1
|
Bian J, Ding H, Hu A, Wang J. Circ_0001361/miR-490-5p/IGF2 Axis Regulates the Viability and Apoptosis of Neuroblastoma Cells. Neurochem Res 2024; 49:3060-3068. [PMID: 39110291 DOI: 10.1007/s11064-024-04225-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 06/17/2024] [Accepted: 08/03/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Circular RNAs (circRNAs) are involved in the neuroblastoma (NB) development. Objectie: The study aimed to determine the biological behaviors of circ_0001361 and explore its underlying mechanism in NB. METHODS The circ_0001361, miR-490-5p, and IGF2 levels were measured using quantitative real-time polymerase chain reaction. Cellular processes were analyzed using MTT assay or fluorescence-activated cell sorting (FACS). Phosphorylated (p)-PI3K, p-AKT, Bax, and caspase-3 were tested by western blot. Dual-luciferase reporter analysis together with RNA pull-down analysis were utilized to evaluate the correlation of miR-490-5p and circ_0001361 or IGF2. RESULTS The results in this study illustrated that an elevation of circ_0001361 levels was observed in NB. Depletion of circ_0001361 suppressed the viability but facilitated apoptosis of NB cells. Circ_0001361 sponged miR-490-5p, which targeted to regulate IGF2. Inhibition of miR-490-5p rescued the effect induced by circ_0001361 knockdown, while deletion of IGF2 rescued the effect induced by the miR-490-5p inhibitor. CONCLUSIONS In summary, a loss of circ_0001361 inhibited NB progression via targeting the miR-490-5p/IGF2 axis, suggesting that circ_0001361 may be a novel therapeutical target of NB.
Collapse
Affiliation(s)
- Jian Bian
- Department of General Surgery, Children's Hospital Affiliated to Soochow University, Suzhou, China
- Department of General Surgery, Anhui Provincial Children's Hospital, Hefei, China
| | - Hao Ding
- Department of Clinical Nutrition, Anhui Provincial Children's Hospital, Hefei, China
| | - Anla Hu
- Department of Nutrition and Food Hygiene, School of Public Health, Anhui Medical University, Hefei, China
| | - Jian Wang
- Department of General Surgery, Children's Hospital Affiliated to Soochow University, Suzhou, China.
| |
Collapse
|
2
|
Miao J, Zhang Y, Su C, Zheng Q, Guo J. Insulin-Like Growth Factor Signaling in Alzheimer's Disease: Pathophysiology and Therapeutic Strategies. Mol Neurobiol 2024:10.1007/s12035-024-04457-1. [PMID: 39240280 DOI: 10.1007/s12035-024-04457-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia among the elderly population, posing a significant public health challenge due to limited therapeutic options that merely delay cognitive decline. AD is associated with impaired energy metabolism and reduced neurotrophic signaling. The insulin-like growth factor (IGF) signaling pathway, crucial for central nervous system (CNS) development, metabolism, repair, cognition, and emotion regulation, includes IGF-1, IGF-2, IGF-1R, IGF-2R, insulin receptor (IR), and six insulin-like growth factor binding proteins (IGFBPs). Research has identified abnormalities in IGF signaling in individuals with AD and AD models. Dysregulated expression of IGFs, receptors, IGFBPs, and disruptions in downstream phosphoinositide 3-kinase-protein kinase B (PI3K/AKT) and mitogen-activated protein kinase (MAPK) pathways collectively increase AD susceptibility. Studies suggest modulating the IGF pathway may ameliorate AD pathology and cognitive decline. This review explores the CNS pathophysiology of IGF signaling in AD progression and assesses the potential of targeting the IGF system as a novel therapeutic strategy. Further research is essential to elucidate how aberrant IGF signaling contributes to AD development, understand underlying molecular mechanisms, and evaluate the safety and efficacy of IGF-based treatments.
Collapse
Affiliation(s)
- Jie Miao
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yanli Zhang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
- Department of Neurology, Sixth Hospital of Shanxi Medical University (General Hospital of Tisco), Taiyuan, 030001, Shanxi, China
| | - Chen Su
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Qiandan Zheng
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Junhong Guo
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
3
|
Kato C, Ueda K, Morimoto S, Takahashi S, Nakamura S, Ozawa F, Ito D, Daté Y, Okada K, Kobayashi N, Nakahara J, Okano H. Proteomic insights into extracellular vesicles in ALS for therapeutic potential of Ropinirole and biomarker discovery. Inflamm Regen 2024; 44:32. [PMID: 38997748 PMCID: PMC11241965 DOI: 10.1186/s41232-024-00346-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/26/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) hold the potential for elucidating the pathogenesis of amyotrophic lateral sclerosis (ALS) and serve as biomarkers. Notably, the comparative and longitudinal alterations in the protein profiles of EVs in serum (sEVs) and cerebrospinal fluid (CSF; cEVs) of sporadic ALS (SALS) patients remain uncharted. Ropinirole hydrochloride (ROPI; dopamine D2 receptor [D2R] agonist), a new anti-ALS drug candidate identified through induced pluripotent stem cell (iPSC)-based drug discovery, has been suggested to inhibit ALS disease progression in the Ropinirole Hydrochloride Remedy for Amyotrophic Lateral Sclerosis (ROPALS) trial, but its mechanism of action is not well understood. Therefore, we tried to reveal longitudinal changes with disease progression and the effects of ROPI on protein profiles of EVs. METHODS We collected serum and CSF at fixed intervals from ten controls and from 20 SALS patients participating in the ROPALS trial. Comprehensive proteomic analysis of EVs, extracted from these samples, was conducted using liquid chromatography/mass spectrometer (LC/MS). Furthermore, we generated iPSC-derived astrocytes (iPasts) and performed RNA sequencing on astrocytes with or without ROPI treatment. RESULTS The findings revealed notable disparities yet high congruity in sEVs and cEVs protein profiles concerning disease status, time and ROPI administration. In SALS, both sEVs and cEVs presented elevated levels of inflammation-related proteins but reduced levels associated with unfolded protein response (UPR). These results mirrored the longitudinal changes after disease onset and correlated with the revised ALS Functional Rating Scale (ALSFRS-R) at sampling time, suggesting a link to the onset and progression of SALS. ROPI appeared to counteract these changes, attenuating inflammation-related protein levels and boosting those tied to UPR in SALS, proposing an anti-ALS impact on EV protein profiles. Reverse translational research using iPasts indicated that these changes may partly reflect the DRD2-dependent neuroinflammatory inhibitory effects of ROPI. We have also identified biomarkers that predict diagnosis and disease progression by machine learning-driven biomarker search. CONCLUSIONS Despite the limited sample size, this study pioneers in reporting time-series proteomic alterations in serum and CSF EVs from SALS patients, offering comprehensive insights into SALS pathogenesis, ROPI-induced changes, and potential prognostic and diagnostic biomarkers.
Collapse
Affiliation(s)
- Chris Kato
- Keio University Regenerative Medicine Research Center, Kanagawa, 210-0821, Japan
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
- Division of Neurodegenerative Disease Research, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Koji Ueda
- Cancer Proteomics Group, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Satoru Morimoto
- Keio University Regenerative Medicine Research Center, Kanagawa, 210-0821, Japan.
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan.
- Division of Neurodegenerative Disease Research, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan.
| | - Shinichi Takahashi
- Keio University Regenerative Medicine Research Center, Kanagawa, 210-0821, Japan
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
- Department of Neurology and Cerebrovascular Medicine, Saitama Medical University International Medical Center, Saitama, 350-1298, Japan
| | - Shiho Nakamura
- Keio University Regenerative Medicine Research Center, Kanagawa, 210-0821, Japan
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
- Division of Neurodegenerative Disease Research, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Fumiko Ozawa
- Keio University Regenerative Medicine Research Center, Kanagawa, 210-0821, Japan
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
- Division of Neurodegenerative Disease Research, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Daisuke Ito
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
- Department of Neurology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Yugaku Daté
- Department of Neurology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Kensuke Okada
- Department of Neurology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Naoki Kobayashi
- Keio University Regenerative Medicine Research Center, Kanagawa, 210-0821, Japan
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Jin Nakahara
- Department of Neurology, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Hideyuki Okano
- Keio University Regenerative Medicine Research Center, Kanagawa, 210-0821, Japan.
- Department of Physiology, Keio University School of Medicine, Tokyo, 160-8582, Japan.
- Division of Neurodegenerative Disease Research, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan.
| |
Collapse
|
4
|
Mo H, Kim J, Kim JY, Kim JW, Han H, Choi SH, Rim YA, Ju JH. Intranasal administration of induced pluripotent stem cell-derived cortical neural stem cell-secretome as a treatment option for Alzheimer's disease. Transl Neurodegener 2023; 12:50. [PMID: 37946307 PMCID: PMC10634159 DOI: 10.1186/s40035-023-00384-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common neurodegenerative disorder in the elderly, resulting in gradual destruction of cognitive abilities. Research on the development of various AD treatments is underway; however, no definitive treatment has been developed yet. Herein, we present induced pluripotent stem cell (iPSC)-derived cortical neural stem cell secretome (CNSC-SE) as a new treatment candidate for AD and explore its efficacy. METHODS We first assessed the effects of CNSC-SE treatment on neural maturation and electromagnetic signal during cortical nerve cell differentiation. Then to confirm the efficacy in vivo, CNSC-SE was administered to the 5×FAD mouse model through the nasal cavity (5 μg/g, once a week, 4 weeks). The cell-mediated effects on nerve recovery, amyloid beta (Aβ) plaque aggregation, microglial and astrocyte detection in the brain, and neuroinflammatory responses were investigated. Metabolomics analysis of iPSC-derived CNSC-SE revealed that it contained components that could exert neuro-protective effects or amplify cognitive restorative effects. RESULTS Human iPSC-derived CNSC-SE increased neuronal proliferation and dendritic structure formation in vitro. Furthermore, CNSC-SE-treated iPSC-derived cortical neurons acquired electrical network activity and action potential bursts. The 5×FAD mice treated with CNSC-SE showed memory restoration and reduced Aβ plaque accumulation. CONCLUSIONS Our findings suggest that the iPSC-derived CNSC-SE may serve as a potential, non-invasive therapeutic option for AD in reducing amyloid infiltration and restoring memory.
Collapse
Affiliation(s)
- Hyunkyung Mo
- CiSTEM Laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Department of Biomedicine and Health Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Juryun Kim
- YiPSCELL, Inc, Omnibus Park, Banpo-daero 222, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Jennifer Yejean Kim
- Department of Biology, Georgetown University, 3700 O St NW, Washington, DC, 20057, USA
| | - Jang Woon Kim
- CiSTEM Laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Heeju Han
- CiSTEM Laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Department of Biomedicine and Health Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Si Hwa Choi
- CiSTEM Laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Department of Biomedicine and Health Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Yeri Alice Rim
- CiSTEM Laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| | - Ji Hyeon Ju
- CiSTEM Laboratory, Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
- YiPSCELL, Inc, Omnibus Park, Banpo-daero 222, Seocho-gu, Seoul, 06591, Republic of Korea.
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| |
Collapse
|
5
|
Baxter RC. Signaling Pathways of the Insulin-like Growth Factor Binding Proteins. Endocr Rev 2023; 44:753-778. [PMID: 36974712 PMCID: PMC10502586 DOI: 10.1210/endrev/bnad008] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/25/2023] [Accepted: 03/15/2023] [Indexed: 03/29/2023]
Abstract
The 6 high-affinity insulin-like growth factor binding proteins (IGFBPs) are multifunctional proteins that modulate cell signaling through multiple pathways. Their canonical function at the cellular level is to impede access of insulin-like growth factor (IGF)-1 and IGF-2 to their principal receptor IGF1R, but IGFBPs can also inhibit, or sometimes enhance, IGF1R signaling either through their own post-translational modifications, such as phosphorylation or limited proteolysis, or by their interactions with other regulatory proteins. Beyond the regulation of IGF1R activity, IGFBPs have been shown to modulate cell survival, migration, metabolism, and other functions through mechanisms that do not appear to involve the IGF-IGF1R system. This is achieved by interacting directly or functionally with integrins, transforming growth factor β family receptors, and other cell-surface proteins as well as intracellular ligands that are intermediates in a wide range of pathways. Within the nucleus, IGFBPs can regulate the diverse range of functions of class II nuclear hormone receptors and have roles in both cell senescence and DNA damage repair by the nonhomologous end-joining pathway, thus potentially modifying the efficacy of certain cancer therapeutics. They also modulate some immune functions and may have a role in autoimmune conditions such as rheumatoid arthritis. IGFBPs have been proposed as attractive therapeutic targets, but their ubiquity in the circulation and at the cellular level raises many challenges. By understanding the diversity of regulatory pathways with which IGFBPs interact, there may still be therapeutic opportunities based on modulation of IGFBP-dependent signaling.
Collapse
Affiliation(s)
- Robert C Baxter
- Kolling Institute of Medical Research, University of Sydney, Royal North Shore Hospital,St Leonards, NSW 2065, Australia
| |
Collapse
|
6
|
Effect of Interleukin-1β on Gene Expression Signatures in Schwann Cells Associated with Neuropathic Pain. Neurochem Res 2021; 46:2958-2968. [PMID: 34264480 DOI: 10.1007/s11064-021-03400-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 05/25/2021] [Accepted: 07/09/2021] [Indexed: 10/20/2022]
Abstract
Interleukin-1β (IL-1β) plays a critical role in the development of neuropathic pain through activation of Schwann cells (SCs) after nerve injury. Here, we applied an RNA sequencing (RNA-seq) approach to identify the effect of IL-1β on gene signatures of a rat SC line (RSC96) and the potential molecular mechanisms underlying the development of neuropathic pain. RNA-seq data demonstrated a total of 57 significantly differentially expressed genes (DEGs) with 35 up-regulated and 22 down-regulated between SCs treated with IL-1β, and control SCs without treatment. Bioinformatics analysis showed that key upregulated DEGs included those associated with immune and inflammation-related processes, neurotrophin production and SC proliferation. Five proteins encoded by key upregulated DEGs (Ceacam1, Hap1, Irs3, Lgi4 and Mif) were further verified by Western blot. Consistent with the RNA-Seq results, the expression of key genes was confirmed in SCs by immunofluorescence of the chronic constriction injury (CCI) sciatic nerve in rats. Furthermore, we demonstrated that treatment with IL-1β resulted in an increase in p38/ERK phosphorylation, and activators of p38/ERK enhanced the effect of IL-1β on the expression some of the key genes, whereas p38/ERK inhibitors reversed these effects. In conclusion, the present study highlights key genes involved in the development of neuropathic pain through activation of SCs after nerve injury. Identification of these genes and subsequent evidence of their mediation by IL-1β treatment promote our understanding of molecular mechanisms of nerve injury induced neuropathic pain, and highlight potential molecular targets for the treatment of neuropathic pain.
Collapse
|
7
|
Ge L, Zhuo Y, Wu P, Liu Y, Qi L, Teng X, Duan D, Chen P, Lu M. Olfactory ensheathing cells facilitate neurite sprouting and outgrowth by secreting high levels of hevin. J Chem Neuroanat 2019; 104:101728. [PMID: 31783092 DOI: 10.1016/j.jchemneu.2019.101728] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/25/2019] [Accepted: 11/25/2019] [Indexed: 02/06/2023]
Abstract
Transplantation of olfactory ensheathing cells (OECs) has been shown to enhance synapse formation. However, the mechanisms underlying this effect are not completely understood. We performed profiling of the OEC and astrocyte secretomes via a proteomics approach, in case hevin secreted by astrocytes might be involved in the formation of synapses. Semi-quantitative proteomic analysis revealed that 25 proteins were highly expressed, and 22 were weakly expressed in OEC conditioned medium compared with astrocyte conditioned medium. These molecules are highly associated with neural differentiation and regeneration, enzyme regulatory activity, and growth factor binding. The quantification data of clusterin, fibronectin, hevin, insulin-like growth factor binding protein 2 and secreted protein acidic and rich in cysteine were further confirmed by western blotting. Moreover, the addition of hevin in the culture medium improved neurite sprouting and outgrowth of differentiated neural stem cells. The greater expression of hevin in OEC conditioned medium than in astrocyte conditioned medium was associated with a greater capacity of synaptic formation. Thus, our results indicate that soluble factors secreted by OECs provide a permissive environment for nerve repair, and hevin is one of the key molecules facilitating neurite sprouting and outgrowth.
Collapse
Affiliation(s)
- Lite Ge
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China; Hunan Provincial Key Laboratory of Neurorestoratology, the Second Affiliated Hospital (the 921st Hospital of PLA), Hunan Normal University, Changsha,China
| | - Yi Zhuo
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China; Hunan Provincial Key Laboratory of Neurorestoratology, the Second Affiliated Hospital (the 921st Hospital of PLA), Hunan Normal University, Changsha,China
| | - Pei Wu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Yisong Liu
- Hunan Key Laboratory of Traditional Chinese Veterinary Medicine, Hunan Agricultural University, Changsha, China
| | - Linyu Qi
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Xiaohua Teng
- Hunan Provincial Key Laboratory of Neurorestoratology, the Second Affiliated Hospital (the 921st Hospital of PLA), Hunan Normal University, Changsha,China
| | - Da Duan
- Hunan Provincial Key Laboratory of Neurorestoratology, the Second Affiliated Hospital (the 921st Hospital of PLA), Hunan Normal University, Changsha,China.
| | - Ping Chen
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China.
| | - Ming Lu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China; Hunan Provincial Key Laboratory of Neurorestoratology, the Second Affiliated Hospital (the 921st Hospital of PLA), Hunan Normal University, Changsha,China.
| |
Collapse
|
8
|
Khan S. IGFBP-2 Signaling in the Brain: From Brain Development to Higher Order Brain Functions. Front Endocrinol (Lausanne) 2019; 10:822. [PMID: 31824433 PMCID: PMC6883226 DOI: 10.3389/fendo.2019.00822] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 11/08/2019] [Indexed: 12/13/2022] Open
Abstract
Insulin-like growth factor-binding protein-2 (IGFBP-2) is a pleiotropic polypeptide that functions as autocrine and/or paracrine growth factors. IGFBP-2 is the most abundant of the IGFBPs in the cerebrospinal fluid (CSF), and developing brain showed the highest expression of IGFBP-2. IGFBP-2 expressed in the hippocampus, cortex, olfactory lobes, cerebellum, and amygdala. IGFBP-2 mRNA expression is seen in meninges, blood vessels, and in small cell-body neurons (interneurons) and astrocytes. The expression pattern of IGFBP-2 is often developmentally regulated and cell-specific. Biological activities of IGFBP-2 which are independent of their abilities to bind to insulin-like growth factors (IGFs) are mediated by the heparin binding domain (HBD). To execute IGF-independent functions, some IGFBPs have shown to bind with their putative receptors or to translocate inside the cells. Thus, IGFBP-2 functions can be mediated both via insulin-like growth factor receptor-1 (IGF-IR) and independent of IGF-Rs. In this review, I suggest that IGFBP-2 is not only involved in the growth, development of the brain but also with the regulation of neuronal plasticity to modulate high-level cognitive operations such as spatial learning and memory and information processing. Hence, IGFBP-2 serves as a neurotrophic factor which acts via metaplastic signaling from embryonic to adult stages.
Collapse
|
9
|
Costa RA, Martins RST, Capilla E, Anjos L, Power DM. Vertebrate SLRP family evolution and the subfunctionalization of osteoglycin gene duplicates in teleost fish. BMC Evol Biol 2018; 18:191. [PMID: 30545285 PMCID: PMC6293640 DOI: 10.1186/s12862-018-1310-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 11/27/2018] [Indexed: 02/07/2023] Open
Abstract
Background Osteoglycin (OGN, a.k.a. mimecan) belongs to cluster III of the small leucine-rich proteoglycans (SLRP) of the extracellular matrix (ECM). In vertebrates OGN is a characteristic ECM protein of bone. In the present study we explore the evolution of SLRP III and OGN in teleosts that have a skeleton adapted to an aquatic environment. Results The SLRP gene family has been conserved since the separation of chondrichthyes and osteichthyes. Few gene duplicates of the SLRP III family exist even in the teleosts that experienced a specific whole genome duplication. One exception is ogn for which duplicate copies were identified in fish genomes. The ogn promoter sequence and in vitro mesenchymal stem cell (MSC) cultures suggest the duplicate ogn genes acquired divergent functions. In gilthead sea bream (Sparus aurata) ogn1 was up-regulated during osteoblast and myocyte differentiation in vitro, while ogn2 was severely down-regulated during bone-derived MSCs differentiation into adipocytes in vitro. Conclusions Overall, the phylogenetic analysis indicates that the SLRP III family in vertebrates has been under conservative evolutionary pressure. The retention of the ogn gene duplicates in teleosts was linked with the acquisition of different functions. The acquisition by OGN of functions other than that of a bone ECM protein occurred early in the vertebrate lineage. Electronic supplementary material The online version of this article (10.1186/s12862-018-1310-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- R A Costa
- Comparative Endocrinology and Integrative Biology Group, Centre of Marine Sciences, University of Algarve, Campus of Gambelas, 8005-139, Faro, Portugal
| | - R S T Martins
- Comparative Endocrinology and Integrative Biology Group, Centre of Marine Sciences, University of Algarve, Campus of Gambelas, 8005-139, Faro, Portugal.
| | - E Capilla
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, 08028, Barcelona, Spain
| | - L Anjos
- Comparative Endocrinology and Integrative Biology Group, Centre of Marine Sciences, University of Algarve, Campus of Gambelas, 8005-139, Faro, Portugal
| | - D M Power
- Comparative Endocrinology and Integrative Biology Group, Centre of Marine Sciences, University of Algarve, Campus of Gambelas, 8005-139, Faro, Portugal.
| |
Collapse
|
10
|
Lind AL, Emami Khoonsari P, Sjödin M, Katila L, Wetterhall M, Gordh T, Kultima K. Spinal Cord Stimulation Alters Protein Levels in the Cerebrospinal Fluid of Neuropathic Pain Patients: A Proteomic Mass Spectrometric Analysis. Neuromodulation 2017; 19:549-62. [PMID: 27513633 DOI: 10.1111/ner.12473] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 06/07/2016] [Accepted: 06/09/2016] [Indexed: 01/03/2023]
Abstract
OBJECTIVES Electrical neuromodulation by spinal cord stimulation (SCS) is a well-established method for treatment of neuropathic pain. However, the mechanism behind the pain relieving effect in patients remains largely unknown. In this study, we target the human cerebrospinal fluid (CSF) proteome, a little investigated aspect of SCS mechanism of action. METHODS Two different proteomic mass spectrometry protocols were used to analyze the CSF of 14 SCS responsive neuropathic pain patients. Each patient acted as his or her own control and protein content was compared when the stimulator was turned off for 48 hours, and after the stimulator had been used as normal for three weeks. RESULTS Eighty-six proteins were statistically significantly altered in the CSF of neuropathic pain patients using SCS, when comparing the stimulator off condition to the stimulator on condition. The top 12 of the altered proteins are involved in neuroprotection (clusterin, gelsolin, mimecan, angiotensinogen, secretogranin-1, amyloid beta A4 protein), synaptic plasticity/learning/memory (gelsolin, apolipoprotein C1, apolipoprotein E, contactin-1, neural cell adhesion molecule L1-like protein), nociceptive signaling (neurosecretory protein VGF), and immune regulation (dickkopf-related protein 3). CONCLUSION Previously unknown effects of SCS on levels of proteins involved in neuroprotection, nociceptive signaling, immune regulation, and synaptic plasticity are demonstrated. These findings, in the CSF of neuropathic pain patients, expand the picture of SCS effects on the neurochemical environment of the human spinal cord. An improved understanding of SCS mechanism may lead to new tracks of investigation and improved treatment strategies for neuropathic pain.
Collapse
Affiliation(s)
- Anne-Li Lind
- Department of Surgical Sciences, Anaesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
| | - Payam Emami Khoonsari
- Department of Medical Sciences, Cancer Pharmacology and Computational Medicine, Uppsala University, Uppsala, Sweden
| | - Marcus Sjödin
- Department of Chemistry-BMC, Analytical Chemistry, Uppsala University, Uppsala//GE Healthcare, Sweden
| | - Lenka Katila
- Department of Surgical Sciences, Anaesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
| | - Magnus Wetterhall
- Department of Chemistry-BMC, Analytical Chemistry, Uppsala University, Uppsala//GE Healthcare, Sweden
| | - Torsten Gordh
- Department of Surgical Sciences, Anaesthesiology and Intensive Care, Uppsala University, Uppsala, Sweden
| | - Kim Kultima
- Department of Medical Sciences, Cancer Pharmacology and Computational Medicine, Uppsala University, Uppsala, Sweden
| |
Collapse
|
11
|
Mercado AT, Yeh JM, Chin TY, Chen WS, Chen-Yang YW, Chen CY. The effect of chemically modified electrospun silica nanofiber on the mRNA and miRNA expression profile of neural stem cell differentiation. J Biomed Mater Res A 2016; 104:2730-43. [DOI: 10.1002/jbm.a.35819] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Accepted: 06/21/2016] [Indexed: 01/30/2023]
Affiliation(s)
- Augustus T. Mercado
- Department of Bioscience Technology; Chung Yuan Christian University; Chung-Li 32023 Taiwan
- Department of Chemistry; Chung Yuan Christian University; Chung-Li 32023 Taiwan
| | - Jui-Ming Yeh
- Department of Chemistry; Chung Yuan Christian University; Chung-Li 32023 Taiwan
- Center for Biomedical Technology, Chung Yuan Christian University; Chung-Li 32023 Taiwan
| | - Ting Yu Chin
- Department of Bioscience Technology; Chung Yuan Christian University; Chung-Li 32023 Taiwan
- Center for Biomedical Technology, Chung Yuan Christian University; Chung-Li 32023 Taiwan
| | - Wen Shuo Chen
- Department of Chemistry; Chung Yuan Christian University; Chung-Li 32023 Taiwan
- Center for Biomedical Technology, Chung Yuan Christian University; Chung-Li 32023 Taiwan
| | - Yui Whei Chen-Yang
- Department of Chemistry; Chung Yuan Christian University; Chung-Li 32023 Taiwan
- Center for Biomedical Technology, Chung Yuan Christian University; Chung-Li 32023 Taiwan
| | - Chung-Yung Chen
- Department of Bioscience Technology; Chung Yuan Christian University; Chung-Li 32023 Taiwan
- Center for Biomedical Technology, Chung Yuan Christian University; Chung-Li 32023 Taiwan
| |
Collapse
|
12
|
Terauchi A, Johnson-Venkatesh EM, Bullock B, Lehtinen MK, Umemori H. Retrograde fibroblast growth factor 22 (FGF22) signaling regulates insulin-like growth factor 2 (IGF2) expression for activity-dependent synapse stabilization in the mammalian brain. eLife 2016; 5. [PMID: 27083047 PMCID: PMC4868541 DOI: 10.7554/elife.12151] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 04/14/2016] [Indexed: 02/06/2023] Open
Abstract
Communication between pre- and postsynaptic cells promotes the initial organization of synaptic specializations, but subsequent synaptic stabilization requires transcriptional regulation. Here we show that fibroblast growth factor 22 (FGF22), a target-derived presynaptic organizer in the mouse hippocampus, induces the expression of insulin-like growth factor 2 (IGF2) for the stabilization of presynaptic terminals. FGF22 is released from CA3 pyramidal neurons and organizes the differentiation of excitatory nerve terminals formed onto them. Local application of FGF22 on the axons of dentate granule cells (DGCs), which are presynaptic to CA3 pyramidal neurons, induces IGF2 in the DGCs. IGF2, in turn, localizes to DGC presynaptic terminals and stabilizes them in an activity-dependent manner. IGF2 application rescues presynaptic defects of Fgf22(-/-) cultures. IGF2 is dispensable for the initial presynaptic differentiation, but is required for the following presynaptic stabilization both in vitro and in vivo. These results reveal a novel feedback signal that is critical for the activity-dependent stabilization of presynaptic terminals in the mammalian hippocampus.
Collapse
Affiliation(s)
- Akiko Terauchi
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, United States
| | - Erin M Johnson-Venkatesh
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, United States
| | - Brenna Bullock
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, United States
| | - Maria K Lehtinen
- Department of Pathology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, United States
| | - Hisashi Umemori
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, United States
| |
Collapse
|
13
|
Deckx S, Heymans S, Papageorgiou AP. The diverse functions of osteoglycin: a deceitful dwarf, or a master regulator of disease? FASEB J 2016; 30:2651-61. [PMID: 27080639 DOI: 10.1096/fj.201500096r] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 04/05/2016] [Indexed: 12/21/2022]
Abstract
Small leucine-rich proteoglycans are emerging as important regulatory proteins within the extracellular matrix, where they exert both structural and nonstructural functions and hence are modulators of numerous biological processes, such as inflammation, fibrosis, and cell proliferation. One proteoglycan in particular, osteoglycin (OGN), also known as mimecan, shows great structural and functional diversity in normal physiology and in disease states, therefore making it a very interesting candidate for the development of novel therapeutic strategies. Unfortunately, the literature on OGN is confusing, as it has different names, and different transcript and protein variants have been identified. This review will give a clear overview of the different structures and functions of OGN that have been identified to date, portray its central role in pathophysiology, and highlight the importance of posttranslational processing, such as glycosylation, for the diversity of its functions.-Deckx, S., Heymans, S., Papageorgiou, A.-P. The diverse functions of osteoglycin: a deceitful dwarf, or a master regulator of disease?
Collapse
Affiliation(s)
- Sophie Deckx
- Department of Cardiology, Maastricht University, Maastricht, The Netherlands; and Center for Molecular and Vascular Biology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Stephane Heymans
- Department of Cardiology, Maastricht University, Maastricht, The Netherlands; and Center for Molecular and Vascular Biology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Anna-Pia Papageorgiou
- Department of Cardiology, Maastricht University, Maastricht, The Netherlands; and Center for Molecular and Vascular Biology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| |
Collapse
|
14
|
Ito K, Sanosaka T, Igarashi K, Ideta-Otsuka M, Aizawa A, Uosaki Y, Noguchi A, Arakawa H, Nakashima K, Takizawa T. Identification of genes associated with the astrocyte-specific gene Gfap during astrocyte differentiation. Sci Rep 2016; 6:23903. [PMID: 27041678 PMCID: PMC4819225 DOI: 10.1038/srep23903] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/16/2016] [Indexed: 01/15/2023] Open
Abstract
Chromosomes and genes are non-randomly arranged within the mammalian cell nucleus, and gene clustering is of great significance in transcriptional regulation. However, the relevance of gene clustering and their expression during the differentiation of neural precursor cells (NPCs) into astrocytes remains unclear. We performed a genome-wide enhanced circular chromosomal conformation capture (e4C) to screen for genes associated with the astrocyte-specific gene glial fibrillary acidic protein (Gfap) during astrocyte differentiation. We identified 18 genes that were specifically associated with Gfap and expressed in NPC-derived astrocytes. Our results provide additional evidence for the functional significance of gene clustering in transcriptional regulation during NPC differentiation.
Collapse
Affiliation(s)
- Kenji Ito
- Department of Pediatrics, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Tsukasa Sanosaka
- Stem Cell Biology and Medicine, Department of Stem cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Katsuhide Igarashi
- Life Science Tokyo Advanced Research Center (L-StaR), Pharmacy and Pharmaceutical Science, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-5801, Japan
| | - Maky Ideta-Otsuka
- Life Science Tokyo Advanced Research Center (L-StaR), Pharmacy and Pharmaceutical Science, Hoshi University, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-5801, Japan
| | - Akira Aizawa
- Department of Pediatrics, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Yuichi Uosaki
- Department of Pediatrics, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Azumi Noguchi
- Department of Pediatrics, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Hirokazu Arakawa
- Department of Pediatrics, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Kinichi Nakashima
- Stem Cell Biology and Medicine, Department of Stem cell Biology and Medicine, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takumi Takizawa
- Department of Pediatrics, Graduate School of Medicine, Gunma University, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| |
Collapse
|
15
|
Sugino IK, Sun Q, Springer C, Cheewatrakoolpong N, Liu T, Li H, Zarbin MA. Two Bioactive Molecular Weight Fractions of a Conditioned Medium Enhance RPE Cell Survival on Age-Related Macular Degeneration and Aged Bruch's Membrane. Transl Vis Sci Technol 2016; 5:8. [PMID: 26933521 PMCID: PMC4771074 DOI: 10.1167/tvst.5.1.8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 01/01/2016] [Indexed: 12/30/2022] Open
Abstract
PURPOSE To characterize molecular weight fractions of bovine corneal endothelial cell conditioned medium (CM) supporting retinal pigment epithelium (RPE) cell survival on aged and age-related macular degeneration (AMD) Bruch's membrane. METHODS CM was subject to size separation using centrifugal filters. Retentate and filtrate fractions were tested for bioactivity by analyzing RPE survival on submacular Bruch's membrane of aged and AMD donor eyes and behavior on collagen I-coated tissue culture wells. Protein and peptide composition of active fractions was determined by mass spectrometry. RESULTS Two bioactive fractions, 3-kDa filtrate and a 10-50-kDa fraction, were necessary for RPE survival on aged and AMD Bruch's membrane. The 3-kDa filtrate, but not the 10-50-kDa fraction, supported RPE growth on collagen 1-coated tissue culture plates. Mass spectrometry of the 10-50-kDa fraction identified 175 extracellular proteins, including growth factors and extracellular matrix molecules. Transforming growth factor (TGF)β-2 was identified as unique to active CM. Peptides representing 29 unique proteins were identified in the 3-KDa filtrate. CONCLUSIONS These results indicate there is a minimum of two bioactive molecules in CM, one found in the 3-kDa filtrate and one in the 10-50-kDa fraction, and that bioactive molecules in both fractions must be present to ensure RPE survival on Bruch's membrane. Mass spectrometry analysis suggested proteins to test in future studies to identify proteins that may contribute to CM bioactivity. TRANSLATIONAL RELEVANCE Results of this study are the first steps in development of an adjunct to cell-based therapy to ensure cell transplant survival and functionality in AMD patients.
Collapse
Affiliation(s)
- Ilene K Sugino
- Institute of Ophthalmology and Visual Science Rutgers, New Jersey Medical School, Newark, NJ, USA
| | - Qian Sun
- Institute of Ophthalmology and Visual Science Rutgers, New Jersey Medical School, Newark, NJ, USA
| | - Carola Springer
- Institute of Ophthalmology and Visual Science Rutgers, New Jersey Medical School, Newark, NJ, USA
| | | | - Tong Liu
- Department of Biochemistry and Molecular Biology, Center for Advanced Proteomics Research, Neuroproteomics Core Facility, Rutgers, New Jersey Medical School, Newark, NJ, USA
| | - Hong Li
- Department of Biochemistry and Molecular Biology, Center for Advanced Proteomics Research, Neuroproteomics Core Facility, Rutgers, New Jersey Medical School, Newark, NJ, USA
| | - Marco A Zarbin
- Institute of Ophthalmology and Visual Science Rutgers, New Jersey Medical School, Newark, NJ, USA
| |
Collapse
|
16
|
Lee RS, Pirooznia M, Guintivano J, Ly M, Ewald ER, Tamashiro KL, Gould TD, Moran TH, Potash JB. Search for common targets of lithium and valproic acid identifies novel epigenetic effects of lithium on the rat leptin receptor gene. Transl Psychiatry 2015; 5:e600. [PMID: 26171981 PMCID: PMC5068731 DOI: 10.1038/tp.2015.90] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 05/21/2015] [Accepted: 06/01/2015] [Indexed: 12/24/2022] Open
Abstract
Epigenetics may have an important role in mood stabilizer action. Valproic acid (VPA) is a histone deacetylase inhibitor, and lithium (Li) may have downstream epigenetic actions. To identify genes commonly affected by both mood stabilizers and to assess potential epigenetic mechanisms that may be involved in their mechanism of action, we administered Li (N = 12), VPA (N = 12), and normal chow (N = 12) to Brown Norway rats for 30 days. Genomic DNA and mRNA were extracted from the hippocampus. We used the mRNA to perform gene expression analysis on Affymetrix microarray chips, and for genes commonly regulated by both Li and VPA, we validated expression levels using quantitative real-time PCR. To identify potential mechanisms underlying expression changes, genomic DNA was bisulfite treated for pyrosequencing of key CpG island 'shores' and promoter regions, and chromatin was prepared from both hippocampal tissue and a hippocampal-derived cell line to assess modifications of histones. For most genes, we found little evidence of DNA methylation changes in response to the medications. However, we detected histone H3 methylation and acetylation in the leptin receptor gene, Lepr, following treatment with both drugs. VPA-mediated effects on histones are well established, whereas the Li effects constitute a novel mechanism of transcriptional derepression for this drug. These data support several shared transcriptional targets of Li and VPA, and provide evidence suggesting leptin signaling as an epigenetic target of two mood stabilizers. Additional work could help clarify whether leptin signaling in the brain has a role in the therapeutic action of Li and VPA in bipolar disorder.
Collapse
Affiliation(s)
- R S Lee
- Johns Hopkins Mood Disorders Center of the Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA,Johns Hopkins Mood Disorders Center of the Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 1068, Baltimore, MD 21205, USA. E-mail:
| | - M Pirooznia
- Johns Hopkins Mood Disorders Center of the Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - J Guintivano
- Johns Hopkins Mood Disorders Center of the Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA,Graduate Program in Human Genetics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - M Ly
- Johns Hopkins Mood Disorders Center of the Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - E R Ewald
- Johns Hopkins Mood Disorders Center of the Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - K L Tamashiro
- Johns Hopkins Mood Disorders Center of the Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - T D Gould
- Departments of Psychiatry, Pharmacology, and Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - T H Moran
- Johns Hopkins Mood Disorders Center of the Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - J B Potash
- Department of Psychiatry, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|