1
|
Luteijn RD, van Terwisga SR, Ver Eecke JE, Onia L, Zaver SA, Woodward JJ, Wubbolts RW, Raulet DH, van Kuppeveld FJM. The activation of the adaptor protein STING depends on its interactions with the phospholipid PI4P. Sci Signal 2024; 17:eade3643. [PMID: 38470955 PMCID: PMC11003704 DOI: 10.1126/scisignal.ade3643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 02/22/2024] [Indexed: 03/14/2024]
Abstract
Activation of the endoplasmic reticulum (ER)-resident adaptor protein STING, a component of a cytosolic DNA-sensing pathway, induces the transcription of genes encoding type I interferons (IFNs) and other proinflammatory factors. Because STING is activated at the Golgi apparatus, control of the localization and activation of STING is important in stimulating antiviral and antitumor immune responses. Through a genome-wide CRISPR interference screen, we found that STING activation required the Golgi-resident protein ACBD3, which promotes the generation of phosphatidylinositol 4-phosphate (PI4P) at the trans-Golgi network, as well as other PI4P-associated proteins. Appropriate localization and activation of STING at the Golgi apparatus required ACBD3 and the PI4P-generating kinase PI4KB. In contrast, STING activation was enhanced when the lipid-shuttling protein OSBP, which removes PI4P from the Golgi apparatus, was inhibited by the US Food and Drug Administration-approved antifungal itraconazole. The increase in the abundance of STING-activating phospholipids at the trans-Golgi network resulted in the increased production of IFN-β and other cytokines in THP-1 cells. Furthermore, a mutant STING that could not bind to PI4P failed to traffic from the ER to the Golgi apparatus in response to a STING agonist, whereas forced relocalization of STING to PI4P-enriched areas elicited STING activation in the absence of stimulation with a STING agonist. Thus, PI4P is critical for STING activation, and manipulating PI4P abundance may therapeutically modulate STING-dependent immune responses.
Collapse
Affiliation(s)
- Rutger D Luteijn
- Virology Section, Infectious Diseases and Immunology Division, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Sypke R van Terwisga
- Virology Section, Infectious Diseases and Immunology Division, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Jill E Ver Eecke
- Virology Section, Infectious Diseases and Immunology Division, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Liberty Onia
- Department of Molecular and Cell Biology, and Cancer Research Laboratory, Division of Immunology and Molecular Medicine, University of California, Berkeley, CA, USA
| | - Shivam A Zaver
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Joshua J Woodward
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Richard W Wubbolts
- Centre for Cell Imaging, Division of Cell Biology, Metabolism and Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - David H Raulet
- Department of Molecular and Cell Biology, and Cancer Research Laboratory, Division of Immunology and Molecular Medicine, University of California, Berkeley, CA, USA
| | - Frank J M van Kuppeveld
- Virology Section, Infectious Diseases and Immunology Division, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
2
|
Jia J, Tang S, Yue X, Jing S, Zhu L, Tan C, Gao J, Du Y, Lee I, Qian Y. An A-Kinase Anchoring Protein (ACBD3) Coordinates Traffic-Induced PKA Activation At The Golgi. J Biol Chem 2023; 299:104696. [PMID: 37044218 DOI: 10.1016/j.jbc.2023.104696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 03/23/2023] [Accepted: 03/25/2023] [Indexed: 04/14/2023] Open
Abstract
KDEL receptor (KDELR) is a key protein that recycles escaped ER resident proteins from the Golgi apparatus back to the ER and maintains a dynamic balance between these two organelles in the early secretory pathway. Studies have shown that this retrograde transport pathway is partly regulated by two KDELR-interacting proteins, Acyl-CoA-binding domain-containing 3 (ACBD3), and cyclic AMP-dependent protein kinase A (PKA). However, whether Golgi-localized ACBD3, which was first discovered as a PKA-anchoring protein in mitochondria, directly interacts with PKA at the Golgi and coordinates its signaling in Golgi-to-ER traffic has remained unclear. In this study, we showed that the GOLD domain of ACBD3 directly interacts with the regulatory subunit II (RII) of PKA and effectively recruits PKA holoenzyme to the Golgi. Forward trafficking of proteins from the ER triggers activation of PKA by releasing the catalytic subunit from RII. Furthermore, we determined that depletion of ACBD3 reduces the Golgi fraction of RII, resulting in moderate, but constitutive activation of PKA and KDELR retrograde transport, independent of cargo influx from the ER. Taken together, these data demonstrate that ACBD3 coordinates the protein secretory pathway at the Golgi by facilitating KDELR/PKA-containing protein complex formation.
Collapse
Affiliation(s)
- Jie Jia
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Shuocheng Tang
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Xihua Yue
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Shuaiyang Jing
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Lianhui Zhu
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Chuanting Tan
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jingkai Gao
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yulei Du
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Intaek Lee
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China.
| | - Yi Qian
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China.
| |
Collapse
|
3
|
Yue X, Qian Y, Zhu L, Gim B, Bao M, Jia J, Jing S, Wang Y, Tan C, Bottanelli F, Ziltener P, Choi S, Hao P, Lee I. ACBD3 modulates KDEL receptor interaction with PKA for its trafficking via tubulovesicular carrier. BMC Biol 2021; 19:194. [PMID: 34493279 PMCID: PMC8424950 DOI: 10.1186/s12915-021-01137-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 08/30/2021] [Indexed: 11/10/2022] Open
Abstract
Background KDEL receptor helps establish cellular equilibrium in the early secretory pathway by recycling leaked ER-chaperones to the ER during secretion of newly synthesized proteins. Studies have also shown that KDEL receptor may function as a signaling protein that orchestrates membrane flux through the secretory pathway. We have recently shown that KDEL receptor is also a cell surface receptor, which undergoes highly complex itinerary between trans-Golgi network and the plasma membranes via clathrin-mediated transport carriers. Ironically, however, it is still largely unknown how KDEL receptor is distributed to the Golgi at steady state, since its initial discovery in late 1980s. Results We used a proximity-based in vivo tagging strategy to further dissect mechanisms of KDEL receptor trafficking. Our new results reveal that ACBD3 may be a key protein that regulates KDEL receptor trafficking via modulation of Arf1-dependent tubule formation. We demonstrate that ACBD3 directly interact with KDEL receptor and form a functionally distinct protein complex in ArfGAPs-independent manner. Depletion of ACBD3 results in re-localization of KDEL receptor to the ER by inducing accelerated retrograde trafficking of KDEL receptor. Importantly, this is caused by specifically altering KDEL receptor interaction with Protein Kinase A and Arf1/ArfGAP1, eventually leading to increased Arf1-GTP-dependent tubular carrier formation at the Golgi. Conclusions These results suggest that ACBD3 may function as a negative regulator of PKA activity on KDEL receptor, thereby restricting its retrograde trafficking in the absence of KDEL ligand binding. Since ACBD3 was originally identified as PAP7, a PBR/PKA-interacting protein at the Golgi/mitochondria, we propose that Golgi-localization of KDEL receptor is likely to be controlled by its interaction with ACBD3/PKA complex at steady state, providing a novel insight for establishment of cellular homeostasis in the early secretory pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01137-7.
Collapse
Affiliation(s)
- Xihua Yue
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Yi Qian
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Lianhui Zhu
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Bopil Gim
- School of Physical Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Mengjing Bao
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Jie Jia
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Shuaiyang Jing
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yijing Wang
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Chuanting Tan
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Francesca Bottanelli
- Institut für Biochemie, Freie Universität Berlin, Thielallee 63, 14195, Berlin, Germany
| | - Pascal Ziltener
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
| | - Sunkyu Choi
- Proteomics Core, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Piliang Hao
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Intaek Lee
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China. .,Shanghai Institute for Advanced Immunochemical Studies, Shanghai, China.
| |
Collapse
|
4
|
Hiser C, Montgomery BL, Ferguson-Miller S. TSPO protein binding partners in bacteria, animals, and plants. J Bioenerg Biomembr 2021; 53:463-487. [PMID: 34191248 PMCID: PMC8243069 DOI: 10.1007/s10863-021-09905-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/12/2021] [Indexed: 12/11/2022]
Abstract
The ancient membrane protein TSPO is phylogenetically widespread from archaea and bacteria to insects, vertebrates, plants, and fungi. TSPO’s primary amino acid sequence is only modestly conserved between diverse species, although its five transmembrane helical structure appears mainly conserved. Its cellular location and orientation in membranes have been reported to vary between species and tissues, with implications for potential diverse binding partners and function. Most TSPO functions relate to stress-induced changes in metabolism, but in many cases it is unclear how TSPO itself functions—whether as a receptor, a sensor, a transporter, or a translocator. Much evidence suggests that TSPO acts indirectly by association with various protein binding partners or with endogenous or exogenous ligands. In this review, we focus on proteins that have most commonly been invoked as TSPO binding partners. We suggest that TSPO was originally a bacterial receptor/stress sensor associated with porphyrin binding as its most ancestral function and that it later developed additional stress-related roles in eukaryotes as its ability to bind new partners evolved.
Collapse
Affiliation(s)
- Carrie Hiser
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA. .,Department of Energy Plant Research Laboratory, Michigan State University, East Lansing, MI, 48824, USA.
| | - Beronda L Montgomery
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA.,Department of Energy Plant Research Laboratory, Michigan State University, East Lansing, MI, 48824, USA.,Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, 48824, USA
| | - Shelagh Ferguson-Miller
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA
| |
Collapse
|
5
|
Leydig cell aging: Molecular mechanisms and treatments. VITAMINS AND HORMONES 2021; 115:585-609. [PMID: 33706963 DOI: 10.1016/bs.vh.2020.12.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Late-onset hypogonadism, resulting from deficiency in serum testosterone (T), affects the health and quality of life of millions of aging men. T is synthesized by Leydig cells (LCs) in response to luteinizing hormone (LH). LH binds LC plasma membrane receptors, inducing the formation of a supramolecular complex of cytosolic and mitochondrial proteins, the Steroidogenic InteracTomE (SITE). SITE proteins are involved in targeting cholesterol to CYP11A1 in the mitochondria, the first enzyme of the steroidogenic cascade. Cholesterol translocation is the rate-determining step in T formation. With aging, LC defects occur that include changes in SITE, an increasingly oxidative intracellular environment, and reduced androgen formation and serum T levels. T replacement therapy (TRT) will restore T levels, but reported side effects make it desirable to develop additional strategies for increasing T. One approach is to target LC protein-protein interactions and thus increase T production by the hypofunctional Leydig cells themselves.
Collapse
|
6
|
Abstract
The field of cAMP signaling is witnessing exciting developments with the recognition that cAMP is compartmentalized and that spatial regulation of cAMP is critical for faithful signal coding. This realization has changed our understanding of cAMP signaling from a model in which cAMP connects a receptor at the plasma membrane to an intracellular effector in a linear pathway to a model in which cAMP signals propagate within a complex network of alternative branches and the specific functional outcome strictly depends on local regulation of cAMP levels and on selective activation of a limited number of branches within the network. In this review, we cover some of the early studies and summarize more recent evidence supporting the model of compartmentalized cAMP signaling, and we discuss how this knowledge is starting to provide original mechanistic insight into cell physiology and a novel framework for the identification of disease mechanisms that potentially opens new avenues for therapeutic interventions. SIGNIFICANCE STATEMENT: cAMP mediates the intracellular response to multiple hormones and neurotransmitters. Signal fidelity and accurate coordination of a plethora of different cellular functions is achieved via organization of multiprotein signalosomes and cAMP compartmentalization in subcellular nanodomains. Defining the organization and regulation of subcellular cAMP nanocompartments is necessary if we want to understand the complex functional ramifications of pharmacological treatments that target G protein-coupled receptors and for generating a blueprint that can be used to develop precision medicine interventions.
Collapse
Affiliation(s)
- Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Anna Zerio
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Miguel J Lobo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
7
|
Islinger M, Costello JL, Kors S, Soupene E, Levine TP, Kuypers FA, Schrader M. The diversity of ACBD proteins - From lipid binding to protein modulators and organelle tethers. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118675. [PMID: 32044385 PMCID: PMC7057175 DOI: 10.1016/j.bbamcr.2020.118675] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/30/2020] [Accepted: 02/05/2020] [Indexed: 12/12/2022]
Abstract
Members of the large multigene family of acyl-CoA binding domain containing proteins (ACBDs) share a conserved motif required for binding of Coenzyme A esterified fatty acids of various chain length. These proteins are present in the three kingdoms of life, and despite their predicted roles in cellular lipid metabolism, knowledge about the precise functions of many ACBD proteins remains scarce. Interestingly, several ACBD proteins are now suggested to function at organelle contact sites, and are recognized as host interaction proteins for different pathogens including viruses and bacteria. Here, we present a thorough phylogenetic analysis of the ACBD family and discuss their structure and evolution. We summarize recent findings on the various functions of animal and fungal ACBDs with particular focus on peroxisomes, the role of ACBD proteins at organelle membranes, and their increasing recognition as targets for pathogens.
Collapse
Affiliation(s)
- Markus Islinger
- Institute of Neuroanatomy, Medical Faculty Manheim, University of Heidelberg, 68167 Mannheim, Germany
| | - Joseph L Costello
- College of Life and Environmental Sciences, Biosciences, University of Exeter, Exeter EX4 4QD, Devon, UK
| | - Suzan Kors
- College of Life and Environmental Sciences, Biosciences, University of Exeter, Exeter EX4 4QD, Devon, UK
| | - Eric Soupene
- Children's Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | | | - Frans A Kuypers
- Children's Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | - Michael Schrader
- College of Life and Environmental Sciences, Biosciences, University of Exeter, Exeter EX4 4QD, Devon, UK.
| |
Collapse
|
8
|
Young PA, Senkal CE, Suchanek AL, Grevengoed TJ, Lin DD, Zhao L, Crunk AE, Klett EL, Füllekrug J, Obeid LM, Coleman RA. Long-chain acyl-CoA synthetase 1 interacts with key proteins that activate and direct fatty acids into niche hepatic pathways. J Biol Chem 2018; 293:16724-16740. [PMID: 30190326 PMCID: PMC6204890 DOI: 10.1074/jbc.ra118.004049] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/24/2018] [Indexed: 12/17/2022] Open
Abstract
Fatty acid channeling into oxidation or storage modes depends on physiological conditions and hormonal signaling. However, the directionality of this channeling may also depend on the association of each of the five acyl-CoA synthetase isoforms with specific protein partners. Long-chain acyl-CoA synthetases (ACSLs) catalyze the conversion of long-chain fatty acids to fatty acyl-CoAs, which are then either oxidized or used in esterification reactions. In highly oxidative tissues, ACSL1 is located on the outer mitochondrial membrane (OMM) and directs fatty acids into mitochondria for β-oxidation. In the liver, however, about 50% of ACSL1 is located on the endoplasmic reticulum (ER) where its metabolic function is unclear. Because hepatic fatty acid partitioning is likely to require the interaction of ACSL1 with other specific proteins, we used an unbiased protein interaction technique, BioID, to discover ACSL1-binding partners in hepatocytes. We targeted ACSL1 either to the ER or to the OMM of Hepa 1-6 cells as a fusion protein with the Escherichia coli biotin ligase, BirA*. Proteomic analysis identified 98 proteins that specifically interacted with ACSL1 at the ER, 55 at the OMM, and 43 common to both subcellular locations. We found subsets of peroxisomal and lipid droplet proteins, tethering proteins, and vesicle proteins, uncovering a dynamic role for ACSL1 in organelle and lipid droplet interactions. Proteins involved in lipid metabolism were also identified, including acyl-CoA-binding proteins and ceramide synthase isoforms 2 and 5. Our results provide fundamental and detailed insights into protein interaction networks that control fatty acid metabolism.
Collapse
Affiliation(s)
| | - Can E Senkal
- the Department of Medicine, Stony Brook University, Stony Brook, New York 11794, and
| | | | | | | | | | | | - Eric L Klett
- From the Departments of Nutrition and
- Medicine, University of North Carolina, Chapel Hill, North Carolina 27599
| | - Joachim Füllekrug
- the Molecular Cell Biology Laboratory, Internal Medicine IV, Heidelberg University Hospital, Otto-Meyerhof-Zentrum, University of Heidelberg, 69120 Heidelberg, Germany
| | - Lina M Obeid
- the Department of Medicine, Stony Brook University, Stony Brook, New York 11794, and
| | | |
Collapse
|
9
|
Gatliff J, East DA, Singh A, Alvarez MS, Frison M, Matic I, Ferraina C, Sampson N, Turkheimer F, Campanella M. A role for TSPO in mitochondrial Ca 2+ homeostasis and redox stress signaling. Cell Death Dis 2017; 8:e2896. [PMID: 28640253 PMCID: PMC5520880 DOI: 10.1038/cddis.2017.186] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/08/2017] [Accepted: 03/23/2017] [Indexed: 12/20/2022]
Abstract
The 18 kDa translocator protein TSPO localizes on the outer mitochondrial membrane (OMM). Systematically overexpressed at sites of neuroinflammation it is adopted as a biomarker of brain conditions. TSPO inhibits the autophagic removal of mitochondria by limiting PARK2-mediated mitochondrial ubiquitination via a peri-organelle accumulation of reactive oxygen species (ROS). Here we describe that TSPO deregulates mitochondrial Ca2+ signaling leading to a parallel increase in the cytosolic Ca2+ pools that activate the Ca2+-dependent NADPH oxidase (NOX) thereby increasing ROS. The inhibition of mitochondrial Ca2+ uptake by TSPO is a consequence of the phosphorylation of the voltage-dependent anion channel (VDAC1) by the protein kinase A (PKA), which is recruited to the mitochondria, in complex with the Acyl-CoA binding domain containing 3 (ACBD3). Notably, the neurotransmitter glutamate, which contributes neuronal toxicity in age-dependent conditions, triggers this TSPO-dependent mechanism of cell signaling leading to cellular demise. TSPO is therefore proposed as a novel OMM-based pathway to control intracellular Ca2+ dynamics and redox transients in neuronal cytotoxicity.
Collapse
Affiliation(s)
- Jemma Gatliff
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK
| | - Daniel A East
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK
- Regina Elena-National Cancer Institute, 00144 Rome, Italy
| | - Aarti Singh
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK
| | - Maria Soledad Alvarez
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK
| | - Michele Frison
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK
- Department of Neuroimaging, Institute of Psychiatry, King's College London, London, UK
| | - Ivana Matic
- Department of Biology, University of Rome ‘TorVergata’, 00133 Rome, Italy
| | - Caterina Ferraina
- Regina Elena-National Cancer Institute, 00144 Rome, Italy
- Department of Biology, University of Rome ‘TorVergata’, 00133 Rome, Italy
| | - Natalie Sampson
- Division of Experimental Urology, Medical University of Innsbruck, A6020 Innsbruck, Austria
| | - Federico Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, King's College London, London, UK
| | - Michelangelo Campanella
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, University of London, Royal College Street, London NW1 0TU, UK
- Department of Biology, University of Rome ‘TorVergata’, 00133 Rome, Italy
- University College London Consortium for Mitochondrial Research, Gower Street, WC1E 6BT London, UK
| |
Collapse
|
10
|
SCAP/SREBP pathway is required for the full steroidogenic response to cyclic AMP. Proc Natl Acad Sci U S A 2016; 113:E5685-93. [PMID: 27601673 DOI: 10.1073/pnas.1611424113] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Luteinizing hormone (LH) stimulates steroidogenesis largely through a surge in cyclic AMP (cAMP). Steroidogenic rates are also critically dependent on the availability of cholesterol at mitochondrial sites of synthesis. This cholesterol is provided by cellular uptake of lipoproteins, mobilization of intracellular lipid, and de novo synthesis. Whether and how these pathways are coordinated by cAMP are poorly understood. Recent phosphoproteomic analyses of cAMP-dependent phosphorylation sites in MA10 Leydig cells suggested that cAMP regulates multiple steps in these processes, including activation of the SCAP/SREBP pathway. SCAP [sterol-regulatory element-binding protein (SREBP) cleavage-activating protein] acts as a cholesterol sensor responsible for regulating intracellular cholesterol balance. Its role in cAMP-mediated control of steroidogenesis has not been explored. We used two CRISPR (clustered regularly interspaced short palindromic repeat)-Cas9 (CRISPR associated protein 9) knockout approaches to test the role of SCAP in steroidogenesis. Our results demonstrate that SCAP is required for progesterone production induced by concurrent inhibition of the cAMP phosphodiesterases PDE4 and PDE8. These inhibitors increased SCAP phosphorylation, SREBP2 activation, and subsequent expression of cholesterol biosynthetic genes, whereas SCAP deficiency largely prevented these effects. Reexpression of SCAP in SCAP-deficient cells restored SREBP2 protein expression and partially restored steroidogenic responses, confirming the requirement of SCAP-SREBP2 in steroidogenesis. Inhibitors of 3-hydroxy-3-methylglutaryl-Coenzyme A reductase and isoprenylation attenuated, whereas exogenously provided cholesterol augmented, PDE inhibitor-induced steroidogenesis, suggesting that the cholesterol substrate needed for steroidogenesis is provided by both de novo synthesis and isoprenylation-dependent mechanisms. Overall, these results demonstrate a novel role for LH/cAMP in SCAP/SREBP activation and subsequent regulation of steroidogenesis.
Collapse
|
11
|
Fan J, Li X, Issop L, Culty M, Papadopoulos V. ACBD2/ECI2-Mediated Peroxisome-Mitochondria Interactions in Leydig Cell Steroid Biosynthesis. Mol Endocrinol 2016; 30:763-82. [PMID: 27167610 DOI: 10.1210/me.2016-1008] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Fatty acid metabolism and steroid biosynthesis are 2 major pathways shared by peroxisomes and mitochondria. Both organelles are in close apposition to the endoplasmic reticulum, with which they communicate via interorganelle membrane contact sites to promote cellular signaling and the exchange of ions and lipids. To date, no convincing evidence of the direct contact between peroxisomes and mitochondria was reported in mammalian cells. Hormone-induced, tightly controlled steroid hormone biosynthesis requires interorganelle interactions. Using immunofluorescent staining and live-cell imaging, we found that dibutyryl-cAMP treatment of MA-10 mouse tumor Leydig cells rapidly induces peroxisomes to approach mitochondria and form peroxisome-mitochondrial contact sites/fusion, revealed by the subcellular distribution of the endogenous acyl-coenzyme A-binding domain (ACBD)2/ECI2 isoform A generated by alternative splicing, and further validated using a proximity ligation assay. This event occurs likely via a peroxisome-like structure, which is mediated by peroxisomal and mitochondrial matrix protein import complexes: peroxisomal import receptor peroxisomal biogenesis factor 5 (PEX5), and the mitochondrial import receptor subunit translocase of outer mitochondrial membrane 20 homolog (yeast) protein. Similar results were obtained using the mLTC-1 mouse tumor Leydig cells. Ectopic expression of the ACBD2/ECI2 isoform A in MA-10 cells led to increased basal and hormone-stimulated steroid formation, indicating that ACBD2/ECI2-mediated peroxisomes-mitochondria interactions favor in the exchange of metabolites and/or macromolecules between these 2 organelles in support of steroid biosynthesis. Considering the widespread occurrence of the ACBD2/ECI2 protein, we propose that this protein might serve as a tool to assist in understanding the contact between peroxisomes and mitochondria.
Collapse
Affiliation(s)
- Jinjiang Fan
- The Research Institute of the McGill University Health Centre (J.F., X.L., L.I., M.C., V.P.) and Departments of Medicine (J.F., L.I., M.C., V.P.), Biochemistry (X.L., V.P.), and Pharmacology and Therapeutics (M.C., V.P.), McGill University, Montréal, Québec, Canada H4A 3J1
| | - Xinlu Li
- The Research Institute of the McGill University Health Centre (J.F., X.L., L.I., M.C., V.P.) and Departments of Medicine (J.F., L.I., M.C., V.P.), Biochemistry (X.L., V.P.), and Pharmacology and Therapeutics (M.C., V.P.), McGill University, Montréal, Québec, Canada H4A 3J1
| | - Leeyah Issop
- The Research Institute of the McGill University Health Centre (J.F., X.L., L.I., M.C., V.P.) and Departments of Medicine (J.F., L.I., M.C., V.P.), Biochemistry (X.L., V.P.), and Pharmacology and Therapeutics (M.C., V.P.), McGill University, Montréal, Québec, Canada H4A 3J1
| | - Martine Culty
- The Research Institute of the McGill University Health Centre (J.F., X.L., L.I., M.C., V.P.) and Departments of Medicine (J.F., L.I., M.C., V.P.), Biochemistry (X.L., V.P.), and Pharmacology and Therapeutics (M.C., V.P.), McGill University, Montréal, Québec, Canada H4A 3J1
| | - Vassilios Papadopoulos
- The Research Institute of the McGill University Health Centre (J.F., X.L., L.I., M.C., V.P.) and Departments of Medicine (J.F., L.I., M.C., V.P.), Biochemistry (X.L., V.P.), and Pharmacology and Therapeutics (M.C., V.P.), McGill University, Montréal, Québec, Canada H4A 3J1
| |
Collapse
|
12
|
Manna PR, Stetson CL, Slominski AT, Pruitt K. Role of the steroidogenic acute regulatory protein in health and disease. Endocrine 2016; 51:7-21. [PMID: 26271515 PMCID: PMC4707056 DOI: 10.1007/s12020-015-0715-6] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 08/03/2015] [Indexed: 01/10/2023]
Abstract
Steroid hormones are an important class of regulatory molecules that are synthesized in steroidogenic cells of the adrenal, ovary, testis, placenta, brain, and skin, and influence a spectrum of developmental and physiological processes. The steroidogenic acute regulatory protein (STAR) predominantly mediates the rate-limiting step in steroid biosynthesis, i.e., the transport of the substrate of all steroid hormones, cholesterol, from the outer to the inner mitochondrial membrane. At the inner membrane, cytochrome P450 cholesterol side chain cleavage enzyme cleaves the cholesterol side chain to form the first steroid, pregnenolone, which is converted by a series of enzymes to various steroid hormones in specific tissues. Both basic and clinical evidence have demonstrated the crucial involvement of the STAR protein in the regulation of steroid biosynthesis. Multiple levels of regulation impinge on STAR action. Recent findings demonstrate that hormone-sensitive lipase, through its action on the hydrolysis of cholesteryl esters, plays an important role in regulating STAR expression and steroidogenesis which involve the liver X receptor pathway. Activation of the latter influences macrophage cholesterol efflux that is a key process in the prevention of atherosclerotic cardiovascular disease. Appropriate regulation of steroid hormones is vital for proper functioning of many important biological activities, which are also paramount for geriatric populations to live longer and healthier. This review summarizes the current level of understanding on tissue-specific and hormone-induced regulation of STAR expression and steroidogenesis, and provides insights into a number of cholesterol and/or steroid coupled physiological and pathophysiological consequences.
Collapse
Affiliation(s)
- Pulak R Manna
- Department of Immunology and Molecular Microbiology, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA.
| | - Cloyce L Stetson
- Department of Dermatology, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Andrzej T Slominski
- Department of Dermatology, VA Medical Center, University of Alabama Birmingham, Birmingham, AL, 35294, USA
| | - Kevin Pruitt
- Department of Immunology and Molecular Microbiology, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| |
Collapse
|
13
|
Repalli J. Translocator protein (TSPO) role in aging and Alzheimer's disease. Curr Aging Sci 2015; 7:168-75. [PMID: 25495567 PMCID: PMC4435228 DOI: 10.2174/1874609808666141210103146] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 10/31/2014] [Accepted: 12/09/2014] [Indexed: 01/22/2023]
Abstract
Cellular damage and deregulated apoptotic cell death lead to functional impairment, and a main
consequence of these events is aging. Cellular damage is initiated by different stress/risk factors such as
oxidative stress, inflammation, and heavy metals. These stress/risk factors affect the cellular homeostasis
by altering methylation status of several aging and Alzheimer’s disease associated genes; these effects can
be manifested immediately after exposure to stress and at later stages of life. However, when cellular damage
exceeds certain threshold levels apoptosis is initiated. This review discusses the stress factors involved
in cellular damage and the role and potential of TSPO-mediated cell death in aging as well as in Alzheimer’s disease,
which is also characterized by extensive cell death. Mitochondrial-mediated apoptotic death through the release of cytochrome
c is regulated by TSPO, and increased expression of this protein is observed in both elderly people and in patients
with Alzheimer’s disease. TSPO forms and mediates opening of the mitochondrial membrane pore, mPTP and oxidizes
cardiolipin, and these events lead to the leakage of apoptotic death mediators, such as cytochrome c, resulting in cell
death. However, TSPO has many proposed functions and can also increase steroid synthesis, which leads to inhibition of
inflammation and inhibition of the release of apoptotic factors, thereby decreasing cell damage and promoting cell survival.
Thus, TSPO mediates apoptosis and decreases the cell damage, which in turn dictates the process of aging as well
as the functionality of organs such as the brain. TSPO modulation with ligands in the Alzheimer’s disease mouse model
showed improvement in behavioral symptoms, and studies in Drosophila species showed increased cell survival and prolonged
lifespan in flies after TSPO inhibition. These data suggest that since effects/signs of stress can manifest at any
time, prevention through change in lifestyle and TSPO modulation could be potential strategies for altering both the aging
process and the progression of Alzheimer’s disease.
Collapse
Affiliation(s)
- Jayanthi Repalli
- NYU Langone Medical Center,180 Varick Street, Room 802, New York, NY, 10014, USA.
| |
Collapse
|
14
|
Chen Y, Mathias L, Falero-Perez JM, Kim SF. PKA-mediated phosphorylation of Dexras1 suppresses iron trafficking by inhibiting S-nitrosylation. FEBS Lett 2015; 589:3212-9. [PMID: 26358293 DOI: 10.1016/j.febslet.2015.08.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 08/21/2015] [Accepted: 08/26/2015] [Indexed: 01/26/2023]
Abstract
Dexras1 is a small GTPase and plays a central role in neuronal iron trafficking. We have shown that stimulation of glutamate receptors activates neuronal nitric oxide synthase, leading to S-nitrosylation of Dexras1 and a physiological increase in iron uptake. Here we report that Dexras1 is phosphorylated by protein kinase A (PKA) on serine 253, leading to a suppression of iron influx. These effects were directly associated with the levels of S-nitrosylated Dexras1, whereby PKA activation reduced Dexras1 S-nitrosylation in a dose dependent manner. Moreover, we found that adiponectin modulates Dexras1 via PKA. Hence these findings suggest the involvement of the PKA pathway in modulating glutamate-mediated ROS in neurons, and hint to a functional crosstalk between S-nitrosylation and phosphorylation.
Collapse
Affiliation(s)
- Yong Chen
- Department of Psychiatry, and Systems Pharmacology and Translational Therapeutics, Center for Neurobiology and Behavior, The Perlman School of Medicine at the University of Pennsylvania, 125 S 31st St. TRL Rm 2207, Philadelphia, PA 19104, United States
| | - Lauren Mathias
- Department of Psychiatry, and Systems Pharmacology and Translational Therapeutics, Center for Neurobiology and Behavior, The Perlman School of Medicine at the University of Pennsylvania, 125 S 31st St. TRL Rm 2207, Philadelphia, PA 19104, United States
| | - Juliana M Falero-Perez
- Department of Psychiatry, and Systems Pharmacology and Translational Therapeutics, Center for Neurobiology and Behavior, The Perlman School of Medicine at the University of Pennsylvania, 125 S 31st St. TRL Rm 2207, Philadelphia, PA 19104, United States
| | - Sangwon F Kim
- Department of Psychiatry, and Systems Pharmacology and Translational Therapeutics, Center for Neurobiology and Behavior, The Perlman School of Medicine at the University of Pennsylvania, 125 S 31st St. TRL Rm 2207, Philadelphia, PA 19104, United States.
| |
Collapse
|
15
|
Börnigen D, Moon YS, Rahnavard G, Waldron L, McIver L, Shafquat A, Franzosa EA, Miropolsky L, Sweeney C, Morgan XC, Garrett WS, Huttenhower C. A reproducible approach to high-throughput biological data acquisition and integration. PeerJ 2015; 3:e791. [PMID: 26157642 PMCID: PMC4493686 DOI: 10.7717/peerj.791] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 02/04/2015] [Indexed: 12/25/2022] Open
Abstract
Modern biological research requires rapid, complex, and reproducible integration of multiple experimental results generated both internally and externally (e.g., from public repositories). Although large systematic meta-analyses are among the most effective approaches both for clinical biomarker discovery and for computational inference of biomolecular mechanisms, identifying, acquiring, and integrating relevant experimental results from multiple sources for a given study can be time-consuming and error-prone. To enable efficient and reproducible integration of diverse experimental results, we developed a novel approach for standardized acquisition and analysis of high-throughput and heterogeneous biological data. This allowed, first, novel biomolecular network reconstruction in human prostate cancer, which correctly recovered and extended the NFκB signaling pathway. Next, we investigated host-microbiome interactions. In less than an hour of analysis time, the system retrieved data and integrated six germ-free murine intestinal gene expression datasets to identify the genes most influenced by the gut microbiota, which comprised a set of immune-response and carbohydrate metabolism processes. Finally, we constructed integrated functional interaction networks to compare connectivity of peptide secretion pathways in the model organisms Escherichia coli, Bacillus subtilis, and Pseudomonas aeruginosa.
Collapse
Affiliation(s)
- Daniela Börnigen
- Biostatistics Department, Harvard School of Public Health, Boston, MA, USA.,The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yo Sup Moon
- Biostatistics Department, Harvard School of Public Health, Boston, MA, USA
| | - Gholamali Rahnavard
- Biostatistics Department, Harvard School of Public Health, Boston, MA, USA.,The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Levi Waldron
- Biostatistics Department, Harvard School of Public Health, Boston, MA, USA.,City University of New York School of Public Health, Hunter College, New York, NY, USA
| | - Lauren McIver
- Biostatistics Department, Harvard School of Public Health, Boston, MA, USA
| | - Afrah Shafquat
- Biostatistics Department, Harvard School of Public Health, Boston, MA, USA
| | - Eric A Franzosa
- Biostatistics Department, Harvard School of Public Health, Boston, MA, USA.,The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Larissa Miropolsky
- Biostatistics Department, Harvard School of Public Health, Boston, MA, USA
| | | | - Xochitl C Morgan
- Biostatistics Department, Harvard School of Public Health, Boston, MA, USA.,The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Wendy S Garrett
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Curtis Huttenhower
- Biostatistics Department, Harvard School of Public Health, Boston, MA, USA.,The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
16
|
Aghazadeh Y, Zirkin BR, Papadopoulos V. Pharmacological regulation of the cholesterol transport machinery in steroidogenic cells of the testis. VITAMINS AND HORMONES 2015; 98:189-227. [PMID: 25817870 DOI: 10.1016/bs.vh.2014.12.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Reduced serum testosterone (T), or hypogonadism, is estimated to affect about 5 million American men, including both aging and young men. Low serum T has been linked to mood changes, worsening cognition, fatigue, depression, decreased lean body mass and bone mineral density, increased visceral fat, metabolic syndrome, decreased libido, and sexual dysfunction. Administering exogenous T, known as T-replacement therapy (TRT), reverses many of the symptoms of low T levels. However, this treatment can result in luteinizing hormone suppression which, in turn, can lead to reduced sperm numbers and infertility, making TRT inappropriate for men who wish to father children. Additionally, TRT may result in supraphysiologic T levels, skin irritation, and T transfer to others upon contact; and there may be increased risk of prostate cancer and cardiovascular disease, particularly in aging men. Therefore, the development of alternate therapies for treating hypogonadism would be highly desirable. To do so requires greater understanding of the series of steps leading to T formation and how they are regulated, and the identification of key steps that are amenable to pharmacological modulation so as to induce T production. We review herein our current understanding of mechanisms underlying the pharmacological induction of T formation in hypogonadal testis.
Collapse
Affiliation(s)
- Yasaman Aghazadeh
- The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Barry R Zirkin
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Vassilios Papadopoulos
- The Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; Department of Medicine, McGill University, Montreal, Quebec, Canada; Department of Biochemistry, McGill University, Montreal, Quebec, Canada; Department of Pharmacology & Therapeutics, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
17
|
Abstract
The purpose of this article is to review fundamentals in adrenal gland histophysiology. Key findings regarding the important signaling pathways involved in the regulation of steroidogenesis and adrenal growth are summarized. We illustrate how adrenal gland morphology and function are deeply interconnected in which novel signaling pathways (Wnt, Sonic hedgehog, Notch, β-catenin) or ionic channels are required for their integrity. Emphasis is given to exploring the mechanisms and challenges underlying the regulation of proliferation, growth, and functionality. Also addressed is the fact that while it is now well-accepted that steroidogenesis results from an enzymatic shuttle between mitochondria and endoplasmic reticulum, key questions still remain on the various aspects related to cellular uptake and delivery of free cholesterol. The significant progress achieved over the past decade regarding the precise molecular mechanisms by which the two main regulators of adrenal cortex, adrenocorticotropin hormone (ACTH) and angiotensin II act on their receptors is reviewed, including structure-activity relationships and their potential applications. Particular attention has been given to crucial second messengers and how various kinases, phosphatases, and cytoskeleton-associated proteins interact to ensure homeostasis and/or meet physiological demands. References to animal studies are also made in an attempt to unravel associated clinical conditions. Many of the aspects addressed in this article still represent a challenge for future studies, their outcome aimed at providing evidence that the adrenal gland, through its steroid hormones, occupies a central position in many situations where homeostasis is disrupted, thus highlighting the relevance of exploring and understanding how this key organ is regulated. © 2014 American Physiological Society. Compr Physiol 4:889-964, 2014.
Collapse
Affiliation(s)
- Nicole Gallo-Payet
- Division of Endocrinology, Department of Medicine, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, and Centre de Recherche Clinique Étienne-Le Bel of the Centre Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, Quebec, Canada
| | | |
Collapse
|
18
|
Mitochondrial cholesterol: mechanisms of import and effects on mitochondrial function. J Bioenerg Biomembr 2014; 48:137-51. [PMID: 25425472 DOI: 10.1007/s10863-014-9592-6] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 11/14/2014] [Indexed: 12/23/2022]
Abstract
Mitochondria require cholesterol for biogenesis and membrane maintenance, and for the synthesis of steroids, oxysterols and hepatic bile acids. Multiple pathways mediate the transport of cholesterol from different subcellular pools to mitochondria. In steroidogenic cells, the steroidogenic acute regulatory protein (StAR) interacts with a mitochondrial protein complex to mediate cholesterol delivery to the inner mitochondrial membrane for conversion to pregnenolone. In non-steroidogenic cells, several members of a protein family defined by the presence of a StAR-related lipid transfer (START) domain play key roles in the delivery of cholesterol to mitochondrial membranes. Subdomains of the endoplasmic reticulum (ER), termed mitochondria-associated ER membranes (MAM), form membrane contact sites with mitochondria and may contribute to the transport of ER cholesterol to mitochondria, either independently or in conjunction with lipid-transfer proteins. Model systems of mitochondria enriched with cholesterol in vitro and mitochondria isolated from cells with (patho)physiological mitochondrial cholesterol accumulation clearly demonstrate that mitochondrial cholesterol levels affect mitochondrial function. Increased mitochondrial cholesterol levels have been observed in several diseases, including cancer, ischemia, steatohepatitis and neurodegenerative diseases, and influence disease pathology. Hence, a deeper understanding of the mechanisms maintaining mitochondrial cholesterol homeostasis may reveal additional targets for therapeutic intervention. Here we give a brief overview of mitochondrial cholesterol import in steroidogenic cells, and then focus on cholesterol trafficking pathways that deliver cholesterol to mitochondrial membranes in non-steroidogenic cells. We also briefly discuss the consequences of increased mitochondrial cholesterol levels on mitochondrial function and their potential role in disease pathology.
Collapse
|
19
|
Liu G, Middleton RJ, Hatty CR, Kam WW, Chan R, Pham T, Harrison‐Brown M, Dodson E, Veale K, Banati RB. The 18 kDa translocator protein, microglia and neuroinflammation. Brain Pathol 2014; 24:631-53. [PMID: 25345894 PMCID: PMC8029074 DOI: 10.1111/bpa.12196] [Citation(s) in RCA: 166] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 08/19/2014] [Indexed: 12/17/2022] Open
Abstract
The 18 kDa translocator protein (TSPO), previously known as the peripheral benzodiazepine receptor, is expressed in the injured brain. It has become known as an imaging marker of "neuroinflammation" indicating active disease, and is best interpreted as a nondiagnostic biomarker and disease staging tool that refers to histopathology rather than disease etiology. The therapeutic potential of TSPO as a drug target is mostly based on the understanding that it is an outer mitochondrial membrane protein required for the translocation of cholesterol, which thus regulates the rate of steroid synthesis. This pivotal role together with the evolutionary conservation of TSPO has underpinned the belief that any loss or mutation of TSPO should be associated with significant physiological deficits or be outright incompatible with life. However, against prediction, full Tspo knockout mice are viable and across their lifespan do not show the phenotype expected if cholesterol transport and steroid synthesis were significantly impaired. Thus, the "translocation" function of TSPO remains to be better substantiated. Here, we discuss the literature before and after the introduction of the new nomenclature for TSPO and review some of the newer findings. In light of the controversy surrounding the function of TSPO, we emphasize the continued importance of identifying compounds with confirmed selectivity and suggest that TSPO expression is analyzed within specific disease contexts rather than merely equated with the reified concept of "neuroinflammation."
Collapse
Affiliation(s)
- Guo‐Jun Liu
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
- Brain & Mind Research InstituteThe University of SydneyNSWAustralia
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
| | - Ryan J. Middleton
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
| | - Claire R. Hatty
- Brain & Mind Research InstituteThe University of SydneyNSWAustralia
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
| | - Winnie Wai‐Ying Kam
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
- Brain & Mind Research InstituteThe University of SydneyNSWAustralia
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
| | - Ronald Chan
- Brain & Mind Research InstituteThe University of SydneyNSWAustralia
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
| | - Tien Pham
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
| | - Meredith Harrison‐Brown
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
| | - Eoin Dodson
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
| | - Kelly Veale
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
| | - Richard B. Banati
- Life SciencesAustralian Nuclear Science and Technology OrganisationNSWAustralia
- Brain & Mind Research InstituteThe University of SydneyNSWAustralia
- Discipline of Medical Imaging & Radiation SciencesFaculty of Health SciencesThe University of SydneyNSWAustralia
- National Imaging Facility and Ramaciotti Brain Imaging CentreSydneyNSWAustralia
| |
Collapse
|
20
|
Azarashvili T, Baburina Y, Grachev D, Krestinina O, Papadopoulos V, Lemasters JJ, Odinokova I, Reiser G. Carbenoxolone induces permeability transition pore opening in rat mitochondria via the translocator protein TSPO and connexin43. Arch Biochem Biophys 2014; 558:87-94. [PMID: 24995971 DOI: 10.1016/j.abb.2014.06.027] [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: 04/08/2014] [Revised: 06/18/2014] [Accepted: 06/23/2014] [Indexed: 01/09/2023]
Abstract
Ca(2+)-induced permeability transition pore (mPTP) opening in isolated rat brain mitochondria is promoted through targeting of connexin43. After a threshold Ca(2+) load, mitochondrial membrane potential drops and efflux of accumulated Ca(2+) from the mitochondrial matrix occurs, indicating the mPTP opening. Specific antibodies were used to assess the role of the translocator protein (18kDa; TSPO) and connexin43 in swelling of isolated rat liver and brain mitochondria induced by carbenoxolone and the endogenous TSPO ligand protoporphyrin IX. Mitochondrial membrane potential, Ca(2+) transport and oxygen consumption were determined using selective electrodes. All the parameters were detected simultaneously in a chamber with the selective electrodes. The phosphorylation state of mitochondrial protein targets was assessed. We report that Ca(2+)-induced mitochondrial swelling was strengthened in the presence of both carbenoxolone and protoporphyrin IX. The carbenoxolone- and protoporphyrin IX-accelerated mPTP induction in brain mitochondria was completely prevented by antibodies specific for the mitochondrial translocator protein (TSPO). The anti-TSPO antibodies were more effective than anti-сonnexin43 antibodies. Moreover, carbenoxolone-stimulated phosphorylation of mitochondrial proteins was inhibited by anti-TSPO antibodies. Taken together, the data suggests that, in addition to acting via connexion43, carbenoxolone may exert its effect on mPTP via mitochondrial outer membrane TSPO.
Collapse
Affiliation(s)
- Tamara Azarashvili
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya Str., Pushchino, Moscow Region 142290, Russia; Institut für Neurobiochemie, Otto-von-Guericke-Universität Magdeburg, Medizinische Fakultät, Leipziger Str. 44, 39120 Magdeburg, Germany.
| | - Yulia Baburina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya Str., Pushchino, Moscow Region 142290, Russia.
| | - Dmitry Grachev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya Str., Pushchino, Moscow Region 142290, Russia.
| | - Olga Krestinina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya Str., Pushchino, Moscow Region 142290, Russia.
| | - Vassilios Papadopoulos
- The Research Institute of the McGill University Health Center, 2155 Guy Street, Suite 500, Montreal, Quebec H3H 2R9, Canada.
| | - John J Lemasters
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya Str., Pushchino, Moscow Region 142290, Russia; Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, DD504 Drug Discovery Bldg., 70 President St., MSC 140, Charleston, SC 29425, USA; Department of Biochemistry & Molecular Biology, Medical University of South Carolina, DD504 Drug Discovery Bldg., 70 President St., MSC 140, Charleston, SC 29425, USA.
| | - Irina Odinokova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya Str., Pushchino, Moscow Region 142290, Russia.
| | - Georg Reiser
- Institut für Neurobiochemie, Otto-von-Guericke-Universität Magdeburg, Medizinische Fakultät, Leipziger Str. 44, 39120 Magdeburg, Germany.
| |
Collapse
|
21
|
Di Benedetto G, Scalzotto E, Mongillo M, Pozzan T. Mitochondrial Ca²⁺ uptake induces cyclic AMP generation in the matrix and modulates organelle ATP levels. Cell Metab 2013; 17:965-975. [PMID: 23747252 DOI: 10.1016/j.cmet.2013.05.003] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 03/08/2013] [Accepted: 04/08/2013] [Indexed: 01/09/2023]
Abstract
While the role of mitochondrial Ca²⁺ homeostasis in cell pathophysiology is widely accepted, the possibility that cAMP regulates mitochondrial functions has only recently received experimental support. The site of cAMP production, its targets, and its functions in the organelles remain uncertain. Using a variety of genetic/pharmacological tools, we here demonstrate that the mitochondrial inner membrane is impermeable to cytosolic cAMP, while an autonomous cAMP signaling toolkit is expressed in the matrix. We demonstrate that rises in matrix Ca²⁺ powerfully stimulate cAMP increases within mitochondria and that matrix cAMP levels regulate their ATP synthesizing efficiency. In cardiomyocyte cultures, mitochondrial cAMP can be increased by treatments that augment the frequency and amplitude of Ca²⁺ oscillations within the cytosol and organelles, revealing that mitochondria can integrate an oscillatory Ca²⁺ signal to increase cAMP in their matrix. The present data reveal the existence, within mitochondria, of a hitherto unknown crosstalk between Ca²⁺ and cAMP.
Collapse
Affiliation(s)
- Giulietta Di Benedetto
- Institute of Neuroscience, Italian National Research Council (CNR), Viale G. Colombo 3, 35121 Padova, Italy; Venetian Institute of Molecular Medicine (VIMM), Via Orus 2, 35129 Padova, Italy.
| | - Elisa Scalzotto
- Venetian Institute of Molecular Medicine (VIMM), Via Orus 2, 35129 Padova, Italy
| | - Marco Mongillo
- Institute of Neuroscience, Italian National Research Council (CNR), Viale G. Colombo 3, 35121 Padova, Italy; Venetian Institute of Molecular Medicine (VIMM), Via Orus 2, 35129 Padova, Italy; Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121 Padova, Italy
| | - Tullio Pozzan
- Institute of Neuroscience, Italian National Research Council (CNR), Viale G. Colombo 3, 35121 Padova, Italy; Venetian Institute of Molecular Medicine (VIMM), Via Orus 2, 35129 Padova, Italy; Department of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121 Padova, Italy
| |
Collapse
|
22
|
Cooke M, Di Cónsoli H, Maloberti P, Cornejo Maciel F. Expression and function of OXE receptor, an eicosanoid receptor, in steroidogenic cells. Mol Cell Endocrinol 2013; 371:71-8. [PMID: 23159987 DOI: 10.1016/j.mce.2012.11.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Revised: 11/05/2012] [Accepted: 11/06/2012] [Indexed: 10/27/2022]
Abstract
Hormonal regulation of steroidogenesis involves arachidonic acid (AA) metabolism through the 5-lipoxygenase pathway. One of the products, 5-hydroperoxy-eicosatetraenoic acid (5-HpETE), acts as a modulator of the activity of the steroidogenic acute regulatory (StAR) protein promoter. Besides, an oxoeicosanoid receptor of the leukotriene receptor family named OXE-R is a membrane protein with high affinity and response to 5-HpETE, among other AA derivatives. The aim of our work was to elucidate whether this receptor may be involved in steroidogenesis. RT-PCR and western blot analysis demonstrated the presence of the mRNA and protein of the receptor in human H295R adrenocortical cells. The treatment of H295R or MA-10 cells (murine Leydig cell line) with 8Br-cAMP together with docosahexaenoic acid (DHA, an antagonist of the receptor) partially reduced StAR induction and steroidogenesis. On the contrary, 5-oxo-ETE - the prototypical agonist, with higher affinity and potency on the receptor - increased cAMP-dependent steroid production, StAR mRNA and protein levels. These results lead us to conclude that AA might modulate StAR induction and steroidogenesis, at least in part, through 5-HpETE production and activation of a membrane receptor, such as the OXE-R.
Collapse
Affiliation(s)
- Mariana Cooke
- INBIOMED - UBA/CONICET, Department of Biochemistry, School of Medicine, University of Buenos Aires, Paraguay 2155, C1121ABG Buenos Aires, Argentina
| | | | | | | |
Collapse
|
23
|
Abstract
Adrenal gonadal, placental and brain mitochondria contain several steroidogenic enzymes, notably the cholesterol side chain cleavage enzyme, P450scc, which is the enzymatic rate-limiting step in steroidogenesis which determines cellular steroidogenic capacity. Even before this step, the access of cholesterol to this enzyme system is both rate-limiting and the site of acute regulation via the steroidogenic acute regulatory protein (StAR) which interacts with a complex multi-component 'transduceosome' on the outer mitochondrial membrane (OMM). The components of the transduceosome include the 18 kDa translocator protein (TSPO), the voltage-dependent anion channel (VDAC-1), TSPO-associated protein 7 (PAP7, ACBD3 for acyl-CoA-binding-domain 3), and protein kinase A regulatory subunit 1α (PKAR1A). The precise fashion in which these proteins interact and move cholesterol from the OMM to P450scc, and the means by which cholesterol is loaded into the OMM, remain unclear. Human deficiency diseases have been described for StAR and for P450scc. Mitochondria also contain several 'downstream' steroidogenic enzymes.
Collapse
Affiliation(s)
- Vassilios Papadopoulos
- The Research Institute of the McGill University Health Centre, Department of Medicine, McGill University, Montreal, Quebec H3G 1A4, Canada.
| | | |
Collapse
|
24
|
Chen Y, Patel V, Bang S, Cohen N, Millar J, Kim SF. Maturation and activity of sterol regulatory element binding protein 1 is inhibited by acyl-CoA binding domain containing 3. PLoS One 2012; 7:e49906. [PMID: 23166793 PMCID: PMC3498211 DOI: 10.1371/journal.pone.0049906] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 10/15/2012] [Indexed: 12/23/2022] Open
Abstract
Imbalance of lipid metabolism has been linked with pathogenesis of a variety of human pathological conditions such as diabetes, obesity, cancer and neurodegeneration. Sterol regulatory element binding proteins (SREBPs) are the master transcription factors controlling the homeostasis of fatty acids and cholesterol in the body. Transcription, expression, and activity of SREBPs are regulated by various nutritional, hormonal or stressful stimuli, yet the molecular and cellular mechanisms involved in these adaptative responses remains elusive. In the present study, we found that overexpressed acyl-CoA binding domain containing 3 (ACBD3), a Golgi-associated protein, dramatically inhibited SREBP1-sensitive promoter activity of fatty acid synthase (FASN). Moreover, lipid deprivation-stimulated SREBP1 maturation was significantly attenuated by ACBD3. With cell fractionation, gene knockdown and immunoprecipitation assays, it was showed that ACBD3 blocked intracellular maturation of SREBP1 probably through directly binding with the lipid regulator rather than disrupted SREBP1-SCAP-Insig1 interaction. Further investigation revealed that acyl-CoA domain-containing N-terminal sequence of ACBD3 contributed to its inhibitory effects on the production of nuclear SREBP1. In addition, mRNA and protein levels of FASN and de novo palmitate biosynthesis were remarkably reduced in cells overexpressed with ACBD3. These findings suggest that ACBD3 plays an essential role in maintaining lipid homeostasis via regulating SREBP1's processing pathway and thus impacting cellular lipogenesis.
Collapse
Affiliation(s)
- Yong Chen
- Department of Psychiatry, Center for Neurobiology and Behavior, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Vishala Patel
- Metabolic Tracer Resource, Institute of Diabetes, Obesity & Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Sookhee Bang
- Department of Psychiatry, Center for Neurobiology and Behavior, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Natalie Cohen
- Department of Psychiatry, Center for Neurobiology and Behavior, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - John Millar
- Metabolic Tracer Resource, Institute of Diabetes, Obesity & Metabolism, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Sangwon F. Kim
- Department of Psychiatry, Center for Neurobiology and Behavior, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
25
|
Mishra J, Gautam M, Dadhich R, Kowtharapu BS, Majumdar SS. Peritubular cells may modulate Leydig cell–mediated testosterone production through a nonclassic pathway. Fertil Steril 2012; 98:1308-17.e1. [DOI: 10.1016/j.fertnstert.2012.07.1124] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Revised: 07/18/2012] [Accepted: 07/19/2012] [Indexed: 10/28/2022]
|
26
|
Aghazadeh Y, Rone MB, Blonder J, Ye X, Veenstra TD, Hales DB, Culty M, Papadopoulos V. Hormone-induced 14-3-3γ adaptor protein regulates steroidogenic acute regulatory protein activity and steroid biosynthesis in MA-10 Leydig cells. J Biol Chem 2012; 287:15380-94. [PMID: 22427666 DOI: 10.1074/jbc.m112.339580] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cholesterol is the sole precursor of steroid hormones in the body. The import of cholesterol to the inner mitochondrial membrane, the rate-limiting step in steroid biosynthesis, relies on the formation of a protein complex that assembles at the outer mitochondrial membrane called the transduceosome. The transduceosome contains several mitochondrial and cytosolic components, including the steroidogenic acute regulatory protein (STAR). Human chorionic gonadotropin (hCG) induces de novo synthesis of STAR, a process shown to parallel maximal steroid production. In the hCG-dependent steroidogenic MA-10 mouse Leydig cell line, the 14-3-3γ protein was identified in native mitochondrial complexes by mass spectrometry and immunoblotting, and its levels increased in response to hCG treatment. The 14-3-3 proteins bind and regulate the activity of many proteins, acting via target protein activation, modification and localization. In MA-10 cells, cAMP induces 14-3-3γ expression parallel to STAR expression. Silencing of 14-3-3γ expression potentiates hormone-induced steroidogenesis. Binding motifs of 14-3-3γ were identified in components of the transduceosome, including STAR. Immunoprecipitation studies demonstrate a hormone-dependent interaction between 14-3-3γ and STAR that coincides with reduced 14-3-3γ homodimerization. The binding site of 14-3-3γ on STAR was identified to be Ser-194 in the STAR-related sterol binding lipid transfer (START) domain, the site phosphorylated in response to hCG. Taken together, these results demonstrate that 14-3-3γ negatively regulates steroidogenesis by binding to Ser-194 of STAR, thus keeping STAR in an unfolded state, unable to induce maximal steroidogenesis. Over time 14-3-3γ homodimerizes and dissociates from STAR, allowing this protein to induce maximal mitochondrial steroid formation.
Collapse
Affiliation(s)
- Yasaman Aghazadeh
- The Research Institute of the McGill University Health Centre and the Department of Medicine, McGill University, Montreal, Quebec H3G 1A4, Canada
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Lucki NC, Bandyopadhyay S, Wang E, Merrill AH, Sewer MB. Acid ceramidase (ASAH1) is a global regulator of steroidogenic capacity and adrenocortical gene expression. Mol Endocrinol 2012; 26:228-43. [PMID: 22261821 PMCID: PMC3275158 DOI: 10.1210/me.2011-1150] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 12/14/2011] [Indexed: 11/19/2022] Open
Abstract
In H295R human adrenocortical cells, ACTH rapidly activates ceramide (Cer) and sphingosine (SPH) turnover with a concomitant increase in SPH-1-phosphate secretion. These bioactive lipids modulate adrenocortical steroidogenesis, primarily by acting as second messengers in the protein kinase A/cAMP-dependent pathway. Acid ceramidase (ASAH1) directly regulates the intracellular balance of Cer, SPH, and SPH-1-phosphate by catalyzing the hydrolysis of Cer into SPH. ACTH/cAMP signaling stimulates ASAH1 transcription and activity, supporting a role for this enzyme in glucocorticoid production. Here, the role of ASAH1 in regulating steroidogenic capacity was examined using a tetracycline-inducible ASAH1 short hairpin RNA H295R human adrenocortical stable cell line. We show that ASAH1 suppression increases the transcription of multiple steroidogenic genes, including Cytochrome P450 monooxygenase (CYP)17A1, CYP11B1/2, CYP21A2, steroidogenic acute regulatory protein, hormone-sensitive lipase, 18-kDa translocator protein, and the melanocortin-2 receptor. Induced gene expression positively correlated with enhanced histone H3 acetylation at target promoters. Repression of ASAH1 expression also induced the expression of members of the nuclear receptor nuclear receptor subfamily 4 (NR4A) family while concomitantly suppressing the expression of dosage-sensitive sex reversal, adrenal hypoplasia critical region, on chromosome X, gene 1. ASAH1 knockdown altered the expression of genes involved in sphingolipid metabolism and changed the cellular amounts of distinct sphingolipid species. Finally, ASAH1 silencing increased basal and cAMP-dependent cortisol and dehydroepiandrosterone secretion, establishing ASAH1 as a pivotal regulator of steroidogenic capacity in the human adrenal cortex.
Collapse
Affiliation(s)
- Natasha C Lucki
- School of Biology, Georgia Institute of Technology, Atlanta, Georgia 30332-0230, USA
| | | | | | | | | |
Collapse
|
28
|
O'Rourke B, Van Eyk JE, Foster DB. Mitochondrial protein phosphorylation as a regulatory modality: implications for mitochondrial dysfunction in heart failure. CONGESTIVE HEART FAILURE (GREENWICH, CONN.) 2011; 17:269-82. [PMID: 22103918 PMCID: PMC4067253 DOI: 10.1111/j.1751-7133.2011.00266.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Phosphorylation of mitochondrial proteins has been recognized for decades, and the regulation of pyruvate- and branched-chain α-ketoacid dehydrogenases by an atypical kinase/phosphatase cascade is well established. More recently, the development of new mass spectrometry-based technologies has led to the discovery of many novel phosphorylation sites on a variety of mitochondrial targets. The evidence suggests that the major classes of kinase and several phosphatases may be present at the mitochondrial outer membrane, intermembrane space, inner membrane, and matrix, but many questions remain to be answered as to the location, timing, and reversibility of these phosphorylation events and whether they are functionally relevant. The authors review phosphorylation as a mitochondrial regulatory strategy and highlight its possible role in the pathophysiology of cardiac hypertrophy and failure.
Collapse
Affiliation(s)
- Brian O'Rourke
- Department of Medicine, Division of Cardiology, The Johns Hopkins University, Baltimore, MD 21205-2195, USA.
| | | | | |
Collapse
|
29
|
Chung S, Son GH, Kim K. Adrenal peripheral oscillator in generating the circadian glucocorticoid rhythm. Ann N Y Acad Sci 2011; 1220:71-81. [DOI: 10.1111/j.1749-6632.2010.05923.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
30
|
Azarashvili TS, Odinokova IV, Krestinina OV, Baburina YL, Grachev DE, Teplova VV, Holmuhamedov EL. Role of phosphorylation of porin (VDAC) in regulation of mitochondrial outer membrane under normal conditions and alcohol intoxication. BIOCHEMISTRY MOSCOW SUPPLEMENT SERIES A-MEMBRANE AND CELL BIOLOGY 2011. [DOI: 10.1134/s1990747811010028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
31
|
Lucki NC, Sewer MB. The interplay between bioactive sphingolipids and steroid hormones. Steroids 2010; 75:390-9. [PMID: 20138078 PMCID: PMC2854287 DOI: 10.1016/j.steroids.2010.01.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2009] [Revised: 01/25/2010] [Accepted: 01/26/2010] [Indexed: 01/02/2023]
Abstract
Steroid hormones regulate various physiological processes including development, reproduction, and metabolism. These regulatory molecules are synthesized from cholesterol in endocrine organs - such as the adrenal glands and gonads - via a multi-step enzymatic process that is catalyzed by the cytochrome P450 superfamily of monooxygenases and hydroxysteroid dehydrogenases. Steroidogenesis is induced by trophic peptide hormones primarily via the activation of a cAMP/protein kinase A (PKA)-dependent pathway. However, other signaling molecules, including cytokines and growth factors, control the steroid hormone biosynthetic pathway. More recently, sphingolipids, including ceramide, sphingosine-1-phosphate, and sphingosine, have been found to modulate steroid hormone secretion at multiple levels. In this review, we provide a brief overview of the mechanisms by which sphingolipids regulate steroidogenesis. In addition, we discuss how steroid hormones control sphingolipid metabolism. Finally, we outline evidence supporting the emerging role of bioactive sphingolipids in various nuclear processes and discuss a role for nuclear sphingolipid metabolism in the control of gene transcription.
Collapse
Affiliation(s)
- Natasha C. Lucki
- School of Biology and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 310 Ferst Dr., Atlanta, GA 30332
| | - Marion B. Sewer
- Skaggs School of Pharmacy & Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Dr. MC0704, La Jolla, CA 92093
| |
Collapse
|
32
|
Acyl-coenzyme A binding domain containing 3 (ACBD3; PAP7; GCP60): an emerging signaling molecule. Prog Lipid Res 2010; 49:218-34. [PMID: 20043945 DOI: 10.1016/j.plipres.2009.12.003] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Golgi body-mediated signaling has been linked to its fragmentation and regeneration during the mitotic cycle of the cell. During this process, Golgi-resident proteins are released to the cytosol and interact with other signaling molecules to regulate various cellular processes. Acyl-coenzyme A binding domain containing 3 protein (ACBD3) is a Golgi protein involved in several signaling events. ACBD3 protein was previously known as peripheral-type benzodiazepine receptor and cAMP-dependent protein kinase associated protein 7 (PAP7), Golgi complex-associated protein of 60kDa (GCP60), Golgi complex-associated protein 1 (GOCAP1), and Golgi phosphoprotein 1 (GOLPH1). In this review, we present the gene ontology of ACBD3, its relations to other Acyl-coenzyme A binding domain containing (ACBD) proteins, and its biological function in steroidogenesis, apoptosis, neurogenesis, and embryogenesis. We also discuss the role of ACBD3 in asymmetric cell division and cancer. New findings about ACBD3 may help understand this newly characterized signaling molecule and stimulate further research into its role in molecular endocrinology, neurology, and stem cell biology.
Collapse
|
33
|
Mechanisms of protein kinase A anchoring. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2010; 283:235-330. [PMID: 20801421 DOI: 10.1016/s1937-6448(10)83005-9] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The second messenger cyclic adenosine monophosphate (cAMP), which is produced by adenylyl cyclases following stimulation of G-protein-coupled receptors, exerts its effect mainly through the cAMP-dependent serine/threonine protein kinase A (PKA). Due to the ubiquitous nature of the cAMP/PKA system, PKA signaling pathways underlie strict spatial and temporal control to achieve specificity. A-kinase anchoring proteins (AKAPs) bind to the regulatory subunit dimer of the tetrameric PKA holoenzyme and thereby target PKA to defined cellular compartments in the vicinity of its substrates. AKAPs promote the termination of cAMP signals by recruiting phosphodiesterases and protein phosphatases, and the integration of signaling pathways by binding additional signaling proteins. AKAPs are a heterogeneous family of proteins that only display similarity within their PKA-binding domains, amphipathic helixes docking into a hydrophobic groove formed by the PKA regulatory subunit dimer. This review summarizes the current state of information on compartmentalized cAMP/PKA signaling with a major focus on structural aspects, evolution, diversity, and (patho)physiological functions of AKAPs and intends to outline newly emerging directions of the field, such as the elucidation of AKAP mutations and alterations of AKAP expression in human diseases, and the validation of AKAP-dependent protein-protein interactions as new drug targets. In addition, alternative PKA anchoring mechanisms employed by noncanonical AKAPs and PKA catalytic subunit-interacting proteins are illustrated.
Collapse
|
34
|
Binding of NIR-conPK and NIR-6T to astrocytomas and microglial cells: evidence for a protein related to TSPO. PLoS One 2009; 4:e8271. [PMID: 20020060 PMCID: PMC2792720 DOI: 10.1371/journal.pone.0008271] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Accepted: 11/12/2009] [Indexed: 12/02/2022] Open
Abstract
PK 11195 and DAA1106 bind with high-affinity to the translocator protein (TSPO, formerly known as the peripheral benzodiazepine receptor). TSPO expression in glial cells increases in response to cytokines and pathological stimuli. Accordingly, [11C]-PK 11195 and [11C]-DAA1106 are recognized molecular imaging (MI) agents capable of monitoring changes in TSPO expression occurring in vivo and in response to various neuropathologies. Here we tested the pharmacological characteristics and TSPO-monitoring potential of two novel MI agents: NIR-conPK and NIR-6T. NIR-conPK is an analogue of PK 11195 conjugated to the near-infrared (NIR) emitting fluorophore: IRDye 800CW. NIR-6T is a DAA1106 analogue also conjugated to IRDye 800CW. We found that NIR-6T competed for [3H]-PK 11195 binding in astrocytoma cell homogenates with nanomolar affinity, but did not exhibit specific binding in intact astrocytoma cells in culture, indicating that NIR-6T is unlikely to constitute a useful MI agent for monitoring TSPO expression in intact cells. Conversely, we found that NIR-conPK did not compete for [3H]-PK 11195 binding in astrocytoma cell homogenate, but exhibited specific binding in intact astrocytoma cells in culture with nanomolar affinity, suggesting that NIR-conPK binds to a protein distinct, but related to, TSPO. Accordingly, treating intact astrocytoma cells and microglia in culture with cytokines led to significant changes in the amount of NIR-conPK specific binding without corresponding change in TSPO expression. Remarkably, the cytokine-induced changes in the protein targeted by NIR-conPK in intact microglia were selective, since IFN-γ (but not TNFα and TGFβ) increased the amount of NIR-conPK specific binding in these cells. Together these results suggest that NIR-conPK binds to a protein that is related to TSPO, and expressed by astrocytomas and microglia. Our results also suggest that the expression of this protein is increased by specific cytokines, and thus allows for the monitoring of a particular subtype of microglia activation.
Collapse
|
35
|
Guillaumot D, Guillon S, Déplanque T, Vanhee C, Gumy C, Masquelier D, Morsomme P, Batoko H. The Arabidopsis TSPO-related protein is a stress and abscisic acid-regulated, endoplasmic reticulum-Golgi-localized membrane protein. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2009; 60:242-56. [PMID: 19548979 DOI: 10.1111/j.1365-313x.2009.03950.x] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The Arabidopsis gene At2g47770 encodes a membrane-bound protein designated AtTSPO (Arabidopsis thaliana TSPO-related). AtTSPO is related to the bacterial outer membrane tryptophan-rich sensory protein (TspO) and the mammalian mitochondrial 18-kDa translocator protein (18 kDa TSPO), members of the group of TspO/MBR domain-containing membrane proteins. In this study we show that AtTSPO is mainly detected in dry seeds, but can be induced in vegetative tissues by osmotic or salt stress or abscisic acid (ABA) treatment, corroborating available transcriptome data. Using subcellular fractionation, immunocytochemistry and fluorescent protein tagging approaches we present evidence that AtTSPO is targeted to the secretory pathway in plants. Induced or constitutively expressed AtTSPO can be detected in the endoplasmic reticulum and the Golgi stacks of plant cells. AtTSPO tagged with fluorescent protein in transgenic plants (Arabidopsis and tobacco) was mainly detected in the Golgi stacks of leaf epidermal cells. Constitutive expression of AtTSPO resulted in increased sensitivity to NaCl, but not to osmotic stress, and in reduced greening of cultured Arabidopsis cells under light growing conditions. Transgenic Arabidopsis plants overexpressing AtTSPO were more sensitive to ABA-induced growth inhibition, indicating that constitutive expression of AtTSPO may enhance ABA sensitivity. AtTSPO is rapidly downregulated during seed imbibition, and the ABA-dependent induction in plant is transient. Downregulation of AtTSPO seems to be boosted by treatment with aminolevulinic acid. Taken together, these results suggest that AtTSPO is a highly regulated protein, induced by abiotic stress to modulate, at least in part, transient intracellular ABA-dependent stress perception and/or signalling.
Collapse
Affiliation(s)
- Damien Guillaumot
- Institut des Sciences de la Vie, Molecular Physiology Group, Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Foster DB, Van Eyk JE, Marbán E, O'Rourke B. Redox signaling and protein phosphorylation in mitochondria: progress and prospects. J Bioenerg Biomembr 2009; 41:159-68. [PMID: 19440831 DOI: 10.1007/s10863-009-9217-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
As we learn more about the factors that govern cardiac mitochondrial bioenergetics, fission and fusion, as well as the triggers of apoptotic and necrotic cell death, there is growing appreciation that these dynamic processes are finely-tuned by equally dynamic post-translational modification of proteins in and around the mitochondrion. In this minireview, we discuss the evidence that S-nitrosylation, glutathionylation and phosphorylation of mitochondrial proteins have important bioenergetic consequences. A full accounting of these targets, and the functional impact of their modifications, will be necessary to determine the extent to which these processes underlie ischemia/reperfusion injury, cardioprotection by pre/post-conditioning, and the pathogenesis of heart failure.
Collapse
Affiliation(s)
- D Brian Foster
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Ross Research Building, Room 847, 720 Rutland Avenue, Baltimore, MD 21205, USA.
| | | | | | | |
Collapse
|
37
|
Krestinina OV, Grachev DE, Odinokova IV, Reiser G, Evtodienko YV, Azarashvili TS. Effect of peripheral benzodiazepine receptor (PBR/TSPO) ligands on opening of Ca2+-induced pore and phosphorylation of 3.5-kDa polypeptide in rat brain mitochondria. BIOCHEMISTRY (MOSCOW) 2009; 74:421-9. [PMID: 19463096 DOI: 10.1134/s0006297909040105] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The effect of nanomolar concentrations of PBR/TSPO ligands--Ro 5-4864, PK11195, and PPIX--on Ca2+-induced permeability transition pore (PTP) opening in isolated rat brain mitochondria was investigated. PBR/TSPO agonist Ro 5-4864 (100 nM) and endogenous ligand PPIX (1 microM) were shown to stimulate PTP opening, while antagonist PK11195 (100 nM) suppressed this process. Correlation between PBR ligand action on PTP opening and phosphorylation of a 3.5 kDa polypeptide was investigated. In intact brain mitochondria, incorporation of [gamma-(32)P]ATP into 3.5 kDa peptide was decreased in the presence of Ro 5-4864 and PPIX and increased in the presence of PK11195. At threshold Ca2+ concentrations leading to PTP opening, PBR/TSPO ligands were found to stimulate dephosphorylation of the 3.5 kDa peptide. Specific anti-PBR/TSPO antibody prevented both PTP opening and dephosphorylation of the 3.5-kDa peptide. The peptide was identified as subunit c of F(o)F(1)-ATPase by Western blot using specific anti-subunit c antibody. The results suggest that subunit c of F(o)F(1)-ATPase could be an additional target for PBR/TSPO ligands action, is subjected to Ca2+- and TSPO-dependent phosphorylation/dephosphorylation, and is involved in PTP operation in mitochondria.
Collapse
Affiliation(s)
- O V Krestinina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | | | | | | | | | | |
Collapse
|
38
|
Rone MB, Fan J, Papadopoulos V. Cholesterol transport in steroid biosynthesis: role of protein-protein interactions and implications in disease states. Biochim Biophys Acta Mol Cell Biol Lipids 2009; 1791:646-58. [PMID: 19286473 DOI: 10.1016/j.bbalip.2009.03.001] [Citation(s) in RCA: 273] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2008] [Revised: 02/28/2009] [Accepted: 03/03/2009] [Indexed: 12/20/2022]
Abstract
The transfer of cholesterol from the outer to the inner mitochondrial membrane is the rate-limiting step in hormone-induced steroid formation. To ensure that this step is achieved efficiently, free cholesterol must accumulate in excess at the outer mitochondrial membrane and then be transferred to the inner membrane. This is accomplished through a series of steps that involve various intracellular organelles, including lysosomes and lipid droplets, and proteins such as the translocator protein (18 kDa, TSPO) and steroidogenic acute regulatory (StAR) proteins. TSPO, previously known as the peripheral-type benzodiazepine receptor, is a high-affinity drug- and cholesterol-binding mitochondrial protein. StAR is a hormone-induced mitochondria-targeted protein that has been shown to initiate cholesterol transfer into mitochondria. Through the assistance of proteins such as the cAMP-dependent protein kinase regulatory subunit Ialpha (PKA-RIalpha) and the PKA-RIalpha- and TSPO-associated acyl-coenzyme A binding domain containing 3 (ACBD3) protein, PAP7, cholesterol is transferred to and docked at the outer mitochondrial membrane. The TSPO-dependent import of StAR into mitochondria, and the association of TSPO with the outer/inner mitochondrial membrane contact sites, drives the intramitochondrial cholesterol transfer and subsequent steroid formation. The focus of this review is on (i) the intracellular pathways and protein-protein interactions involved in cholesterol transport and steroid biosynthesis and (ii) the roles and interactions of these proteins in endocrine pathologies and neurological diseases where steroid synthesis plays a critical role.
Collapse
Affiliation(s)
- Malena B Rone
- The Research Institute of the McGill University Health Centre and Department of Medicine, McGill University, 1650 Cedar Avenue, Montreal, Quebec, Canada H3G 1A4
| | | | | |
Collapse
|
39
|
Dyson MT, Kowalewski MP, Manna PR, Stocco DM. The differential regulation of steroidogenic acute regulatory protein-mediated steroidogenesis by type I and type II PKA in MA-10 cells. Mol Cell Endocrinol 2009; 300:94-103. [PMID: 19111595 PMCID: PMC2692359 DOI: 10.1016/j.mce.2008.11.029] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2008] [Revised: 11/19/2008] [Accepted: 11/24/2008] [Indexed: 10/21/2022]
Abstract
Following tropic hormone challenge, steroidogenic tissues utilize PKA to phosphorylate unique subsets of proteins necessary to facilitate steroidogenesis. This includes the PKA-dependent expression and activation of the steroidogenic acute regulatory protein (STAR), which mediates the rate-limiting step of steroidogenesis by inducing the transfer of cholesterol from the outer to the inner mitochondrial membrane. Since both type I and type II PKA are present in steroidogenic tissues, we have utilized cAMP analog pairs that preferentially activate each PKA subtype in order to examine their impact on STAR synthesis and activity. In MA-10 mouse Leydig tumor cells Star gene expression is more dependent upon type I PKA, while the post-transcriptional regulation of STAR appears subject to type II PKA. These experiments delineate the discrete effects that type I and type II PKA exert on STAR-mediated steroidogenesis, and suggest complimentary roles for each subtype in coordinating steroidogenesis.
Collapse
Affiliation(s)
| | | | | | - Douglas M. Stocco
- To whom correspondence should be addressed: Dr. Douglas Stocco, Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center. 3601 4th Street, Lubbock, TX 79430, Phone: (806)-743-2505, Fax: (806) 743-2990, E-mail:
| |
Collapse
|
40
|
Abidi P, Leers-Sucheta S, Cortez Y, Han J, Azhar S. Evidence that age-related changes in p38 MAP kinase contribute to the decreased steroid production by the adrenocortical cells from old rats. Aging Cell 2008; 7:168-78. [PMID: 18241324 DOI: 10.1111/j.1474-9726.2007.00364.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The current studies were initiated to investigate whether excessive oxidative stress exerts its antisteroidogenic action through modulation of oxidant-sensitive mitogen-activated protein kinase (MAPK) signaling pathways. Western blot analysis indicated that aging caused increased phosphorylation and activation of rat adrenal p38 MAPK, but not the ERK1/2 or JNK1/2. Lipid peroxidation measurements (an index of cellular oxidative stress) indicated that adrenal membranes from young animals contained only minimal levels of endogenous thiobarbituric acid-reactive substances (TBARS), and exposure of membranes to enzymatic and non-enzymatic pro-oxidants enhanced TBARS formation approximately 12- and 20-fold, respectively. The adrenal membranes from old animals showed much more susceptibility to lipid peroxidation and exhibited roughly 4- to 6-fold higher TBARS formation than young controls both under basal conditions and in response to pro-oxidants. Qualitatively similar results were obtained when lipid peroxide formation was measured using a sensitive FOXRS (ferrous oxidation-xylenol orange-reactive substances) technique. We next tested whether aging-induced excessive oxidative insult alters steroidogenesis through modulation of MAPK signaling pathway. Treatment of adrenocortical cells from old rats with specific p38 MAPK inhibitors restored Bt(2)cAMP-stimulated steroidogenesis approximately 60-70% of the value seen in cells of young animals. Likewise, pretreatment of cells with reactive oxygen species (ROS) scavengers MnTMPyP and N-acetyl cysteine also partially rescued age-induced loss of steroid production. In contrast, simultaneous treatment of cells with ROS scavengers and p38 MAPK inhibitor did not produce any additional effect suggesting that both types of inhibitors exert their stimulatory action through inhibition of p38 MAPK activation. Collectively, these results indicate that p38 MAPK functions as a signaling effector in oxidative stress-induced inhibition of steroidogenesis during aging.
Collapse
Affiliation(s)
- Parveen Abidi
- Geriatric Research, Education and Clinical Center, Department of Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | | | | | | | | |
Collapse
|
41
|
Abstract
Steroid hormones are essential regulators of a vast number of physiological processes. The biosynthesis of these chemical messengers occurs in specialized steroidogenic tissues via a multi-step process that is catalyzed by members of the cytochrome P450 superfamily of monooxygenases and hydroxysteroid dehydrogenases. Though numerous signaling mediators, including cytokines and growth factors control steroidogenesis, trophic peptide hormones are the primary regulators of steroid hormone production. These peptide hormones activate a cAMP/cAMP-dependent kinase (PKA) signaling pathway, however, studies have shown that crosstalk between multiple signal transduction pathways and signaling molecules modulates optimal steroidogenic capacity. Sphingolipids such as ceramide, sphingosine, sphingosine-1-phosphate, sphingomyelin, and gangliosides have been shown to control the steroid hormone biosynthetic pathway at multiple levels, including regulating steroidogenic gene expression and activity as well as acting as second messengers in signaling cascades. In this review, we provide an overview of recent studies that have investigated the role of sphingolipids in adrenal, gonadal, and neural steroidogenesis.
Collapse
Affiliation(s)
- Natasha C Lucki
- School of Biology and Parker H, Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 310 Ferst Drive, Atlanta, GA 30332-0230, USA
| | | |
Collapse
|
42
|
Dyson MT, Jones JK, Kowalewski MP, Manna PR, Alonso M, Gottesman ME, Stocco DM. Mitochondrial A-kinase anchoring protein 121 binds type II protein kinase A and enhances steroidogenic acute regulatory protein-mediated steroidogenesis in MA-10 mouse leydig tumor cells. Biol Reprod 2007; 78:267-77. [PMID: 17989356 DOI: 10.1095/biolreprod.107.064238] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The expression of the steroidogenic acute regulatory protein (STAR) is regulated by PKA in response to trophic hormone stimulation through the second messenger cAMP. However, in steroidogenic cells, the concentrations of hormone necessary to maximally induce cAMP synthesis and PKA activity are often significantly higher than is necessary to achieve maximum steroidogenesis. One general mechanism believed to make PKA signaling more effective is the use of A-kinase anchoring proteins (AKAPs) to recruit PKA to discrete subcellular compartments, which coordinates and focuses PKA action with respect to its substrates. The characterization of AKAP121 has suggested that it enhances the posttranscriptional regulation of STAR by recruiting both Star mRNA and PKA to the mitochondria, thereby permitting more effective translation and phosphorylation of STAR. Testing this hypothesis revealed that cAMP-induced STAR expression and steroidogenesis closely followed AKAP121 abundance when this AKAP was silenced or overexpressed in MA-10 cells but that these changes were effected posttranscriptionally. Moreover, silencing AKAP121 expression in these cells specifically altered the localization of type II PKA regulatory subunit alpha (PKAR2A) at the mitochondria but did not affect its relative expression within the cell. Affinity purification experiments showed that PKAR2A preferentially associated with AKAP121, and cAMP analogs that activate type II PKA induced STAR phosphorylation more efficiently than analogs stimulating type I PKA. This suggests that AKAP121 and PKAR2A serve to enhance steroidogenesis by directing the synthesis and activation of STAR at the mitochondria in response to cAMP.
Collapse
Affiliation(s)
- Matthew T Dyson
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Sbodio JI, Machamer CE. Identification of a redox-sensitive cysteine in GCP60 that regulates its interaction with golgin-160. J Biol Chem 2007; 282:29874-81. [PMID: 17711851 DOI: 10.1074/jbc.m705794200] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Golgin-160 is ubiquitously expressed in vertebrates. It localizes to the cytoplasmic side of the Golgi and has a large C-terminal coiled-coil domain. The noncoiled-coil N-terminal head domain contains Golgi targeting information, a cryptic nuclear localization signal, and three caspase cleavage sites. Caspase cleavage of the golgin-160 head domain generates different fragments that can translocate to the nucleus by exposing the nuclear localization signal. We have previously shown that GCP60, a Golgi resident protein, interacts weakly with the golgin-160 head domain but has a strong interaction with one of the caspase-generated golgin-160 fragments (residues 140-311). This preferential interaction increases the Golgi retention of the golgin-160 fragment in cells overexpressing GCP60. Here we studied the interaction of golgin-160-(140-311) with GCP60 and identified a single cysteine residue in GCP60 (Cys-463) that is critical for the interaction of the two proteins. Mutation of the cysteine blocked the interaction in vitro and disrupted the ability to retain the golgin-160 fragment at the Golgi in cells. We also found that Cys-463 is redox-sensitive; in its reduced form, interaction with golgin-160 was diminished or abolished, whereas oxidation of the Cys-463 by hydrogen peroxide restored the interaction. In addition, incubation with a nitric oxide donor promoted this interaction in vitro. These findings suggest that nuclear translocation of golgin-160-(140-311) is a highly coordinated event regulated not only by cleavage of the golgin-160 head but also by the oxidation state of GCP60.
Collapse
Affiliation(s)
- Juan I Sbodio
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | |
Collapse
|
44
|
Li D, Urs AN, Allegood J, Leon A, Merrill AH, Sewer MB. Cyclic AMP-stimulated interaction between steroidogenic factor 1 and diacylglycerol kinase theta facilitates induction of CYP17. Mol Cell Biol 2007; 27:6669-85. [PMID: 17664281 PMCID: PMC2099220 DOI: 10.1128/mcb.00355-07] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In the human adrenal cortex, adrenocorticotropin (ACTH) activates CYP17 transcription by promoting the binding of the nuclear receptor steroidogenic factor 1 (SF1) (Ad4BP, NR5A1) to the promoter. We recently found that sphingosine is an antagonist for SF1 and inhibits cyclic AMP (cAMP)-dependent CYP17 gene transcription. The aim of the current study was to identify phospholipids that bind to SF1 and to characterize the mechanism by which ACTH/cAMP regulates the biosynthesis of this molecule(s). Using tandem mass spectrometry, we show that in H295R human adrenocortical cells, SF1 is bound to phosphatidic acid (PA). Activation of the ACTH/cAMP signal transduction cascade rapidly increases nuclear diacylglycerol kinase (DGK) activity and PA production. PA stimulates SF1-dependent transcription of CYP17 reporter plasmids, promotes coactivator recruitment, and induces the mRNA expression of CYP17 and several other steroidogenic genes. Inhibition of DGK activity attenuates the binding of SF1 to the CYP17 promoter, and silencing of DGK-theta expression inhibits cAMP-dependent CYP17 transcription. LXXLL motifs in DGK-theta mediate a direct interaction of SF1 with the kinase and may facilitate binding of PA to the receptor. We conclude that ACTH/cAMP stimulates PA production in the nucleus of H295R cells and that this increase in PA concentrations facilitates CYP17 induction.
Collapse
Affiliation(s)
- Donghui Li
- School of Biology, Georgia Institute of Technology, Atlanta, GA 30332-0230, USA
| | | | | | | | | | | |
Collapse
|
45
|
Liu J, Rone MB, Papadopoulos V. Protein-Protein Interactions Mediate Mitochondrial Cholesterol Transport and Steroid Biosynthesis. J Biol Chem 2006; 281:38879-93. [PMID: 17050526 DOI: 10.1074/jbc.m608820200] [Citation(s) in RCA: 186] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Transport of cholesterol into the mitochondria is the rate-determining, hormone-sensitive step in steroid biosynthesis. Here we report that the mechanism underlying mitochondrial cholesterol transport involves the formation of a macromolecular signaling complex composed of the outer mitochondrial membrane translocator protein (TSPO), previously known as peripheral-type benzodiazepine receptor; the TSPO-associated protein PAP7, which binds and brings to mitochondria the regulatory subunit RIalpha of the cAMP-dependent protein kinase (PKARIalpha); and the hormone-induced PKA substrate, steroidogenic acute regulatory protein (StAR). Hormone treatment of MA-10 Leydig cells induced the co-localization of TSPO, PAP7, PKARIalpha, and StAR in mitochondria, visualized by confocal microscopy, and the formation in living cells of a high molecular weight multimeric complex identified using photoactivable amino acids. The hormone-induced recruitment of exogenous TSPO in this complex was found to parallel the increased presence of 7-azi-5alpha-cholestan-3beta-ol in the samples. Co-expression of Tspo, Pap7, PkarIalpha, and Star genes resulted in the stimulation of steroid formation in both steroidogenic MA-10 and non-steroidogenic COS-F2-130 cells that were engineered to metabolize cholesterol. Disruption of these protein-protein interactions and specifically the PKARIalpha-PAP7 and PAP7-TSPO interactions, using PAP7 mutants where the N0 area homologous to dual A-kinase-anchoring protein-1 or the acyl-CoA signature motif were deleted or using the peptide Ht31 known to disrupt the anchoring of PKA, inhibited both basal and hormone-induced steroidogenesis. These results suggest that the initiation of cAMP-induced protein-protein interactions results in the formation of a multivalent scaffold in the outer mitochondrial membrane that mediates the effect of hormones on mitochondrial cholesterol transport and steroidogenesis.
Collapse
Affiliation(s)
- Jun Liu
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, D. C. 20057, USA
| | | | | |
Collapse
|
46
|
Herman M, Kang SS, Lee S, James P, Rivier C. Systemic Administration of Alcohol to Adult Rats Inhibits Leydig Cell Activity: Time Course of Effect and Role of Nitric Oxide. Alcohol Clin Exp Res 2006; 30:1479-91. [PMID: 16930210 DOI: 10.1111/j.1530-0277.2006.00179.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Alcohol has been shown to interfere with testosterone (T) release from Leydig cells. However, the mechanisms responsible for this phenomenon, which may include decreased activity of the luteinizing hormone-releasing hormone (LHRH)-LH axis, as well as a direct influence of the drug on the testes, are not fully understood. In this work, we investigated the influence of alcohol, administered intragastrically (i.g.) or delivered via vapors, on Leydig cell activity and T release. Leydig cell function was studied by measuring changes in the levels of the steroidogenic proteins steroidogenic acute regulatory (StAR), the peripheral-type benzodiazepine receptor (PBR), and the cytochrome P450 side-chain cleavage enzyme (P450scc). Testosterone release was studied under basal conditions or in response to human chorionic gonadotropin (hCG). Finally, to identify potential factors mediating the influence of alcohol, we measured the testicular variant of the neuronal nitric oxide (NO) synthase (NOS), TnNOS, in semipurified Leydig cells. METHODS Adult male Sprague-Dawley rats were either injected with alcohol i.g. once or exposed to alcohol vapors (4 h/d) for 1 or 5 days. Controls received the vehicle (i.g. model) or were kept in boxes through which no vapors were circulated. Following these treatments, one series of experiments was devoted to investigate Leydig cell responsiveness by measuring plasma T levels before or after the intravenous injection of hCG (1 U/kg). In another series of experiments, we used semipurified Leydig cell preparations to measure StAR, PBR, P450scc, and TnNOS by Western blot analysis. RESULTS In the i.g. model, the T response to hCG was blunted for 12 hours following alcohol injection, but showed a rebound at 48 hours. Levels of StAR protein and of PBR, but not of P450scc, were significantly decreased within 10 minutes of drug administration. While StAR then remained depressed for 24 hours, PBR values were variable over this time course. By 48 hours, StAR, PBR, and P450scc levels had increased above control values. Both StAR and PBR levels showed correlations with plasma T levels. In the alcohol vapor models, both regimens of the drug also significantly depressed StAR and PBR protein concentrations, blunted the T response to hCG, and did not alter P450scc. Finally, we observed that alcohol delivered i.g. or via vapors up-regulated TnNOS levels in Leydig cells, but that blockade of NO formation failed to restore a normal T response to hCG. CONCLUSIONS Collectively, these results suggest that (a) the ability of Leydig cells to release T does not show a simple correlation with changes in StAR, PBR, and P450scc levels; (b) the time course of the alcohol-induced changes were protein-specific; and (c) despite the ability of alcohol to stimulate TnNOS expression, NO does not appear to mediate the inhibitory influence of this drug on testicular steroidogenesis in the models that we studied.
Collapse
Affiliation(s)
- Melissa Herman
- Clayton Foundation Laboratories for Peptide Biology, The Salk Institute, La Jolla, California 92037, USA
| | | | | | | | | |
Collapse
|
47
|
Sbodio JI, Hicks SW, Simon D, Machamer CE. GCP60 preferentially interacts with a caspase-generated golgin-160 fragment. J Biol Chem 2006; 281:27924-31. [PMID: 16870622 DOI: 10.1074/jbc.m603276200] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Golgin-160, a ubiquitous protein in vertebrates, localizes to the cytoplasmic face of the Golgi complex. Golgin-160 has a large coiled-coil C-terminal domain and a non-coiled-coil N-terminal ("head") domain. The head domain contains important motifs, including a nuclear localization signal, a Golgi targeting domain, and three aspartates that are recognized by caspases during apoptosis. Some of the caspase cleavage products accumulate in the nucleus when overexpressed. Expression of a non-cleavable form of golgin-160 impairs apoptosis induced by some pro-apoptotic stimuli; thus cleavage of golgin-160 appears to play a role in apoptotic signaling. We used a yeast two-hybrid assay to screen for interactors of the golgin-160 head and identified GCP60 (Golgi complex-associated protein of 60 kDa). Further analysis demonstrated that GCP60 interacts preferentially with one of the golgin-160 caspase cleavage fragments (residues 140-311). This strong interaction prevented the golgin-160 fragment from accumulating in the nucleus when this fragment and GCP60 were overexpressed. In addition, cells overexpressing GCP60 were more sensitive to apoptosis induced by staurosporine, suggesting that nuclear-localized golgin-160-(140-311) might promote cell survival. Our results suggest a potential mechanism for regulating the nuclear translocation and potential functions of golgin-160 fragments.
Collapse
Affiliation(s)
- Juan I Sbodio
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | |
Collapse
|
48
|
Carlson CR, Lygren B, Berge T, Hoshi N, Wong W, Taskén K, Scott JD. Delineation of Type I Protein Kinase A-selective Signaling Events Using an RI Anchoring Disruptor. J Biol Chem 2006; 281:21535-21545. [PMID: 16728392 DOI: 10.1074/jbc.m603223200] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Control of specificity in cAMP signaling is achieved by A-kinase anchoring proteins (AKAPs), which assemble cAMP effectors such as protein kinase A (PKA) into multiprotein signaling complexes in the cell. AKAPs tether the PKA holoenzymes at subcellular locations to favor the phosphorylation of selected substrates. PKA anchoring is mediated by an amphipathic helix of 14-18 residues on each AKAP that binds to the R subunit dimer of the PKA holoenzymes. Using a combination of bioinformatics and peptide array screening, we have developed a high affinity-binding peptide called RIAD (RI anchoring disruptor) with >1000-fold selectivity for type I PKA over type II PKA. Cell-soluble RIAD selectively uncouples cAMP-mediated inhibition of T cell function and inhibits progesterone synthesis at the mitochondria in steroid-producing cells. This study suggests that these processes are controlled by the type I PKA holoenzyme and that RIAD can be used as a tool to define anchored type I PKA signaling events.
Collapse
Affiliation(s)
- Cathrine Rein Carlson
- Biotechnology Centre of Oslo, University of Oslo, PB 1125 Blindern, N-0317, Oslo, Norway; Howard Hughes Medical Institute, Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239.
| | - Birgitte Lygren
- Biotechnology Centre of Oslo, University of Oslo, PB 1125 Blindern, N-0317, Oslo, Norway; Howard Hughes Medical Institute, Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239
| | - Torunn Berge
- Biotechnology Centre of Oslo, University of Oslo, PB 1125 Blindern, N-0317, Oslo, Norway
| | - Naoto Hoshi
- Howard Hughes Medical Institute, Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239
| | - Wei Wong
- Howard Hughes Medical Institute, Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239
| | - Kjetil Taskén
- Biotechnology Centre of Oslo, University of Oslo, PB 1125 Blindern, N-0317, Oslo, Norway
| | - John D Scott
- Howard Hughes Medical Institute, Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239
| |
Collapse
|
49
|
Veenman L, Gavish M. The peripheral-type benzodiazepine receptor and the cardiovascular system. Implications for drug development. Pharmacol Ther 2006; 110:503-24. [PMID: 16337685 DOI: 10.1016/j.pharmthera.2005.09.007] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2005] [Accepted: 09/27/2005] [Indexed: 11/16/2022]
Abstract
Peripheral-type benzodiazepine receptors (PBRs) are abundant in the cardiovascular system. In the cardiovascular lumen, PBRs are present in platelets, erythrocytes, lymphocytes, and mononuclear cells. In the walls of the cardiovascular system, PBR can be found in the endothelium, the striated cardiac muscle, the vascular smooth muscles, and the mast cells. The subcellular location of PBR is primarily in mitochondria. The PBR complex includes the isoquinoline binding protein (IBP), voltage-dependent anion channel (VDAC), and adenine nucleotide transporter (ANT). Putative endogenous ligands for PBR include protoporphyrin IX, diazepam binding inhibitor (DBI), triakontatetraneuropeptide (TTN), and phospholipase A2 (PLA2). Classical synthetic ligands for PBR are the isoquinoline 1-(2-chlorophenyl)-N-methyl-N-(1-methyl-propyl)-3-isoquinolinecarboxamide (PK 11195) and the benzodiazepine 7-chloro-5-(4-chlorophenyl)-1,3-dihydro-1-methyl-2H-1,4-benzodiazepin-2-one (Ro5 4864). Novel PBR ligands include N,N-di-n-hexyl 2-(4-fluorophenyl)indole-3-acetamide (FGIN-1-27) and 7-chloro-N,N,5-trimethyl-4-oxo-3-phenyl-3,5-dihydro-4H-pyridazino[4,5-b]indole-1-acetamide (SSR180575), both possessing steroidogenic properties, but while FGIN-1-27 is pro-apoptotic, SSR180575 is anti-apoptotic. Putative PBR functions include regulation of steroidogenesis, apoptosis, cell proliferation, the mitochondrial membrane potential, the mitochondrial respiratory chain, voltage-dependent calcium channels, responses to stress, and microglial activation. PBRs in blood vessel walls appear to take part in responses to trauma such as ischemia. The irreversible PBR antagonist, SSR180575, was found to reduce damage correlated with ischemia. Stress, anxiety disorders, and neurological disorders, as well as their treatment, can affect PBR levels in blood cells. PBRs in blood cells appear to play roles in several aspects of the immune response, such as phagocytosis and the secretion of interleukin-2, interleukin-3, and immunoglobulin A (IgA). Thus, alterations in PBR density in blood cells may have immunological consequences in the affected person. In conclusion, PBR in the cardiovascular system may represent a new target for drug development.
Collapse
Affiliation(s)
- Leo Veenman
- Rappaport Family Institute for Research in the Medical Sciences, Technion-Israel Institute of Technology, Department of Pharmacology, Ephron Street, P.O. Box 9649, Bat-Galim, Haifa 31096, Israel
| | | |
Collapse
|
50
|
Herman M, Rivier C. Activation of a neural brain-testicular pathway rapidly lowers Leydig cell levels of the steroidogenic acute regulatory protein and the peripheral-type benzodiazepine receptor while increasing levels of neuronal nitric oxide synthase. Endocrinology 2006; 147:624-33. [PMID: 16239298 DOI: 10.1210/en.2005-0879] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Activation of a neural brain-testicular pathway by the intracerebroventricular injection of the beta-adrenergic agonist isoproterenol (ISO), the hypothalamic peptide corticotropin-releasing factor (CRF), or alcohol (EtOH) rapidly decreases the testosterone (T) response to human chorionic gonadotropin. To elucidate the intratesticular mechanisms responsible for this phenomenon, we investigated the influence of intracerebroventricular-injected ISO, CRF, or EtOH on levels of the steroidogenic acute regulatory (StAR) protein, the peripheral-type benzodiazepine receptor (PBR), and the cytochrome P450 side-chain cleavage enzyme in semipurified Leydig cells. ISO (10 microg), CRF (5 microg), or EtOH (5 microl of 200 proof, a dose that does not induce neuronal damage nor leaks to the periphery) rapidly decreased StAR and PBR but not cytochrome P450 side-chain cleavage enzyme protein levels. Levels of the variant of the neuronal nitric oxide synthase (nNOS) that is restricted to Leydig cells, TnNOS, significantly increased in response to ISO, CRF, and EtOH over the time course of altered StAR/PBR concentrations. However, pretreatment of the rats with N(w)nitro-arginine methylester, which blocked ISO-induced increases in TnNOS, neither restored the T response to human chorionic gonadotropin nor prevented the decreases in StAR and PBR. These results provide evidence of concomitant changes in Leydig cell StAR and PBR levels in live rats. They also indicate that activation of a neural brain-testicular pathway rapidly decreases concentrations of these steroidogenic proteins while up-regulating testicular NO production. However, additional studies are necessary to elucidate the functional role played by this gas in our model.
Collapse
Affiliation(s)
- Melissa Herman
- The Clayton Foundation Laboratories for Peptide Biology, The Salk Institute, La Jolla, California 92037, USA
| | | |
Collapse
|