1
|
Hu S, Liu B, Shang J, Guo Q, Lu T, Zhou X, Zhou X, Wang X. Targeting PTGDS Promotes ferroptosis in peripheral T cell lymphoma through regulating HMOX1-mediated iron metabolism. Br J Cancer 2025; 132:384-400. [PMID: 39706989 PMCID: PMC11833084 DOI: 10.1038/s41416-024-02919-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 10/30/2024] [Accepted: 11/22/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Peripheral T cell lymphoma (PTCL) is characterized by high heterogeneity, strong aggressiveness, and extremely poor prognosis. Ferroptosis, a novel form of programmed cell death, has been involved in tumor development and targeting ferroptosis holds great potential for tumor therapy. METHODS Lentiviral transfection was performed to regulate gene expression, followed by Tandem mass tag (TMT)-mass spectrometry and RNA-sequencing. Tumor xenograft models were established for in vivo experiments. RESULTS High expression of prostaglandin D2 synthase (PTGDS) was closely associated with poor prognosis of PTCL patients. PTGDS knockdown and AT56 treatment significantly inhibited the progression of PTCL through regulating cell viability, proliferation, apoptosis, cell cycle and invasion in vitro and in vivo. We further revealed that targeting PTGDS promoted ferroptosis process and enhanced the sensitivity of PTCL cells to ferroptosis inducers Sorafenib in vitro and in vivo. Mechanically, PTGDS interacted with heme-degrading enzymes HMOX1, and targeting PTGDS increased the level of iron and induced ferroptosis in PTCL through promoting HMOX1-mediated heme catabolism and ferritin autophagy process. Through the construction of H25A mutation, the specific gene site of HMOX1 corresponding to its role was identified. CONCLUSIONS Taken together, our findings firstly identified that targeting PTGDS promotes the ferroptosis in PTCL through regulating HMOX1-mediated iron metabolism, and highlighted novel therapeutic strategies to improve the efficacy of ferroptosis-targeted therapy in PTCL patients.
Collapse
MESH Headings
- Ferroptosis/drug effects
- Ferroptosis/genetics
- Humans
- Heme Oxygenase-1/metabolism
- Heme Oxygenase-1/genetics
- Animals
- Mice
- Iron/metabolism
- Lymphoma, T-Cell, Peripheral/pathology
- Lymphoma, T-Cell, Peripheral/metabolism
- Lymphoma, T-Cell, Peripheral/genetics
- Lymphoma, T-Cell, Peripheral/drug therapy
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
- Female
- Male
- Cell Proliferation
- Intramolecular Oxidoreductases/genetics
- Intramolecular Oxidoreductases/metabolism
- Intramolecular Oxidoreductases/antagonists & inhibitors
- Gene Expression Regulation, Neoplastic
- Prognosis
- Sorafenib/pharmacology
Collapse
Affiliation(s)
- Shunfeng Hu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Bingyu Liu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Juanjuan Shang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Qianqian Guo
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Tiange Lu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China
| | - Xiaoli Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Taishan Scholars Program of Shandong Province, Jinan, Shandong, 250021, China.
- Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong, 250021, China.
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Taishan Scholars Program of Shandong Province, Jinan, Shandong, 250021, China.
- Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong, 250021, China.
- National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| |
Collapse
|
2
|
Chen B, Guo L, Wang L, Wu P, Zheng X, Tan C, Xie N, Sun X, Zhou M, Huang H, Hao N, Lei Y, Yan K, Wu D, Du Y. Leveraging cell death patterns to predict metastasis in prostate adenocarcinoma and targeting PTGDS for tumor suppression. Sci Rep 2024; 14:21680. [PMID: 39289451 PMCID: PMC11408614 DOI: 10.1038/s41598-024-72985-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 09/12/2024] [Indexed: 09/19/2024] Open
Abstract
Metastasis is the major cause of treatment failure in patients with prostate adenocarcinoma (PRAD). Diverse programmed cell death (PCD) patterns play an important role in tumor metastasis and hold promise as predictive indicators for PRAD metastasis. Using the LASSO Cox regression method, we developed PCD score (PCDS) based on differentially expressed genes (DEGs) associated with PCD. Clinical correlation, external validation, functional enrichment analysis, mutation landscape analysis, tumor immune environment analysis, and immunotherapy analysis were conducted. The role of Prostaglandin D2 Synthase (PTGDS) in PRAD was examined through in vitro experiments, single-cell, and Mendelian randomization (MR) analysis. PCDS is elevated in patients with higher Gleason scores, higher T stage, biochemical recurrence (BCR), and higher prostate-specific antigen (PSA) levels. Individuals with higher PCDS are prone to metastasis, metastasis after BCR, BCR, and castration resistance. Moreover, PRAD patients with low PCDS responded positively to immunotherapy. Random forest analysis and Mendelian randomization analysis identified PTGDS as the top gene associated with PRAD metastasis and in vitro experiments revealed that PTGDS was considerably downregulated in PRAD cells against normal prostate cells. Furthermore, the overexpression of PTGDS was found to suppress the migration, invasion, proliferationof DU145 and LNCaP cells. To sum up, PCDS may be a useful biomarker for forecasting the possibility of metastasis, recurrence, castration resistance, and the efficacy of immunotherapy in PRAD patients. Additionally, PTGDS was identified as a viable therapeutic target for the management of PRAD.
Collapse
Affiliation(s)
- Bohong Chen
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Li Guo
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Lihui Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi Province, China
| | - Peiqiang Wu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Xinyu Zheng
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Congzhu Tan
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Na Xie
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Xinyue Sun
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Mingguo Zhou
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Haoxiang Huang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Na Hao
- Department of Breast Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 716000, Shaanxi Province, China
| | - Yangyang Lei
- Yan'an University, Yan'an, 710061, Shaanxi Province, China
| | - Kun Yan
- Department of Urology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Dapeng Wu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
- Department of Urology, First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, 710061, Shaanxi Province, China.
| | - Yuefeng Du
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
- Department of Urology, First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, 710061, Shaanxi Province, China.
| |
Collapse
|
3
|
Pundlik SS, Barik A, Venkateshvaran A, Sahoo SS, Jaysingh MA, Math RGH, Lal H, Hashmi MA, Ramanathan A. Senescent cells inhibit mouse myoblast differentiation via the SASP-lipid 15d-PGJ 2 mediated modification and control of HRas. eLife 2024; 13:RP95229. [PMID: 39196610 PMCID: PMC11357351 DOI: 10.7554/elife.95229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024] Open
Abstract
Senescent cells are characterized by multiple features such as increased expression of senescence-associated β-galactosidase activity (SA β-gal) and cell cycle inhibitors such as p21 or p16. They accumulate with tissue damage and dysregulate tissue homeostasis. In the context of skeletal muscle, it is known that agents used for chemotherapy such as Doxorubicin (Doxo) cause buildup of senescent cells, leading to the inhibition of tissue regeneration. Senescent cells influence the neighboring cells via numerous secreted factors which form the senescence-associated secreted phenotype (SASP). Lipids are emerging as a key component of SASP that can control tissue homeostasis. Arachidonic acid-derived lipids have been shown to accumulate within senescent cells, specifically 15d-PGJ2, which is an electrophilic lipid produced by the non-enzymatic dehydration of the prostaglandin PGD2. This study shows that 15d-PGJ2 is also released by Doxo-induced senescent cells as an SASP factor. Treatment of skeletal muscle myoblasts with the conditioned medium from these senescent cells inhibits myoblast fusion during differentiation. Inhibition of L-PTGDS, the enzyme that synthesizes PGD2, diminishes the release of 15d-PGJ2 by senescent cells and restores muscle differentiation. We further show that this lipid post-translationally modifies Cys184 of HRas in C2C12 mouse skeletal myoblasts, causing a reduction in the localization of HRas to the Golgi, increased HRas binding to Ras Binding Domain (RBD) of RAF Kinase (RAF-RBD), and activation of cellular Mitogen Activated Protein (MAP) kinase-Extracellular Signal Regulated Kinase (Erk) signaling (but not the Akt signaling). Mutating C184 of HRas prevents the ability of 15d-PGJ2 to inhibit the differentiation of muscle cells and control the activity of HRas. This work shows that 15d-PGJ2 released from senescent cells could be targeted to restore muscle homeostasis after chemotherapy.
Collapse
Affiliation(s)
- Swarang Sachin Pundlik
- Metabolic Regulation of Cell Fate (RCF), Institute for Stem Cell Science and Regenerative Medicine (InStem), Bangalore Life Science ClusterBengaluruIndia
- Manipal Academy of Higher Education (MAHE)ManipalIndia
| | - Alok Barik
- Metabolic Regulation of Cell Fate (RCF), Institute for Stem Cell Science and Regenerative Medicine (InStem), Bangalore Life Science ClusterBengaluruIndia
| | - Ashwin Venkateshvaran
- Metabolic Regulation of Cell Fate (RCF), Institute for Stem Cell Science and Regenerative Medicine (InStem), Bangalore Life Science ClusterBengaluruIndia
| | - Snehasudha Subhadarshini Sahoo
- Metabolic Regulation of Cell Fate (RCF), Institute for Stem Cell Science and Regenerative Medicine (InStem), Bangalore Life Science ClusterBengaluruIndia
- University of North Carolina at Chapel HillChapel HillUnited States
| | - Mahapatra Anshuman Jaysingh
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata (IISER-K)MohanpurIndia
- Division of Biology and Biomedical Sciences, Washington University in St LouisSt LouisUnited States
| | | | - Heera Lal
- Metabolic Regulation of Cell Fate (RCF), Institute for Stem Cell Science and Regenerative Medicine (InStem), Bangalore Life Science ClusterBengaluruIndia
- Manipal Academy of Higher Education (MAHE)ManipalIndia
| | - Maroof Athar Hashmi
- Metabolic Regulation of Cell Fate (RCF), Institute for Stem Cell Science and Regenerative Medicine (InStem), Bangalore Life Science ClusterBengaluruIndia
- Manipal Academy of Higher Education (MAHE)ManipalIndia
| | - Arvind Ramanathan
- Metabolic Regulation of Cell Fate (RCF), Institute for Stem Cell Science and Regenerative Medicine (InStem), Bangalore Life Science ClusterBengaluruIndia
| |
Collapse
|
4
|
Farrag Y, Farrag M, Varela-García M, Torrijos-Pulpón C, Capuozzo M, Ottaiano A, Lago F, Mera A, Pino J, Gualillo O. Adipokines as potential pharmacological targets for immune inflammatory rheumatic diseases: Focus on rheumatoid arthritis, osteoarthritis, and intervertebral disc degeneration. Pharmacol Res 2024; 205:107219. [PMID: 38763327 DOI: 10.1016/j.phrs.2024.107219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/16/2024] [Accepted: 05/16/2024] [Indexed: 05/21/2024]
Abstract
Adipokines are a heterogeneous group of signalling molecules secreted prevalently by adipose tissue. Initially considered as regulators of energy metabolism and appetite, adipokines have been recognized for their substantial involvement in musculoskeletal disorders, including osteoarthritis, rheumatoid arthritis, and many others. Understanding the role of adipokines in rheumatic inflammatory and autoimmune diseases, as well as in other musculoskeletal diseases such as intervertebral disc degeneration, is crucial for the development of novel therapeutic strategies. Targeting adipokines, or their signalling pathways, may offer new opportunities for the treatment and management of these conditions. By modulating adipokines levels or activity, it may be possible to regulate inflammation, to maintain bone health, and preserve muscle mass, thereby improving the outcomes and quality of life for individuals affected by musculoskeletal diseases. The aim of this review article is to update the reader on the multifaceted role of adipokines in the main rheumatic diseases such as osteoarthritis and rheumatoid arthritis and to unravel the complex interplay among adipokines, cartilage metabolism, bone remodelling and muscles, which will pave the way for innovative therapeutic intervention in the future. For completeness, the role of adipokines in intervertebral disc degeneration will be also addressed.
Collapse
Affiliation(s)
- Yousof Farrag
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), The NEIRID Group (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Santiago University Clinical Hospital, Building C, Travesía da Choupana SIN, Santiago de Compostela 15706, Spain.
| | - Mariam Farrag
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), The NEIRID Group (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Santiago University Clinical Hospital, Building C, Travesía da Choupana SIN, Santiago de Compostela 15706, Spain.
| | - María Varela-García
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), The NEIRID Group (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Santiago University Clinical Hospital, Building C, Travesía da Choupana SIN, Santiago de Compostela 15706, Spain.
| | - Carlos Torrijos-Pulpón
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), The NEIRID Group (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Santiago University Clinical Hospital, Building C, Travesía da Choupana SIN, Santiago de Compostela 15706, Spain.
| | - Maurizio Capuozzo
- Pharmaceutical Department, ASL-Napoli-3 Sud, Via Marittima 3, Ercolano 80056, Italy.
| | - Alessando Ottaiano
- Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Via M. Semmola, Naples 80131, Italy.
| | - Francisca Lago
- Molecular and Cellular Cardiology Group, SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), Research Laboratory 7, Santiago University Clinical Hospital, Santiago de Compostela 15706, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain.
| | - Antonio Mera
- SERGAS, Servizo Galego de Saude, Santiago University Clinical Hospital, Division of Rheumatology, Santiago de Compostela 15706, Spain.
| | - Jesus Pino
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), The NEIRID Group (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Santiago University Clinical Hospital, Building C, Travesía da Choupana SIN, Santiago de Compostela 15706, Spain; International PhD School, University of Santiago de Compostela (EDIUS), Santiago de Compostela 15706, Spain; University of Santiago de Compostela, Department of Surgery and Medical Surgical Specialties, Santiago University Clinical Hospital, Trav. Choupana s/n, 15706, Santiago de Compostela, Spain.
| | - Oreste Gualillo
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), The NEIRID Group (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Santiago University Clinical Hospital, Building C, Travesía da Choupana SIN, Santiago de Compostela 15706, Spain; International PhD School, University of Santiago de Compostela (EDIUS), Santiago de Compostela 15706, Spain.
| |
Collapse
|
5
|
Trimarco A, Audano M, Marca RL, Cariello M, Falco M, Pedretti S, Imperato G, Cestaro A, Podini P, Dina G, Quattrini A, Massimino L, Caruso D, Mitro N, Taveggia C. Prostaglandin D2 synthase controls Schwann cells metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.29.582775. [PMID: 38496560 PMCID: PMC10942270 DOI: 10.1101/2024.02.29.582775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
We previously reported that in the absence of Prostaglandin D2 synthase (L-PGDS) peripheral nerves are hypomyelinated in development and that with aging they present aberrant myelin sheaths. We now demonstrate that L-PGDS expressed in Schwann cells is part of a coordinated program aiming at preserving myelin integrity. In vivo and in vitro lipidomic, metabolomic and transcriptomic analyses confirmed that myelin lipids composition, Schwann cells energetic metabolism and key enzymes controlling these processes are altered in the absence of L-PGDS. Moreover, Schwann cells undergo a metabolic rewiring and turn to acetate as the main energetic source. Further, they produce ketone bodies to ensure glial cell and neuronal survival. Importantly, we demonstrate that all these changes correlate with morphological myelin alterations and describe the first physiological pathway implicated in preserving PNS myelin. Collectively, we posit that myelin lipids serve as a reservoir to provide ketone bodies, which together with acetate represent the adaptive substrates Schwann cells can rely on to sustain the axo-glial unit and preserve the integrity of the PNS.
Collapse
|
6
|
Pan L, Trimarco A, Zhang AJ, Fujimori K, Urade Y, Sun LO, Taveggia C, Zhang Y. Oligodendrocyte-lineage cell exocytosis and L-type prostaglandin D synthase promote oligodendrocyte development and myelination. eLife 2023; 12:e77441. [PMID: 36779701 PMCID: PMC9946447 DOI: 10.7554/elife.77441] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 02/12/2023] [Indexed: 02/14/2023] Open
Abstract
In the developing central nervous system, oligodendrocyte precursor cells (OPCs) differentiate into oligodendrocytes, which form myelin around axons. Oligodendrocytes and myelin are essential for the function of the central nervous system, as evidenced by the severe neurological symptoms that arise in demyelinating diseases such as multiple sclerosis and leukodystrophy. Although many cell-intrinsic mechanisms that regulate oligodendrocyte development and myelination have been reported, it remains unclear whether interactions among oligodendrocyte-lineage cells (OPCs and oligodendrocytes) affect oligodendrocyte development and myelination. Here, we show that blocking vesicle-associated membrane protein (VAMP) 1/2/3-dependent exocytosis from oligodendrocyte-lineage cells impairs oligodendrocyte development, myelination, and motor behavior in mice. Adding oligodendrocyte-lineage cell-secreted molecules to secretion-deficient OPC cultures partially restores the morphological maturation of oligodendrocytes. Moreover, we identified L-type prostaglandin D synthase as an oligodendrocyte-lineage cell-secreted protein that promotes oligodendrocyte development and myelination in vivo. These findings reveal a novel autocrine/paracrine loop model for the regulation of oligodendrocyte and myelin development.
Collapse
Affiliation(s)
- Lin Pan
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| | - Amelia Trimarco
- Division of Neuroscience, IRCCS, San Raffaele HospitalMilanItaly
| | - Alice J Zhang
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| | - Ko Fujimori
- Department of Pathobiochemistry, Osaka Medical and Pharmaceutical UniversityOsakaJapan
| | - Yoshihiro Urade
- Hirono Satellite, Isotope Science Center, The University of TokyoFukushimaJapan
| | - Lu O Sun
- Department of Molecular Biology, University of Texas Southwestern Medical CenterDallasUnited States
| | - Carla Taveggia
- Division of Neuroscience, IRCCS, San Raffaele HospitalMilanItaly
| | - Ye Zhang
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
- Brain Research Institute, University of California, Los AngelesLos AngelesUnited States
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los AngelesLos AngelesUnited States
- Molecular Biology Institute, University of California, Los AngelesLos AngelesUnited States
| |
Collapse
|
7
|
Zhu Y, Xiao H, Yu T, Cai D, Zhou Q, Zhou G, Wang L. An immune risk score predicts survival of patients with diffuse large B-cell lymphoma. Leuk Res 2023; 125:107008. [PMID: 36630885 DOI: 10.1016/j.leukres.2022.107008] [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: 11/16/2022] [Revised: 12/22/2022] [Accepted: 12/28/2022] [Indexed: 12/30/2022]
Abstract
PURPOSE Providing accurate prognostic models is necessary for diffuse large B-cell lymphoma, but there are still many uncertainties. So far, none of the models include immune cells. Therefore, an immune risk score was constructed to predict the survival of patients. METHODS CIBERSORTx was chosen to estimate the proportion of 22 human immune cell subsets from public datasets and generate an immune risk score to predict patients' survival in a training cohort using the least absolute shrinkage and selection operator (LASSO) Cox regression model. RESULTS The prognostic model had high predictive ability in the training and validation cohorts. Subjects in the training cohort with high scores had a worse prognosis compared with subjects with low scores. The same result was also found in the three validation cohorts. Multivariable analysis suggested that the immune risk score was an independent prognostic factor. The merged score, including the immune risk score and the international prognostic index (IPI) risk category, had better predictive accuracy. CONCLUSIONS Our immune risk score promises to be a complement to current prognostic models.
Collapse
Affiliation(s)
- Yu Zhu
- Department of Hematology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Han Xiao
- Department of Hematology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Tingyu Yu
- Department of Hematology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Duo Cai
- Department of Hematology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Qiao Zhou
- Department of Hematology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Guangyu Zhou
- Department of Hematology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Li Wang
- Department of Hematology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China..
| |
Collapse
|
8
|
Islam MA, Khairnar R, Fleishman J, Thompson K, Kumar S. Lipocalin-Type Prostaglandin D 2 Synthase Protein- A Central Player in Metabolism. Pharm Res 2022; 39:2951-2963. [PMID: 35799081 DOI: 10.1007/s11095-022-03329-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/29/2022] [Indexed: 11/28/2022]
Abstract
Lipocalin-type prostaglandin D synthase was previously known as β-trace protein (BTP), a low-molecular-weight glycoprotein that is heavily expressed in human cerebrospinal fluid. Nevertheless, it is also seen to be expressed in numerous other tissues including the kidney, liver, lung, heart, adipose, muscle, and pancreas. Functionally, L-PGDS behaves like a lipocalin type protein where it helps in binding and transportation of small lipophilic substances, such as steroids, retinoids, and other lipophilic ligands. Enzymatically, L-PGDS functions as a prostaglandin synthase where it helps in the production of PGD2 by catalyzing the isomerization of PGH2, a common precursor of the two series of prostaglandins. PGD2 regulates its physiological function through two individual receptors named DP1 and DP2. L-PGDS has been a central player in many diseases, its role in metabolism including diabetes, fatty liver disease, and obesity has gathered a large attention. In this review, we summarize the current state of knowledge about L-PGDS and it's signaling in adipose, hepatic, skeletal muscle, and pancreas tissues, which are core targets for metabolic studies. Modulation of L-PGDS signaling can be considered as a potential future therapeutic target for the treatment of insulin resistance as well as fatty liver disease.
Collapse
Affiliation(s)
- Md Asrarul Islam
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, SAH 141A, 8000 Utopia Parkway, Queens, NY, 11439, USA
| | - Rhema Khairnar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, SAH 141A, 8000 Utopia Parkway, Queens, NY, 11439, USA
| | - Joshua Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, SAH 141A, 8000 Utopia Parkway, Queens, NY, 11439, USA
| | - Kamala Thompson
- Department of Biology, Chemistry, and Environmental Studies, Molloy College, Rockville Centre, NY, 11571, USA
| | - Sunil Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, SAH 141A, 8000 Utopia Parkway, Queens, NY, 11439, USA.
| |
Collapse
|
9
|
Hu S, Ren S, Cai Y, Liu J, Han Y, Zhao Y, Yang J, Zhou X, Wang X. Glycoprotein PTGDS promotes tumorigenesis of diffuse large B-cell lymphoma by MYH9-mediated regulation of Wnt-β-catenin-STAT3 signaling. Cell Death Differ 2022; 29:642-656. [PMID: 34743203 PMCID: PMC8901925 DOI: 10.1038/s41418-021-00880-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 09/20/2021] [Indexed: 02/06/2023] Open
Abstract
Glycoprotein prostaglandin D2 synthase (PTGDS) is a member of the lipocalin superfamily and plays dual roles in prostaglandins metabolism and lipid transport. PTGDS has been involved in various cellular processes including the tumorigenesis of solid tumors, yet its role in carcinogenesis is contradictory and the significance of PTGDS in hematological malignancies is ill-defined. Here, we aimed to explore the expression and function of PTGDS in diffuse large B-cell lymphoma (DLBCL), especially the potential role of PTGDS inhibitor, AT56, in lymphoma therapy. Remarkable high expression of PTGDS was found in DLBCL, which was significantly correlated with poor prognosis. PTGDS overexpression and rhPTGDS were found to promote cell proliferation. Besides, in vitro and in vivo studies indicated that PTGDS knockdown and AT56 treatment exerted an anti-tumor effect by regulating cell viability, proliferation, apoptosis, cell cycle, and invasion, and enhanced the drug sensitivity to adriamycin and bendamustine through promoting DNA damage. Moreover, the co-immunoprecipitation-based mass spectrum identified the interaction between PTGDS and MYH9, which was found to promote DLBCL progression. PTGDS inhibition led to reduced expression of MYH9, and then declined activation of the Wnt-β-catenin-STAT3 pathway through influencing the ubiquitination and degradation of GSK3-β in DLBCL. The rescue experiment demonstrated that PTGDS exerted an oncogenic role through regulating MYH9 and then the Wnt-β-catenin-STAT3 pathway. Based on point mutation of glycosylation sites, we confirmed the N-glycosylation of PTGDS in Asn51 and Asn78 and found that abnormal glycosylation of PTGDS resulted in its nuclear translocation, prolonged half-life, and enhanced cell proliferation. Collectively, our findings identified for the first time that glycoprotein PTGDS promoted tumorigenesis of DLBCL through MYH9-mediated regulation of Wnt-β-catenin-STAT3 signaling, and highlighted the potential role of AT56 as a novel therapeutic strategy for DLBCL treatment.
Collapse
Affiliation(s)
- Shunfeng Hu
- grid.27255.370000 0004 1761 1174Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 250021 Jinan, Shandong China
| | - Shuai Ren
- grid.27255.370000 0004 1761 1174Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 250021 Jinan, Shandong China
| | - Yiqing Cai
- grid.27255.370000 0004 1761 1174Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 250021 Jinan, Shandong China
| | - Jiarui Liu
- grid.27255.370000 0004 1761 1174Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 250021 Jinan, Shandong China
| | - Yang Han
- grid.27255.370000 0004 1761 1174Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 250021 Jinan, Shandong China
| | - Yi Zhao
- grid.27255.370000 0004 1761 1174Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 250021 Jinan, Shandong China
| | - Juan Yang
- grid.27255.370000 0004 1761 1174Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 250021 Jinan, Shandong China
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 250021, Jinan, Shandong, China. .,Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021, Jinan, Shandong, China. .,School of Medicine, Shandong University, 250012, Jinan, Shandong, China. .,Shandong Provincial Engineering Research Center of Lymphoma, 250021, Jinan, Shandong, China. .,Branch of National Clinical Research Center for Hematologic Diseases, 250021, Jinan, Shandong, China. .,National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, 251006, Suzhou, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, 250021, Jinan, Shandong, China. .,Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021, Jinan, Shandong, China. .,School of Medicine, Shandong University, 250012, Jinan, Shandong, China. .,Shandong Provincial Engineering Research Center of Lymphoma, 250021, Jinan, Shandong, China. .,Branch of National Clinical Research Center for Hematologic Diseases, 250021, Jinan, Shandong, China. .,National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, 251006, Suzhou, China.
| |
Collapse
|
10
|
Li C, Li J, Loreno EG, Miriyala S, Panchatcharam M, Sun H. Protective Effect of Low-Dose Alcohol Consumption against Post-Ischemic Neuronal Apoptosis: Role of L-PGDS. Int J Mol Sci 2021; 23:ijms23010133. [PMID: 35008575 PMCID: PMC8745720 DOI: 10.3390/ijms23010133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/15/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke is one of the leading causes of permanent disability and death in adults worldwide. Apoptosis is a major element contributing to post-ischemic neuronal death. We previously found that low-dose alcohol consumption (LAC) protects against neuronal apoptosis in the peri-infarct cortex following transient focal cerebral ischemia. Lipocalin-type prostaglandin D2 synthase (L-PGDS), which is mainly localized in the central nervous system (CNS), was previously shown to inhibit neuronal apoptosis. Therefore, we determined whether L-PGDS is involved in the protective effect of LAC against post-ischemic neuronal apoptosis. Wild-type (WT), CaMKIIαCreERT2/+/L-PGDS+/+, and CaMKIIαCreERT2/+/L-PGDSflox/flox mice on a C57BL/6J background were gavage fed with ethanol or volume-matched water once a day for 8 weeks. Tamoxifen (2 mg/day) was given intraperitoneally to CaMKIIαCreERT2/+/L-PGDS+/+ and CaMKIIαCreERT2/+/L-PGDSflox/flox mice for 5 days during the fourth week. AT-56 (30 mg/kg/day), a selective inhibitor of L-PGDS, was given orally to AT-56-treated WT mice from the fifth week for four weeks. Cerebral ischemia/reperfusion (I/R) injury, TUNEL-positive neurons, and cleaved caspase-3-positive neurons were measured at 24 h of reperfusion after a 90 min unilateral middle cerebral artery occlusion (MCAO). We found that 0.7 g/kg/day but not 2.8 g/kg/day ethanol significantly upregulated L-PGDS in the cerebral cortex. In addition, 0.7 g/kg/day ethanol diminished cerebral ischemia/reperfusion (I/R) injury and TUNEL-positive and cleaved caspase-3-positive neurons in the peri-infarct cortex in WT and CaMKIIαCreERT2/+/L-PGDS+/+ mice. Furthermore, the neuroprotective effect of 0.7 g/kg/day ethanol was alleviated in AT-56-treated WT and CaMKIIαCreERT2/+/L-PGDSflox/flox mice. Our findings suggest that LAC may protect against cerebral I/R injury by suppressing post-ischemic neuronal apoptosis via an upregulated L-PGDS.
Collapse
Affiliation(s)
| | | | | | | | | | - Hong Sun
- Correspondence: ; Tel.: +1-318-675-4566; Fax: +1-318-675-5889
| |
Collapse
|
11
|
Urade Y. Biochemical and Structural Characteristics, Gene Regulation, Physiological, Pathological and Clinical Features of Lipocalin-Type Prostaglandin D 2 Synthase as a Multifunctional Lipocalin. Front Physiol 2021; 12:718002. [PMID: 34744762 PMCID: PMC8569824 DOI: 10.3389/fphys.2021.718002] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/01/2021] [Indexed: 11/13/2022] Open
Abstract
Lipocalin-type prostaglandin (PG) D2 synthase (L-PGDS) catalyzes the isomerization of PGH2, a common precursor of the two series of PGs, to produce PGD2. PGD2 stimulates three distinct types of G protein-coupled receptors: (1) D type of prostanoid (DP) receptors involved in the regulation of sleep, pain, food intake, and others; (2) chemoattractant receptor-homologous molecule expressed on T helper type 2 cells (CRTH2) receptors, in myelination of peripheral nervous system, adipocyte differentiation, inhibition of hair follicle neogenesis, and others; and (3) F type of prostanoid (FP) receptors, in dexamethasone-induced cardioprotection. L-PGDS is the same protein as β-trace, a major protein in human cerebrospinal fluid (CSF). L-PGDS exists in the central nervous system and male genital organs of various mammals, and human heart; and is secreted into the CSF, seminal plasma, and plasma, respectively. L-PGDS binds retinoic acids and retinal with high affinities (Kd < 100 nM) and diverse small lipophilic substances, such as thyroids, gangliosides, bilirubin and biliverdin, heme, NAD(P)H, and PGD2, acting as an extracellular carrier of these substances. L-PGDS also binds amyloid β peptides, prevents their fibril formation, and disaggregates amyloid β fibrils, acting as a major amyloid β chaperone in human CSF. Here, I summarize the recent progress of the research on PGD2 and L-PGDS, in terms of its “molecular properties,” “cell culture studies,” “animal experiments,” and “clinical studies,” all of which should help to understand the pathophysiological role of L-PGDS and inspire the future research of this multifunctional lipocalin.
Collapse
Affiliation(s)
- Yoshihiro Urade
- Center for Supporting Pharmaceutical Education, Daiichi University of Pharmacy, Fukuoka, Japan.,Isotope Science Center, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
12
|
Li L, Lai EY, Cao X, Welch WJ, Wilcox CS. Endothelial prostaglandin D 2 opposes angiotensin II contractions in mouse isolated perfused intracerebral microarterioles. J Renin Angiotensin Aldosterone Syst 2020; 21:1470320320966177. [PMID: 33094663 PMCID: PMC7585895 DOI: 10.1177/1470320320966177] [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] [Indexed: 12/29/2022] Open
Abstract
Hypothesis: A lack of contraction of cerebral microarterioles to Ang II (“resilience”) depends on cyclooxygenase (COX) and lipocalin type prostaglandin D sythase L-PGDS producing PGD2 that activates prostaglandin D type 1 receptors (DP1Rs) and nitric oxide synthase (NOS). Materials & Methods: Contractions were assessed in isolated, perfused vessels and NO by fluorescence microscopy. Results: The mRNAs of penetrating intraparenchymal cerebral microarterioles versus renal afferent arterioles were >3000-fold greater for L-PGDS and DP1R and 5-fold for NOS III and COX 2. Larger cerebral arteries contracted with Ang II. However, cerebral microarterioles were entirely unresponsive but contracted with endothelin 1 and perfusion pressure. Ang II contractions were evoked in cerebral microarterioles from COX1 –/– mice or after blockade of COX2, L-PGDS or NOS and in deendothelialized vessels but effects of deendothelialization were lost during COX blockade. NO generation with Ang II depended on COX and also was increased by DP1R activation. Conclusion: The resilience of cerebral arterioles to Ang II contractions is specific for intraparenchymal microarterioles and depends on endothelial COX1 and two products that are metabolized by L-PGDS to generate PGD2 that signals via DP1Rs and NO.
Collapse
Affiliation(s)
- L Li
- Hypertension Center and Division of Nephrology and Hypertension, Georgetown University, Washington DC, USA.,Kidney Disease Center, the First Affiliated Hospital and Department of Physiology, School of Basic Medical Science, Zhejiang University School of Medicine, Hangzhou, China
| | - E Y Lai
- Hypertension Center and Division of Nephrology and Hypertension, Georgetown University, Washington DC, USA.,Kidney Disease Center, the First Affiliated Hospital and Department of Physiology, School of Basic Medical Science, Zhejiang University School of Medicine, Hangzhou, China
| | - X Cao
- Kidney Disease Center, the First Affiliated Hospital and Department of Physiology, School of Basic Medical Science, Zhejiang University School of Medicine, Hangzhou, China
| | - W J Welch
- Hypertension Center and Division of Nephrology and Hypertension, Georgetown University, Washington DC, USA
| | - C S Wilcox
- Hypertension Center and Division of Nephrology and Hypertension, Georgetown University, Washington DC, USA
| |
Collapse
|
13
|
Tavares-Ferreira D, Ray PR, Sankaranarayanan I, Mejia GL, Wangzhou A, Shiers S, Uttarkar R, Megat S, Barragan-Iglesias P, Dussor G, Akopian AN, Price TJ. Sex Differences in Nociceptor Translatomes Contribute to Divergent Prostaglandin Signaling in Male and Female Mice. Biol Psychiatry 2020; 91:129-140. [PMID: 33309016 PMCID: PMC8019688 DOI: 10.1016/j.biopsych.2020.09.022] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/22/2020] [Accepted: 09/23/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND There are clinically relevant sex differences in acute and chronic pain mechanisms, but we are only beginning to understand their mechanistic basis. Transcriptome analyses of rodent whole dorsal root ganglion (DRG) have revealed sex differences, mostly in immune cells. We examined the transcriptome and translatome of the mouse DRG with the goal of identifying sex differences. METHODS We used translating ribosome affinity purification sequencing and behavioral pharmacology to test the hypothesis that in Nav1.8-positive neurons, most of which are nociceptors, translatomes would differ by sex. RESULTS We found 80 genes with sex differential expression in the whole DRG transcriptome and 66 genes whose messenger RNAs were sex differentially actively translated (translatome). We also identified different motifs in the 3' untranslated region of messenger RNAs that were sex differentially translated. In further validation studies, we focused on Ptgds, which was increased in the translatome of female mice. The messenger RNA encodes the prostaglandin PGD2 synthesizing enzyme. We observed increased PTGDS protein and PGD2 in female mouse DRG. The PTGDS inhibitor AT-56 caused intense pain behaviors in male mice but was only effective at high doses in female mice. Conversely, female mice responded more robustly to another major prostaglandin, PGE2, than did male mice. PTGDS protein expression was also higher in female cortical neurons, suggesting that DRG findings may be generalizable to other nervous system structures. CONCLUSIONS Our results demonstrate sex differences in nociceptor-enriched translatomes and reveal unexpected sex differences in one of the oldest known nociceptive signaling molecule families, the prostaglandins.
Collapse
Affiliation(s)
- Diana Tavares-Ferreira
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies
| | - Pradipta R. Ray
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies
| | | | - Galo L. Mejia
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies
| | - Andi Wangzhou
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies
| | - Stephanie Shiers
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies
| | - Ruta Uttarkar
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies
| | - Salim Megat
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies
| | | | - Gregory Dussor
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies
| | - Armen N. Akopian
- University of Texas Health San Antonio, Department of Endodontics
| | - Theodore J. Price
- University of Texas at Dallas, Department of Neuroscience and Center for Advanced Pain Studies,correspondence to Theodore J Price – , 800 W Campbell Rd, Richardson TX 75080, USA, 972-883-4311
| |
Collapse
|
14
|
Asirvatham-Jeyaraj N, Jones AD, Burnett R, Fink GD. Brain Prostaglandin D2 Increases Neurogenic Pressor Activity and Mean Arterial Pressure in Angiotensin II-Salt Hypertensive Rats. Hypertension 2019; 74:1499-1506. [PMID: 31587572 DOI: 10.1161/hypertensionaha.119.13175] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
This study tested whether brain L-PGDS (lipocalin-type prostaglandin [PG] D synthase), through prostanoid signaling, might increase neurogenic pressor activity and thereby cause hypertension. Sprague Dawley rats on high-salt diet received either vehicle or Ang II (angiotensin II) infusion. On day 4, the developmental stage of hypertension, brains from different sets of control and Ang II-treated rats were collected for measuring L-PGDS expression, PGD2 levels, and DP1R (type 1 PGD2 receptor) expression. In a different set of 14-day Ang II-salt-treated rats, mini-osmotic pumps were used to infuse either a nonselective COX (cyclooxygenase) inhibitor ketorolac, L-PGDS inhibitor AT56, or DP1R inhibitor BWA868C to test the role of brain COX-PGD2-DP1R signaling in Ang II-salt hypertension. The acute depressor response to ganglion blockade with hexamethonium was used to quantify neurogenic pressor activity. During the developmental stage of Ang II-salt hypertension, L-PGDS expression was higher in cerebrospinal fluid, and PGD2 levels were increased in the choroid plexus, cerebrospinal fluid, and the cardioregulatory brain region rostral ventrolateral medulla. DP1R expression was decreased in rostral ventrolateral medulla. Both brain COX inhibition with ketorolac and L-PGDS inhibition with AT56 lowered mean arterial pressure by altering neurogenic pressor activity compared with vehicle controls. Blockade of DP1R with BWA868C, however, increased the magnitude of Ang II-salt hypertension and significantly increased neurogenic pressor activity. In summary, we establish that the development of Ang II-salt hypertension requires increased COX- and L-PGDS-derived PGD2 production in the brain, making L-PGDS a possible target for treating neurogenic hypertension.
Collapse
Affiliation(s)
- Ninitha Asirvatham-Jeyaraj
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru (N.A.-J.).,Department of Pharmacology and Toxicology (N.A.-J., R.B., G.D.F.), Michigan State University, East Lansing
| | - A Daniel Jones
- Department of Biochemistry and Molecular Biology (A.D.J.), Michigan State University, East Lansing.,Department of Chemistry (A.D.J.), Michigan State University, East Lansing
| | - Robert Burnett
- Department of Pharmacology and Toxicology (N.A.-J., R.B., G.D.F.), Michigan State University, East Lansing
| | - Gregory D Fink
- Department of Pharmacology and Toxicology (N.A.-J., R.B., G.D.F.), Michigan State University, East Lansing
| |
Collapse
|
15
|
Ahmad AS, Ottallah H, Maciel CB, Strickland M, Doré S. Role of the L-PGDS-PGD2-DP1 receptor axis in sleep regulation and neurologic outcomes. Sleep 2019; 42:zsz073. [PMID: 30893431 PMCID: PMC6559173 DOI: 10.1093/sleep/zsz073] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 02/16/2019] [Indexed: 12/18/2022] Open
Abstract
To meet the new challenges of modern lifestyles, we often compromise a good night's sleep. In preclinical models as well as in humans, a chronic lack of sleep is reported to be among the leading causes of various physiologic, psychologic, and neurocognitive deficits. Thus far, various endogenous mediators have been implicated in inter-regulatory networks that collectively influence the sleep-wake cycle. One such mediator is the lipocalin-type prostaglandin D2 synthase (L-PGDS)-Prostaglandin D2 (PGD2)-DP1 receptor (L-PGDS-PGD2-DP1R) axis. Findings in preclinical models confirm that DP1R are predominantly expressed in the sleep-regulating centers. This finding led to the discovery that the L-PGDS-PGD2-DP1R axis is involved in sleep regulation. Furthermore, we showed that the L-PGDS-PGD2-DP1R axis is beneficial in protecting the brain from ischemic stroke. Protein sequence homology was also performed, and it was found that L-PGDS and DP1R share a high degree of homology between humans and rodents. Based on the preclinical and clinical data thus far pertaining to the role of the L-PGDS-PGD2-DP1R axis in sleep regulation and neurologic conditions, there is optimism that this axis may have a high translational potential in human therapeutics. Therefore, here the focus is to review the regulation of the homeostatic component of the sleep process with a special focus on the L-PGDS-PGD2-DP1R axis and the consequences of sleep deprivation on health outcomes. Furthermore, we discuss whether the pharmacological regulation of this axis could represent a tool to prevent sleep disturbances and potentially improve outcomes, especially in patients with acute brain injuries.
Collapse
Affiliation(s)
- Abdullah Shafique Ahmad
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL
- McKnight Brain Institute, University of Florida, Gainesville, FL
| | - Haneen Ottallah
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL
- McKnight Brain Institute, University of Florida, Gainesville, FL
| | - Carolina B Maciel
- Department of Neurology, University of Florida College of Medicine, Gainesville, FL
| | - Michael Strickland
- Division of Biology and Biomedical Sciences, Washington University in Saint Louis, Saint Louis, MO
| | - Sylvain Doré
- Department of Anesthesiology, University of Florida College of Medicine, Gainesville, FL
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL
- McKnight Brain Institute, University of Florida, Gainesville, FL
- Department of Psychiatry, University of Florida, Gainesville, FL
- Department of Pharmaceutics, University of Florida, Gainesville, FL
- Department of Psychology, University of Florida, Gainesville, FL
- Department of Neuroscience, University of Florida, Gainesville, FL
| |
Collapse
|
16
|
Yeh YN, Hsin KY, Zimmer A, Lin LY, Hung MS. A structure-function approach identifies L-PGDS as a mediator responsible for glucocorticoid-induced leptin expression in adipocytes. Biochem Pharmacol 2019; 166:203-211. [PMID: 31129049 DOI: 10.1016/j.bcp.2019.05.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 05/21/2019] [Indexed: 02/01/2023]
Abstract
Leptin is an adipokine predominantly secreted by adipocytes and has many physiological roles, including in energy homeostasis. We identified that AM630, a cannabinoid receptor 2 (CB2) antagonist, down-regulated leptin expression in mature adipocytes differentiated from either stromal vascular fractions isolated from inguinal fat pads of C57BL/6J mice or 3T3-L1 preadipocytes. However, the leptin-suppressive effects of AM630 preserved in CB2-deficient adipocytes indicated the off-target activity of AM630 in leptin expression. Pharmacological and genetic studies, cheminformatics, and docking simulation were applied to identify the potential protein target of AM630 that modulates leptin expression in differentiated primary preadipocytes. Screening of the reported off-targets of AM630 identified a synthetic cannabinoid WIN55212-2 exerting the same function. Target deconvolution and docking simulation suggested that AM630 and WIN55212-2 were both inhibitors of lipocalin-type prostaglandin D2 synthase (L-PGDS). Further studies showed that L-PGDS positively regulates leptin expression. Although glucocorticoid and aldosterone were previously reported to induce expression of both L-PGDS and leptin, our data demonstrated that L-PGDS mediates only glucocorticoid-induced leptin expression in differentiated primary preadipocytes. No effect was observed after aldosterone treatment. This newly discovered glucocorticoid - L-PGDS - leptin pathway may provide insights into current clinical use of glucocorticoid and management of their undesired effects such as obesity.
Collapse
Affiliation(s)
- Yen-Nan Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan; Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Kun-Yi Hsin
- Okinawa Institute of Science and Technology Graduate University, Okinawa 904-0496, Japan; Department of Animal Science, National Chung Hsing University, Taichung 40227, Taiwan
| | - Andreas Zimmer
- Institute for Molecular Psychiatry, University of Bonn, 53113 Bonn, Germany
| | - Lih-Yuan Lin
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Ming-Shiu Hung
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan.
| |
Collapse
|
17
|
Wang TA, Teo CF, Åkerblom M, Chen C, Tynan-La Fontaine M, Greiner VJ, Diaz A, McManus MT, Jan YN, Jan LY. Thermoregulation via Temperature-Dependent PGD 2 Production in Mouse Preoptic Area. Neuron 2019; 103:309-322.e7. [PMID: 31151773 DOI: 10.1016/j.neuron.2019.04.035] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 04/03/2019] [Accepted: 04/24/2019] [Indexed: 12/30/2022]
Abstract
Body temperature control is essential for survival. In mammals, thermoregulation is mediated by the preoptic area of anterior hypothalamus (POA), with ∼30% of its neurons sensitive to brain temperature change. It is still unknown whether and how these temperature-sensitive neurons are involved in thermoregulation, because for eight decades they have only been identified via electrophysiological recording. By combining single-cell RNA-seq with whole-cell patch-clamp recordings, we identified Ptgds as a genetic marker for temperature-sensitive POA neurons. Then, we demonstrated these neurons' role in thermoregulation via chemogenetics. Given that Ptgds encodes the enzyme that synthesizes prostaglandin D2 (PGD2), we further explored its role in thermoregulation. Our study revealed that rising temperature of POA alters the activity of Ptgds-expressing neurons so as to increase PGD2 production. PGD2 activates its receptor DP1 and excites downstream neurons in the ventral medial preoptic area (vMPO) that mediates body temperature decrease, a negative feedback loop for thermoregulation.
Collapse
Affiliation(s)
- Tongfei A Wang
- Departments of Physiology, Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Chin Fen Teo
- Departments of Physiology, Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Malin Åkerblom
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94158, USA; UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Chao Chen
- Departments of Physiology, Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Marena Tynan-La Fontaine
- Departments of Physiology, Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Vanille Juliette Greiner
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94158, USA; UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Aaron Diaz
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Michael T McManus
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94158, USA; UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yuh Nung Jan
- Departments of Physiology, Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lily Y Jan
- Departments of Physiology, Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
18
|
Peinhaupt M, Sturm EM, Heinemann A. Prostaglandins and Their Receptors in Eosinophil Function and As Therapeutic Targets. Front Med (Lausanne) 2017; 4:104. [PMID: 28770200 PMCID: PMC5515835 DOI: 10.3389/fmed.2017.00104] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/27/2017] [Indexed: 02/06/2023] Open
Abstract
Of the known prostanoid receptors, human eosinophils express the prostaglandin D2 (PGD2) receptors DP1 [also D-type prostanoid (DP)] and DP2 (also chemoattractant receptor homologous molecule, expressed on Th2 cells), the prostaglandin E2 receptors EP2 and EP4, and the prostacyclin (PGI2) receptor IP. Prostanoids can bind to either one or multiple receptors, characteristically have a short half-life in vivo, and are quickly degraded into metabolites with altered affinity and specificity for a given receptor subtype. Prostanoid receptors signal mainly through G proteins and naturally activate signal transduction pathways according to the G protein subtype that they preferentially interact with. This can lead to the activation of sometimes opposing signaling pathways. In addition, prostanoid signaling is often cell-type specific and also the combination of expressed receptors can influence the outcome of the prostanoid impulse. Accordingly, it is assumed that eosinophils and their (patho-)physiological functions are governed by a sensitive prostanoid signaling network. In this review, we specifically focus on the functions of PGD2, PGE2, and PGI2 and their receptors on eosinophils. We discuss their significance in allergic and non-allergic diseases and summarize potential targets for drug intervention.
Collapse
Affiliation(s)
- Miriam Peinhaupt
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Eva M Sturm
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - Akos Heinemann
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| |
Collapse
|
19
|
Le Loupp AG, Bach-Ngohou K, Bourreille A, Boudin H, Rolli-Derkinderen M, Denis MG, Neunlist M, Masson D. Activation of the prostaglandin D2 metabolic pathway in Crohn's disease: involvement of the enteric nervous system. BMC Gastroenterol 2015; 15:112. [PMID: 26338799 PMCID: PMC4558965 DOI: 10.1186/s12876-015-0338-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 08/24/2015] [Indexed: 02/08/2023] Open
Abstract
Background Recent works provide evidence of the importance of the prostaglandin D2 (PGD2) metabolic pathway in inflammatory bowel diseases. We investigated the expression of PGD2 metabolic pathway actors in Crohn’s disease (CD) and the ability of the enteric nervous system (ENS) to produce PGD2 in inflammatory conditions. Methods Expression of key actors involved in the PGD2 metabolic pathway and its receptors was analyzed using quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) in colonic mucosal biopsies of patients from three groups: controls, quiescent and active CD patients. To determine the ability of the ENS to secrete PGD2 in proinflammatory conditions, Lipocalin-type prostaglandin D synthase (L-PGDS) expression by neurons and glial cells was analyzed by immunostaining. PGD2 levels were determined in a medium of primary culture of ENS and neuro-glial coculture model treated by lipopolysaccharide (LPS). Results In patients with active CD, inflamed colonic mucosa showed significantly higher COX2 and L-PGDS mRNA expression, and significantly higher PGD2 levels than healthy colonic mucosa. On the contrary, peroxysome proliferator-activated receptor Gamma (PPARG) expression was reduced in inflamed colonic mucosa of CD patients with active disease. Immunostaining showed that L-PGDS was expressed in the neurons of human myenteric and submucosal plexi. A rat ENS primary culture model confirmed this expression. PGD2 levels were significantly increased on primary culture of ENS treated with LPS. This production was abolished by AT-56, a specific competitive L-PGDS inhibitor. The neuro-glial coculture model revealed that each component of the ENS, ECG and neurons, could contribute to PGD2 production. Conclusions Our results highlight the activation of the PGD2 metabolic pathway in Crohn’s disease. This study supports the hypothesis that in Crohn’s disease, enteric neurons and glial cells form a functional unit reacting to inflammation by producing PGD2.
Collapse
Affiliation(s)
- Anne-Gaelle Le Loupp
- INSERM Unité 913, 1 rue Gaston Veil, Nantes, F-44035, France. .,Université Nantes, 1 quai de Tourville, BP 13522, Nantes, F-44035, France. .,Institut des Maladies de l'Appareil Digestif, 1 place Alexis Ricordeau, Nantes, F-44093, France. .,Laboratoire de Biochimie, Institut de Biologie, CHU de Nantes, 1 place Alexis Ricordeau, Nantes, F-44093, France.
| | - Kalyane Bach-Ngohou
- INSERM Unité 913, 1 rue Gaston Veil, Nantes, F-44035, France. .,Université Nantes, 1 quai de Tourville, BP 13522, Nantes, F-44035, France. .,Institut des Maladies de l'Appareil Digestif, 1 place Alexis Ricordeau, Nantes, F-44093, France. .,Laboratoire de Biochimie, Institut de Biologie, CHU de Nantes, 1 place Alexis Ricordeau, Nantes, F-44093, France.
| | - Arnaud Bourreille
- INSERM Unité 913, 1 rue Gaston Veil, Nantes, F-44035, France. .,Université Nantes, 1 quai de Tourville, BP 13522, Nantes, F-44035, France. .,Institut des Maladies de l'Appareil Digestif, 1 place Alexis Ricordeau, Nantes, F-44093, France. .,Laboratoire de Biochimie, Institut de Biologie, CHU de Nantes, 1 place Alexis Ricordeau, Nantes, F-44093, France.
| | - Hélène Boudin
- INSERM Unité 913, 1 rue Gaston Veil, Nantes, F-44035, France. .,Université Nantes, 1 quai de Tourville, BP 13522, Nantes, F-44035, France. .,Institut des Maladies de l'Appareil Digestif, 1 place Alexis Ricordeau, Nantes, F-44093, France.
| | - Malvyne Rolli-Derkinderen
- INSERM Unité 913, 1 rue Gaston Veil, Nantes, F-44035, France. .,Université Nantes, 1 quai de Tourville, BP 13522, Nantes, F-44035, France. .,Institut des Maladies de l'Appareil Digestif, 1 place Alexis Ricordeau, Nantes, F-44093, France.
| | - Marc G Denis
- INSERM Unité 913, 1 rue Gaston Veil, Nantes, F-44035, France. .,Université Nantes, 1 quai de Tourville, BP 13522, Nantes, F-44035, France. .,Institut des Maladies de l'Appareil Digestif, 1 place Alexis Ricordeau, Nantes, F-44093, France. .,Laboratoire de Biochimie, Institut de Biologie, CHU de Nantes, 1 place Alexis Ricordeau, Nantes, F-44093, France.
| | - Michel Neunlist
- INSERM Unité 913, 1 rue Gaston Veil, Nantes, F-44035, France. .,Université Nantes, 1 quai de Tourville, BP 13522, Nantes, F-44035, France. .,Institut des Maladies de l'Appareil Digestif, 1 place Alexis Ricordeau, Nantes, F-44093, France.
| | - Damien Masson
- INSERM Unité 913, 1 rue Gaston Veil, Nantes, F-44035, France. .,Université Nantes, 1 quai de Tourville, BP 13522, Nantes, F-44035, France. .,Institut des Maladies de l'Appareil Digestif, 1 place Alexis Ricordeau, Nantes, F-44093, France. .,Laboratoire de Biochimie, Institut de Biologie, CHU de Nantes, 1 place Alexis Ricordeau, Nantes, F-44093, France.
| |
Collapse
|
20
|
Zhao W, Jiang B, Hu H, Zhang S, Lv S, Yuan J, Qian Y, Zou Y, Li X, Jiang H, Liu F, Shao C, Gong Y. Lack of CUL4B leads to increased abundance of GFAP-positive cells that is mediated by PTGDS in mouse brain. Hum Mol Genet 2015; 24:4686-97. [PMID: 26025376 DOI: 10.1093/hmg/ddv200] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Accepted: 05/26/2015] [Indexed: 01/05/2023] Open
Abstract
Astrocytes are the most abundant cell type in the mammalian brain and are important for the functions of the central nervous system. Glial fibrillary acidic protein (GFAP) is regarded as a hallmark of mature astrocytes, though some GFPA-positive cells may act as neural stem cells. Missense heterozygous mutations in GFAP cause Alexander disease that manifests leukodystrophy and intellectual disability. Here, we show that CUL4B, a scaffold protein that assembles E3 ubiquitin ligase, represses the expression of GFAP in neural progenitor cells (NPCs) during brain development. Lack of Cul4b in NPCs in cultures led to increased generation of astrocytes, marked by GFAP and S100β. The GFAP+ cells were also found to be more abundant in the brains of nervous system-specific Cul4b knockout mice in vivo. Moreover, we demonstrated that the increased generation of GFAP+ cells from Cul4b-null NPCs was mediated by an upregulation of prostaglandin D2 synthase PTGDS. We showed that the increased GFAP expression can be attenuated by pharmacological inhibition of the PTGDS enzymatic activity or by shRNA-mediated knockdown of Ptgds. Importantly, exogenously added PTGDS could promote the generation of GFAP+ cells from wild-type NPCs. We further observed that Ptgds is targeted and repressed by the CUL4B/PRC2 complex. Together, our results demonstrate CUL4B as a negative regulator of GFAP expression during neural development.
Collapse
Affiliation(s)
- Wei Zhao
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Baichun Jiang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Huili Hu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Shuqian Zhang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Shuaishuai Lv
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Jupeng Yuan
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Yanyan Qian
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Yongxin Zou
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Xi Li
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Hong Jiang
- Institute of Medical Psychology, Shandong University School of Medicine, Jinan, Shandong 250012, China and
| | - Fang Liu
- Department of Neuroscience, Centre for Addiction and Mental Health, Toronto, Ontario, Canada M5T 1R8
| | - Changshun Shao
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China,
| | - Yaoqin Gong
- The Key Laboratory of Experimental Teratology, Ministry of Education and Institute of Molecular Medicine and Genetics, Shandong University School of Medicine, Jinan, Shandong 250012, China,
| |
Collapse
|
21
|
Urbanet R, Nguyen Dinh Cat A, Feraco A, Venteclef N, El Mogrhabi S, Sierra-Ramos C, Alvarez de la Rosa D, Adler GK, Quilliot D, Rossignol P, Fallo F, Touyz RM, Jaisser F. Adipocyte Mineralocorticoid Receptor Activation Leads to Metabolic Syndrome and Induction of Prostaglandin D2 Synthase. Hypertension 2015; 66:149-57. [PMID: 25966493 DOI: 10.1161/hypertensionaha.114.04981] [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: 12/04/2014] [Accepted: 03/13/2015] [Indexed: 11/16/2022]
Abstract
Metabolic syndrome is a major risk factor for the development of diabetes mellitus and cardiovascular diseases. Pharmacological antagonism of the mineralocorticoid receptor (MR), a ligand-activated transcription factor, limits metabolic syndrome in preclinical models, but mechanistic studies are lacking to delineate the role of MR activation in adipose tissue. In this study, we report that MR expression is increased in visceral adipose tissue in a preclinical mouse model of metabolic syndrome and in obese patients. In vivo conditional upregulation of MR in mouse adipocytes led to increased weight and fat mass, insulin resistance, and metabolic syndrome features without affecting blood pressure. We identified prostaglandin D2 synthase as a novel MR target gene in adipocytes and AT56, a specific inhibitor of prostaglandin D2 synthase enzymatic activity, blunted adipogenic aldosterone effects. Moreover, translational studies showed that expression of MR and prostaglandin D2 synthase is strongly correlated in adipose tissues from obese patients.
Collapse
Affiliation(s)
- Riccardo Urbanet
- From the INSERM, UMR_S 1138, Teams 1 (R.U., A.F., S.E.M., F.J.) and 8 (N.V.), Centre de Recherche des Cordeliers, UPMC Univ Paris 06, Université Paris Descartes, Paris, France; Department of Medicine (DIMED), University of Padova, Padova, Italy (R.U., F.F.); Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (A.N.D.C., R.M.T.); Laboratory of Cardiovascular Endocrinology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Pisana, Rome, Italy (A.F.); Department of Physiology and Institute of Biomedical Technologies, University of La Laguna, Tenerife, Spain (D.A.D.l.R.); Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.K.A.); Department of Nutrition, Nancy University Hospital, INSERM U954, Medical Faculty and CHU of Nancy, Vandoeuvre-les-Nancy, France (D.Q.); and INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, and Université de Lorraine, and Investigation Network Initiative Cardiovascular and Renal Clinical Trialists (INI-CRCT) French Clinical Research Infrastructure Network (F-CRIN), Nancy, France (P.R., F.J.)
| | - Aurelie Nguyen Dinh Cat
- From the INSERM, UMR_S 1138, Teams 1 (R.U., A.F., S.E.M., F.J.) and 8 (N.V.), Centre de Recherche des Cordeliers, UPMC Univ Paris 06, Université Paris Descartes, Paris, France; Department of Medicine (DIMED), University of Padova, Padova, Italy (R.U., F.F.); Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (A.N.D.C., R.M.T.); Laboratory of Cardiovascular Endocrinology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Pisana, Rome, Italy (A.F.); Department of Physiology and Institute of Biomedical Technologies, University of La Laguna, Tenerife, Spain (D.A.D.l.R.); Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.K.A.); Department of Nutrition, Nancy University Hospital, INSERM U954, Medical Faculty and CHU of Nancy, Vandoeuvre-les-Nancy, France (D.Q.); and INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, and Université de Lorraine, and Investigation Network Initiative Cardiovascular and Renal Clinical Trialists (INI-CRCT) French Clinical Research Infrastructure Network (F-CRIN), Nancy, France (P.R., F.J.)
| | - Alessandra Feraco
- From the INSERM, UMR_S 1138, Teams 1 (R.U., A.F., S.E.M., F.J.) and 8 (N.V.), Centre de Recherche des Cordeliers, UPMC Univ Paris 06, Université Paris Descartes, Paris, France; Department of Medicine (DIMED), University of Padova, Padova, Italy (R.U., F.F.); Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (A.N.D.C., R.M.T.); Laboratory of Cardiovascular Endocrinology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Pisana, Rome, Italy (A.F.); Department of Physiology and Institute of Biomedical Technologies, University of La Laguna, Tenerife, Spain (D.A.D.l.R.); Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.K.A.); Department of Nutrition, Nancy University Hospital, INSERM U954, Medical Faculty and CHU of Nancy, Vandoeuvre-les-Nancy, France (D.Q.); and INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, and Université de Lorraine, and Investigation Network Initiative Cardiovascular and Renal Clinical Trialists (INI-CRCT) French Clinical Research Infrastructure Network (F-CRIN), Nancy, France (P.R., F.J.)
| | - Nicolas Venteclef
- From the INSERM, UMR_S 1138, Teams 1 (R.U., A.F., S.E.M., F.J.) and 8 (N.V.), Centre de Recherche des Cordeliers, UPMC Univ Paris 06, Université Paris Descartes, Paris, France; Department of Medicine (DIMED), University of Padova, Padova, Italy (R.U., F.F.); Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (A.N.D.C., R.M.T.); Laboratory of Cardiovascular Endocrinology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Pisana, Rome, Italy (A.F.); Department of Physiology and Institute of Biomedical Technologies, University of La Laguna, Tenerife, Spain (D.A.D.l.R.); Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.K.A.); Department of Nutrition, Nancy University Hospital, INSERM U954, Medical Faculty and CHU of Nancy, Vandoeuvre-les-Nancy, France (D.Q.); and INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, and Université de Lorraine, and Investigation Network Initiative Cardiovascular and Renal Clinical Trialists (INI-CRCT) French Clinical Research Infrastructure Network (F-CRIN), Nancy, France (P.R., F.J.)
| | - Soumaya El Mogrhabi
- From the INSERM, UMR_S 1138, Teams 1 (R.U., A.F., S.E.M., F.J.) and 8 (N.V.), Centre de Recherche des Cordeliers, UPMC Univ Paris 06, Université Paris Descartes, Paris, France; Department of Medicine (DIMED), University of Padova, Padova, Italy (R.U., F.F.); Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (A.N.D.C., R.M.T.); Laboratory of Cardiovascular Endocrinology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Pisana, Rome, Italy (A.F.); Department of Physiology and Institute of Biomedical Technologies, University of La Laguna, Tenerife, Spain (D.A.D.l.R.); Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.K.A.); Department of Nutrition, Nancy University Hospital, INSERM U954, Medical Faculty and CHU of Nancy, Vandoeuvre-les-Nancy, France (D.Q.); and INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, and Université de Lorraine, and Investigation Network Initiative Cardiovascular and Renal Clinical Trialists (INI-CRCT) French Clinical Research Infrastructure Network (F-CRIN), Nancy, France (P.R., F.J.)
| | - Catalina Sierra-Ramos
- From the INSERM, UMR_S 1138, Teams 1 (R.U., A.F., S.E.M., F.J.) and 8 (N.V.), Centre de Recherche des Cordeliers, UPMC Univ Paris 06, Université Paris Descartes, Paris, France; Department of Medicine (DIMED), University of Padova, Padova, Italy (R.U., F.F.); Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (A.N.D.C., R.M.T.); Laboratory of Cardiovascular Endocrinology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Pisana, Rome, Italy (A.F.); Department of Physiology and Institute of Biomedical Technologies, University of La Laguna, Tenerife, Spain (D.A.D.l.R.); Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.K.A.); Department of Nutrition, Nancy University Hospital, INSERM U954, Medical Faculty and CHU of Nancy, Vandoeuvre-les-Nancy, France (D.Q.); and INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, and Université de Lorraine, and Investigation Network Initiative Cardiovascular and Renal Clinical Trialists (INI-CRCT) French Clinical Research Infrastructure Network (F-CRIN), Nancy, France (P.R., F.J.)
| | - Diego Alvarez de la Rosa
- From the INSERM, UMR_S 1138, Teams 1 (R.U., A.F., S.E.M., F.J.) and 8 (N.V.), Centre de Recherche des Cordeliers, UPMC Univ Paris 06, Université Paris Descartes, Paris, France; Department of Medicine (DIMED), University of Padova, Padova, Italy (R.U., F.F.); Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (A.N.D.C., R.M.T.); Laboratory of Cardiovascular Endocrinology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Pisana, Rome, Italy (A.F.); Department of Physiology and Institute of Biomedical Technologies, University of La Laguna, Tenerife, Spain (D.A.D.l.R.); Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.K.A.); Department of Nutrition, Nancy University Hospital, INSERM U954, Medical Faculty and CHU of Nancy, Vandoeuvre-les-Nancy, France (D.Q.); and INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, and Université de Lorraine, and Investigation Network Initiative Cardiovascular and Renal Clinical Trialists (INI-CRCT) French Clinical Research Infrastructure Network (F-CRIN), Nancy, France (P.R., F.J.)
| | - Gail K Adler
- From the INSERM, UMR_S 1138, Teams 1 (R.U., A.F., S.E.M., F.J.) and 8 (N.V.), Centre de Recherche des Cordeliers, UPMC Univ Paris 06, Université Paris Descartes, Paris, France; Department of Medicine (DIMED), University of Padova, Padova, Italy (R.U., F.F.); Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (A.N.D.C., R.M.T.); Laboratory of Cardiovascular Endocrinology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Pisana, Rome, Italy (A.F.); Department of Physiology and Institute of Biomedical Technologies, University of La Laguna, Tenerife, Spain (D.A.D.l.R.); Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.K.A.); Department of Nutrition, Nancy University Hospital, INSERM U954, Medical Faculty and CHU of Nancy, Vandoeuvre-les-Nancy, France (D.Q.); and INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, and Université de Lorraine, and Investigation Network Initiative Cardiovascular and Renal Clinical Trialists (INI-CRCT) French Clinical Research Infrastructure Network (F-CRIN), Nancy, France (P.R., F.J.)
| | - Didier Quilliot
- From the INSERM, UMR_S 1138, Teams 1 (R.U., A.F., S.E.M., F.J.) and 8 (N.V.), Centre de Recherche des Cordeliers, UPMC Univ Paris 06, Université Paris Descartes, Paris, France; Department of Medicine (DIMED), University of Padova, Padova, Italy (R.U., F.F.); Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (A.N.D.C., R.M.T.); Laboratory of Cardiovascular Endocrinology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Pisana, Rome, Italy (A.F.); Department of Physiology and Institute of Biomedical Technologies, University of La Laguna, Tenerife, Spain (D.A.D.l.R.); Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.K.A.); Department of Nutrition, Nancy University Hospital, INSERM U954, Medical Faculty and CHU of Nancy, Vandoeuvre-les-Nancy, France (D.Q.); and INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, and Université de Lorraine, and Investigation Network Initiative Cardiovascular and Renal Clinical Trialists (INI-CRCT) French Clinical Research Infrastructure Network (F-CRIN), Nancy, France (P.R., F.J.)
| | - Patrick Rossignol
- From the INSERM, UMR_S 1138, Teams 1 (R.U., A.F., S.E.M., F.J.) and 8 (N.V.), Centre de Recherche des Cordeliers, UPMC Univ Paris 06, Université Paris Descartes, Paris, France; Department of Medicine (DIMED), University of Padova, Padova, Italy (R.U., F.F.); Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (A.N.D.C., R.M.T.); Laboratory of Cardiovascular Endocrinology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Pisana, Rome, Italy (A.F.); Department of Physiology and Institute of Biomedical Technologies, University of La Laguna, Tenerife, Spain (D.A.D.l.R.); Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.K.A.); Department of Nutrition, Nancy University Hospital, INSERM U954, Medical Faculty and CHU of Nancy, Vandoeuvre-les-Nancy, France (D.Q.); and INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, and Université de Lorraine, and Investigation Network Initiative Cardiovascular and Renal Clinical Trialists (INI-CRCT) French Clinical Research Infrastructure Network (F-CRIN), Nancy, France (P.R., F.J.)
| | - Francesco Fallo
- From the INSERM, UMR_S 1138, Teams 1 (R.U., A.F., S.E.M., F.J.) and 8 (N.V.), Centre de Recherche des Cordeliers, UPMC Univ Paris 06, Université Paris Descartes, Paris, France; Department of Medicine (DIMED), University of Padova, Padova, Italy (R.U., F.F.); Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (A.N.D.C., R.M.T.); Laboratory of Cardiovascular Endocrinology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Pisana, Rome, Italy (A.F.); Department of Physiology and Institute of Biomedical Technologies, University of La Laguna, Tenerife, Spain (D.A.D.l.R.); Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.K.A.); Department of Nutrition, Nancy University Hospital, INSERM U954, Medical Faculty and CHU of Nancy, Vandoeuvre-les-Nancy, France (D.Q.); and INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, and Université de Lorraine, and Investigation Network Initiative Cardiovascular and Renal Clinical Trialists (INI-CRCT) French Clinical Research Infrastructure Network (F-CRIN), Nancy, France (P.R., F.J.)
| | - Rhian M Touyz
- From the INSERM, UMR_S 1138, Teams 1 (R.U., A.F., S.E.M., F.J.) and 8 (N.V.), Centre de Recherche des Cordeliers, UPMC Univ Paris 06, Université Paris Descartes, Paris, France; Department of Medicine (DIMED), University of Padova, Padova, Italy (R.U., F.F.); Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (A.N.D.C., R.M.T.); Laboratory of Cardiovascular Endocrinology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Pisana, Rome, Italy (A.F.); Department of Physiology and Institute of Biomedical Technologies, University of La Laguna, Tenerife, Spain (D.A.D.l.R.); Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.K.A.); Department of Nutrition, Nancy University Hospital, INSERM U954, Medical Faculty and CHU of Nancy, Vandoeuvre-les-Nancy, France (D.Q.); and INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, and Université de Lorraine, and Investigation Network Initiative Cardiovascular and Renal Clinical Trialists (INI-CRCT) French Clinical Research Infrastructure Network (F-CRIN), Nancy, France (P.R., F.J.)
| | - Frédéric Jaisser
- From the INSERM, UMR_S 1138, Teams 1 (R.U., A.F., S.E.M., F.J.) and 8 (N.V.), Centre de Recherche des Cordeliers, UPMC Univ Paris 06, Université Paris Descartes, Paris, France; Department of Medicine (DIMED), University of Padova, Padova, Italy (R.U., F.F.); Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom (A.N.D.C., R.M.T.); Laboratory of Cardiovascular Endocrinology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Pisana, Rome, Italy (A.F.); Department of Physiology and Institute of Biomedical Technologies, University of La Laguna, Tenerife, Spain (D.A.D.l.R.); Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (G.K.A.); Department of Nutrition, Nancy University Hospital, INSERM U954, Medical Faculty and CHU of Nancy, Vandoeuvre-les-Nancy, France (D.Q.); and INSERM, Centre d'Investigations Cliniques-Plurithématique 1433, CHU de Nancy, and Université de Lorraine, and Investigation Network Initiative Cardiovascular and Renal Clinical Trialists (INI-CRCT) French Clinical Research Infrastructure Network (F-CRIN), Nancy, France (P.R., F.J.).
| |
Collapse
|
22
|
Kumar S, Palaia T, Hall CE, Ragolia L. Role of Lipocalin-type prostaglandin D2 synthase (L-PGDS) and its metabolite, prostaglandin D2, in preterm birth. Prostaglandins Other Lipid Mediat 2015; 118-119:28-33. [PMID: 25964109 DOI: 10.1016/j.prostaglandins.2015.04.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 04/14/2015] [Accepted: 04/27/2015] [Indexed: 01/07/2023]
Abstract
The objective of the study was to investigate the role of prostaglandin D2 during pregnancy and its mediator Lipocalin-type prostaglandin D2 synthase (L-PGDS) as a predictor of preterm birth (PTB). Transgenic L-PGDS (+/+), L-PGDS (-/-) and C57BL/6 control pregnant mice models were used to determine the effect of DP1 and DP2 receptor antagonists in lipopolysaccharide (LPS)-induced PTB mice. In addition, L-PGDS levels were measured in the cervicovaginal secretions (CVS) of 370 pregnant women using ELISA and further processed for isoform detection using 2-D gel electrophoresis. Our results found that C57BL/6 control mice (n = 26), transgenic L-PGDS (+/+) (n = 26), demonstrated an 89% and 100% preterm birth in LPS (intraperitoneal injection, 20mg/kg) induced mice model respectively. Interestingly, the incidence of PTB was significantly reduced to 40% in L-PGDS (-/-) knockout mice (n = 26). DP1 and DP2 receptor antagonists (0.264 μg/day, dose of 0.1 μg/μl with the flow of 0.11 μl/h for 28 day using Alzet pumps) were used to investigate the effect in LPS-induced PTB in C57BL/6 mice and found 3.3-fold increase in viable pups after LPS-induction. In addition, L-PGDS levels were measured in CVS samples and found that PTB women (n = 296) had two-fold higher levels compared to full term births (n = 74) and established a significant inverse correlation between levels of L-PGDS and days to expected delivery by using 370 preterm birth CVS samples. Elevated L-PGDS levels in the CVS of women may be considered as a potential biomarker for PTB in future. Secondly, the use of DP1 and DP2 receptor antagonists may represent novel tocolytic agents for the treatment of PTB.
Collapse
Affiliation(s)
- Sunil Kumar
- Department of Biomedical Research, Winthrop University Hospital, Mineola, NY 11501, United States
| | - Thomas Palaia
- Department of Biomedical Research, Winthrop University Hospital, Mineola, NY 11501, United States
| | - Christopher E Hall
- Department of Biomedical Research, Winthrop University Hospital, Mineola, NY 11501, United States
| | - Louis Ragolia
- Department of Biomedical Research, Winthrop University Hospital, Mineola, NY 11501, United States; Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY 11794, United States.
| |
Collapse
|
23
|
Prostaglandin D2 synthase/GPR44: a signaling axis in PNS myelination. Nat Neurosci 2014; 17:1682-92. [PMID: 25362470 DOI: 10.1038/nn.3857] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 10/07/2014] [Indexed: 12/18/2022]
Abstract
Neuregulin 1 type III is processed following regulated intramembrane proteolysis, which allows communication from the plasma membrane to the nucleus. We found that the intracellular domain of neuregulin 1 type III upregulated the prostaglandin D2 synthase (L-pgds, also known as Ptgds) gene, which, together with the G protein-coupled receptor Gpr44, forms a previously unknown pathway in PNS myelination. Neuronal L-PGDS is secreted and produces the PGD2 prostanoid, a ligand of Gpr44. We found that mice lacking L-PGDS were hypomyelinated. Consistent with this, specific inhibition of L-PGDS activity impaired in vitro myelination and caused myelin damage. Furthermore, in vivo ablation and in vitro knockdown of glial Gpr44 impaired myelination. Finally, we identified Nfatc4, a key transcription factor for myelination, as one of the downstream effectors of PGD2 activity in Schwann cells. Thus, L-PGDS and Gpr44 are previously unknown components of an axo-glial interaction that controls PNS myelination and possibly myelin maintenance.
Collapse
|
24
|
White CA, Ghazan-Shahi S, Adams MA. β-Trace protein: a marker of GFR and other biological pathways. Am J Kidney Dis 2014; 65:131-46. [PMID: 25446025 DOI: 10.1053/j.ajkd.2014.06.038] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 06/27/2014] [Indexed: 11/11/2022]
Abstract
β-Trace protein (BTP), also known as lipocalin prostaglandin D2 synthase (L-PGDS; encoded by the PTGDS gene), is a low-molecular-weight glycoprotein and an emerging novel marker of glomerular filtration rate. BTP is an important constituent of cerebral spinal fluid and is found in much lower concentrations in blood. Its serum origin and renal handling remain poorly understood. Unlike serum creatinine, BTP is not physiologically inert. It possesses both ligand-binding and enzymatic properties. BTP catalyzes the conversion of prostaglandin H2 (PGH2) to PGD2. PGD2 is an eicosanoid involved in a variety of important physiologic processes, including platelet aggregation, vasodilation, inflammation, adipogenesis, and bone remodeling. Several studies now have documented BTP's strong association with glomerular filtration rate, end-stage renal disease, cardiovascular disease, and death in a variety of different patient populations. This review provides an overview of the biochemistry, physiology and metabolism, biological functions, and measurement of BTP; summarizes the evidence for BTP as a marker of both kidney function and cardiovascular disease; and then considers the interplay between its biological properties, serum concentration, and patient outcomes.
Collapse
Affiliation(s)
- Christine A White
- Division of Nephrology, Department of Medicine, Queen's University, Kingston, Canada.
| | - Sassan Ghazan-Shahi
- Division of Nephrology, Department of Medicine, Queen's University, Kingston, Canada
| | - Michael A Adams
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| |
Collapse
|
25
|
Huang HL, Huang YC, Lee WY, Yeh CN, Lin KJ, Yu CS. 18F-glutathione conjugate as a PET tracer for imaging tumors that overexpress L-PGDS enzyme. PLoS One 2014; 9:e104118. [PMID: 25111383 PMCID: PMC4128654 DOI: 10.1371/journal.pone.0104118] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 07/06/2014] [Indexed: 12/17/2022] Open
Abstract
Lipocalin-type prostaglandin D synthase (L-PGDS) has been correlated with the progression of neurological disorders. The present study aimed at evaluating the imaging potency of a glutathione conjugate of fluorine-18-labeled fluorobutyl ethacrynic amide ([18F]FBuEA-GS) for brain tumors. Preparation of [18F]FBuEA-GS has been modified from the -4-tosylate derivative via radiofluorination in 5% radiochemical yield. The mixture of nonradioactive FBuEA-GS derived from a parallel preparation has be resolved to two isomers in a ratio of 9∶1 using analytic chiral reversed phase high performance liquid chromatography (RP-HPLC). The two fluorine-18-labeled isomers purified through nonchiral semipreparative RP-HPLC as a mixture were studied by assessing the binding affinity toward L-PGDS through a gel filtration HPLC, by analyzing radiotracer accumulation in C6 glioma cells, and by evaluating the imaging of radiotracer in a C6 glioma rat with positron emission tomography. The inhibition percentage of the production of PGD2 from PGH2 at the presence of 200 µM of FBuEA-GS and 4-Dibenzo[a,d]cyclohepten-5-ylidene-1-[4-(2H-tetrazol-5-yl)butyl]piperidine (AT-56) were 74.1±4.8% and 97.6±16.0%, respectively. [18F]FBuEA-GS bound L-PGDS (16.3–21.7%) but not the isoform, microsomal prostaglandin E synthase 1. No binding to GST-alpha and GST-pi was observed. The binding strength between [18F]FBuEA-GS and L-PGDS has been evaluated using analytic gel filtration HPLC at the presence of various concentrations of the cold competitor FBuEA-GS. The contrasted images indicated that the radiotracer accumulation in tumor lesions is probably related to the overexpression of L-PGDS.
Collapse
Affiliation(s)
- Ho-Lien Huang
- Department of Biomedical Engineering and Environmental Sciences, National Tsinghua University, Hsinchu, Taiwan
| | - Ying-Cheng Huang
- Department of Neurosurgery, Chang-Gung memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Wei-Yuan Lee
- Department of Biomedical Engineering and Environmental Sciences, National Tsinghua University, Hsinchu, Taiwan
| | - Chun-Nan Yeh
- Department of Surgery, Chang-Gung memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Kun-Ju Lin
- Department of Nuclear Medicine, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan, Taiwan
| | - Chung-Shan Yu
- Department of Biomedical Engineering and Environmental Sciences, National Tsinghua University, Hsinchu, Taiwan
- Institute of Nuclear Engineering and Sciences, National Tsinghua University, Hsinchu, Taiwan
- * E-mail:
| |
Collapse
|
26
|
Dittrich AM, Meyer HA, Hamelmann E. The role of lipocalins in airway disease. Clin Exp Allergy 2013; 43:503-11. [PMID: 23600540 DOI: 10.1111/cea.12025] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The pathogenesis of allergic airway inflammation and disease is complex and still not fully understood. Many cells, factors and mediators are involved in the different aspects of induction, maintenance and persistence of airway inflammation. The heterogeneity and redundancy of this system is one of the main reasons why novel therapeutic targets focusing on the pathogenesis of asthma only hesitantly reach the market and clinical application. Thus, it seems mandatory that we proceed in our efforts to better understand this micro cosmos to succeed in the development of safe and effective drugs for the treatment of more severe and refractory forms of asthma and chronic obstructive pulmonary disease. One of the more recently discovered mediators in the context of airway inflammation are the lipocalins (Lcns). They are a family of proteins that share functional and structural similarities and are involved in the transport of small hydrophobic molecules such as steroids and lipids into the cell. Lcns are found in many different cell types from plants and bacteria through invertebrate cells to cells of vertebrate origin. The purpose of this review is to summarize the role of Lcns in airway diseases, focusing on allergic and infectious inflammation. In particular, we will summarize the present knowledge about Lipocalin 1 and Lipocalin 2, where exciting new discoveries in the recent years have highlighted their role in pulmonary disease and infection. This new class of proteins is another putative candidate for the development of novel drugs against airway inflammation.
Collapse
Affiliation(s)
- A M Dittrich
- Junior Research Group, Allergic Sensitization, Medical School Hannover, Hannover, Germany
| | | | | |
Collapse
|
27
|
Zhao G, Yu R, Deng J, Zhao Q, Li Y, Joo M, van Breemen RB, Christman JW, Xiao L. Pivotal role of reactive oxygen species in differential regulation of lipopolysaccharide-induced prostaglandins production in macrophages. Mol Pharmacol 2013; 83:167-78. [PMID: 23071105 PMCID: PMC3533474 DOI: 10.1124/mol.112.080762] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Accepted: 10/15/2012] [Indexed: 12/29/2022] Open
Abstract
Gram-negative bacterial endotoxin lipopolysaccharide (LPS) triggers the production of inflammatory cytokines, reactive oxygen species (ROS), and prostaglandins (PGs) by pulmonary macrophages. Here, we investigated if ROS influenced PGs production in response to LPS treatment in mouse bone marrow-derived macrophages (BMDM). We observed that pretreatment of BMDM with two structurally unrelated ROS scavengers, MnTMPyP and EUK-134, not only prevented LPS-induced ROS accumulation, but also attenuated the LPS-induced PGD(2), but not PGE(2), production. Conversely LPS-induced PGD(2), but not PGE(2), production, was potentiated with the cotreatment of BMDM with H(2)O(2). These data suggest that ROS differentially regulate PGD(2) and PGE(2) production in BMDM. In addition, selective inhibition of the ROS generator NADPH oxidase (NOX) using either pharmacologic inhibitors or its p47(phox) subunit deficient mouse BMDM also attenuated LPS-induced PGD(2), but not PGE(2) production, suggesting the critical role of NOX-generated ROS in LPS-induced PGD(2) production in BMDM. We further found that both hematopoietic PGD synthase (H-PGDS) siRNA and its inhibitor HQL-79, but not lipocalin PGDS (L-PGDS) siRNA and its inhibitor AT-56, significantly attenuated LPS-induced PGD(2) production, suggesting that H-PGDS, but not L-PGDS, mediates LPS-induced PGD(2) production in BMDM. Furthermore, data from our in vitro cell-free enzymatic studies showed that coincubation of the recombinant H-PGDS with either MnTMPyP, EUK-134, or catalase significantly decreased PGD(2) production, whereas coincubation with H(2)O(2) significantly increased PGD(2) production. Taken together, our results show that LPS-induced NOX-generated ROS production differentially and specifically regulates the H-PGDS-mediated production of PGD(2), but not PGE(2), in mouse BMDM.
Collapse
Affiliation(s)
- Guiqing Zhao
- Department of Medicine, M/C: 719 University of Illinois at Chicago 840 S. Wood Street Chicago, IL 60612, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Systematic interaction analysis of human lipocalin-type prostaglandin D synthase with small lipophilic ligands. Biochem J 2012; 446:279-89. [DOI: 10.1042/bj20120324] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
L-PGDS [lipocalin-type PG (prostaglandin) D synthase] is a multi-functional protein, acting as a PGD2-producing enzyme and a lipid-transporter. In the present study, we focus on the function of L-PGDS as an extracellular transporter for small lipophilic molecules. We characterize the binding mechanism of human L-PGDS for the molecules, especially binding affinity stoichiometry and driving force, using tryptophan fluorescence quenching, ICD (induced circular dichroism) and ITC (isothermal titration calorimetry). The tryptophan fluorescence quenching measurements revealed that haem metabolites such as haemin, biliverdin and bilirubin bind to L-PGDS with significantly higher affinities than the other small lipophilic ligands examined, showing dissociation constant (Kd) values from 17.0 to 20.9 nM. We focused particularly on the extra-specificities of haem metabolites and L-PGDS. The ITC and ICD data revealed that two molecules of the haem metabolites bind to L-PGDS with high and low affinities, showing Kd values from 2.8 to 18.1 nM and from 0.209 to 1.63 μM respectively. The thermodynamic parameters for the interactions revealed that the contributions of enthalpy and entropy change were considerably different for each haem metabolite even when the Gibbs energy change was the same. Thus we believe that the binding energy of haem metabolites to L-PGDS is optimized by balancing enthalpy and entropy change.
Collapse
|
29
|
Fujimori K, Maruyama T, Kamauchi S, Urade Y. Activation of adipogenesis by lipocalin-type prostaglandin D synthase-generated Δ¹²-PGJ₂ acting through PPARγ-dependent and independent pathways. Gene 2012; 505:46-52. [PMID: 22664386 DOI: 10.1016/j.gene.2012.05.052] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Revised: 04/12/2012] [Accepted: 05/28/2012] [Indexed: 10/28/2022]
Abstract
Lipocalin-type prostaglandin (PG) D synthase (L-PGDS)-produced PGD(2) accelerates adipogenesis. In this study, we investigated the molecular mechanism of PGD(2)-mediated activation of adipogenesis in mouse adipocytic 3T3-L1 cells. LC/MS analysis showed that Δ(12)-PGJ(2), one of the PGD(2) metabolites, was predominantly produced in the differentiated 3T3-L1 cells. Δ(12)-PGJ(2) enhanced the expression of adipogenic genes in a Δ(12)-PGJ(2)-concentration-dependent manner. Suppression of the expression of the adipogenic genes by L-PGDS siRNA or AT-56, an L-PGDS inhibitor, was cleared by the addition of Δ(12)-PGJ(2). Moreover, the production of adiponectin and leptin was increased by treatment with Δ(12)-PGJ(2). Furthermore, the results of a mammalian two-hybrid assay demonstrated that Δ(12)-PGJ(2) enhanced the PPARγ-mediated transcription activity. However, Δ(12)-PGJ(2)-activated expression of adipogenic genes such as fatty acid binding protein 4 (aP2) and stearoyl-CoA desaturase was inhibited only at 38% and 42%, respectively, by treatment with GW9662, a PPARγ antagonist in 3T3-L1 cells, although Troglitazone-mediated activation of the expression of these adipogenic genes was completely suppressed by GW9662, suggesting the existence of a PPARγ-independent mechanism for Δ(12)-PGJ(2)-activated adipogenesis. These results, taken together, indicate that Δ(12)-PGJ(2) is a dominant metabolite of L-PGDS-produced PGD(2) during adipogenesis and acts as an activator for adipogenesis through both PPARγ-dependent and -independent mechanisms in 3T3-L1 cells.
Collapse
Affiliation(s)
- Ko Fujimori
- Laboratory of Biodefense and Regulation, Osaka University of Pharmaceutical Sciences, 4-20-1 Nasahara, Takatsuki, Osaka 569-1094, Japan.
| | | | | | | |
Collapse
|
30
|
PGH1, the precursor for the anti-inflammatory prostaglandins of the 1-series, is a potent activator of the pro-inflammatory receptor CRTH2/DP2. PLoS One 2012; 7:e33329. [PMID: 22442685 PMCID: PMC3307725 DOI: 10.1371/journal.pone.0033329] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 02/07/2012] [Indexed: 12/19/2022] Open
Abstract
Prostaglandin H1 (PGH1) is the cyclo-oxygenase metabolite of dihomo-γ-linolenic acid (DGLA) and the precursor for the 1-series of prostaglandins which are often viewed as “anti-inflammatory”. Herein we present evidence that PGH1 is a potent activator of the pro-inflammatory PGD2 receptor CRTH2, an attractive therapeutic target to treat allergic diseases such as asthma and atopic dermatitis. Non-invasive, real time dynamic mass redistribution analysis of living human CRTH2 transfectants and Ca2+ flux studies reveal that PGH1 activates CRTH2 as PGH2, PGD2 or PGD1 do. The PGH1 precursor DGLA and the other PGH1 metabolites did not display such effect. PGH1 specifically internalizes CRTH2 in stable CRTH2 transfectants as assessed by antibody feeding assays. Physiological relevance of CRTH2 ligation by PGH1 is demonstrated in several primary human hematopoietic lineages, which endogenously express CRTH2: PGH1 mediates migration of and Ca2+ flux in Th2 lymphocytes, shape change of eosinophils, and their adhesion to human pulmonary microvascular endothelial cells under physiological flow conditions. All these effects are abrogated in the presence of the CRTH2 specific antagonist TM30089. Together, our results identify PGH1 as an important lipid intermediate and novel CRTH2 agonist which may trigger CRTH2 activation in vivo in the absence of functional prostaglandin D synthase.
Collapse
|
31
|
Urade Y, Hayaishi O. Prostaglandin D2 and sleep/wake regulation. Sleep Med Rev 2012; 15:411-8. [PMID: 22024172 DOI: 10.1016/j.smrv.2011.08.003] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Revised: 08/09/2011] [Accepted: 08/11/2011] [Indexed: 11/19/2022]
Abstract
Prostaglandin (PG) D2 is the most potent endogenous sleep-promoting substance. PGD2 is produced by lipocalin-type PGD synthase localized in the leptomeninges, choroid plexus, and oligodendrocytes in the brain, and is secreted into the cerebrospinal fluid as a sleep hormone. PGD2 stimulates DP1 receptors localized in the leptomeninges under the basal forebrain and the hypothalamus. As a consequence, adenosine is released as a paracrine sleep-promoting molecule to activate adenosine A2A receptor-expressing sleep-promoting neurons and to inhibit adenosine A1 receptor-possessing arousal neurons. PGD2 activates a center of non-rapid eye movement (NREM) sleep regulation in the ventrolateral preoptic area, probably mediated by adenosine signaling, which activation inhibits the histaminergic arousal center in the tuberomammillary nucleus via descending GABAergic and galaninergic projections. The administration of a lipocalin-type PGD synthase inhibitor (SeCl4), DP1 antagonist (ONO-4127Na) or adenosine A2A receptor antagonist (caffeine) suppresses both NREM and rapid eye movement (REM) sleep, indicating that the PGD2-adenosine system is crucial for the maintenance of physiological sleep.
Collapse
Affiliation(s)
- Yoshihiro Urade
- Department of Molecular Behavioral Biology, Osaka Bioscience Institute, 6-2-4, Furuedai, Suita, Osaka 565 0874, Japan.
| | | |
Collapse
|
32
|
Yu R, Xiao L, Zhao G, Christman JW, van Breemen RB. Competitive enzymatic interactions determine the relative amounts of prostaglandins E2 and D2. J Pharmacol Exp Ther 2011; 339:716-25. [PMID: 21865441 PMCID: PMC3199988 DOI: 10.1124/jpet.111.185405] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 08/22/2011] [Indexed: 12/12/2022] Open
Abstract
Prostaglandins (PGs) are a family of cellular messengers exerting diverse homeostatic and pathophysiologic effects. Recently, several studies reported significant increases of PGI(2) and PGF(2α) after the inhibition of microsomal PGE synthase-1 (mPGES-1) expression, which indicated that PGH(2) metabolism might be redistributed when the PGE(2) pathway is blocked. To address the determinants that govern the relative amounts of PGs, we developed an in vitro cell-free method, based on liquid chromatography-tandem mass spectrometry, to measure the exact amounts of these PGs formed in response to the addition of recombinant isomerases and their selective inhibitors. Our in vitro cell-free assay results were confirmed in cells using bone marrow-derived macrophage. Initially, we determined the in vitro stability of PGH(2) and noted that there was spontaneous nonenzymatic conversion to PGD(2) and PGE(2). mPGES-1 markedly increased the conversion to PGE(2) and decreased conversion to PGD(2). Reciprocally, the addition of hematopoietic or lipocalin PGD synthase resulted in a relative increase of PGD(2) and decrease of PGE(2). A detailed titration study showed that the ratio of PGE(2)/PGD(2) was closely correlated with the ratio of PGE synthase/PGD synthase. Our redistribution results also provide the foundation for understanding how PGH(2) metabolism is redistributed by the presence of distal isomerases or by blocking the major metabolic outlet, which could determine the relative benefits and risks resulting from interdiction in nonrated-limiting components of PG synthesis pathways.
Collapse
Affiliation(s)
- Rui Yu
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois College of Pharmacy, 833 S. Wood Street, Chicago, IL 60612-7231, USA
| | | | | | | | | |
Collapse
|
33
|
Smith WL, Urade Y, Jakobsson PJ. Enzymes of the cyclooxygenase pathways of prostanoid biosynthesis. Chem Rev 2011; 111:5821-65. [PMID: 21942677 PMCID: PMC3285496 DOI: 10.1021/cr2002992] [Citation(s) in RCA: 364] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- William L Smith
- Department of Biological Chemistry, University of Michigan Medical School, 1150 West Medical Center Drive, 5301 MSRB III, Ann Arbor, Michigan 48109-5606, USA.
| | | | | |
Collapse
|
34
|
Nagata N, Kusakari Y, Fukunishi Y, Inoue T, Urade Y. Catalytic mechanism of the primary human prostaglandin F2α synthase, aldo-keto reductase 1B1--prostaglandin D2 synthase activity in the absence of NADP(H). FEBS J 2011; 278:1288-98. [PMID: 21306562 DOI: 10.1111/j.1742-4658.2011.08049.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Aldo-keto reductase 1B1 and 1B3 (AKR1B1 and AKR1B3) are the primary human and mouse prostaglandin F(2α) (PGF(2α)) synthases, respectively, which catalyze the NADPH-dependent reduction of PGH(2), a common intermediate of various prostanoids, to form PGF(2α). In this study, we found that AKR1B1 and AKR1B3, but not AKR1B7 and AKR1C3, also catalyzed the isomerization of PGH(2) to PGD(2) in the absence of NADPH or NADP(+). Both PGD(2) and PGF(2α) synthase activities of AKR1B1 and AKR1B3 completely disappeared in the presence of NADP(+) or after heat treatment of these enzymes at 100 °C for 5 min. The K(m), V(max), pK and optimum pH values of the PGD(2) synthase activities of AKR1B1 and AKR1B3 were 23 and 18 μM, 151 and 57 nmol·min(-1)·(mg protein)(-1), 7.9 and 7.6, and pH 8.5 for both AKRs, respectively, and those of PGF(2α) synthase activity were 29 and 33 μM, 169 and 240 nmol·min(-1)·(mg protein)(-1), 6.2 and 5.4, and pH 5.5 and pH 5.0, respectively, in the presence of 0.5 mm NADPH. Site-directed mutagenesis of the catalytic tetrad of AKR1B1, composed of Tyr, Lys, His and Asp, revealed that the triad of Asp43, Lys77 and His110, but not Tyr48, acts as a proton donor in most AKR activities, and is crucial for PGD(2) and PGF(2α) synthase activities. These results, together with molecular docking simulation of PGH(2) to the crystallographic structure of AKR1B1, indicate that His110 acts as a base in concert with Asp43 and Lys77 and as an acid to generate PGD(2) and PGF(2α) in the absence of NADPH or NADP(+) and in the presence of NADPH, respectively.
Collapse
Affiliation(s)
- Nanae Nagata
- Department of Molecular Behavioral Biology, Osaka Bioscience Institute, Suita, Osaka, Japan
| | | | | | | | | |
Collapse
|
35
|
|
36
|
Yoshihiro Urade, Osamu Hayaishi. Crucial role of prostaglandin D2 and adenosine in sleep regulation: experimental evidence from pharmacological approaches to gene-knockout mice. FUTURE NEUROLOGY 2010. [DOI: 10.2217/fnl.10.18] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Prostaglandin (PG) D2 is the most potent endogenous sleep-promoting substance reported thus far. Its mechanism of action has been extensively studied at the molecular level. PGD2 is produced by lipocalin-type PGD synthase, which is predominantly localized in the leptomeninges, choroid plexus and oligodendrocytes in the brain; it is secreted into the cerebrospinal fluid and stimulates DP1 receptors localized in the arachnoid membrane of the ventral surface from the basal forebrain to the hypothalamus, increasing the extracellular concentration of adenosine as a paracrine sleep-promoting molecule. Adenosine diffuses into the brain parenchyma, suppresses cholinergic arousal neurons in the basal forebrain via adenosine A1 receptors, activates sleep-active neurons in the ventrolateral preoptic area via adenosine A2A receptors and concomitantly suppresses the histaminergic arousal center in the tuberomammillary nucleus through GABAergic and galaninergic inhibitory projections. Administration of an inhibitor of lipocalin-type PGD synthase (SeCl4), an antagonist of DP1 receptors (ONO-4127Na) or an antagonist of adenosine A2A receptors (caffeine) results in sleep inhibition in rats and mice. These results indicate that the PGD2–adenosine system is crucial for the maintenance of physiological sleep.
Collapse
|
37
|
Nagata N, Fujimori K, Okazaki I, Oda H, Eguchi N, Uehara Y, Urade Y. De novo synthesis, uptake and proteolytic processing of lipocalin-type prostaglandin D synthase, β-trace, in the kidneys. FEBS J 2009; 276:7146-58. [DOI: 10.1111/j.1742-4658.2009.07426.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
38
|
Kumasaka T, Aritake K, Ago H, Irikura D, Tsurumura T, Yamamoto M, Miyano M, Urade Y, Hayaishi O. Structural basis of the catalytic mechanism operating in open-closed conformers of lipocalin type prostaglandin D synthase. J Biol Chem 2009; 284:22344-22352. [PMID: 19546224 PMCID: PMC2755957 DOI: 10.1074/jbc.m109.018341] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Revised: 06/15/2009] [Indexed: 11/06/2022] Open
Abstract
Lipocalin type prostaglandin D synthase (L-PGDS) is a multifunctional protein acting as a somnogen (PGD2)-producing enzyme, an extracellular transporter of various lipophilic ligands, and an amyloid-beta chaperone in human cerebrospinal fluid. In this study, we determined the crystal structures of two different conformers of mouse L-PGDS, one with an open cavity of the beta-barrel and the other with a closed cavity due to the movement of the flexible E-F loop. The upper compartment of the central large cavity contains the catalytically essential Cys65 residue and its network of hydrogen bonds with the polar residues Ser45, Thr67, and Ser81, whereas the lower compartment is composed of hydrophobic amino acid residues that are highly conserved among other lipocalins. SH titration analysis combined with site-directed mutagenesis revealed that the Cys65 residue is activated by its interaction with Ser45 and Thr67 and that the S45A/T67A/S81A mutant showed less than 10% of the L-PGDS activity. The conformational change between the open and closed states of the cavity indicates that the mobile calyx contributes to the multiligand binding ability of L-PGDS.
Collapse
Affiliation(s)
- Takashi Kumasaka
- From the Structural Biophysics Laboratory, RIKEN SPring-8 Center, Harima Institute, Kouto, Sayo, Hyogo 679-5148, Japan and
| | - Kosuke Aritake
- the Department of Molecular and Behavioral Biology, Osaka Bioscience Institute, Furuedai, Suita, Osaka 565-0874, Japan
| | - Hideo Ago
- From the Structural Biophysics Laboratory, RIKEN SPring-8 Center, Harima Institute, Kouto, Sayo, Hyogo 679-5148, Japan and
| | - Daisuke Irikura
- From the Structural Biophysics Laboratory, RIKEN SPring-8 Center, Harima Institute, Kouto, Sayo, Hyogo 679-5148, Japan and
- the Department of Molecular and Behavioral Biology, Osaka Bioscience Institute, Furuedai, Suita, Osaka 565-0874, Japan
| | - Toshiharu Tsurumura
- the Department of Molecular and Behavioral Biology, Osaka Bioscience Institute, Furuedai, Suita, Osaka 565-0874, Japan
| | - Masaki Yamamoto
- From the Structural Biophysics Laboratory, RIKEN SPring-8 Center, Harima Institute, Kouto, Sayo, Hyogo 679-5148, Japan and
| | - Masashi Miyano
- From the Structural Biophysics Laboratory, RIKEN SPring-8 Center, Harima Institute, Kouto, Sayo, Hyogo 679-5148, Japan and
| | - Yoshihiro Urade
- the Department of Molecular and Behavioral Biology, Osaka Bioscience Institute, Furuedai, Suita, Osaka 565-0874, Japan
| | - Osamu Hayaishi
- the Department of Molecular and Behavioral Biology, Osaka Bioscience Institute, Furuedai, Suita, Osaka 565-0874, Japan
| |
Collapse
|