1
|
Pitchai A, Buhman K, Shannahan JH. Lipid mediators of inhalation exposure-induced pulmonary toxicity and inflammation. Inhal Toxicol 2024; 36:57-74. [PMID: 38422051 PMCID: PMC11022128 DOI: 10.1080/08958378.2024.2318389] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 02/07/2024] [Indexed: 03/02/2024]
Abstract
Many inhalation exposures induce pulmonary inflammation contributing to disease progression. Inflammatory processes are actively regulated via mediators including bioactive lipids. Bioactive lipids are potent signaling molecules involved in both pro-inflammatory and resolution processes through receptor interactions. The formation and clearance of lipid signaling mediators are controlled by multiple metabolic enzymes. An imbalance of these lipids can result in exacerbated and sustained inflammatory processes which may result in pulmonary damage and disease. Dysregulation of pulmonary bioactive lipids contribute to inflammation and pulmonary toxicity following exposures. For example, inhalation of cigarette smoke induces activation of pro-inflammatory bioactive lipids such as sphingolipids, and ceramides contributing to chronic obstructive pulmonary disease. Additionally, exposure to silver nanoparticles causes dysregulation of inflammatory resolution lipids. As inflammation is a common consequence resulting from inhaled exposures and a component of numerous diseases it represents a broadly applicable target for therapeutic intervention. With new appreciation for bioactive lipids, technological advances to reliably identify and quantify lipids have occurred. In this review, we will summarize, integrate, and discuss findings from recent studies investigating the impact of inhaled exposures on pro-inflammatory and resolution lipids within the lung and their contribution to disease. Throughout the review current knowledge gaps in our understanding of bioactive lipids and their contribution to pulmonary effects of inhaled exposures will be presented. New methods being employed to detect and quantify disruption of pulmonary lipid levels following inhalation exposures will be highlighted. Lastly, we will describe how lipid dysregulation could potentially be addressed by therapeutic strategies to address inflammation.
Collapse
Affiliation(s)
- Arjun Pitchai
- School of Health Sciences, College of Health and Human Sciences, Purdue University, West Lafayette, IN, United States
| | - Kimberly Buhman
- Department of Nutrition, College of Health and Human Sciences, Purdue University, West Lafayette, IN, United States
| | - Jonathan H. Shannahan
- School of Health Sciences, College of Health and Human Sciences, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
2
|
Zou F, Li Y, Zhang S, Zhang J. DP1 (Prostaglandin D 2 Receptor 1) Activation Protects Against Vascular Remodeling and Vascular Smooth Muscle Cell Transition to Myofibroblasts in Angiotensin II-Induced Hypertension in Mice. Hypertension 2022; 79:1203-1215. [PMID: 35354317 DOI: 10.1161/hypertensionaha.121.17584] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Vascular smooth muscle cell (VSMC) phenotype transition plays an essential role in vascular remodeling. PGD2 (Prostaglandin D2) is involved in cardiovascular inflammation. In this study, we aimed to investigates the role of DP1 (PGD2 receptor 1) on VSMC phenotype transition in vascular remodeling after Ang II (angiotensin II) infusion in mice. METHODS VSMC-specific DP1 knockout mice and DP1flox/flox mice were infused with Ang II for 28 days and systolic blood pressure was measured by noninvasive tail-cuff system. The arterial samples were applied to an unbiased proteome analysis. DP1f/f Myh11 (myosin heavy chain 11) CREERT2 R26mTmG/+ mice were generated for VSMC lineage tracing. Multiple genetic and pharmacological approaches were used to investigate DP1-mediated signaling in phenotypic transition of VSMCs in response to Ang II administration. RESULTS DP1 knockout promoted vascular media thickness and increased systolic blood pressure after Ang II infusion by impairing Epac (exchange protein directly activated by cAMP)-1-mediated Rap-1 (Ras-related protein 1) activation. The DP1 agonist facilitated the interaction of myocardin-related transcription factor A and G-actin, which subsequently inhibited the VSMC transition to myofibroblasts through the suppression of RhoA (Ras homolog family member A)/ROCK-1 (Rho associated coiled-coil containing protein kinase 1) activity. Moreover, Epac-1 overexpression by lentivirus blocked the progression of vascular fibrosis in DP1 deficient mice in response to Ang II infusion. CONCLUSIONS Our finding revealed a protective role of DP1 in VSMC switch to myofibroblasts by impairing the phosphorylation of MRTF (myocardin-related transcription factor)-A by ROCK-1 through Epac-1/Rap-1/RhoA pathway and thus inhibited the expression of collagen I, fibronectin, ED-A (extra domain A) fibronectin, and vinculin. Thus, DP1 activation has therapeutic potential for vascular fibrosis in hypertension.
Collapse
Affiliation(s)
- Fangdi Zou
- Department of Pharmacology, School of Basic Medical Sciences (F.Z., Y.L., S.Z., J.Z.), Tianjin Medical University, China.,School of Pharmacy (F.Z.), Tianjin Medical University, China
| | - Yong Li
- Department of Pharmacology, School of Basic Medical Sciences (F.Z., Y.L., S.Z., J.Z.), Tianjin Medical University, China
| | - Shijie Zhang
- Department of Pharmacology, School of Basic Medical Sciences (F.Z., Y.L., S.Z., J.Z.), Tianjin Medical University, China
| | - Jian Zhang
- Department of Pharmacology, School of Basic Medical Sciences (F.Z., Y.L., S.Z., J.Z.), Tianjin Medical University, China.,School of Pharmacy, East China University of Science and Technology, Shanghai, China (J.Z.)
| |
Collapse
|
3
|
Lee BR, Paing MH, Sharma-Walia N. Cyclopentenone Prostaglandins: Biologically Active Lipid Mediators Targeting Inflammation. Front Physiol 2021; 12:640374. [PMID: 34335286 PMCID: PMC8320392 DOI: 10.3389/fphys.2021.640374] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 05/10/2021] [Indexed: 12/13/2022] Open
Abstract
Cyclopentenone prostaglandins (cyPGs) are biologically active lipid mediators, including PGA2, PGA1, PGJ2, and its metabolites. cyPGs are essential regulators of inflammation, cell proliferation, apoptosis, angiogenesis, cell migration, and stem cell activity. cyPGs biologically act on multiple cellular targets, including transcription factors and signal transduction pathways. cyPGs regulate the inflammatory response by interfering with NF-κB, AP-1, MAPK, and JAK/STAT signaling pathways via both a group of nuclear receptor peroxisome proliferator-activated receptor-gamma (PPAR-γ) dependent and PPAR-γ independent mechanisms. cyPGs promote the resolution of chronic inflammation associated with cancers and pathogen (bacterial, viral, and parasitic) infection. cyPGs exhibit potent effects on viral infections by repressing viral protein synthesis, altering viral protein glycosylation, inhibiting virus transmission, and reducing virus-induced inflammation. We summarize their anti-proliferative, pro-apoptotic, cytoprotective, antioxidant, anti-angiogenic, anti-inflammatory, pro-resolution, and anti-metastatic potential. These properties render them unique therapeutic value, especially in resolving inflammation and could be used in adjunct with other existing therapies. We also discuss other α, β -unsaturated carbonyl lipids and cyPGs like isoprostanes (IsoPs) compounds.
Collapse
|
4
|
Nomogram Personalizes and Visualizes the Overall Survival of Patients with Triple-Negative Breast Cancer Based on the Immune Genome. BIOMED RESEARCH INTERNATIONAL 2020; 2020:4029062. [PMID: 33299869 PMCID: PMC7709499 DOI: 10.1155/2020/4029062] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/02/2020] [Accepted: 11/12/2020] [Indexed: 02/05/2023]
Abstract
Background Triple-negative breast cancer (TNBC) is usually poorly differentiated, highly invasive, susceptible to distant metastasis, and less responsive to endocrine and targeted therapy. However, immunotherapy is a promising treatment for TNBC patients recently. Methods The prognostic value of immune-related genes (IRGs) was explored by using RNA sequencing and microarray data of 123 and 107 TNBC patients from TCGA and GEO databases, respectively. Results In TCGA database, GO and KEGG pathway analysis of 119 differential IRGs indicated that they actively participate in the interaction of cytokines and receptors. A nomogram model constructed by the prognosis-related CCL25, IL29, TDGF3, GPR44, and GREM2 in the IRGs could personalize and visualize the 1-, 2-, 3-, 4-, and 5-year overall survival (OS) of TNBC patients. Moreover, TNBC patients could be defined as low-risk (risk score < 194) or high-risk (risk score ≥ 194) cohorts based on the risk score derived from the nomogram model. The results could be validated by the GSE58812 dataset. Furthermore, the risk score was an independent risk factor for TNBC patients (HR = 1.019, 95% CI 1.012-1.027, p < 0.001) and was positively related to stage (p = 0.017). Interestingly, the risk score could reflect the infiltration of B cells, CD4+ T cells, CD8+ T cells, dendritic cells, and neutrophils. Conclusion These findings provided a reference for personalized OS prediction in TNBC patients and might be potential immune biomarkers for designing novel therapy.
Collapse
|
5
|
Jara CP, Mendes NF, Prado TPD, de Araújo EP. Bioactive Fatty Acids in the Resolution of Chronic Inflammation in Skin Wounds. Adv Wound Care (New Rochelle) 2020; 9:472-490. [PMID: 32320357 DOI: 10.1089/wound.2019.1105] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Significance: Optimal skin wound healing is crucial for maintaining tissue homeostasis, particularly in response to an injury. The skin immune system is under regulation of mediators such as bioactive lipids and cytokines that can initiate an immune response with controlled inflammation, followed by efficient resolution. However, nutritional deficiency impacts wound healing by hindering fibroblast proliferation, collagen synthesis, and epithelialization, among other crucial functions. In this way, the correct nutritional support of bioactive lipids and of other essential nutrients plays an important role in the outcome of the wound healing process. Recent Advances and Critical Issues: Several studies have revealed the potential role of lipids as a treatment for the healing of skin wounds. Unsaturated fatty acids such as linoleic acid, α-linolenic acid, oleic acid, and most of their bioactive products have shown an effective role as a topical treatment of chronic skin wounds. Their effect, when the treatment starts at day 0, has been observed mainly in the inflammatory phase of the wound healing process. Moreover, some of them were associated with different dressings and were tested for clinical purposes, including pluronic gel, nanocapsules, collagen films and matrices, and polymeric bandages. Therefore, future research is still needed to evaluate these dressing technologies in association with different bioactive fatty acids in a wound healing context. Future Directions: This review summarizes the main results of the available clinical trials and basic research studies and provides evidence-based conclusions. Together, current data encourage the use of bioactive fatty acids for an optimal wound healing resolution.
Collapse
Affiliation(s)
- Carlos Poblete Jara
- Faculty of Nursing, University of Campinas, Campinas, Brazil
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Natália Ferreira Mendes
- Faculty of Nursing, University of Campinas, Campinas, Brazil
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Thais Paulino do Prado
- Faculty of Nursing, University of Campinas, Campinas, Brazil
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Eliana Pereira de Araújo
- Faculty of Nursing, University of Campinas, Campinas, Brazil
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| |
Collapse
|
6
|
Marone G, Galdiero MR, Pecoraro A, Pucino V, Criscuolo G, Triassi M, Varricchi G. Prostaglandin D 2 receptor antagonists in allergic disorders: safety, efficacy, and future perspectives. Expert Opin Investig Drugs 2018; 28:73-84. [PMID: 30513028 DOI: 10.1080/13543784.2019.1555237] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Prostaglandin D2 (PGD2) is a major cyclooxygenase mediator that is synthesized by activated human mast cells and other immune cells. The biological effects of PGD2 are mediated by D-prostanoid (DP1), DP2 (CRTH2) and thromboxane prostanoid (TP) receptors that are expressed on several immune and non-immune cells involved in allergic inflammation. PGD2 exerts various proinflammatory effects relevant to the pathophysiology of allergic disorders. Several selective, orally active, DP2 receptor antagonists and a small number of DP1 receptor antagonists are being developed for the treatment of allergic disorders. AREAS COVERED The role of DP2 and DP1 receptor antagonists in the treatment of asthma and allergic rhinitis. EXPERT OPINION Head-to-head studies that compare DP1 antagonists with the standard treatment for allergic rhinitis are necessary to verify the role of these novel drugs as mono- or combination therapies. Further clinical trials are necessary to verify whether DP2 antagonists as monotherapies or, more likely, as add-on therapies, will be effective for the treatment of different phenotypes of adult and childhood asthma. Long-term studies are necessary to evaluate the safety of targeted anti-PGD2 treatments.
Collapse
Affiliation(s)
- Giancarlo Marone
- a Department of Public Health , University of Naples Federico II , Naples , Italy.,b Monaldi Hospital Pharmacy , Naples , Italy
| | - Maria Rosaria Galdiero
- c Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI) , University of Naples Federico II , Naples , Italy.,d WAO Center of Excellence , Naples , Italy
| | - Antonio Pecoraro
- c Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI) , University of Naples Federico II , Naples , Italy.,d WAO Center of Excellence , Naples , Italy
| | - Valentina Pucino
- e William Harvey Research Institute, Barts and The London School of Medicine &Dentistry , Queen Mary University of London , London , UK
| | - Gjada Criscuolo
- c Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI) , University of Naples Federico II , Naples , Italy.,d WAO Center of Excellence , Naples , Italy
| | - Maria Triassi
- a Department of Public Health , University of Naples Federico II , Naples , Italy
| | - Gilda Varricchi
- c Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI) , University of Naples Federico II , Naples , Italy.,d WAO Center of Excellence , Naples , Italy
| |
Collapse
|
7
|
Sadam H, Pihlak A, Kivil A, Pihelgas S, Jaago M, Adler P, Vilo J, Vapalahti O, Neuman T, Lindholm D, Partinen M, Vaheri A, Palm K. Prostaglandin D2 Receptor DP1 Antibodies Predict Vaccine-induced and Spontaneous Narcolepsy Type 1: Large-scale Study of Antibody Profiling. EBioMedicine 2018; 29:47-59. [PMID: 29449194 PMCID: PMC5925455 DOI: 10.1016/j.ebiom.2018.01.043] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 01/23/2018] [Accepted: 01/31/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Neuropathological findings support an autoimmune etiology as an underlying factor for loss of orexin-producing neurons in spontaneous narcolepsy type 1 (narcolepsy with cataplexy; sNT1) as well as in Pandemrix influenza vaccine-induced narcolepsy type 1 (Pdmx-NT1). The precise molecular target or antigens for the immune response have, however, remained elusive. METHODS Here we have performed a comprehensive antigenic repertoire analysis of sera using the next-generation phage display method - mimotope variation analysis (MVA). Samples from 64 children and adolescents were analyzed: 10 with Pdmx-NT1, 6 with sNT1, 16 Pandemrix-vaccinated, 16 H1N1 infected, and 16 unvaccinated healthy individuals. The diagnosis of NT1 was defined by the American Academy of Sleep Medicine international criteria of sleep disorders v3. FINDINGS Our data showed that although the immunoprofiles toward vaccination were generally similar in study groups, there were also striking differences in immunoprofiles between sNT1 and Pdmx-NT1 groups as compared with controls. Prominent immune response was observed to a peptide epitope derived from prostaglandin D2 receptor (DP1), as well as peptides homologous to B cell lymphoma 6 protein. Further validation confirmed that these can act as true antigenic targets in discriminating NT1 diseased along with a novel epitope of hemagglutinin of H1N1 to delineate exposure to H1N1. INTERPRETATION We propose that DP1 is a novel molecular target of autoimmune response and presents a potential diagnostic biomarker for NT1. DP1 is involved in the regulation of non-rapid eye movement (NREM) sleep and thus alterations in its functions could contribute to the disturbed sleep regulation in NT1 that warrants further studies. Together our results also show that MVA is a helpful method for finding novel peptide antigens to classify human autoimmune diseases, possibly facilitating the design of better therapies.
Collapse
Affiliation(s)
- Helle Sadam
- Protobios Llc, Mäealuse 4, 12618 Tallinn, Estonia; Department of Gene Technology, Tallinn University of Technology, Akadeemia Tee 15, 12618 Tallinn, Estonia
| | - Arno Pihlak
- Protobios Llc, Mäealuse 4, 12618 Tallinn, Estonia; Department of Gene Technology, Tallinn University of Technology, Akadeemia Tee 15, 12618 Tallinn, Estonia
| | - Anri Kivil
- Protobios Llc, Mäealuse 4, 12618 Tallinn, Estonia
| | | | | | - Priit Adler
- Institute of Computer Science, University of Tartu, Liivi 2-314, 50409 Tartu, Estonia; Quretec LLC, Ülikooli 6a, 51003 Tartu, Estonia
| | - Jaak Vilo
- Institute of Computer Science, University of Tartu, Liivi 2-314, 50409 Tartu, Estonia; Quretec LLC, Ülikooli 6a, 51003 Tartu, Estonia
| | - Olli Vapalahti
- Department of Virology, Medicum, Haartmaninkatu 3, 00014 University of Helsinki, Finland; Department of Veterinary Biosciences, University of Helsinki, Agnes Sjöbergin Katu 2, 00014 University of Helsinki, Finland; Virology and Immunology, HUSLAB, Helsinki University Hospital, 00290 Helsinki, Finland
| | - Toomas Neuman
- Protobios Llc, Mäealuse 4, 12618 Tallinn, Estonia; IPDx Immunoprofiling Diagnostics GmbH, Deutscher Platz 5e, 04103 Leipzig, Germany
| | - Dan Lindholm
- Department of Biochemistry and Developmental Biology, Medicum, Haartmaninkatu 8, 00014 University of Helsinki, Finland; Minerva Foundation Medical Research Institute, Tukholmankatu 8, 00290 Helsinki, Finland
| | - Markku Partinen
- Finnish Narcolepsy Research Center, Helsinki Sleep Clinic, Vitalmed Research Center, Valimotie 21, 00380, Helsinki, Finland
| | - Antti Vaheri
- Department of Virology, Medicum, Haartmaninkatu 3, 00014 University of Helsinki, Finland
| | - Kaia Palm
- Protobios Llc, Mäealuse 4, 12618 Tallinn, Estonia; Department of Gene Technology, Tallinn University of Technology, Akadeemia Tee 15, 12618 Tallinn, Estonia.
| |
Collapse
|
8
|
Sasaki F, Koga T, Saeki K, Okuno T, Kazuno S, Fujimura T, Ohkawa Y, Yokomizo T. Biochemical and immunological characterization of a novel monoclonal antibody against mouse leukotriene B4 receptor 1. PLoS One 2017; 12:e0185133. [PMID: 28922396 PMCID: PMC5602668 DOI: 10.1371/journal.pone.0185133] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 09/05/2017] [Indexed: 01/27/2023] Open
Abstract
Leukotriene B4 (LTB4) receptor 1 (BLT1) is a G protein-coupled receptor expressed in various leukocyte subsets; however, the precise expression of mouse BLT1 (mBLT1) has not been reported because a mBLT1 monoclonal antibody (mAb) has not been available. In this study, we present the successful establishment of a hybridoma cell line (clone 7A8) that produces a high-affinity mAb for mBLT1 by direct immunization of BLT1-deficient mice with mBLT1-overexpressing cells. The specificity of clone 7A8 was confirmed using mBLT1-overexpressing cells and mouse peripheral blood leukocytes that endogenously express BLT1. Clone 7A8 did not cross-react with human BLT1 or other G protein-coupled receptors, including human chemokine (C-X-C motif) receptor 4. The 7A8 mAb binds to the second extracellular loop of mBLT1 and did not affect LTB4 binding or intracellular calcium mobilization by LTB4. The 7A8 mAb positively stained Gr-1-positive granulocytes, CD11b-positive granulocytes/monocytes, F4/80-positive monocytes, CCR2-high and CCR2-low monocyte subsets in the peripheral blood and a CD4-positive T cell subset, Th1 cells differentiated in vitro from naïve CD4-positive T cells. This mAb was able to detect Gr-1-positive granulocytes and monocytes in the spleens of naïve mice by immunohistochemistry. Finally, intraperitoneal administration of 7A8 mAb depleted granulocytes and monocytes in the peripheral blood. We have therefore succeeded in generating a high-affinity anti-mBLT1 mAb that is useful for analyzing mBLT1 expression in vitro and in vivo.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Murine-Derived/chemistry
- Antibodies, Monoclonal, Murine-Derived/immunology
- Antibodies, Monoclonal, Murine-Derived/pharmacology
- CHO Cells
- Calcium Signaling/drug effects
- Cell Differentiation/immunology
- Cricetinae
- Cricetulus
- Granulocytes/immunology
- Leukotriene B4/immunology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Monocytes/immunology
- Protein Structure, Secondary
- Receptors, Leukotriene B4/antagonists & inhibitors
- Receptors, Leukotriene B4/chemistry
- Receptors, Leukotriene B4/immunology
- Th1 Cells/immunology
Collapse
Affiliation(s)
- Fumiyuki Sasaki
- Department of Biochemistry, Juntendo University School of Medicine, Tokyo, Japan
| | - Tomoaki Koga
- Department of Biochemistry, Juntendo University School of Medicine, Tokyo, Japan
| | - Kazuko Saeki
- Department of Biochemistry, Juntendo University School of Medicine, Tokyo, Japan
| | - Toshiaki Okuno
- Department of Biochemistry, Juntendo University School of Medicine, Tokyo, Japan
| | - Saiko Kazuno
- Laboratory of Proteomics and Biomolecular Science Research Support Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tsutomu Fujimura
- Laboratory of Bioanalytical Chemistry, Tohoku Medical and Pharmaceutical University, Miyagi, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Takehiko Yokomizo
- Department of Biochemistry, Juntendo University School of Medicine, Tokyo, Japan
- * E-mail:
| |
Collapse
|