Triple a Patient Cells Suffering from Mitotic Defects Fail to Localize PGRMC1 to Mitotic Kinetochore Fibers

Total Page:16

File Type:pdf, Size:1020Kb

Triple a Patient Cells Suffering from Mitotic Defects Fail to Localize PGRMC1 to Mitotic Kinetochore Fibers Jühlen et al. Cell Div (2018) 13:8 https://doi.org/10.1186/s13008-018-0041-5 Cell Division RESEARCH Open Access Triple A patient cells sufering from mitotic defects fail to localize PGRMC1 to mitotic kinetochore fbers Ramona Jühlen1,2* , Dana Landgraf1, Angela Huebner1 and Katrin Koehler1* Abstract Background: Membrane-associated progesterone receptors are restricted to the endoplasmic reticulum and are shown to regulate the activity of cytochrome P450 enzymes which are involved in steroidogenesis or drug detoxifca- tion. PGRMC1 and PGRMC2 belong to the membrane-associated progesterone receptor family and are of interest due to their suspected role during cell cycle. PGRMC1 and PGRMC2 are thought to bind to each other; thereby suppress- ing entry into mitosis. We could previously report that PGRMC2 interacts with the nucleoporin ALADIN which when mutated results in the autosomal recessive disorder triple A syndrome. ALADIN is a novel regulator of mitotic control- ler Aurora kinase A and depletion of this nucleoporin leads to microtubule instability. Results: In the current study, we present that proliferation is decreased when ALADIN, PGRMC1 or PGRMC2 are over- expressed. Furthermore, we fnd that depletion of ALADIN results in mislocalization of Aurora kinase A and PGRMC1 in metaphase cells. Additionally, PGRMC2 is over-expressed in triple A patient fbroblasts. Conclusion: Our results emphasize the possibility that loss of the regulatory association between ALADIN and PGRMC2 gives rise to a depletion of PGRMC1 at kinetochore fbers. This observation may explain part of the symp- toms seen in triple A syndrome patients. Keywords: ALADIN, Kinetochore fbers, Mitosis, Nuclear pore complex, PGRMC1, PGRMC2, Triple A syndrome Background was published in 2015 [7]. In cooperation with Carvalhal ALADIN is a scafold nucleoporin (NUP) anchored et al. we proposed ALADIN as novel regulator of mitotic within the nuclear pore complex by the transmembrane kinase Aurora kinase A and showed that depletion of the NUP NDC1 [1, 2]. ALADIN seems to be involved in nucleoporin resulted in impaired mitotic spindle assem- building the structural scafold backbone of the nuclear bly and chromosomal alignment at the metaphase plate pore complex [3]. Over the last years it has been shown [7]. Furthermore, we could recently document that ALA- that nuclear pore complexes and its NUPs have fun- DIN is necessary for murine oocyte maturation and for damental functions beyond nucleocytoplasmic trans- specifc stages during meiosis [8]. port and control cellular dysfunction in a variety of Mutations in the human AAAS gene, coding for the cellular pathways, especially during mitosis [4–6]. Te protein ALADIN (alacrima-achalasia-adrenal insuf- frst report that ALADIN plays a role during cell division fciency neurologic disorder), lead to the autosomal recessive disorder named triple A syndrome [9, 10]. Triple A patients present with three distinct symptoms: *Correspondence: [email protected]; katrin. absent adrenal glucocorticoid and mineralocorticoid koehler@uniklinikum‑dresden.de synthesis (adrenal insufciency), impaired movement 1 Klinik und Poliklinik für Kinder‑ und Jugendmedizin, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, of the stomach cardia (achalasia) and loss of tear pro- 01307 Dresden, Germany duction (alacrima) [11]. Tese symptoms are highly 2 Present Address: Institute for Molecular Biology and Medicine, Université heterogeneous and are accompanied by progressive Libre de Bruxelles, 6041 Charleroi, Belgium © The Author(s) 2018. This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creat​iveco​mmons​.org/licen​ses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creat​iveco​mmons​.org/ publi​cdoma​in/zero/1.0/) applies to the data made available in this article, unless otherwise stated. Jühlen et al. Cell Div (2018) 13:8 Page 2 of 14 impairments of the central, peripheral or autonomous (p58) [28]. PGRMC1 and PGRMC2 are reported to inter- nervous system [11]. Most mutations in AAAS result act and furthermore, to bind to each other during meta- in a mis-localization of ALADIN to the cytoplasm [12, phase, thereby suppressing entry into cell cycle [27]. 13]. Interestingly, in a large scale interactome mapping of Previously, we identifed PGRMC2 as novel interac- the centrosome-cilium interface the nucleoporin ALA- tor for the nucleoporin ALADIN and provided new DIN and both membrane-associated progesterone recep- insights into the molecular function of the nucleoporin tors PGRMC1 and PGRMC2 have been identifed to in the pathogenesis of triple A syndrome [14]. PGRMC2 localize to the cilium transition zone [31, 32]. Te centro- belongs to the group of membrane-associated proges- some is an important regulator of cell cycle progression terone receptors. Tese receptors mainly localize to the and mitotic spindle assembly [33]. Furthermore, ALA- endoplasmic reticulum (ER) and are thought to regu- DIN is strongly dephosphorylated during mitotic exit, late the activity of microsomal cytochrome (CYP) P450 whereas PGRMC1 and PGRMC2 are phosphorylated enzymes which are involved in steroidogenesis or drug during early mitotic exit [34, 35]. Obviously, ALADIN, detoxifcation [15]. Te frst identifed membrane- PGRMC1 and PGRMC2 seem to have critical roles for associated progesterone receptor, PGRMC1, gained the formation and function of the mitotic spindle appara- wide-spread attention due to its several implications in tus in somatic cells. cancerogenesis [16–19]. Te mixed-function oxidase Here, we report that proliferation in human adrenal system of CYP P450 enzymes requires a donor trans- cells is decreased after increased expression of ALADIN, ferring electrons from NADPH to reduce the enzyme’s PGRMC1 or PGRMC2. We show that PGRMC1 local- prosthetic heme group [20]. PGRMC1 and PGRMC2 izes to the microtubule kinetochore-fbers in metaphase contain a CYP b5-similar heme-binding domain which and to the midbody in telophase of human adrenal cells makes them possible electron donors for CYP P450 and fbroblasts. Further, we present that PGRMC1 and enzymes. [19]. Indeed, PGRMC1 forms stable protein– Aurora kinase A are mislocalized in metaphase triple protein complexes with CYP51A1, CYP7A1, CYP21A2 A patient fbroblasts. We observed that patient fbro- and CYP3A4 [21]. Additionally, PGRMC1 is able to acti- blasts present with increased expression of PGRMC2 vate CYP19 aromatase [22]. PGRMC1 is shown to physi- and we hypothesize that a depletion of ALADIN in these ologically afect cholesterol/steroid biosynthesis and cells leads to a dysregulation of PGRMC2 and displaces metabolism [19]. It is known that PGRMC2 has similar PGRMC1 at the metaphase spindle. interaction potential, alters activity of CYP3A4 as pos- sible electron donor, and binds CYP21A2 [23, 24]. Most Results recently, both PGRMC1 and PGRMC2 were identifed Adrenal cell proliferation is decreased as putative interacting partners of ferrochelatase, an upon over‑expression of ALADIN, PGRMC1 or PGRMC2 enzyme catalyzing the terminal step in the heme biosyn- and down‑regulation of ALADIN thetic pathway, thereby possibly controlling heme release It is well documented that triple A patient fbroblasts as chaperone or sensor [25]. Interaction of ALADIN with show decreased cellular proliferation and doubling PGRMC2 at the perinuclear ER could infuence CYP time [36]. To test whether depletion of ALADIN also P450 enzyme activity through electron transfer from results in decreased cellular proliferation in adrenal NADPH and/or control heme synthesis. In triple A syn- cells we used inducible adrenocortical NCI-H295R1- drome, altered CYP P450 enzyme activity would consec- TR cells stably expressing AAAS shRNA (AAAS utively contribute to adrenal atrophy [14]. knock-down) and monitored cellular proliferation Human PGRMC1 and PGRMC2 share 67% of their using live cell imaging for at least 65 h. AAAS knock- protein sequence [15, 26]. Defciency of either PGRMC1 down resulted in decreased proliferation in adrenal or PGRMC2 decreases the anti-apoptotic and anti- cells (growth constant k (slope of linear regression mitotic action of progesterone [27]. Additionally, line) = 0.044) compared to control cells expressing a increased expression of PGRMC1 or PGRMC2 inhibits scrambled shRNA (k = 0.062) (wild-type cells: k = 0.2) entry into cell cycle [27, 28]. On the one hand, PGRMC1 (Fig. 1a). Surprisingly, in live cell imaging stable over- is distributed with β- and γ-tubulin to the mitotic bipo- expression of N-terminal-GFP-tagged ALADIN in lar spindle and spindle poles in metaphase cells and on adrenocortical NCI-H295R cells also impaired cellu- the other hand, with Aurora kinase B in meiotic cells lar proliferation (k = 0.082) compared to over-expres- [29, 30]. Furthermore, PGRMC1 is thought to regulate sion of GFP alone in these cells (k = 0.305) (wild-type microtubule stability [28]. PGRMC2 is shown to local- cells: k = 0.16) (Fig. 1b). Furthermore, during live cell ize to the mitotic spindles in metaphase and anaphase imaging we observed that transient over-expression of cells and shall interact with cyclin-dependent kinase 11B C-terminal-GFP-tagged PGRMC2 in adrenocortical Jühlen et al. Cell
Recommended publications
  • Insulin Receptor Plasma Membrane Levels Increased by the Progesterone Receptor Membrane Component 1 S
    Supplemental material to this article can be found at: http://molpharm.aspetjournals.org/content/suppl/2018/04/19/mol.117.110510.DC1 1521-0111/94/1/665–673$35.00 https://doi.org/10.1124/mol.117.110510 MOLECULAR PHARMACOLOGY Mol Pharmacol 94:665–673, July 2018 Copyright ª 2018 by The American Society for Pharmacology and Experimental Therapeutics Insulin Receptor Plasma Membrane Levels Increased by the Progesterone Receptor Membrane Component 1 s Kaia K. Hampton, Katie Anderson, Hilaree Frazier, Olivier Thibault, and Rolf J. Craven Department of Pharmacology and Nutritional Sciences, Markey Cancer Center, University of Kentucky College of Medicine, Lexington, Kentucky Received September 7, 2017; accepted April 13, 2018 Downloaded from ABSTRACT The insulin receptor (IR) is a ligand-activated receptor tyrosine therapeutic applications because a small-molecule PGRMC1 kinase that has a key role in metabolism, cellular survival, and ligand, AG205, also decreases plasma membrane IR levels. proliferation. Progesterone receptor membrane component However, PGRMC1 knockdown via short hairpin RNA expres- 1 (PGRMC1) promotes cellular signaling via receptor trafficking sion and AG205 treatment potentiated insulin-mediated phos- and is essential for some elements of tumor growth and phorylation of the IR signaling mediator AKT. Finally, PGRMC1 metastasis. In the present study, we demonstrate that PGRMC1 also increased plasma membrane levels of two key glucose molpharm.aspetjournals.org coprecipitates with IR. Furthermore, we show that PGRMC1 transporters, GLUT-4 and GLUT-1. Our data support a role for increases plasma membrane IR levels in multiple cell lines and PGRMC1 maintaining plasma membrane pools of the receptor, decreases insulin binding at the cell surface.
    [Show full text]
  • PGRMC1 and PGRMC2 in Uterine Physiology and Disease
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Frontiers - Publisher Connector PERSPECTIVE ARTICLE published: 19 September 2013 doi: 10.3389/fnins.2013.00168 PGRMC1 and PGRMC2 in uterine physiology and disease James K. Pru* and Nicole C. Clark Department of Animal Sciences, School of Molecular Biosciences, Center for Reproductive Biology, Washington State University, Pullman, WA, USA Edited by: It is clear from studies using progesterone receptor (PGR) mutant mice that not all of Sandra L. Petersen, University of the actions of progesterone (P4) are mediated by this receptor. Indeed, many rapid, Massachusetts Amherst, USA non-classical P4 actions have been reported throughout the female reproductive tract. Reviewed by: Progesterone treatment of Pgr null mice results in behavioral changes and in differential Cecily V. Bishop, Oregon National Primate Research Center, USA regulation of genes in the endometrium. Progesterone receptor membrane component Christopher S. Keator, Ross (PGRMC) 1 and PGRMC2 belong to the heme-binding protein family and may serve as University School of Medicine, P4 receptors. Evidence to support this derives chiefly from in vitro culture work using Dominica primary or transformed cell lines that lack the classical PGR. Endometrial expression of *Correspondence: PGRMC1 in menstrual cycling mammals is most abundant during the proliferative phase James K. Pru, Department of Animal Sciences, School of Molecular of the cycle. Because PGRMC2 expression shows the most consistent cross-species Biosciences, Center for expression, with highest levels during the secretory phase, PGRMC2 may serve as a Reproductive Biology, Washington universal non-classical P4 receptor in the uterus.
    [Show full text]
  • Loss of PGRMC1 Delays the Progression of Hepatocellular Carcinoma Via Suppression of Pro-Inflammatory Immune Responses
    cancers Article Loss of PGRMC1 Delays the Progression of Hepatocellular Carcinoma via Suppression of Pro-Inflammatory Immune Responses Sang R. Lee 1,† , Jong Geol Lee 2,†, Jun H. Heo 1, Seong Lae Jo 1, Jihoon Ryu 1 , Globinna Kim 2, Jung-Min Yon 2, Myeong Sup Lee 3, Geun-Shik Lee 4 , Beum-Soo An 5 , Hyun-Jin Shin 1, Dong-Cheol Woo 2 , In-Jeoung Baek 2,* and Eui-Ju Hong 1,* 1 College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Korea; [email protected] (S.R.L.); [email protected] (J.H.H.); [email protected] (S.L.J.); [email protected] (J.R.); [email protected] (H.-J.S.) 2 Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; [email protected] (J.G.L.); [email protected] (G.K.); [email protected] (J.-M.Y.); [email protected] (D.-C.W.) 3 Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; [email protected] 4 College of Veterinary Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Korea; [email protected] 5 Department of Biomaterials Science, College of Natural Resources & Life Science, Pusan National University, Miryang, Gyeongsangnam 50463, Korea; [email protected] * Correspondence: [email protected] (I.-J.B.); [email protected] (E.-J.H.); Tel.: +82-2-3010-2798 (I.-J.B.); Citation: Lee, S.R.; Lee, J.G.; Heo, +82-42-821-6781 (E.-J.H.); Fax: +82-2-3010-4197 (I.-J.B.); +82-42-821-8903 (E.-J.H.) J.H.; Jo, S.L.; Ryu, J.; Kim, G.; Yon, † These authors equally contributed to the study.
    [Show full text]
  • P338. Does Norethisterone Stimulate Human Breast Cancer Cells Proliferation by Promoting PGRMC1 Expression?
    P338. Does Norethisterone stimulate human breast cancer cells proliferation by promoting PGRMC1 expression? M Gu (CN) [1], X Ruan (CN) [2], C Jia (CN) [3], C Yang (CA) [4], P Hardy (CA) [5], A O Mueck (DE) [6] Context : Important studies such as the Women’s Health Initiative (WHI) and the Million Women Study (MWS) showed progestogens may play a important role in the development of breast cancer under hormone therapy in menopausal women. Progesterone receptor membrane component 1 (PGRMC1) has been found to be highly expressed in the tissue of breast cancer patients.in our previous studys,we found norethisterone (NET) can stimulate the proliferation of breast cancer cells which express PGRMC1 in vitro and in vivo,However the mechanism is unclear. Objective: To investigate possible mechanisms for increased breast cancer risk with NET in hormone therapy and oral contraceptives. Methods: Cell viability assay was performed to investigate the proliferation effect of MCF-7 cells stimulated with NET or progesterone (10-5 M~10-11 M).Quantitative PCR and Western blot analysis were used to evaluate the PGRMC1 expression in three groups. The promoter sequence of PGRMC1(2kb) was cloned into pGL3-basic reporter vector. Cells were transfected with plasmids using FuGene. Luciferase activity was determined 48 hours posttransfection with the Dual-Luciferase Reporter Assay System . Results: NET induced MCF-7 cell viability dose-dependently, but this effect was not observed by using progesterone. Our real-time quantitative PCR data displayed the significantly increase mRNA level of PGRMC1 in the MCF-7 cells stimulated with NET dose-dependently. Western blot analysis show that expression of PGRMC1 in protein level significantly increase in the MCF-7 cells stimulated with NET compare to MCF-7 cells stimulated with progesterone.We found NET up-regulated the activity of the PGRMC1 promoter.
    [Show full text]
  • Progesterone Receptor Membrane Component 1 Suppresses the P53
    www.nature.com/scientificreports OPEN Progesterone Receptor Membrane Component 1 suppresses the p53 and Wnt/β-catenin pathways to Received: 30 October 2017 Accepted: 2 February 2018 promote human pluripotent stem Published: xx xx xxxx cell self-renewal Ji Yea Kim1, So Young Kim1, Hong Seo Choi1, Min Kyu Kim1, Hyun Min Lee1, Young-Joo Jang2 & Chun Jeih Ryu1 Progesterone receptor membrane component 1 (PGRMC1) is a multifunctional heme-binding protein involved in various diseases, including cancers and Alzheimer’s disease. Previously, we generated two monoclonal antibodies (MAbs) 108-B6 and 4A68 against surface molecules on human pluripotent stem cells (hPSCs). Here we show that PGRMC1 is the target antigen of both MAbs, and is predominantly expressed on hPSCs and some cancer cells. PGRMC1 is rapidly downregulated during early diferentiation of hPSCs. Although PGRMC1 knockdown leads to a spread-out morphology and impaired self-renewal in hPSCs, PGRMC1 knockdown hPSCs do not show apoptosis and autophagy. Instead, PGRMC1 knockdown leads to diferentiation of hPSCs into multiple lineage cells without afecting the expression of pluripotency markers. PGRMC1 knockdown increases cyclin D1 expression and decreases Plk1 expression in hPSCs. PGRMC1 knockdown also induces p53 expression and stability, suggesting that PGRMC1 maintains hPSC self-renewal through suppression of p53-dependent pathway. Analysis of signaling molecules further reveals that PGRMC1 knockdown promotes inhibitory phosphorylation of GSK-3β and increased expression of Wnt3a and β-catenin, which leads to activation of Wnt/β-catenin signaling. The results suggest that PGRMC1 suppresses the p53 and Wnt/β-catenin pathways to promote self-renewal and inhibit early diferentiation in hPSCs.
    [Show full text]
  • PGRMC1 Phosphorylation Status and Cell Plasticity 1: Glucose Metabolism, Mitochondria
    bioRxiv preprint doi: https://doi.org/10.1101/737718; this version posted August 24, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Title PGRMC1 phosphorylation status and cell plasticity 1: glucose metabolism, mitochondria, and mouse xenograft tumorigenesis Authors Bashar M. Thejer,1,2,3 Partho P. Adhikary,1,2,4 Amandeep Kaur,5,6 Sarah L. Teakel,1 Ashleigh Van Oosterum,7 Ishith Seth,1 Marina Pajic,8,9 Kate M. Hannan,10,11 Megan Pavy,11 Perlita Poh,11 Jalal A. Jazayeri,1 Thiri Zaw,12 Dana Pascovici,12 Marina Ludescher,13 Michael Pawlak,14 Juan C. Cassano,15 Lynne Turnbull,16 Mitra Jazayeri,17 Alexander C. James,18,19,20 Craig P. Coorey,18,21 Tara L. Roberts,18,19,20,21 Simon J. Kinder,22 Ross D. Hannan,9,10,23,24,25 Ellis Patrick,26 Mark P. Molloy,12,27 Elizabeth J. New,5 Tanja N. Fehm,13 Hans Neubauer,13 Ewa M. Goldys,28,29 Leslie A. Weston,30,31 and Michael A. Cahill.1,10,* Affiliations 1. School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW, 2650, Australia. 2. Co-first author. 3. Department of Biology, College of Science, University of Wasit, Wasit, Iraq. 4. Current address: Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, Canada. 5. University of Sydney, School of Chemistry, Sydney, NSW, 2006 Australia.
    [Show full text]
  • The Evolutionary Appearance of Signaling Motifs in PGRMC1
    BioScience Trends Advance Publication P1 Original Article Advance Publication DOI: 10.5582/bst.2017.01009 The evolutionary appearance of signaling motifs in PGRMC1 Michael A. Cahill* School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, Australia. Summary A complex PGRMC1-centred regulatory system controls multiple cell functions. Although PGRMC1 is phosphorylated at several positions, we do not understand the mechanisms regulating its function. PGRMC1 is the archetypal member of the membrane associated progesterone receptor (MAPR) family. Phylogentic comparison of MAPR proteins suggests that the ancestral metazoan "PGRMC-like" MAPR gene resembled PGRMC1/PGRMC2, containing the equivalents of PGRMC1 Y139 and Y180 SH2 target motifs. It later acquired a CK2 site with phosphoacceptor at S181. Separate PGRMC1 and PGRMC2 genes with this "PGRMC-like" structure diverged after the separation of vertebrates from protochordates. Terrestrial tetrapods possess a novel proline-rich PGRMC1 SH3 target motif centred on P64 which in mammals is augmented by a phosphoacceptor at PGRMC1 S54, and in primates by an additional S57 CK2 site. All of these phosphoacceptors are phosphorylated in vivo. This study suggests that an increasingly sophisticated system of PGRMC1-modulated multicellular functional regulation has characterised animal evolution since Precambrian times. Keywords: Phosphorylation, evolution, steroid signalling, kinases, metazoan 1. Introduction that regulates synthesis of sterol precursors, conferring responsiveness to progesterone
    [Show full text]
  • Microrna-181A Suppresses Progestin-Stimulated Breast Cancer
    Li X, Yang C, Ruan X, Croteau S, Hardy P. MicroRNA-181a Suppresses Progestin- Stimu-lated Breast Cancer Cell Growth. J Cancer Treatment Diagn. (2018);2(5):1-5 Journal of Cancer Treatment and Diagnosis Mini Review Open Access MicroRNA-181a Suppresses Progestin-Stimulated Breast Cancer Cell Growth Xue Li1, Chun Yang2, Xiangyan Ruan1, Stéphane Croteau2, Pierre Hardy2* 1Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China 2Departments of Medicine, Pediatrics, Pharmacology and Physiology, University of Montreal, Montreal, Quebec, Canada Article Info ABSTRACT Article Notes Despite all our efforts, breast cancer remains a major public health problem Received: July 23, 2018 threatening women’s health all around the world. The morbidity of breast Accepted: September 4, 2018 cancer is rising in most countries and is going to rise further over the next 20 *Correspondence: years. Hormone treatment is widely used and it is the most efficient method Dr. Pierre Hardy, MD, PhD, Research Center of CHU Sainte- of reducing menopausal symptoms or preventing abortion and pregnancy. Justine, 3175 Côte-Sainte-Catherine, Room 2714 Montréal, However, multiple studies have demonstrated that hormones, especially Québec, H3T 1C5, Canada; Telephone No: (514) 345-4931 synthetic progesterone, remain an indisputable risk factor for breast cancer. (ext. 3656); MicroRNAs are a group of endogenous small non-coding single strand RNAs E-mail: [email protected]. which play regulatory roles in the initiation, development, and progression © 2018 Hardy P. This article is distributed under the terms of of different types of cancer. Evidence from multiple sources indicates that the Creative Commons Attribution 4.0 International License.
    [Show full text]
  • Insights Into the Mechanism of Action of 13-Cis Retinoic Acid
    The Pennsylvania State University The Graduate School College of Medicine INSIGHTS INTO THE MECHANISM OF ACTION OF 13-CIS RETINOIC ACID IN SUPPRESSING SEBACEOUS GLAND FUNCTION A Thesis in Molecular Medicine by Amanda Marie Nelson © 2007 Amanda Marie Nelson Submitted in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy May 2007 The thesis of Amanda Marie Nelson was reviewed and approved* by the following: Diane M. Thiboutot Professor of Dermatology Thesis Advisor Chair of Committee Gary A. Clawson Professor of Pathology, Biochemistry and Molecular Biology Mark Kester Distinguished Professor of Pharmacology Jeffrey M. Peters Associate Professor of Molecular Toxicology Jong K. Yun Assistant Professor of Pharmacology Craig Meyers Professor of Microbiology and Immunology Co-chair, Molecular Medicine Graduate Degree Program *Signatures are on file in the Graduate School iii ABSTRACT Nearly 40-50 million people of all races and ages in the United States have acne, making it the most common skin disease. Although acne is not a serious health threat, severe acne can lead to disfiguring, permanent scarring; increased anxiety; and depression. Isotretinoin (13-cis Retinoic Acid) is the most potent agent that affects each of the pathogenic features of acne: 1) follicular hyperkeratinization, 2) the activity of Propionibacterium acnes, 3) inflammation and 4) increased sebum production. Isotretinoin has been on the market since 1982 and even though it has been prescribed for 25 years, extensive studies into its molecular mechanism of action in human skin and sebaceous glands have not been done. Since isotretinoin is a teratogen, there is a clear need for safe and effective alternative therapeutic agents.
    [Show full text]
  • Progesterone Receptor Membrane Component-1 Regulates Hepcidin Biosynthesis
    Progesterone receptor membrane component-1 regulates hepcidin biosynthesis Xiang Li, … , Donald B. Bloch, Randall T. Peterson J Clin Invest. 2016;126(1):389-401. https://doi.org/10.1172/JCI83831. Research Article Hematology Iron homeostasis is tightly regulated by the membrane iron exporter ferroportin and its regulatory peptide hormone hepcidin. The hepcidin/ferroportin axis is considered a promising therapeutic target for the treatment of diseases of iron overload or deficiency. Here, we conducted a chemical screen in zebrafish to identify small molecules that decrease ferroportin protein levels. The chemical screen led to the identification of 3 steroid molecules, epitiostanol, progesterone, and mifepristone, which decrease ferroportin levels by increasing the biosynthesis of hepcidin. These hepcidin-inducing steroids (HISs) did not activate known hepcidin-inducing pathways, including the BMP and JAK/STAT3 pathways. Progesterone receptor membrane component-1 (PGRMC1) was required for HIS-dependent increases in hepcidin biosynthesis, as PGRMC1 depletion in cultured hepatoma cells and zebrafish blocked the ability of HISs to increase hepcidin mRNA levels. Neutralizing antibodies directed against PGRMC1 attenuated the ability of HISs to induce hepcidin gene expression. Inhibiting the kinases of the SRC family, which are downstream of PGRMC1, blocked the ability of HISs to increase hepcidin mRNA levels. Furthermore, HIS treatment increased hepcidin biosynthesis in mice and humans. Together, these data indicate that PGRMC1 regulates hepcidin gene expression through an evolutionarily conserved mechanism. These studies have identified drug candidates and potential therapeutic targets for the treatment of diseases of abnormal iron metabolism. Find the latest version: https://jci.me/83831/pdf The Journal of Clinical Investigation RESEARCH ARTICLE Progesterone receptor membrane component-1 regulates hepcidin biosynthesis Xiang Li,1,2 David K.
    [Show full text]
  • Progesterone Inhibits Apoptosis in Part by PGRMC1-Regulated Gene Expression
    Molecular and Cellular Endocrinology 320 (2010) 153–161 Contents lists available at ScienceDirect Molecular and Cellular Endocrinology journal homepage: www.elsevier.com/locate/mce Progesterone inhibits apoptosis in part by PGRMC1-regulated gene expression J.J. Peluso a,b,∗,X.Liua, A. Gawkowska a, V. Lodde a,1, C.A. Wu c a Department of Cell Biology, University of Connecticut Health Center, Farmington, CT 06030, United States b Department of Obstetrics and Gynecology, University of Connecticut Health Center, Farmington, CT 06030, United States c Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030, United States article info abstract Article history: Progesterone receptor membrane component-1 (PGRMC1) is present in both the cytoplasm and nucleus Received 4 December 2009 of spontaneously immortalized granulosa cells (SIGCs). PGRMC1 is detected as a monomer in the cyto- Received in revised form 27 January 2010 plasm and a DTT-resistant PGRMC1 dimer in the nucleus. Transfected PGRMC1–GFP localizes mainly to Accepted 1 February 2010 the cytoplasm and does not form a DTT-resistant dimer. Moreover, forced expression of PGRMC1–GFP increases the sensitivity of the SIGCs to progesterone (P4)’s anti-apoptotic action, indicating that the Keywords: PGRMC1 monomer is functional. However, when endogenous PGRMC1 is depleted by siRNA treatment Membrane progesterone receptor and replaced with PGRMC1–GFP, P4 responsiveness is not enhanced, although overall levels of PGRMC1 Apoptosis Progesterone are increased. P4’s anti-apoptotic action is also attenuated by actinomycin D, an inhibitor of RNA synthe- Granulosa cell sis, and P4 activation of PGRMC1 suppresses Bad and increases Bcl2a1d expression.
    [Show full text]
  • PGRMC Proteins Are Coming of Age: a Special Issue on the Role of PGRMC1 and PGRMC2 in Metabolism and Cancer Biology
    cancers Editorial PGRMC Proteins Are Coming of Age: A Special Issue on the Role of PGRMC1 and PGRMC2 in Metabolism and Cancer Biology Michael A. Cahill 1,2,* and Hans Neubauer 3,* 1 School of Biomedical Sciences, Charles Sturt University, WaggaWagga, NSW 2678, Australia 2 ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, Canberra, ACT 2601, Australia 3 Department of Gynecology and Obstetrics, University Women’s Hospital of Dusseldorf, 40225 Duesseldorf, Germany * Correspondence: [email protected] (M.A.C.); [email protected] (H.N.) 1. PGRMC Research History This is a preface by the guest editors of the special issue of Cancers featuring the biology of progesterone (P4) receptor membrane component (PGRMC) proteins as it relates to metabolism and cancer. PGRMC proteins are members of the cytochrome b5-related membrane-associated progesterone receptor (MAPR) family, of which humans have four members: PGRMC1, PGRMC2, Neudesin and Neuferricin [1,2]. In the early 2000s, we were both part of a German Human Genome Project (Proteom- analyse von Brustkrebsgenen, 01 KW0102, 2001–2004) that focused on the development of highly sensitivity proteomics methods for the identification of proteins whose properties Citation: Cahill, M.A; Neubauer, H. were altered between clinical protein samples. Before the rise and supremacy of mass PGRMC Proteins Are Coming of Age: spectrometric proteomics methods, we developed an advanced gel electrophoresis and A Special Issue on the Role of highly sensitive/accurate differential detection method to examine cryoconserved breast PGRMC1 and PGRMC2 in cancer tissues, comparing estrogen receptor positive versus negative patient cohorts [3,4].
    [Show full text]