molecular and Medical Research, Deakin University, Geelong, Victoria 3217, Australia; and
| | - Robert Sertori
- School of Medicine, Deakin University, Geelong, Victoria 3217, Australia; Strategic Research Centre in Molecular and Medical Research, Deakin University, Geelong, Victoria 3217, Australia; and
| | - Clifford Liongue
- School of Medicine, Deakin University, Geelong, Victoria 3217, Australia; Strategic Research Centre in Molecular and Medical Research, Deakin University, Geelong, Victoria 3217, Australia; and
| | - Alister C Ward
- School of Medicine, Deakin University, Geelong, Victoria 3217, Australia; Strategic Research Centre in Molecular and Medical Research, Deakin University, Geelong, Victoria 3217, Australia; and
| |
Collapse
|
193
|
Shi SY, Luk CT, Brunt JJ, Sivasubramaniyam T, Lu SY, Schroer SA, Woo M. Adipocyte-specific deficiency of Janus kinase (JAK) 2 in mice impairs lipolysis and increases body weight, and leads to insulin resistance with ageing. Diabetologia 2014; 57:1016-26. [PMID: 24531222 DOI: 10.1007/s00125-014-3185-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Accepted: 01/21/2014] [Indexed: 12/14/2022]
Abstract
AIMS/HYPOTHESIS The growing obesity epidemic necessitates a better understanding of adipocyte biology and its role in metabolism. The Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway mediates signalling by numerous cytokines and hormones that regulate adipocyte function, illustrating the physiological importance of adipose JAK-STAT. The aim of this study was to investigate potential roles of adipocyte JAK2, an essential player in the JAK-STAT pathway, in adipocyte biology and metabolism. METHODS We generated adipocyte-specific Jak2 knockout (A-Jak2 KO) mice using the Cre-loxP system with Cre expression driven by the Ap2 (also known as Fabp4) promoter. RESULTS Starting at 2-3 months of age, male and female A-Jak2 KO mice gradually gained more body weight than control littermates primarily due to increased adiposity. This was associated with reduced energy expenditure in A-Jak2 KO mice. In perigonadal adipose tissue, the expression of numerous genes involved in lipid metabolism was differentially regulated. In addition, adipose tissue from A-Jak2 KO mice displayed impaired lipolysis in response to isoprenaline, growth hormone and leptin stimulation, suggesting that adipose JAK2 directly modulates the lipolytic program. Impaired lipid homeostasis was also associated with disrupted adipokine secretion. Accordingly, while glucose metabolism was normal at 2 months of age, by 5-6 months of age, A-Jak2 KO mice had whole-body insulin resistance. CONCLUSIONS/INTERPRETATION Our results suggest that adipocyte JAK2 plays a critical role in the regulation of adipocyte biology and whole-body metabolism. Targeting of the JAK-STAT pathway could be a novel therapeutic option for the treatment of obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Sally Yu Shi
- Toronto General Research Institute, 101 College Street, MaRS Centre/TMDT, Room 10-363, Toronto, ON, Canada, M5G 1L7
| | | | | | | | | | | | | |
Collapse
|
194
|
Furigo IC, Kim KW, Nagaishi VS, Ramos-Lobo AM, de Alencar A, Pedroso JAB, Metzger M, Donato J. Prolactin-sensitive neurons express estrogen receptor-α and depend on sex hormones for normal responsiveness to prolactin. Brain Res 2014; 1566:47-59. [PMID: 24751572 DOI: 10.1016/j.brainres.2014.04.018] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 04/10/2014] [Accepted: 04/12/2014] [Indexed: 11/26/2022]
Abstract
Estrogens and prolactin share important target tissues, including the gonads, brain, liver, kidneys and some types of cancer cells. Herein, we sought anatomical and functional evidence of possible crosstalk between prolactin and estrogens in the mouse brain. First, we determined the distribution of prolactin-responsive neurons that express the estrogen receptor α (ERα). A large number of prolactin-induced pSTAT5-immunoreactive neurons expressing ERα mRNA were observed in several brain areas, including the anteroventral periventricular nucleus, medial preoptic nucleus, arcuate nucleus of the hypothalamus, ventrolateral subdivision of the ventromedial nucleus of the hypothalamus (VMH), medial nucleus of the amygdala and nucleus of the solitary tract. However, although the medial preoptic area, periventricular nucleus of the hypothalamus, paraventricular nucleus of the hypothalamus, retrochiasmatic area, dorsomedial subdivision of the VMH, lateral hypothalamic area, dorsomedial nucleus of the hypothalamus and ventral premammillary nucleus contained significant numbers of prolactin-responsive neurons, these areas showed very few pSTAT5-immunoreactive cells expressing ERα mRNA. Second, we evaluated prolactin sensitivity in ovariectomized mice and observed that sex hormones are required for a normal responsiveness to prolactin as ovariectomized mice showed a lower number of prolactin-induced pSTAT5 immunoreactive neurons in all analyzed brain nuclei compared to gonad-intact females. In addition, we performed hypothalamic gene expression analyses to determine possible post-ovariectomy changes in components of prolactin signaling. We observed no significant changes in the mRNA expression of prolactin receptor, STAT5a or STAT5b. In summary, sex hormones exert a permissive role in maintaining the brain's prolactin sensitivity, most likely through post-transcriptional mechanisms.
Collapse
Affiliation(s)
- Isadora C Furigo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Ki Woo Kim
- Department of Pharmacology, Institute of Lifestyle Medicine, and Nuclear Receptor Research Consortium, Yonsei University Wonju College of Medicine, Wonju 220-701, Republic of Korea
| | - Vanessa S Nagaishi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Angela M Ramos-Lobo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Amanda de Alencar
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - João A B Pedroso
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Martin Metzger
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
195
|
Genetic contribution of CISH promoter polymorphisms to susceptibility to tuberculosis in Chinese children. PLoS One 2014; 9:e92020. [PMID: 24632804 PMCID: PMC3954833 DOI: 10.1371/journal.pone.0092020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 02/15/2014] [Indexed: 12/19/2022] Open
Abstract
Tuberculosis (TB) is the leading cause of death due to an infectious disease worldwide, particularly in developing countries. A series of candidate genes have been suggested to be associated with development of TB disease. Among them, the human Cytokine-inducible Src homology 2(SH2) domain protein (CISH) gene has been very recently reported to be involved in T cell activation and differentiation in response to Mycobacterium tuberculosis infection. Here, we studied the association between CISH promoter polymorphisms and pediatric TB. A case-control study enrolled 352 TB patients and 527 healthy controls, who were of Han Chinese ethnicity and aged from 0.2 to 18 years. CISH gene promoter SNPs rs414171, rs622502 and rs809451 were genotyped in all subjects and transcriptional activity, mRNA level, and plasma cytokine level of subjects with different genotypes were further examined. Carriers with rs414171TT homozygotes and rs809451GC heterozygotes had a 1.78-fold (95% CI,1.16–2.74) and 1.86-fold (95% CI, 1.26–2.74) excess risk of developing TB compared to those with wild-type genotypes. A greater risk of TB disease was observed in population carrying C−809451-T−414171-C−622502 haplotype (OR 3.66, 95% CI:2.12–6.32). The G−809451-A−414171-C−622502-containing CISH promoter drove a 5.43-fold increased reporter expression compared to the C−809451-T−414171-C−622502-containing counterpart in Hela cell lines (P = 0.0009). PBMCs carrying rs414171TT homozygotes and rs809451GC heterozygotes showed a reduced CISH mRNA level compared to cells carrying wild type genotypes. Individuals with the rs414171TT genotype had significantly increased IL-12p40 and IL-10 production. In conclusion, CISH promoter rs414171 and rs809451 polymorphisms may play a vital role in mediating individual susceptibility to tuberculosis.
Collapse
|
196
|
Giraud M, Jmari N, Du L, Carallis F, Nieland TJF, Perez-Campo FM, Bensaude O, Root DE, Hacohen N, Mathis D, Benoist C. An RNAi screen for Aire cofactors reveals a role for Hnrnpl in polymerase release and Aire-activated ectopic transcription. Proc Natl Acad Sci U S A 2014; 111:1491-1496. [PMID: 24434558 PMCID: PMC3910647 DOI: 10.1073/pnas.1323535111] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Aire induces the expression of a large set of autoantigen genes in the thymus, driving immunological tolerance in maturing T cells. To determine the full spectrum of molecular mechanisms underlying the Aire transactivation function, we screened an AIRE-dependent gene-expression system with a genome-scale lentiviral shRNA library, targeting factors associated with chromatin architecture/function, transcription, and mRNA processing. Fifty-one functional allies were identified, with a preponderance of factors that impact transcriptional elongation compared with initiation, in particular members of the positive transcription elongation factor b (P-TEFb) involved in the release of "paused" RNA polymerases (CCNT2 and HEXIM1); mRNA processing and polyadenylation factors were also highlighted (HNRNPL/F, SFRS1, SFRS3, and CLP1). Aire's functional allies were validated on transfected and endogenous target genes, including the generation of lentigenic knockdown (KD) mice. We uncovered the effect of the splicing factor Hnrnpl on Aire-induced transcription. Transcripts sensitive to the P-TEFb inhibitor flavopiridol were reduced by Hnrnpl knockdown in thymic epithelial cells, independently of their dependence on Aire, therefore indicating a general effect of Hnrnpl on RNA elongation. This conclusion was substantiated by demonstration of HNRNPL interactions with P-TEFb components (CDK9, CCNT2, HEXIM1, and the small 7SK RNA). Aire-containing complexes include 7SK RNA, the latter interaction disrupted by HNRNPL knockdown, suggesting that HNRNPL may partake in delivering inactive P-TEFb to Aire. Thus, these results indicate that mRNA processing factors cooperate with Aire to release stalled polymerases and to activate ectopic expression of autoantigen genes in the thymus.
Collapse
Affiliation(s)
- Matthieu Giraud
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
- Department of Immunology, Institut Cochin, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Université Paris Descartes, 75014 Paris, France
| | - Nada Jmari
- Department of Immunology, Institut Cochin, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Université Paris Descartes, 75014 Paris, France
| | - Lina Du
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Floriane Carallis
- Department of Immunology, Institut Cochin, Institut National de la Santé et de la Recherche Médicale (INSERM) U1016, Université Paris Descartes, 75014 Paris, France
| | | | - Flor M. Perez-Campo
- Department of Internal Medicine, Hospital U.M. Valdecilla-Instituto de Formación e Investigación Marqués de Valdecilla, University of Cantabria, 39008 Santander, Spain; and
| | - Olivier Bensaude
- Ecole Normale Supérieure, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8197, INSERM U1024, 75005 Paris, France
| | - David E. Root
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142
| | - Nir Hacohen
- The Broad Institute of MIT and Harvard, Cambridge, MA 02142
| | - Diane Mathis
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| | - Christophe Benoist
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
197
|
Stat5 regulates the phosphatidylinositol 3-kinase/Akt1 pathway during mammary gland development and tumorigenesis. Mol Cell Biol 2014; 34:1363-77. [PMID: 24469394 DOI: 10.1128/mcb.01220-13] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Stat5 (signal transducer and activator of transcription 5) is an essential mediator of cytokine receptor signaling and plays important roles in the proliferation of alveolar progenitors and the survival of functionally differentiated epithelial cells in the mammary gland. A deregulated expression and activation of Stat5 leads to precocious alveolar development in the absence of pregnancy hormones, impaired mammary gland remodeling following the cessation of lactation, and mammary tumor formation. We reported previously that Stat5 induces the transcription of the Akt1 gene from a novel promoter. In this report, we provide experimental evidence that Akt1 is an essential mediator for the biological function of Stat5 as a survival factor. Additionally, Stat5 controls the expression of the regulatory and catalytic subunits of the phosphatidylinositol 3-kinase (PI3K) (p85α and p110α), thereby greatly augmenting signaling through the prosurvival PI3K/Akt pathway. In agreement with this model, we observed that the constitutive activation of Stat5 cooperates with the loss of function of the tumor suppressor PTEN by accelerating the formation of preneoplastic lesions and mammary tumors. The mammary gland-specific ablation of Stat5 is sufficient to prevent mammary carcinogenesis in a genuine mouse model for Cowden syndrome. Therefore, targeting the Jak2/Stat5 pathway might be a suitable strategy to prevent breast cancer in patients that carry a mutant PTEN allele.
Collapse
|
198
|
Haricharan S, Li Y. STAT signaling in mammary gland differentiation, cell survival and tumorigenesis. Mol Cell Endocrinol 2014; 382:560-569. [PMID: 23541951 PMCID: PMC3748257 DOI: 10.1016/j.mce.2013.03.014] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 03/18/2013] [Indexed: 01/10/2023]
Abstract
The mammary gland is a unique organ that undergoes extensive and profound changes during puberty, menstruation, pregnancy, lactation and involution. The changes that take place during puberty involve large-scale proliferation and invasion of the fat-pad. During pregnancy and lactation, the mammary cells are exposed to signaling pathways that inhibit apoptosis, induce proliferation and invoke terminal differentiation. Finally, during involution the mammary gland is exposed to milk stasis, programmed cell death and stromal reorganization to clear the differentiated milk-producing cells. Not surprisingly, the signaling pathways responsible for bringing about these changes in breast cells are often subverted during the process of tumorigenesis. The STAT family of proteins is involved in every stage of mammary gland development, and is also frequently implicated in breast tumorigenesis. While the roles of STAT3 and STAT5 during mammary gland development and tumorigenesis are well studied, others members, e.g. STAT1 and STAT6, have only recently been observed to play a role in mammary gland biology. Continued investigation into the STAT protein network in the mammary gland will likely yield new biomarkers and risk factors for breast cancer, and may also lead to novel prophylactic or therapeutic strategies against breast cancer.
Collapse
Affiliation(s)
- S Haricharan
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Y Li
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
199
|
Sangeeta Devi Y, Halperin J. Reproductive actions of prolactin mediated through short and long receptor isoforms. Mol Cell Endocrinol 2014; 382:400-410. [PMID: 24060636 DOI: 10.1016/j.mce.2013.09.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 07/20/2013] [Accepted: 09/13/2013] [Indexed: 10/26/2022]
Abstract
Prolactin (PRL) is a polypeptide hormone with a wide range of physiological functions, and is critical for female reproduction. PRL exerts its action by binding to membrane bound receptor isoforms broadly classified as the long form and the short form receptors. Both receptor isoforms are highly expressed in the ovary as well as in the uterus. Although signaling through the long form is believed to be more predominant, it remains unclear whether activation of this isoform alone is sufficient to support reproductive functions or whether both types of receptor are required. The generation of transgenic mice selectively expressing either the short or the long form of PRL receptor has provided insight into the differential signaling mechanisms and physiological functions of these receptors. This review describes the essential finding that both long and short receptor isoforms are crucial for ovarian functions and female fertility, and highlights novel mechanisms of action for these receptors.
Collapse
Affiliation(s)
- Y Sangeeta Devi
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI-49503, USA.
| | - Julia Halperin
- Centro de Estudios Biomédicos, Biotecnológicos, Ambientales y Diagnóstico (CEBBAD), Universidad Maimónides, Hidalgo 775 6to piso, C1405BCK Ciudad Autónoma de Buenos Aires, Argentina and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Rivadavia 1917, Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
200
|
Walker SR, Xiang M, Frank DA. Distinct roles of STAT3 and STAT5 in the pathogenesis and targeted therapy of breast cancer. Mol Cell Endocrinol 2014; 382:616-621. [PMID: 23531638 PMCID: PMC3732813 DOI: 10.1016/j.mce.2013.03.010] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 03/14/2013] [Indexed: 02/04/2023]
Abstract
The transcription factors STAT3 and STAT5 play important roles in the regulation of mammary gland function during pregnancy, lactation, and involution. Given that STAT3 and STAT5 regulate genes involved in proliferation and survival, it is not surprising that inappropriate activation of STAT3 and STAT5 occurs commonly in breast cancer. Although these proteins are structurally similar, they have divergent and opposing effects on gene expression and cellular phenotype. Notably, when STAT5 and STAT3 are activated simultaneously, STAT5 has a dominant effect, and leads to decreased proliferation and increased sensitivity to cell death. Similarly, in breast cancer, activation of both STAT5 and STAT3 is associated with longer patient survival than activation of STAT3 alone. Pharmacological inhibitors of STAT3 and STAT5 are being developed for cancer therapy, though understanding the activation state and functional interaction of STAT3 and STAT5 in a patient's tumor may be critical for the optimal use of this strategy.
Collapse
Affiliation(s)
- Sarah R Walker
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Michael Xiang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - David A Frank
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|