1
|
Philip M, Kal AKK, Subhahar MB, Karatt TK, Graiban FM, Ajeebsanu MM, Joseph M, Jose SV. Equine metabolic investigation of the phosphodiesterase-4 inhibitor ibudilast as a potential performance enhancer. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2024; 38:e9916. [PMID: 39307998 DOI: 10.1002/rcm.9916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/02/2024] [Accepted: 09/05/2024] [Indexed: 09/25/2024]
Abstract
RATIONALE Phosphodiesterase 4 (PDE4) inhibitors are a newer class of drugs that induce bronchodilation and have anti-inflammatory effects, making them susceptible to misuse as performance enhancers in competitive sports. METHODS This study explores the metabolic conversion of PDE4 inhibitor ibudilast in thoroughbred horses after oral administration and in vitro using equine liver microsomes and Cunninghamella elegans. A liquid chromatography-high resolution mass spectrometry method was used to postulate the plausible structures of the detected metabolites. RESULTS A total of 20 in vivo metabolites were identified under experimental conditions, including 12 Phase I and 8 Phase II conjugated metabolites. Phase I metabolites were predominantly formed through hydroxylation (mono-, di-, and tri-hydroxylation). Demethylated metabolites were also identified during this investigation. Additionally, the research detected Phase II metabolites conjugated with glucuronic and sulfonic acids. CONCLUSIONS The data presented here can assist in detecting the PDE4 inhibitor ibudilast and uncover its illicit use in competitive sports.
Collapse
Affiliation(s)
- Moses Philip
- Equine Forensic Unit, Central Veterinary Research Laboratory, Dubai, United Arab Emirates
| | | | | | - Tajudheen K Karatt
- Equine Forensic Unit, Central Veterinary Research Laboratory, Dubai, United Arab Emirates
| | - Fatma Mohammed Graiban
- Equine Forensic Unit, Central Veterinary Research Laboratory, Dubai, United Arab Emirates
| | | | - Marina Joseph
- Department of Bacteriology, Diagnostic Section, Central Veterinary Research Laboratory, Dubai, United Arab Emirates
| | - Shantymol V Jose
- Department of Bacteriology, Diagnostic Section, Central Veterinary Research Laboratory, Dubai, United Arab Emirates
| |
Collapse
|
2
|
Chen Q, Xia Y, Liu HN, Chi Y, Li X, Shan LS, Dai B, Zhu Y, Wang YT, Miao X, Sun Q. Synthetic approaches and clinical application of representative small-molecule inhibitors of phosphodiesterase. Eur J Med Chem 2024; 277:116769. [PMID: 39163778 DOI: 10.1016/j.ejmech.2024.116769] [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: 05/18/2024] [Revised: 07/25/2024] [Accepted: 08/11/2024] [Indexed: 08/22/2024]
Abstract
Phosphodiesterases (PDEs) constitute a family of enzymes that play a pivotal role in the regulation of intracellular levels of cyclic nucleotides, including cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Dysregulation of PDE activity has been implicated in diverse pathological conditions encompassing cardiovascular disorders, pulmonary diseases, and neurological disorders. Small-molecule inhibitors targeting PDEs have emerged as promising therapeutic agents for the treatment of these ailments, some of which have been approved for their clinical use. Despite their success, challenges such as resistance mechanisms and off-target effects persist, urging continuous research for the development of next-generation PDE inhibitors. The objective of this review is to provide an overview of the synthesis and clinical application of representative approved small-molecule PDE inhibitors, with the aim of offering guidance for further advancements in the development of novel PDE inhibitors.
Collapse
Affiliation(s)
- Qingqing Chen
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Xia
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - He-Nan Liu
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuan Chi
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Xun Li
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Li-Shen Shan
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Bing Dai
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Ying Zhu
- Department of Neurology, The First Hospital of China Medical University, Shenyang, China
| | - Ya-Tao Wang
- Rega Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49-Box 1041, 3000, Leuven, Belgium.
| | - Xinxin Miao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Qian Sun
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
3
|
Zhang C, Xue ZH, Luo WH, Jiang MY, Wu Y. The therapeutic potential of phosphodiesterase 9 (PDE9) inhibitors: a patent review (2018-present). Expert Opin Ther Pat 2024; 34:759-772. [PMID: 38979973 DOI: 10.1080/13543776.2024.2376632] [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: 03/19/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024]
Abstract
INTRODUCTION Phosphodiesterase 9 (PDE9) has been demonstrated as a potential target for neurological disorders and cardiovascular diseases, such as Alzheimer's disease and heart failure. For the last few years, a series of PDE9 inhibitors with structural diversities have been developed and patented by researchers and pharmaceutical companies, providing insights into first-in-class therapies of PDE9 drug candidates. AREA COVERED This review provides an overview of PDE9 inhibitors in patents from 2018 to the present. EXPERT OPINION Only a few of the current PDE9 inhibitors are highly selective over other PDEs, which limits their application in pharmacological and clinical research. The design and development of highly selective PDE9 inhibitors remain the top priority in future research. The advantages of targeting PDE9 rather than other PDEs in treating neurodegenerative diseases need to be explained thoroughly. Besides, application of PDE9 inhibitor-based combination therapies sheds light on treating diabetes and refractory heart diseases. Finally, PDE9 inhibitors should be further explored in clinical indications beyond neurological disorders and cardiovascular diseases.
Collapse
Affiliation(s)
- Chen Zhang
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan, P. R. China
| | - Zhao-Hang Xue
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan, P. R. China
| | - Wei-Hao Luo
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Zhongshan, P. R. China
| | - Mei-Yan Jiang
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, P. R. China
| | - Yinuo Wu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, P. R. China
| |
Collapse
|
4
|
Wei R, Zong F, Dong J, Zhao W, Zhang F, Wang W, Zhao S, Wang Z, Zhang F, Zhang HT. Identification of Phosphodiesterase-7A (PDE7A) as a Novel Target for Reducing Ethanol Consumption in Mice. Int J Neuropsychopharmacol 2024; 27:pyae032. [PMID: 39099166 PMCID: PMC11348009 DOI: 10.1093/ijnp/pyae032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/29/2024] [Indexed: 08/06/2024] Open
Abstract
BACKGROUND Ethanol elicits a rapid stimulatory effect and a subsequent, prolonged sedative response, which are potential predictors of EtOH consumption by decreasing adenosine signaling; this phenomenon also reflects the obvious sex difference. cAMP (cyclic Adenosine Monophosphate)-PKA (Protein Kinase A) signaling pathway modulation can influence the stimulatory and sedative effects induced by EtOH in mice. This study's objective is to clarify the role of phosphodiesterase (PDE) in mediating the observed sex differences in EtOH responsiveness between male and female animals. METHODS EtOH was administered i.p. for 7 days to identify the changes in PDE isoforms in response to EtOH treatment. Additionally, EtOH consumption and preference of male and female C57BL/6J mice were assessed using the drinking-in-the-dark and 2-bottle choice tests. Further, pharmacological inhibition of PDE7A heterozygote knockout mice was performed to investigate its effects on EtOH-induced stimulation and sedation in both male and female mice. Finally, Western blotting analysis was performed to evaluate the alterations in cAMP-PKA/Epac2 pathways. RESULTS EtOH administration resulted in an immediate upregulation in PDE7A expression in female mice, indicating a strong association between PDE7A and EtOH stimulation. Through the pharmacological inhibition of PDE7A KD mice, we have demonstrated for the first time, to our knowledge, that PDE7A selectively attenuates EtOH responsiveness and consumption exclusively in female mice, whichmay be associated with the cAMP-PKA/Epac2 pathway and downstream phosphorylation of CREB and ERK1/2. CONCLUSIONS Inhibition or knockdown of PDE7A attenuates EtOH responsivenessand consumption exclusively in female mice, which is associated with alterations in the cAMP-PKA/Epac2 signaling pathways, thereby highlighting its potential as a novel therapeutic target for alcohol use disorder.
Collapse
Affiliation(s)
- Ran Wei
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
- Weifang Chinese Medical Hospital, Shandong Second Medical University, Weifang, China
| | - Fangjiao Zong
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Jiahao Dong
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
- Weifang People’s Hospital, Shandong Second Medical University, Weifang, China
| | - Wei Zhao
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Fangfang Zhang
- Institude of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, China
| | - Wei Wang
- Institude of Pharmacology, Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, China
| | - Shuang Zhao
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Ziqi Wang
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Fang Zhang
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| | - Han-Ting Zhang
- Department of Pharmacology, Qingdao University School of Pharmacy, Qingdao, China
| |
Collapse
|
5
|
Chen K, Wang D, Qian M, Weng M, Lu Z, Zhang K, Jin Y. Endothelial cell dysfunction and targeted therapeutic drugs in sepsis. Heliyon 2024; 10:e33340. [PMID: 39027563 PMCID: PMC11255673 DOI: 10.1016/j.heliyon.2024.e33340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 06/19/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024] Open
Abstract
Sepsis is a life-threatening organ dysfunction caused by an abnormal host response to microbial infections. During its pathogenesis, vascular endothelial cells (ECs) play a pivotal role as essential components in maintaining microcirculatory homeostasis. This article aims to comprehensively review the multifaceted physiological functions of vascular ECs, elucidate the alterations in their functionality throughout the course of sepsis, and explore recent advancements in research concerning sepsis-related therapeutic drugs targeting ECs.
Collapse
Affiliation(s)
- Kunwei Chen
- Department of Anesthesiology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dongdong Wang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Minyue Qian
- Department of Anesthesiology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mengcao Weng
- Department of Anesthesiology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhongteng Lu
- Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Kai Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yue Jin
- Department of Anesthesiology, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Children's Hospital, National Clinical Research Center for Child Health, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
6
|
Staller DW, Bennett RG, Mahato RI. Therapeutic perspectives on PDE4B inhibition in adipose tissue dysfunction and chronic liver injury. Expert Opin Ther Targets 2024; 28:545-573. [PMID: 38878273 PMCID: PMC11305103 DOI: 10.1080/14728222.2024.2369590] [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: 04/01/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
INTRODUCTION Chronic liver disease (CLD) is a complex disease associated with profound dysfunction. Despite an incredible burden, the first and only pharmacotherapy for metabolic-associated steatohepatitis was only approved in March of this year, indicating a gap in the translation of preclinical studies. There is a body of preclinical work on the application of phosphodiesterase 4 inhibitors in CLD, none of these molecules have been successfully translated into clinical use. AREAS COVERED To design therapies to combat CLD, it is essential to consider the dysregulation of other tissues that contribute to its development and progression. As such, proper therapies must combat this throughout the body rather than focusing only on the liver. To detail this, literature characterizing the pathogenesis of CLD was pulled from PubMed, with a particular focus placed on the role of PDE4 in inflammation and metabolism. Then, the focus is shifted to detailing the available information on existing PDE4 inhibitors. EXPERT OPINION This review gives a brief overview of some of the pathologies of organ systems that are distinct from the liver but contribute to disease progression. The demonstrated efficacy of PDE4 inhibitors in other human inflammatory diseases should earn them further examination for the treatment of CLD.
Collapse
Affiliation(s)
- Dalton W. Staller
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Robert G. Bennett
- Department of Internal Medicine, Division of Diabetes Endocrinology and Metabolism, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
- VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Ram I. Mahato
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
7
|
Alam A, Gul S, Zainab, Khan M, Elhenawy AA, Islam MS, Ali M, Ali Shah SA, Latif A, Ahmad M. Synthesis of 2,4-dihydroxyacetophenone derivatives as potent PDE-1 and -3 inhibitors: in vitro and in silico insights. Future Med Chem 2024; 16:1185-1203. [PMID: 38989989 PMCID: PMC11382721 DOI: 10.1080/17568919.2024.2342707] [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: 10/29/2023] [Accepted: 04/08/2024] [Indexed: 07/12/2024] Open
Abstract
Aim: Synthesis of novel bis-Schiff bases having potent inhibitory activity against phosphodiesterase (PDE-1 and -3) enzymes, potentially offering therapeutic implications for various conditions. Methods: Bis-Schiff bases were synthesized by refluxing 2,4-dihydroxyacetophenone with hydrazine hydrate, followed by treatment of substituted aldehydes with the resulting hydrazone to obtain the product compounds. After structural confirmation, the compounds were screened for their in vitro PDE-1 and -3 inhibitory activities. Results: The prepared compounds exhibited noteworthy inhibitory efficacy against PDE-1 and -3 enzymes by comparing with suramin standard. To clarify the binding interactions between the drugs, PDE-1 and -3 active sites, molecular docking studies were carried out. Conclusion: The potent compounds discovered in this study may be good candidates for drug development.
Collapse
Affiliation(s)
- Aftab Alam
- Department of Chemistry, University of Malakand, P.O. Box 18800, Dir Lower, Pakistan
| | - Sana Gul
- Department of Chemistry, University of Malakand, P.O. Box 18800, Dir Lower, Pakistan
| | - Zainab
- College of Chemistry & Materials Science, Hebei Normal University, Shijiazhuang, 050024, China
| | - Majid Khan
- H.E.J. Research Institute of Chemistry, International Center for Chemical & Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Ahmed A Elhenawy
- Chemistry Department, Faculty of Science, Al-Azhar University, Nasr City, 11884, Cairo, Egypt
| | - Mohammad Shahidul Islam
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Mumtaz Ali
- Department of Chemistry, University of Malakand, P.O. Box 18800, Dir Lower, Pakistan
| | - Syed Adnan Ali Shah
- Faculty of Pharmacy, Universiti Teknologi MARA Puncak Alam Campus, 42300 Bandar Puncak Alam,Selangor D. E., Malaysia
| | - Abdul Latif
- Department of Chemistry, University of Malakand, P.O. Box 18800, Dir Lower, Pakistan
| | - Manzoor Ahmad
- Department of Chemistry, University of Malakand, P.O. Box 18800, Dir Lower, Pakistan
| |
Collapse
|
8
|
Li Q, Liao Q, Qi S, Huang H, He S, Lyu W, Liang J, Qin H, Cheng Z, Yu F, Dong X, Wang Z, Han L, Han Y. Opportunities and perspectives of small molecular phosphodiesterase inhibitors in neurodegenerative diseases. Eur J Med Chem 2024; 271:116386. [PMID: 38614063 DOI: 10.1016/j.ejmech.2024.116386] [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: 02/05/2024] [Revised: 03/19/2024] [Accepted: 04/01/2024] [Indexed: 04/15/2024]
Abstract
Phosphodiesterase (PDE) is a superfamily of enzymes that are responsible for the hydrolysis of two second messengers: cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). PDE inhibition promotes the gene transcription by activating cAMP-response element binding protein (CREB), initiating gene transcription of brain-derived neurotrophic factor (BDNF). The procedure exerts neuroprotective profile, and motor and cognitive improving efficacy. From this point of view, PDE inhibition will provide a promising therapeutic strategy for treating neurodegenerative disorders. Herein, we summarized the PDE inhibitors that have entered the clinical trials or been discovered in recent five years. Well-designed clinical or preclinical investigations have confirmed the effectiveness of PDE inhibitors, such as decreasing Aβ oligomerization and tau phosphorylation, alleviating neuro-inflammation and oxidative stress, modulating neuronal plasticity and improving long-term cognitive impairment.
Collapse
Affiliation(s)
- Qi Li
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China.
| | - Qinghong Liao
- Shandong Kangqiao Biotechnology Co., Ltd, Qingdao, 266033, Shandong, PR China
| | - Shulei Qi
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - He Huang
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Siyu He
- Guizhou Province Engineering Technology Research Center for Chemical Drug R&D, Guizhou Medical University, Guiyang, 550004, Guizhou, PR China
| | - Weiping Lyu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, PR China
| | - Jinxin Liang
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Huan Qin
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Zimeng Cheng
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Fan Yu
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Xue Dong
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China
| | - Ziming Wang
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China; School of Pharmacy, Binzhou Medical University, Yantai, 256699, Shandong, PR China
| | - Lingfei Han
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, PR China
| | - Yantao Han
- Department of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong, PR China.
| |
Collapse
|
9
|
Nongthombam PD, Haobam R. Targeting phosphodiesterase 4 as a potential therapy for Parkinson's disease: a review. Mol Biol Rep 2024; 51:510. [PMID: 38622307 DOI: 10.1007/s11033-024-09484-8] [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: 12/08/2023] [Accepted: 03/26/2024] [Indexed: 04/17/2024]
Abstract
Phosphodiesterases (PDEs) have become a promising therapeutic target for various disorders. PDEs are a vast and diversified family of enzymes that degrade cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP), which have several biochemical and physiological functions. Phosphodiesterase 4 (PDE4) is the most abundant PDE in the central nervous system (CNS) and is extensively expressed in the mammalian brain, where it catalyzes the hydrolysis of intracellular cAMP. An alteration in the balance of PDE4 and cAMP results in the dysregulation of different biological mechanisms involved in neurodegenerative diseases. By inhibiting PDE4 with drugs, the levels of cAMP inside the cells could be stabilized, which may improve the symptoms of mental and neurological disorders such as memory loss, depression, and Parkinson's disease (PD). Though numerous studies have shown that phosphodiesterase 4 inhibitors (PDE4Is) are beneficial in PD, there are presently no approved PDE4I drugs for PD. This review presents an overview of PDE4Is and their effects on PD, their possible underlying mechanism in the restoration/protection of dopaminergic cell death, which holds promise for developing PDE4Is as a treatment strategy for PD. Methods on how these drugs could be effectively delivered to develop as a promising treatment for PD have been suggested.
Collapse
Affiliation(s)
| | - Reena Haobam
- Department of Biotechnology, Manipur University, Canchipur, Imphal, 795003, India.
| |
Collapse
|
10
|
Bae J, Park SJ. The Combination of Oolonghomobisflavan B and Diallyl Disulfide Induces Apoptotic Cell Death via 67-kDa Laminin Receptor/Cyclic Guanosine Monophosphate in Acute Myeloid Leukemia Cells. Curr Issues Mol Biol 2024; 46:2444-2455. [PMID: 38534770 DOI: 10.3390/cimb46030154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/07/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
Diallyl disulfide (DADS) is a well-known principal functional component derived from garlic (Allium sativum) that has various health benefits. Previously, we identified a 67-kDa laminin receptor, a receptor for oolong tea polyphenol oolonghomobisflavan B (OHBFB). However, its molecular mechanisms still remain to be elucidated. Here, we show that DADS synergistically enhanced the effect of the oolong tea polyphenol oolonghomobisflavan B (OHBFB), which induces apoptosis in acute myeloid leukemia (AML) cancer cells without affecting normal human peripheral blood mononuclear cells (PBMCs). The underlying mechanism of OHBFB-induced anti-AML effects involves the upregulation of the 67-kDa laminin receptor/endothelial nitric oxide synthase/cyclic guanosine monophosphate (cGMP)/protein kinase c delta (PKCδ)/acid sphingomyelinase (ASM)/cleaved caspase-3 signaling pathway. In conclusion, we show that the combination of OHBFB and DADS synergistically induced apoptotic cell death in AML cells through activation of 67LR/cGMP/PKCδ/ASM signaling pathway. Moreover, in this mechanism, we demonstrate DADS may reduce the enzyme activity of phosphodiesterase, which is a negative regulator of cGMP that potentiates OHBFB-induced AML apoptotic cell death without affecting normal PBMCs.
Collapse
Affiliation(s)
- Jaehoon Bae
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-gil, Jeongeup-si 56212, Republic of Korea
| | - Su-Jin Park
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-gil, Jeongeup-si 56212, Republic of Korea
| |
Collapse
|
11
|
Stengl M, Schneider AC. Contribution of membrane-associated oscillators to biological timing at different timescales. Front Physiol 2024; 14:1243455. [PMID: 38264332 PMCID: PMC10803594 DOI: 10.3389/fphys.2023.1243455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 12/12/2023] [Indexed: 01/25/2024] Open
Abstract
Environmental rhythms such as the daily light-dark cycle selected for endogenous clocks. These clocks predict regular environmental changes and provide the basis for well-timed adaptive homeostasis in physiology and behavior of organisms. Endogenous clocks are oscillators that are based on positive feedforward and negative feedback loops. They generate stable rhythms even under constant conditions. Since even weak interactions between oscillators allow for autonomous synchronization, coupling/synchronization of oscillators provides the basis of self-organized physiological timing. Amongst the most thoroughly researched clocks are the endogenous circadian clock neurons in mammals and insects. They comprise nuclear clockworks of transcriptional/translational feedback loops (TTFL) that generate ∼24 h rhythms in clock gene expression entrained to the environmental day-night cycle. It is generally assumed that this TTFL clockwork drives all circadian oscillations within and between clock cells, being the basis of any circadian rhythm in physiology and behavior of organisms. Instead of the current gene-based hierarchical clock model we provide here a systems view of timing. We suggest that a coupled system of autonomous TTFL and posttranslational feedback loop (PTFL) oscillators/clocks that run at multiple timescales governs adaptive, dynamic homeostasis of physiology and behavior. We focus on mammalian and insect neurons as endogenous oscillators at multiple timescales. We suggest that neuronal plasma membrane-associated signalosomes constitute specific autonomous PTFL clocks that generate localized but interlinked oscillations of membrane potential and intracellular messengers with specific endogenous frequencies. In each clock neuron multiscale interactions of TTFL and PTFL oscillators/clocks form a temporally structured oscillatory network with a common complex frequency-band comprising superimposed multiscale oscillations. Coupling between oscillator/clock neurons provides the next level of complexity of an oscillatory network. This systemic dynamic network of molecular and cellular oscillators/clocks is suggested to form the basis of any physiological homeostasis that cycles through dynamic homeostatic setpoints with a characteristic frequency-band as hallmark. We propose that mechanisms of homeostatic plasticity maintain the stability of these dynamic setpoints, whereas Hebbian plasticity enables switching between setpoints via coupling factors, like biogenic amines and/or neuropeptides. They reprogram the network to a new common frequency, a new dynamic setpoint. Our novel hypothesis is up for experimental challenge.
Collapse
Affiliation(s)
- Monika Stengl
- Department of Biology, Animal Physiology/Neuroethology, University of Kassel, Kassel, Germany
| | | |
Collapse
|
12
|
Metkar SK, Yan Y, Lu Y, Lu J, Zhu X, Du F, Xu Y. Phosphodiesterase 2 and Its Isoform A as Therapeutic Targets in the Central Nervous System Disorders. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:941-955. [PMID: 37855295 DOI: 10.2174/1871527323666230811093126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 06/15/2023] [Accepted: 07/07/2023] [Indexed: 10/20/2023]
Abstract
Cyclic adenosine monophosphates (cAMP) and cyclic guanosine monophosphate (cGMP) are two essential second messengers, which are hydrolyzed by phosphodiesterase's (PDEs), such as PDE-2. Pharmacological inhibition of PDE-2 (PDE2A) in the central nervous system improves cAMP and cGMP signaling, which controls downstream proteins related to neuropsychiatric, neurodegenerative, and neurodevelopmental disorders. Considering that there are no specific treatments for these disorders, PDE-2 inhibitors' development has gained more attention in the recent decade. There is high demand for developing new-generation drugs targeting PDE2 for treating diseases in the central nervous and peripheral systems. This review summarizes the relationship between PDE-2 with neuropsychiatric, neurodegenerative, and neurodevelopmental disorders as well as its possible treatment, mainly involving inhibitors of PDE2.
Collapse
Affiliation(s)
- Sanjay K Metkar
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Yuqing Yan
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Yue Lu
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| | - Jianming Lu
- Codex BioSolutions Inc. 12358 Parklawn Drive, Suite 250A, Rockville, MD 20852, Maryland
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio 44106; USA
| | - Fu Du
- FD NeuroTechnologies Consulting & Services, Inc., Columbia, MD 21046, Maryland
| | - Ying Xu
- Department of Anesthesiology, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07103, USA
| |
Collapse
|
13
|
Crowley EL, Gooderham MJ. Phosphodiesterase-4 Inhibition in the Management of Psoriasis. Pharmaceutics 2023; 16:23. [PMID: 38258034 PMCID: PMC10819567 DOI: 10.3390/pharmaceutics16010023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/16/2023] [Accepted: 12/17/2023] [Indexed: 01/24/2024] Open
Abstract
Psoriasis is a common chronic immune-mediated disease with many comorbidities and impacts on quality of life. Among the treatments for psoriasis, phosphodiesterase-4 (PDE4) inhibitors are emerging with expanding options. PDE4 inhibitors play a pivotal role in the inflammatory cascade by degrading cyclic adenosine monophosphate (cAMP), contributing to pro-inflammatory mediator production. Apremilast, an oral PDE4 inhibitor, is approved for psoriasis. While effective, its adverse effects can limit its utility. Roflumilast, a topical PDE4 inhibitor, was also recently approved for psoriasis and shows promise in clinical trials. Crisaborole, a PDE4 inhibitor approved for atopic dermatitis, has also been studied in psoriasis. This review summarizes evidence from randomized clinical trials regarding the efficacy and safety of PDE4 inhibitors in psoriasis treatment. By highlighting their potential benefits and limitations, this review provides valuable insights for clinicians and researchers aiming to optimize psoriasis management.
Collapse
Affiliation(s)
- Erika L. Crowley
- Faculty of Medicine, University of British Columbia Okanagan, 3333 University Way, Kelowna, BC V1V 1V7, Canada;
| | - Melinda J. Gooderham
- SKiN Centre for Dermatology, 775 Monaghan Rd, Peterborough, ON K9J 5K2, Canada
- Probity Medical Research, 139 Union St E, Waterloo, ON N2J 1C4, Canada
- Department of Medicine, Queen’s University, 99 University Ave, Kingston, ON K7L 3N6, Canada
| |
Collapse
|
14
|
Puertas-Umbert L, Alonso J, Hove-Madsen L, Martínez-González J, Rodríguez C. PDE4 Phosphodiesterases in Cardiovascular Diseases: Key Pathophysiological Players and Potential Therapeutic Targets. Int J Mol Sci 2023; 24:17017. [PMID: 38069339 PMCID: PMC10707411 DOI: 10.3390/ijms242317017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
3',5'-cyclic adenosine monophosphate (cAMP) is a second messenger critically involved in the control of a myriad of processes with significant implications for vascular and cardiac cell function. The temporal and spatial compartmentalization of cAMP is governed by the activity of phosphodiesterases (PDEs), a superfamily of enzymes responsible for the hydrolysis of cyclic nucleotides. Through the fine-tuning of cAMP signaling, PDE4 enzymes could play an important role in cardiac hypertrophy and arrhythmogenesis, while it decisively influences vascular homeostasis through the control of vascular smooth muscle cell proliferation, migration, differentiation and contraction, as well as regulating endothelial permeability, angiogenesis, monocyte/macrophage activation and cardiomyocyte function. This review summarizes the current knowledge and recent advances in understanding the contribution of the PDE4 subfamily to cardiovascular function and underscores the intricate challenges associated with targeting PDE4 enzymes as a therapeutic strategy for the management of cardiovascular diseases.
Collapse
Affiliation(s)
- Lídia Puertas-Umbert
- Institut de Recerca Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (L.P.-U.); (J.A.); (L.H.-M.)
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Judith Alonso
- Institut de Recerca Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (L.P.-U.); (J.A.); (L.H.-M.)
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), 08036 Barcelona, Spain
| | - Leif Hove-Madsen
- Institut de Recerca Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (L.P.-U.); (J.A.); (L.H.-M.)
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), 08036 Barcelona, Spain
| | - José Martínez-González
- Institut de Recerca Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (L.P.-U.); (J.A.); (L.H.-M.)
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), 08036 Barcelona, Spain
| | - Cristina Rodríguez
- Institut de Recerca Sant Pau (IR SANT PAU), 08041 Barcelona, Spain; (L.P.-U.); (J.A.); (L.H.-M.)
- CIBER de Enfermedades Cardiovasculares, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas (IIBB-CSIC), 08036 Barcelona, Spain
| |
Collapse
|
15
|
Bae J, Kumazoe M, Park S, Fujimura Y, Tachibana H. The anti-cancer effect of epigallocatechin-3-O-gallate against multiple myeloma cells is potentiated by 5,7-dimethoxyflavone. FEBS Open Bio 2023; 13:2147-2156. [PMID: 37730921 PMCID: PMC10626272 DOI: 10.1002/2211-5463.13708] [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: 04/21/2023] [Revised: 08/21/2023] [Accepted: 09/19/2023] [Indexed: 09/22/2023] Open
Abstract
(-)-Epigallocatechin-3-O-gallate (EGCG) is one of the major components of green tea polyphenol. Previous studies have shown that EGCG induces cancer-specific cell death in vitro and in vivo without causing severe side effects. However, the anti-cancer effect of EGCG alone is limited. 5,7-dimethoxyflavone (5,7-DMF), one of the principal functional components of black ginger (Kaempferia parviflora), also exerts anti-cancer effects. Here, we show that 5,7-DMF synergistically enhances the anti-cancer effect of EGCG in multiple myeloma cells by potentiating EGCG-induced intracellular cyclic guanosine monophosphate (cGMP) production. Moreover, the combination of EGCG and 5,7-DMF induces apoptotic cell death in multiple myeloma cells, and this is accompanied by activation of the cGMP/acid sphingomyelinase (ASM)/cleaved caspase-3 pathway. In conclusion, we have shown that 5,7-DMF enhances the anti-cancer effect of EGCG by upregulating cGMP in multiple myeloma cells.
Collapse
Affiliation(s)
- Jaehoon Bae
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of AgricultureKyushu UniversityFukuokaJapan
- Functional Biomaterial Research CenterKorea Research Institute of Bioscience and BiotechnologyJeongeup‐siKorea
| | - Motofumi Kumazoe
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of AgricultureKyushu UniversityFukuokaJapan
| | - Su‐Jin Park
- Functional Biomaterial Research CenterKorea Research Institute of Bioscience and BiotechnologyJeongeup‐siKorea
| | - Yoshinori Fujimura
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of AgricultureKyushu UniversityFukuokaJapan
| | - Hirofumi Tachibana
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of AgricultureKyushu UniversityFukuokaJapan
| |
Collapse
|
16
|
Gambaryan S, Mohagaonkar S, Nikolaev VO. Regulation of the renin-angiotensin-aldosterone system by cyclic nucleotides and phosphodiesterases. Front Endocrinol (Lausanne) 2023; 14:1239492. [PMID: 37674612 PMCID: PMC10478253 DOI: 10.3389/fendo.2023.1239492] [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: 06/13/2023] [Accepted: 08/02/2023] [Indexed: 09/08/2023] Open
Abstract
The renin-angiotensin-aldosterone system (RAAS) is one of the key players in the regulation of blood volume and blood pressure. Dysfunction of this system is connected with cardiovascular and renal diseases. Regulation of RAAS is under the control of multiple intracellular mechanisms. Cyclic nucleotides and phosphodiesterases are the major regulators of this system since they control expression and activity of renin and aldosterone. In this review, we summarize known mechanisms by which cyclic nucleotides and phosphodiesterases regulate renin gene expression, secretion of renin granules from juxtaglomerular cells and aldosterone production from zona glomerulosa cells of adrenal gland. We also discuss several open questions which deserve future attention.
Collapse
Affiliation(s)
- Stepan Gambaryan
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Saint Petersburg, Russia
| | - Sanika Mohagaonkar
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Viacheslav O. Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Hamburg, Germany
| |
Collapse
|
17
|
Vielmuth F, Radeva MY, Yeruva S, Sigmund AM, Waschke J. cAMP: A master regulator of cadherin-mediated binding in endothelium, epithelium and myocardium. Acta Physiol (Oxf) 2023; 238:e14006. [PMID: 37243909 DOI: 10.1111/apha.14006] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/05/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023]
Abstract
Regulation of cadherin-mediated cell adhesion is crucial not only for maintaining tissue integrity and barrier function in the endothelium and epithelium but also for electromechanical coupling within the myocardium. Therefore, loss of cadherin-mediated adhesion causes various disorders, including vascular inflammation and desmosome-related diseases such as the autoimmune blistering skin dermatosis pemphigus and arrhythmogenic cardiomyopathy. Mechanisms regulating cadherin-mediated binding contribute to the pathogenesis of diseases and may also be used as therapeutic targets. Over the last 30 years, cyclic adenosine 3',5'-monophosphate (cAMP) has emerged as one of the master regulators of cell adhesion in endothelium and, more recently, also in epithelial cells as well as in cardiomyocytes. A broad spectrum of experimental models from vascular physiology and cell biology applied by different generations of researchers provided evidence that not only cadherins of endothelial adherens junctions (AJ) but also desmosomal contacts in keratinocytes and the cardiomyocyte intercalated discs are central targets in this scenario. The molecular mechanisms involve protein kinase A- and exchange protein directly activated by cAMP-mediated regulation of Rho family GTPases and S665 phosphorylation of the AJ and desmosome adaptor protein plakoglobin. In line with this, phosphodiesterase 4 inhibitors such as apremilast have been proposed as a therapeutic strategy to stabilize cadherin-mediated adhesion in pemphigus and may also be effective to treat other disorders where cadherin-mediated binding is compromised.
Collapse
Affiliation(s)
- Franziska Vielmuth
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Mariya Y Radeva
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Sunil Yeruva
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Anna M Sigmund
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, LMU Munich, Munich, Germany
| |
Collapse
|
18
|
Yue Y, Wang Y, Zhang B, Zeng J, Wang Q, Wang C, Peng S. Whole-Genome Methylation Sequencing of Large Yellow Croaker (Larimichthys crocea) Liver Under Hypoxia and Acidification Stress. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2023; 25:567-579. [PMID: 37450059 DOI: 10.1007/s10126-023-10226-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 06/08/2023] [Indexed: 07/18/2023]
Abstract
Large yellow croaker (Larimichthys crocea) is an important aquaculture species in China. This study analysed whole-genome methylation differences in liver tissues of young fish under different hypoxic and acidification conditions. Differentially methylated regions (DMRs) and differentially methylated genes (DMGs) were identified. Gene ontology (GO) and Kyoto encyclopaedia of genes and genomes (KEGG) enrichment analyses of DMGs were conducted to explore the mechanism of coping with hypoxic acidification. The main methylation type was CG, accounting for > 70% of total methylation, significantly higher than CHG and CHH methylation types. GO enrichment analysis of DMGs revealed strong enrichment of nervous system development, cell periphery, plasma membrane, cell junction organisation, cell junction, signalling receptor activity, molecular sensor activity, cell-linked tissue junction organisation, cell-cell adhesion and nervous system development. KEGG enrichment analysis of DMR-related genes identified cell adhesion molecules, cortisol synthesis and secretion and aldosterone synthesis and secretion as the three key pathways regulating the physiological responses to hypoxia and acidification. Long-term hypoxic and acidification stress affected the immune system, nervous system and stress responses of large yellow croaker. Whole-genome sequencing analysis of exposed tissues was used to investigate changes that occur in L. crocea in response to hypoxic and acidic conditions at the DNA methylation level. The findings contribute to our comprehensive understanding of functional methylation in large yellow croaker and will support future research on the response mechanisms of this species under different environmental pressures.
Collapse
Affiliation(s)
- Yanfeng Yue
- Key Laboratory of Marine and Estuarine Fisheries, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, 200090, People's Republic of China
| | - Yabing Wang
- Key Laboratory of Marine and Estuarine Fisheries, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, 200090, People's Republic of China
| | - Bianbian Zhang
- Key Laboratory of Marine and Estuarine Fisheries, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, 200090, People's Republic of China
| | - Jiao Zeng
- Key Laboratory of Marine and Estuarine Fisheries, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, 200090, People's Republic of China
| | - Qian Wang
- Key Laboratory of Marine and Estuarine Fisheries, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, 200090, People's Republic of China
| | - Cuihua Wang
- Key Laboratory of Marine and Estuarine Fisheries, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, 200090, People's Republic of China.
| | - Shiming Peng
- Key Laboratory of Marine and Estuarine Fisheries, Ministry of Agriculture, East China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Shanghai, 200090, People's Republic of China.
| |
Collapse
|
19
|
Kang DH, Ahn S, Chae JW, Song JS. Differential effects of two phosphodiesterase 4 inhibitors against lipopolysaccharide-induced neuroinflammation in mice. BMC Neurosci 2023; 24:39. [PMID: 37525115 PMCID: PMC10391911 DOI: 10.1186/s12868-023-00810-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 07/06/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND Several phosphodiesterase 4 (PDE4) inhibitors have emerged as potential therapeutics for central nervous system (CNS) diseases. This study investigated the pharmacological effects of two selective PDE4 inhibitors, roflumilast and zatolmilast, against lipopolysaccharide-induced neuroinflammation. RESULTS In BV-2 cells, the PDE4 inhibitor roflumilast reduced the production of nitric oxide and tumor necrosis factor-α (TNF-α) by inhibiting NF-κB phosphorylation. Moreover, mice administered roflumilast had significantly reduced TNF-α, interleukin-1β (IL-1β), and IL-6 levels in plasma and brain tissues. By contrast, zatolmilast, a PDE4D inhibitor, showed no anti-neuroinflammatory effects in vitro or in vivo. Next, in vitro and in vivo pharmacokinetic studies of these compounds in the brain were performed. The apparent permeability coefficients of 3 µM roflumilast and zatolmilast were high (> 23 × 10-6 cm/s) and moderate (3.72-7.18 × 10-6 cm/s), respectively, and increased in a concentration-dependent manner in the MDR1-MDCK monolayer. The efflux ratios were < 1.92, suggesting that these compounds are not P-glycoprotein substrates. Following oral administration, both roflumilast and zatolmilast were slowly absorbed and eliminated, with time-to-peak drug concentrations of 2-2.3 h and terminal half-lives of 7-20 h. Assessment of their brain dispositions revealed the unbound brain-to-plasma partition coefficients of roflumilast and zatolmilast to be 0.17 and 0.18, respectively. CONCLUSIONS These findings suggest that roflumilast, but not zatolmilast, has the potential for use as a therapeutic agent against neuroinflammatory diseases.
Collapse
Affiliation(s)
- Dong Ho Kang
- Data Convergence Drug Research Center, Therapeutics & Biotechnology Division, Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Korea
- College of Pharmacy, Chungnam National University, Daejeon, Korea
| | - Sunjoo Ahn
- Data Convergence Drug Research Center, Therapeutics & Biotechnology Division, Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Korea
| | - Jung Woo Chae
- College of Pharmacy, Chungnam National University, Daejeon, Korea
| | - Jin Sook Song
- Data Convergence Drug Research Center, Therapeutics & Biotechnology Division, Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Korea.
| |
Collapse
|
20
|
Yousaf H, Rehmat S, Jameel M, Ibrahim R, Hashmi SN, Makhdoom EUH, Iwaszkiewicz J, Saadi SM, Tariq M, Baig SM, Toft M, Fatima A, Iqbal Z. A homozygous founder variant in PDE2A causes paroxysmal dyskinesia with intellectual disability. Clin Genet 2023. [PMID: 37317634 DOI: 10.1111/cge.14386] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/22/2023] [Accepted: 05/28/2023] [Indexed: 06/16/2023]
Abstract
Intellectual developmental disorder with paroxysmal dyskinesia or seizures (IDDPADS, OMIM#619150) is an ultra-rare childhood-onset autosomal recessive movement disorder manifesting paroxysmal dyskinesia, global developmental delay, impaired cognition, progressive psychomotor deterioration and/or drug-refractory seizures. We investigated three consanguineous Pakistani families with six affected individuals presenting overlapping phenotypes partially consistent with the reported characteristics of IDDPADS. Whole exome sequencing identified a novel missense variant in Phosphodiesterase 2A (PDE2A): NM_002599.4: c.1514T > C p.(Phe505Ser) that segregated with the disease status of individuals in these families. Retrospectively, we performed haplotype analysis that revealed a 3.16 Mb shared haplotype at 11q13.4 among three families suggesting a founder effect in this region. Moreover, we also observed abnormal mitochondrial morphology in patient fibroblasts compared to controls. Belonging to diverse age groups (13 years-60 years), patients presented paroxysmal dyskinesia, developmental delay, cognitive abnormalities, speech impairment, and drug-refractory seizures with variable onset of disease (as early as 3 months of age to 7 years). Together with the previous reports, we observed that intellectual disability, progressive psychomotor deterioration, and drug-refractory seizures are consistent outcomes of the disease. However, permanent choreodystonia showed variability. We also noticed that the later onset of paroxysmal dyskinesia manifests severe attacks in terms of duration. Being the first report from Pakistan, we add to the clinical and mutation spectrum of PDE2A-related recessive disease raising the total number of patients from six to 12 and variants from five to six. Together, with our findings, the role of PDE2A is strengthened in critical physio-neurological processes.
Collapse
Affiliation(s)
- Hammad Yousaf
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| | - Shagufta Rehmat
- Center for Regenerative Medicine and Stem Cell Research (CRM), The Aga Khan University, Karachi, Pakistan
| | - Muhammad Jameel
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
- Center for Regenerative Medicine and Stem Cell Research (CRM), The Aga Khan University, Karachi, Pakistan
| | - Rabab Ibrahim
- Department of Biological and Biomedical Sciences, The Aga Khan University, Karachi, Pakistan
| | - Sohana Nadeem Hashmi
- Department of Biological and Biomedical Sciences, The Aga Khan University, Karachi, Pakistan
| | - Ehtisham Ul Haq Makhdoom
- Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Justyna Iwaszkiewicz
- Molecular Modeling Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Saadia Maryam Saadi
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| | - Muhammad Tariq
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
| | - Shahid M Baig
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Pakistan
- Department of Biological and Biomedical Sciences, The Aga Khan University, Karachi, Pakistan
| | - Mathias Toft
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Ambrin Fatima
- Department of Biological and Biomedical Sciences, The Aga Khan University, Karachi, Pakistan
| | - Zafar Iqbal
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
21
|
Yamashita M, Kumazoe M, Onda H, Hiroi S, Shimada Y, Fujimura Y, Tachibana H. PPAR/PDK4 pathway is involved in the anticancer effects of cGMP in pancreatic cancer. Biochem Biophys Res Commun 2023; 672:154-160. [PMID: 37354608 DOI: 10.1016/j.bbrc.2023.06.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 06/14/2023] [Indexed: 06/26/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a type of cancer with a high mortality rate. Current treatments for PDACs often have side effects, and drug resistance in cancer stem cells (CSCs) would be also a problem. Cyclic guanosine monophosphate (cGMP) suppresses the mitochondrial function of PDACs and inhibits their CSC properties. Metabolic regulation plays a crucial role in the maintenance of CSC phenotype, and we hypothesized that cGMP induction suppresses cancer stem cell properties in the cancer cell through energy-related signaling pathways. We demonstrated that induction of cGMP upregulated the PPARα/PDK4 pathway and suppressed CSC properties in PDAC, and patients with pancreatic cancer with high PDK4 gene expression had a better prognosis than those with low gene expression. Therefore, these mechanisms may provide new therapeutic targets for the eradication of pancreatic CSCs.
Collapse
Affiliation(s)
- Mai Yamashita
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | - Motofumi Kumazoe
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | - Hiroaki Onda
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | - Shun Hiroi
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | - Yu Shimada
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | - Yoshinori Fujimura
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan
| | - Hirofumi Tachibana
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, 819-0395, Japan.
| |
Collapse
|
22
|
Xu Y, Qiu Y, Zhang Y, Li X. A cAMP phosphodiesterase is essential for sclerotia formation and virulence in Sclerotinia sclerotiorum. FRONTIERS IN PLANT SCIENCE 2023; 14:1175552. [PMID: 37324679 PMCID: PMC10264682 DOI: 10.3389/fpls.2023.1175552] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/12/2023] [Indexed: 06/17/2023]
Abstract
Sclerotinia sclerotiorum is a plant pathogenic fungus that causes white mold or stem rot diseases. It affects mostly dicotyledonous crops, resulting in significant economic losses worldwide. Sclerotia formation is a special feature of S. sclerotiorum, allowing its survival in soil for extended periods and facilitates the spread of the pathogen. However, the detailed molecular mechanisms of how sclerotia are formed and how virulence is achieved in S. sclerotiorum are not fully understood. Here, we report the identification of a mutant that cannot form sclerotia using a forward genetics approach. Next-generation sequencing of the mutant's whole genome revealed candidate genes. Through knockout experiments, the causal gene was found to encode a cAMP phosphodiesterase (SsPDE2). From mutant phenotypic examinations, we found that SsPDE2 plays essential roles not only in sclerotia formation, but also in the regulation of oxalic acid accumulation, infection cushion functionality and virulence. Downregulation of SsSMK1 transcripts in Sspde2 mutants revealed that these morphological defects are likely caused by cAMP-dependent inhibition of MAPK signaling. Moreover, when we introduced HIGS construct targeting SsPDE2 in Nicotiana benthamiana, largely compromised virulence was observed against S. sclerotiorum. Taken together, SsPDE2 is indispensable for key biological processes of S. sclerotiorum and can potentially serve as a HIGS target to control stem rot in the field.
Collapse
Affiliation(s)
- Yan Xu
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Botany, University of British Columbia, Vancouver, BC, Canada
| | - Yilan Qiu
- Department of Life Science, Hunan Normal University, Changsha, China
| | - Yuelin Zhang
- Department of Botany, University of British Columbia, Vancouver, BC, Canada
| | - Xin Li
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
- Department of Botany, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
23
|
Paolocci E, Zaccolo M. Compartmentalised cAMP signalling in the primary cilium. Front Physiol 2023; 14:1187134. [PMID: 37256063 PMCID: PMC10226274 DOI: 10.3389/fphys.2023.1187134] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/25/2023] [Indexed: 06/01/2023] Open
Abstract
cAMP is a universal second messenger that relies on precise spatio-temporal regulation to control varied, and often opposing, cellular functions. This is achieved via selective activation of effectors embedded in multiprotein complexes, or signalosomes, that reside at distinct subcellular locations. cAMP is also one of many pathways known to operate within the primary cilium. Dysfunction of ciliary signaling leads to a class of diseases known as ciliopathies. In Autosomal Dominant Polycystic Kidney Disease (ADPKD), a ciliopathy characterized by the formation of fluid-filled kidney cysts, upregulation of cAMP signaling is known to drive cystogenesis. For decades it has been debated whether the primary cilium is an independent cAMP sub-compartment, or whether it shares a diffusible pool of cAMP with the cell body. Recent studies now suggest it is a specific pool of cAMP generated in the cilium that propels cyst formation in ADPKD, supporting the notion that this antenna-like organelle is a compartment within which cAMP signaling occurs independently from cAMP signaling in the bulk cytosol. Here we present examples of cAMP function in the cilium which suggest this mysterious organelle is home to more than one cAMP signalosome. We review evidence that ciliary membrane localization of G-Protein Coupled Receptors (GPCRs) determines their downstream function and discuss how optogenetic tools have contributed to establish that cAMP generated in the primary cilium can drive cystogenesis.
Collapse
|
24
|
Erofeeva N, Meshalkina D, Firsov M. Multiple Roles of cAMP in Vertebrate Retina. Cells 2023; 12:cells12081157. [PMID: 37190066 DOI: 10.3390/cells12081157] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/09/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
cAMP is a key regulatory molecule that controls many important processes in the retina, including phototransduction, cell development and death, growth of neural processes, intercellular contacts, retinomotor effects, and so forth. The total content of cAMP changes in the retina in a circadian manner following the natural light cycle, but it also shows local and even divergent changes in faster time scales in response to local and transient changes in the light environment. Changes in cAMP might also manifest or cause various pathological processes in virtually all cellular components of the retina. Here we review the current state of knowledge and understanding of the regulatory mechanisms by which cAMP influences the physiological processes that occur in various retinal cells.
Collapse
Affiliation(s)
- Natalia Erofeeva
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 St. Petersburg, Russia
| | - Darya Meshalkina
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 St. Petersburg, Russia
| | - Michael Firsov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 194223 St. Petersburg, Russia
| |
Collapse
|
25
|
Shekarian M, Salehi I, Raoufi S, Asadbegi M, Kourosh-Arami M, Komaki A. Neuroprotective effects of vinpocetine, as a phosphodiesterase 1 inhibitor, on long-term potentiation in a rat model of Alzheimer's disease. BMC Neurosci 2023; 24:20. [PMID: 36927298 PMCID: PMC10018848 DOI: 10.1186/s12868-023-00790-8] [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: 12/13/2022] [Accepted: 03/08/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND Vinpocetine (Vin) is known as a phosphodiesterase 1 inhibitor (PDE1-I) drug with multilateral effects, including antioxidant and anti-inflammatory activity. In this research, we investigated the neuroprotective and therapeutic effects of Vin through hippocampal synaptic plasticity on a rat's model of Alzheimer's disease (AD) induced by an intracerebroventricular (ICV) injection of beta-amyloid (Aβ). METHODS Sixty adult male Wistar rats were randomly divided into six groups: 1. control, 2. sham, 3. Aβ, 4. pretreatment (Vin + Aβ): Vin (4 mg/kg, gavage) for 30 days and then, inducing an AD model by an ICV injection of Aβ(1-42), 5. treatment (Aβ + Vin): inducing an AD model and then receiving Vin for 30 days by gavage, and 7. pretreatment + treatment (Vin + Aβ + Vin): receiving Vin by gavage for 30 days before and 30 days after the induction of an AD model. After these procedures, via stereotaxic surgery, the stimulating electrodes were placed at the perforant pathway (PP) and the recording electrodes were implanted in the dentate gyrus. RESULTS Excitatory postsynaptic potential (EPSP) slope and population spike (PS) amplitude in the Aβ group meaningfully diminished compared to the control group after the induction of long-term potentiation (LTP). CONCLUSIONS Vin could significantly prevent the Aβ effects on LTP. It can be concluded that pretreatment and treatment with Vin can be neuroprotective against harmful consequences of Aβ on hippocampal synaptic plasticity.
Collapse
Affiliation(s)
- Meysam Shekarian
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/518, Iran
| | - Iraj Salehi
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/518, Iran
| | - Safoura Raoufi
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/518, Iran
| | - Masoumeh Asadbegi
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/518, Iran
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Masoumeh Kourosh-Arami
- Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Komaki
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/518, Iran.
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
26
|
Ma Y, Zhang F, Zhong Y, Huang Y, Yixizhuoma, Jia Q, Zhang S. A label-free LC/MS-based enzymatic activity assay for the detection of PDE5A inhibitors. Front Chem 2023; 11:1097027. [PMID: 36860644 PMCID: PMC9968969 DOI: 10.3389/fchem.2023.1097027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 01/30/2023] [Indexed: 02/15/2023] Open
Abstract
Phosphodiesterase type 5 (PDE5), a cyclic nucleotide phosphodiesterase, controls the duration of the cyclic guanosine monophosphate (cGMP) signal by hydrolyzing cGMP to GMP. Inhibiting the activity of PDE5A has proven to be an effective strategy for treating pulmonary arterial hypertension and erectile dysfunction. Current enzymatic activity assay methods for PDE5A mainly use fluorescent or isotope-labeled substrates, which are expensive and inconvenient. Here, we developed an LC/MS-based enzymatic activity assay for PDE5A without labeling, which detects the enzymatic activity of PDE5A by quantifying the substrate cGMP and product GMP at a concentration of 100 nM. The accuracy of this method was verified by a fluorescently labeled substrate. Moreover, a new inhibitor of PDE5A was identified by this method and virtual screening. It inhibited PDE5A with an IC50 value of 870 nM. Overall, the proposed strategy provides a new method for screening PDE5A inhibitors.
Collapse
Affiliation(s)
- Yufeng Ma
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China,Department of Pharmacy, Medical College of Qinghai University, Xining, China
| | - Fengsen Zhang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China,Department of Pharmacy, Medical College of Qinghai University, Xining, China
| | - Yijing Zhong
- Department of Pharmacy, Medical College of Qinghai University, Xining, China
| | - Yongchun Huang
- Department of Pharmacy, Medical College of Qinghai University, Xining, China
| | - Yixizhuoma
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China
| | - Qiangqiang Jia
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China,*Correspondence: Qiangqiang Jia, ; Shoude Zhang,
| | - Shoude Zhang
- State Key Laboratory of Plateau Ecology and Agriculture, Qinghai University, Xining, China,Department of Pharmacy, Medical College of Qinghai University, Xining, China,*Correspondence: Qiangqiang Jia, ; Shoude Zhang,
| |
Collapse
|
27
|
James AS, Eteng OE, Dosumu OA, Moses CA, Ogbonna CU, Adeleye OA, Ugwor EI, Omilo BC, Fabunmi RF, Olakitan AM, Ugbaja RN. Morin Augmented Myocardial eNOS/cGMP/PKG Signaling Pathway and Abated Oxidative and Inflammo-apoptotic Responses in Diethyl Phthalate and Bisphenol-S Co-Exposed Male Albino Rats. Inflammation 2023; 46:175-189. [PMID: 35900689 DOI: 10.1007/s10753-022-01720-2] [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: 04/19/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/27/2022]
Abstract
Cardiac failure accounts for many deaths worldwide. Increasing experimental evidence suggests that exposure to chemicals such as bisphenol-S (BPS) and diethyl phthalate (DEP) exacerbate cardiac injuries. Morin is a flavonoid with reported cardioprotective activity. This study evaluated the modulation of pathways relevant to cardiac endothelial function in rats exposed to BPS and DEP mixture (Mix). Thirty male albino rats were distributed across five groups (n = 6): control received dimethyl sulfoxide (DMSO) as vehicle, Mix dissolved in DMSO, Mix + morin (25 mg/kg), Mix + morin (50 mg/kg), and morin (50 mg/kg). After 21 days of oral exposure at 1 ml/kg bodyweight of the Mix and treatment with morin, the animals were sacrificed, and their hearts were excised for biochemical, histological, immunohistochemical, and gene expression analyses. Exposure to the Mix caused a significant increase in oxidative stress indices (H2O2, malondialdehyde, DNA fragmentation, and advanced oxidation protein products). Also, arginase, phosphodiesterase 5', and the relative expression of TNF-α, interleukin-1β, Bax, androgen receptor, and vascular endothelial growth factor were markedly increased. In contrast, nitric oxide, reduced glutathione, interleukin-10 levels, superoxide dismutase, catalase, and glutathione peroxidase activities decreased significantly. Furthermore, p-NF-kB-p65 expression increased markedly in the Mix-exposed group. Morin treatment significantly reversed these perturbations in a dose-dependent manner in most instances. This study concludes that morin might offer a cardioprotective effect by enhancing the cardiac endothelial system and attenuating oxidative stress, inflammation, and apoptosis elicited by BPS and DEP co-exposure in male Wistar rats.
Collapse
Affiliation(s)
- Adewale Segun James
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Ogun State, Abeokuta, 2240, PMB, Nigeria.
- Department of Chemical Sciences (Biochemistry Program), Faculty of Science, Augustine University, Ilara-Epe, PMB 1010, Lagos State, Nigeria.
| | - Ofem Effiom Eteng
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Ogun State, Abeokuta, 2240, PMB, Nigeria
| | - Oluwatosin Adebisi Dosumu
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Ogun State, Abeokuta, 2240, PMB, Nigeria
| | - Ceasar Antiya Moses
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Ogun State, Abeokuta, 2240, PMB, Nigeria
| | - Chukwuka Uzoamaka Ogbonna
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Ogun State, Abeokuta, 2240, PMB, Nigeria
| | - Oladokun Abdulwasiu Adeleye
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Ogun State, Abeokuta, 2240, PMB, Nigeria
| | - Emmanuel Ifeanyichukwu Ugwor
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Ogun State, Abeokuta, 2240, PMB, Nigeria
| | - Blessing Chukwueku Omilo
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Ogun State, Abeokuta, 2240, PMB, Nigeria
| | - Risikat Funmilayo Fabunmi
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Ogun State, Abeokuta, 2240, PMB, Nigeria
| | - Aduragbemi Moses Olakitan
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Ogun State, Abeokuta, 2240, PMB, Nigeria
| | - Regina Ngozi Ugbaja
- Department of Biochemistry, College of Biosciences, Federal University of Agriculture, Ogun State, Abeokuta, 2240, PMB, Nigeria
| |
Collapse
|
28
|
Structural Characterization of Murine Phosphodiesterase 5 Isoforms and Involvement of Cysteine Residues in Supramolecular Assembly. Int J Mol Sci 2023; 24:ijms24021108. [PMID: 36674621 PMCID: PMC9862819 DOI: 10.3390/ijms24021108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/22/2022] [Accepted: 12/24/2022] [Indexed: 01/09/2023] Open
Abstract
Phosphodiesterases (PDEs) are a superfamily of evolutionarily conserved cyclic nucleotide (cAMP/cGMP)-hydrolyzing enzymes, components of transduction pathways regulating crucial aspects of cell life. Within this family, the cGMP-dependent PDE5 is the major hydrolyzing enzyme in many mammalian tissues, where it regulates a number of cellular and tissular processes. Using Kluyveromyces lactis as a model organism, the murine PDE5A1, A2 and A3 isoforms were successfully expressed and studied, evidencing, for the first time, a distinct role of each isoform in the control, modulation and maintenance of the cellular redox metabolism. Moreover, we demonstrated that the short N-terminal peptide is responsible for the tetrameric assembly of MmPDE5A1 and for the mitochondrial localization of MmPDE5A2. We also analyzed MmPDE5A1, A2 and A3 using small-angle X-ray scattering (SAXS), transmission electron microscopy (TEM), structural mass spectrometry (MS) and polyacrylamide gel electrophoresis in their native conditions (native-PAGE) and in the presence of redox agents. These analyses pointed towards the role of a few specific cysteines in the isoforms' oligomeric assembly and the loss of enzymatic activity when modified.
Collapse
|
29
|
Qiu ZE, Chen L, Hou XC, Sheng J, Xu JB, Xu JW, Gao DD, Huang ZX, Lei TL, Huang ZY, Peng L, Yang HL, Lin QH, Zhu YX, Guan WJ, Lun ZR, Zhou WL, Zhang YL. Toxoplasma gondii infection triggers ongoing inflammation mediated by increased intracellular Cl - concentration in airway epithelium. J Infect 2023; 86:47-59. [PMID: 36334726 DOI: 10.1016/j.jinf.2022.10.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 10/03/2022] [Accepted: 10/28/2022] [Indexed: 12/12/2022]
Abstract
Toxoplasma gondii is a widespread parasitic protozoan causing toxoplasmosis including pulmonary toxoplasmosis. As the first line of host defense, airway epithelial cells play critical roles in orchestrating pulmonary innate immunity. However, the mechanism underlying the airway inflammation induced by the T. gondii infection remains largely unclear. This study demonstrated that after infection with T. gondii, the major anion channel located in the apical membranes of airway epithelial cells, cystic fibrosis transmembrane conductance regulator (CFTR), was degraded by the parasite-secreted cysteine proteases. The intracellular Cl- concentration ([Cl-]i) was consequently elevated, leading to activation of nuclear factor-κB (NF-κB) signaling via serum/glucocorticoid regulated kinase 1. Furthermore, the heightened [Cl-]i and activated NF-κB signaling could be sustained in a positive feedback regulatory manner resulting from decreased intracellular cAMP level through NF-κB-mediated up-regulation of phosphodiesterase 4. Conversely, the sulfur-containing compound allicin conferred anti-inflammatory effects on pulmonary toxoplasmosis by decreasing [Cl-]i via activation of CFTR. These results suggest that the intracellular Cl- dynamically modulated by T. gondii mediates sustained airway inflammation, which provides a potential therapeutic target against pulmonary toxoplasmosis.
Collapse
Affiliation(s)
- Zhuo-Er Qiu
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Lei Chen
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Xiao-Chun Hou
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Jie Sheng
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Jian-Bang Xu
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China; State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, P. R. China
| | - Jia-Wen Xu
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Dong-Dong Gao
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China; Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou 510500, P. R. China
| | - Ze-Xin Huang
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Tian-Lun Lei
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Zi-Yang Huang
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Lei Peng
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Hai-Long Yang
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Qin-Hua Lin
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Yun-Xin Zhu
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Wei-Jie Guan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, P. R. China
| | - Zhao-Rong Lun
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Wen-Liang Zhou
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China.
| | - Yi-Lin Zhang
- School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China.
| |
Collapse
|
30
|
Abdelrahman RS, Nashar EME, Alghamdi MA, Al-Khater KM, Taha RI. Phosphodiesterase1 inhibitor "Vinpocetine" ameliorates the inflammation, apoptosis and oxidative stress induced by cyclophosphamide in urinary bladder: an experimental study. Int Urol Nephrol 2023; 55:129-139. [PMID: 35817991 DOI: 10.1007/s11255-022-03246-w] [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: 02/16/2022] [Accepted: 06/08/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND Hemorrhagic cystitis often develops in patients treated with cyclophosphamide (CP). Vincamine (vinca alkaloid) is the source of the synthetic derivative vinpocetine (Vinpo). Worldwide, Vinpo is used as a cerebroprotective drug. As it has anti-oxidant, anti-thrombotic and anti-inflammatory effects but the power of Vinpo to prevent CP induced cystitis has not been studied. AIM OF STUDY This research was planned to explore the effect of Vinpo (10-30 mg/kg, orally) administered 1 or 4 h before inducing cystitis by CP injection (300 mg/kg, i.p.) on the urinary bladder of mice. RESULTS Administration of Vinpo 30 mg/kg, 4 h before CP injection ameliorated inflammatory markers. It reduced inducible nitric oxide synthase (iNOS), tumor necrosis factor- α (TNF-α), and BCL2 Associated X (Bax) expression in the bladder and increased the total antioxidant capacity level. Histological examination of the bladder has further supported these results. The present study suggests a protective effect of Vinpo (30 mg/kg, 4 h before CP injection) against CP-induced bladder inflammation. CONCLUSION This proposes that Vinpo 30 mg/kg may become a promising pharmacological drug to prevent urinary adverse effects in patients treated with chemotherapy using CP.
Collapse
Affiliation(s)
- Rehab Sabri Abdelrahman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, Al-Madina Al-Munawwarah, Riyadh, 30001, Saudi Arabia
| | - Eman Mohamad El Nashar
- Department of Anatomy, College of Medicine, King Khalid University, King Khalid University Post Office Box: 960, Abha, Postal Code: 61421, Saudi Arabia.
- Department of Histology and Cell Biology College of Medicine, Benha University, Benha, Egypt.
| | - Mansour Abdullah Alghamdi
- Department of Anatomy, College of Medicine, King Khalid University, Abha, 61421, Saudi Arabia
- Genomics and Personalized Medicine Unit, College of Medicine, King Khalid University, Abha, 61421, Saudi Arabia
| | - Khulood Mohammed Al-Khater
- Department of Anatomy, College of Medicine, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia
| | - Reham Ismail Taha
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
31
|
Olawale F, Olofinsan K, Ogunyemi OM, Karigidi KO, Gyebi GA, Ibrahim IM, Iwaloye O. Deciphering the therapeutic role of Kigelia africana fruit in erectile dysfunction through metabolite profiling and molecular modelling. INFORMATICS IN MEDICINE UNLOCKED 2023. [DOI: 10.1016/j.imu.2023.101190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
|
32
|
Low Expression of Phosphodiesterase 2 (PDE2A) Promotes the Progression by Regulating Mitochondrial Morphology and ATP Content and Predicts Poor Prognosis in Hepatocellular Carcinoma. Cells 2022; 12:cells12010068. [PMID: 36611861 PMCID: PMC9818237 DOI: 10.3390/cells12010068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/18/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022] Open
Abstract
Phosphodiesterase 2 (PDE2A) modulates the levels of cAMP/cGMP and was recently found to be involved in mitochondria function regulation, closely related to multiple types of tumor progression. This study aimed to estimate the prognostic significance and biological effects of PDE2A on hepatocellular carcinoma (HCC). We comprehensively analyzed the PDE2A mRNA expression in HCC based on The Cancer Genome Atlas (TCGA) database and investigated the effects of PDE2A on the proliferation and metastatic capacity of HCC cells. PDE2A was downregulated in 25 cancer types, including HCC. Lower PDE2A expression was a protective factor in HCC and was negatively associated with serum AFP levels, tumor status, vascular invasion, histologic grade, and pathologic stage of HCC. Moreover, tumors with low PDE2A expression displayed a decreased immune function. Then, the ROC curve was used to assess the diagnostic ability of PDE2A in HCC (AUC = 0.823 in TCGA and AUC = 0.901 in GSE76427). Patients with low PDE2A expression exhibited worse outcomes compared with those with high PDE2A expression. Additionally, GO functional annotations demonstrated the involvement of PDE2A in the ECM organization, systems development, and ERK-related pathways, indicating that PDE2A might regulate HCC growth and metastasis. The in vitro experiments confirmed that overexpression of PDE2A inhibited proliferation, colony formation, migration, and invasion in two HCC cell lines (HLF and SNU-368), while inhibition of PDE2A has the opposite results. The mechanism of PDE2A's effect on HCC cells is attributed to the change of mitochondrial morphology and ATP content. These data demonstrated that PDE2A closely participated in the regulation of HCC proliferation and metastasis and can be used as a predictive marker candidate and a potential therapeutic target for HCC.
Collapse
|
33
|
Bae J, Lee K, Park JS, Jung J, Tachibana H, Fujimura Y, Kumazoe M, Lim JS, Cho YC, Lee SJ, Park SJ. Phosphodiesterase 5 Inhibitor Potentiates Epigallocatechin 3-O-Gallate-Induced Apoptotic Cell Death via Activation of the cGMP Signaling Pathway in Caco-2 Cells. Curr Issues Mol Biol 2022; 44:6247-6256. [PMID: 36547087 PMCID: PMC9777077 DOI: 10.3390/cimb44120426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 11/30/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022] Open
Abstract
Epigallocatechin 3-O-gallate (EGCG) is a predominant component in green tea with various health benefits. The 67 kDa laminin receptor (67LR) is a nonintegrin cell surface receptor that is overexpressed in various types of cancer; 67LR was identified a cell surface EGCG target that plays a pivotal role in tumor growth, metastasis, and resistance to chemotherapy. However, the plasma concentration of EGCG is limited, and its molecular mechanisms remain unelucidated in colon cancer. In this study, we found that the phosphodiesterase 5 (PDE5) inhibitor, vardenafil (VDN), potentiates EGCG-induced apoptotic cell death in colon cancer cells. The combination of EGCG and VDN induced apoptosis via activation of the endothelial nitric oxide synthase/cyclic guanosine monophosphate/protein kinase Cδ signaling pathway. In conclusion, the PDE5 inhibitor, VDN, may reduce the intracellular PDE5 enzyme activity that potentiates EGCG-induced apoptotic cell death in Caco-2 cells. These results suggest that PDE5 inhibitors can be used to elevate cGMP levels to induce 67LR-mediated, cancer-specific cell death. Therefore, EGCG may be employed as a therapeutic candidate for colon cancer.
Collapse
Affiliation(s)
- Jaehoon Bae
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-gil, Jeongeup-si 56212, Jeonbuk, Republic of Korea
| | - Kwanwoo Lee
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka 812-8581, Japan
| | - Ji-Sun Park
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-gil, Jeongeup-si 56212, Jeonbuk, Republic of Korea
| | - Jinseok Jung
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-gil, Jeongeup-si 56212, Jeonbuk, Republic of Korea
| | - Hirofumi Tachibana
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka 812-8581, Japan
| | - Yoshinori Fujimura
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka 812-8581, Japan
| | - Motofumi Kumazoe
- Division of Applied Biological Chemistry, Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka 812-8581, Japan
| | - Jae Sung Lim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea
| | - Young-Chang Cho
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea
| | - Seung-Jae Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-gil, Jeongeup-si 56212, Jeonbuk, Republic of Korea
- Correspondence: (S.-J.L.); (S.-J.P.); Tel.: +82-(63)-570-5267 (S.-J.L.); +82-(63)-570-5240 (S.-J.P.)
| | - Su-Jin Park
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-gil, Jeongeup-si 56212, Jeonbuk, Republic of Korea
- Correspondence: (S.-J.L.); (S.-J.P.); Tel.: +82-(63)-570-5267 (S.-J.L.); +82-(63)-570-5240 (S.-J.P.)
| |
Collapse
|
34
|
Aoki I, Shiota M, Tsukada Y, Nakano S, Mori I. cGMP dynamics that underlies thermosensation in temperature-sensing neuron regulates thermotaxis behavior in C. elegans. PLoS One 2022; 17:e0278343. [PMID: 36472979 PMCID: PMC9725164 DOI: 10.1371/journal.pone.0278343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 11/08/2022] [Indexed: 12/12/2022] Open
Abstract
Living organisms including bacteria, plants and animals sense ambient temperature so that they can avoid noxious temperature or adapt to new environmental temperature. A nematode C. elegans can sense innocuous temperature, and navigate themselves towards memorize past cultivation temperature (Tc) of their preference. For this thermotaxis, AFD thermosensory neuron is pivotal, which stereotypically responds to warming by increasing intracellular Ca2+ level in a manner dependent on the remembered past Tc. We aimed to reveal how AFD encodes the information of temperature into neural activities. cGMP synthesis in AFD is crucial for thermosensation in AFD and thermotaxis behavior. Here we characterized the dynamic change of cGMP level in AFD by imaging animals expressing a fluorescence resonance energy transfer (FRET)-based cGMP probe specifically in AFD and found that cGMP dynamically responded to both warming and cooling in a manner dependent on past Tc. Moreover, we characterized mutant animals that lack guanylyl cyclases (GCYs) or phosphodiesterases (PDEs), which synthesize and hydrolyze cGMP, respectively, and uncovered how GCYs and PDEs contribute to cGMP and Ca2+ dynamics in AFD and to thermotaxis behavior.
Collapse
Affiliation(s)
- Ichiro Aoki
- Group of Molecular Neurobiology, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
- Department of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Makoto Shiota
- Department of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Yuki Tsukada
- Group of Molecular Neurobiology, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
- Department of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Shunji Nakano
- Group of Molecular Neurobiology, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
- Department of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| | - Ikue Mori
- Group of Molecular Neurobiology, Neuroscience Institute, Graduate School of Science, Nagoya University, Nagoya, Japan
- Department of Biological Science, Graduate School of Science, Nagoya University, Nagoya, Japan
| |
Collapse
|
35
|
Emerging Potential of the Phosphodiesterase (PDE) Inhibitor Ibudilast for Neurodegenerative Diseases: An Update on Preclinical and Clinical Evidence. Molecules 2022; 27:molecules27238448. [PMID: 36500540 PMCID: PMC9737612 DOI: 10.3390/molecules27238448] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative diseases constitute a broad range of central nervous system disorders, characterized by neuronal degeneration. Alzheimer's disease, Parkinson's disease, amyolotrophic lateral sclerosis (ALS), and progressive forms of multiple sclerosis (MS) are some of the most frequent neurodegenerative diseases. Despite their diversity, these diseases share some common pathophysiological mechanisms: the abnormal aggregation of disease-related misfolded proteins, autophagosome-lysosome pathway dysregulation, impaired ubiquitin-proteasome system, oxidative damage, mitochondrial dysfunction and excessive neuroinflammation. There is still no effective drug that could halt the progression of neurodegenerative diseases, and the current treatments are mainly symptomatic. In this regard, the development of novel multi-target pharmaceutical approaches presents an attractive therapeutic strategy. Ibudilast, an anti-inflammatory drug firstly developed as an asthma treatment, is a cyclic nucleotide phosphodiesterases (PDEs) inhibitor, which mainly acts by increasing the amount of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP), while downregulating the pro-inflammatory factors, such as tumor necrosis factor-α (TNF-α), macrophage migration inhibitory factor (MIF) and Toll-like receptor 4 (TLR-4). The preclinical evidence shows that ibudilast may act neuroprotectively in neurodegenerative diseases, by suppressing neuroinflammation, inhibiting apoptosis, regulating the mitochondrial function and by affecting the ubiquitin-proteasome and autophagosome-lysosome pathways, as well as by attenuating oxidative stress. The clinical trials in ALS and progressive MS also show some promising results. Herein, we aim to provide an update on the emerging preclinical and clinical evidence on the therapeutic potential of ibudilast in these disorders, discuss the potential challenges and suggest the future directions.
Collapse
|
36
|
Attanzio A, Restivo I, Tutone M, Tesoriere L, Allegra M, Livrea MA. Redox Properties, Bioactivity and Health Effects of Indicaxanthin, a Bioavailable Phytochemical from Opuntia ficus indica, L.: A Critical Review of Accumulated Evidence and Perspectives. Antioxidants (Basel) 2022; 11:antiox11122364. [PMID: 36552572 PMCID: PMC9774763 DOI: 10.3390/antiox11122364] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/16/2022] [Accepted: 11/23/2022] [Indexed: 12/05/2022] Open
Abstract
Phytochemicals from plant foods are considered essential to human health. Known for their role in the adaptation of plants to their environment, these compounds can induce adaptive responses in cells, many of which are directed at maintaining the redox tone. Indicaxanthin is a long-known betalain pigment found in the genus Opuntia of cactus pear and highly concentrated in the edible fruits of O. ficus indica, L. whose bioactivity has been overlooked until recently. This review summarizes studies conducted so far in vitro and in vivo, most of which have been performed in our laboratory. The chemical and physicochemical characteristics of Indicaxanthin are reflected in the molecule's reducing properties and antioxidant effects and help explain its ability to interact with membranes, modulate redox-regulated cellular pathways, and possibly bind to protein molecules. Measurement of bioavailability in volunteers has been key to exploring its bioactivity; amounts consistent with dietary intake, or plasma concentration after dietary consumption of cactus pear fruit, have been used in experimental setups mimicking physiological or pathophysiological conditions, in cells and in animals, finally suggesting pharmacological potential and relevance of Indicaxanthin as a nutraceutical. In reporting experimental results, this review also aimed to raise questions and seek insights for further basic research and health promotion applications.
Collapse
|
37
|
Tseng SY, Chang HY, Li YH, Chao TH. Effects of Cilostazol on Angiogenesis in Diabetes through Adiponectin/Adiponectin Receptors/Sirtuin1 Signaling Pathway. Int J Mol Sci 2022; 23:14839. [PMID: 36499166 PMCID: PMC9739574 DOI: 10.3390/ijms232314839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/16/2022] [Accepted: 11/23/2022] [Indexed: 12/02/2022] Open
Abstract
Cilostazol is an antiplatelet agent with vasodilating effects that functions by increasing the intracellular concentration of cyclic adenosine monophosphate. We have previously shown that cilostazol has favorable effects on angiogenesis. However, there is no study to evaluate the effects of cilostazol on adiponectin. We investigated the effects of cilostazol on angiogenesis in diabetes in vitro and in vivo through adiponectin/adiponectin receptors (adipoRs) and the sirtuin 1 (SIRT1)/AMP-activated protein kinase (AMPK) signaling pathway. Human umbilical vein endothelial cells (HUVECs) and human aortic smooth muscle cells (HASMCs) were cocultured under high glucose (HG) conditions. Adiponectin concentrations in the supernatants were significantly increased when HASMCs were treated with cilostazol but not significantly changed when only HUVECs were treated with cilostazol. Cilostazol treatment enhanced the expression of SIRT1 and upregulated the phosphorylation of AMPK in HG-treated HUVECs. By sequential knockdown of adipoRs, SIRT1, and AMPK, our data demonstrated that cilostazol prevented apoptosis and stimulated proliferation, chemotactic motility, and capillary-like tube formation in HG-treated HUVECs through the adipoRs/SIRT1/AMPK signaling pathway. The phosphorylation of downstream signaling molecules, including acetyl-CoA carboxylase (ACC) and endothelial nitric oxide synthase (eNOS), was downregulated when HUVECs were treated with a SIRT1 inhibitor. In streptozotocin-induced diabetic mice, cilostazol treatment could improve blood flow recovery 21-28 days after inducing hindlimb ischemia as well as increase the circulating of CD34+CD45dim cells 14-21 days after operation; moreover, these effects were significantly attenuated by the knockdown of adipoR1 but not adipoR2. The expression of SIRT1 and phosphorylation of AMPK/ACC and Akt/eNOS in ischemic muscles were significantly attenuated by the gene knockdown of adipoRs. Cilostazol improves HG-induced endothelial dysfunction in vascular endothelial cells and enhances angiogenesis in diabetic mice by upregulating the expression of adiponectin/adipoRs and its SIRT1/AMPK downstream signaling pathway.
Collapse
Affiliation(s)
- Shih-Ya Tseng
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
- Department of Biological Science, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| | - Hsien-Yuan Chang
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Yi-Heng Li
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
| | - Ting-Hsing Chao
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan
- Health Management Center, National Cheng Kung University Hospital, Tainan 704, Taiwan
| |
Collapse
|
38
|
Adefegha SA, Oboh G, Adedipe AO. Aqueous extract of
Massularia acuminata
exerts erectogenic effect by modulating critical enzymes and hormones in streptozotocin‐induced erectile dysfunction in rats. Andrologia 2022; 54:e14629. [DOI: 10.1111/and.14629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 09/29/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022] Open
Affiliation(s)
- Stephen Adeniyi Adefegha
- Functional Foods, Nutraceuticals and Phytomedicine Laboratory, Department of Biochemistry Federal University of Technology, Akure (FUTA) Akure Nigeria
| | - Ganiyu Oboh
- Functional Foods, Nutraceuticals and Phytomedicine Laboratory, Department of Biochemistry Federal University of Technology, Akure (FUTA) Akure Nigeria
| | - Abraham Olanrewaju Adedipe
- Functional Foods, Nutraceuticals and Phytomedicine Laboratory, Department of Biochemistry Federal University of Technology, Akure (FUTA) Akure Nigeria
| |
Collapse
|
39
|
Non-Selective PDE4 Inhibition Induces a Rapid and Transient Decrease of Serum Potassium in Mice. BIOLOGY 2022; 11:biology11111582. [PMID: 36358283 PMCID: PMC9687940 DOI: 10.3390/biology11111582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
Abstract
Simple Summary Inhibitors of phosphodiesterase 4 (PDE4), a group of isoenzymes that hydrolyze and inactivate the second messenger cAMP, produce promising therapeutic benefits, including anti-inflammatory and memory-enhancing effects. Here, we report that, unexpectedly, PDE4 inhibitors also reduce serum potassium levels in mice. As both the total potassium content of the body, as well as the distribution of potassium between intra- and extracellular compartments, are critical for normal cellular functions, we further explored this observation. Several structurally distinct PDE4 inhibitors reduce serum potassium levels in mice, suggesting it is a class effect of these drugs. Serum potassium levels decrease within 15 min of drug injection, suggesting that PDE4 inhibition lowers serum potassium levels by promoting a transcellular shift of potassium from the blood into cells. This shift is a characteristically fast process, compared to a loss of total-body potassium via the kidneys or digestive tract (e.g., diarrhea). Indeed, stimulating cAMP synthesis with β-adrenoceptor agonists is known to rapidly shift potassium into cells, and PDE4 inhibitors appear to mimic this process by preventing PDE4-mediated cAMP degradation. Our findings reveal that the various acute physiologic effects of PDE4 inhibitors are paralleled and/or may be affected by reduced serum potassium levels. Abstract The analysis of blood samples from mice treated with the PDE4 inhibitor Roflumilast revealed an unexpected reduction in serum potassium levels, while sodium and chloride levels were unaffected. Treatment with several structurally distinct PAN-PDE4 inhibitors, including Roflumilast, Rolipram, RS25344, and YM976 dose-dependently reduced serum potassium levels, indicating the effect is a class-characteristic property. PDE4 inhibition also induces hypothermia and hypokinesia in mice. However, while general anesthesia abrogates these effects of PDE4 inhibitors, potassium levels decrease to similar extents in both awake as well as in fully anesthetized mice. This suggests that the hypokalemic effects of PDE4 inhibitors occur independently of hypothermia and hypokinesia. PDE4 inhibition reduces serum potassium within 15 min of treatment, consistent with a rapid transcellular shift of potassium. Catecholamines promote the uptake of potassium into the cell via increased cAMP signaling. PDE4 appears to modulate these adrenoceptor-mediated effects, as PDE4 inhibition has no additional effects on serum potassium in the presence of saturating doses of the β-adrenoceptor agonist Isoprenaline or the α2-blocker Yohimbine, and is partially blocked by pre-treatment with the β-blocker Propranolol. Together, these data suggest that PDE4 inhibitors reduce serum potassium levels by modulating the adrenergic regulation of cellular potassium uptake.
Collapse
|
40
|
Sheng J, Zhang S, Wu L, Kumar G, Liao Y, GK P, Fan H. Inhibition of phosphodiesterase: A novel therapeutic target for the treatment of mild cognitive impairment and Alzheimer's disease. Front Aging Neurosci 2022; 14:1019187. [PMID: 36268188 PMCID: PMC9577554 DOI: 10.3389/fnagi.2022.1019187] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and is ranked as the 6th leading cause of death in the US. The prevalence of AD and dementia is steadily increasing and expected cases in USA is 14.8 million by 2050. Neuroinflammation and gradual neurodegeneration occurs in Alzheimer's disease. However, existing medications has limitation to completely abolish, delay, or prevent disease progression. Phosphodiesterases (PDEs) are large family of enzymes to hydrolyze the 3'-phosphodiester links in cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) in signal-transduction pathways for generation of 5'-cyclic nucleotides. It plays vital role to orchestrate several pharmacological activities for proper cell functioning and regulating the levels of cAMP and cGMP. Several evidence has suggested that abnormal cAMP signaling is linked to cognitive problems in neurodegenerative disorders like AD. Therefore, the PDE family has become a widely accepted and multipotential therapeutic target for neurodegenerative diseases. Notably, modulation of cAMP/cGMP by phytonutrients has a huge potential for the management of AD. Natural compounds have been known to inhibit phosphodiesterase by targeting key enzymes of cGMP synthesis pathway, however, the mechanism of action and their therapeutic efficacy has not been explored extensively. Currently, few PDE inhibitors such as Vinpocetine and Nicergoline have been used for treatment of central nervous system (CNS) disorders. Considering the role of flavonoids to inhibit PDE, this review discussed the therapeutic potential of natural compounds with PDE inhibitory activity for the treatment of AD and related dementia.
Collapse
Affiliation(s)
- Jianwen Sheng
- Department of Gastroenterology, The People’s Hospital of Yichun City, Yichun, China
| | - Shanjin Zhang
- Department of Gastroenterology, The People’s Hospital of Yichun City, Yichun, China
| | - Lule Wu
- Department of Gastroenterology, The People’s Hospital of Yichun City, Yichun, China
| | - Gajendra Kumar
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, Hong Kong SAR, China
| | - Yuanhang Liao
- Department of Gastroenterology, The People’s Hospital of Yichun City, Yichun, China
| | - Pratap GK
- Department of Biochemistry, Davangere University, Davangere, India
| | - Huizhen Fan
- Department of Gastroenterology, The People’s Hospital of Yichun City, Yichun, China
| |
Collapse
|
41
|
Scaffold Repurposing Reveals New Nanomolar Phosphodiesterase Type 5 (PDE5) Inhibitors Based on Pyridopyrazinone Scaffold: Investigation of In Vitro and In Silico Properties. Pharmaceutics 2022; 14:pharmaceutics14091954. [PMID: 36145702 PMCID: PMC9501832 DOI: 10.3390/pharmaceutics14091954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/17/2022] Open
Abstract
Inhibition of PDE5 results in elevation of cGMP leading to vascular relaxation and reduction in the systemic blood pressure. Therefore, PDE5 inhibitors are used as antihypertensive and antianginal agents in addition to their major use as male erectile dysfunction treatments. Previously, we developed a novel series of 34 pyridopyrazinone derivatives as anticancer agents (series A–H). Herein, a multi-step in silico approach was preliminary conducted to evaluate the predicted PDE5 inhibitory activity, followed by an in vitro biological evaluation over the enzymatic level and a detailed SAR study. The designed 2D-QSAR model which was carried out to predict the IC50 of the tested compounds revealed series B, D, E and G with nanomolar range of IC50 values (6.00–81.56 nM). A further docking simulation model was performed to investigate the binding modes within the active site of PDE5. Interestingly, most of the tested compounds showed almost the same binding modes of that of reported PDE5 inhibitors. To validate the in silico results, an in vitro enzymatic assay over PDE5 enzyme was performed for a number of the promising candidates with different substitutions. Both series E and G exhibited a potent inhibitory activity (IC50 = 18.13–41.41 nM). Compound 11b (series G, oxadiazole-based derivatives with terminal 4-NO2 substituted phenyl ring and rigid linker) was the most potent analogue with IC50 value of 18.13 nM. Structure–activity relationship (SAR) data attained for various substitutions were rationalized. Furthermore, a molecular dynamic simulation gave insights into the inhibitory activity of the most active compound (11b). Accordingly, this report presents a successful scaffold repurposing approach that reveals compound 11b as a highly potent nanomolar PDE5 inhibitor worthy of further investigation.
Collapse
|
42
|
Lugnier C. The Complexity and Multiplicity of the Specific cAMP Phosphodiesterase Family: PDE4, Open New Adapted Therapeutic Approaches. Int J Mol Sci 2022; 23:10616. [PMID: 36142518 PMCID: PMC9502408 DOI: 10.3390/ijms231810616] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/04/2022] [Accepted: 09/07/2022] [Indexed: 11/19/2022] Open
Abstract
Cyclic nucleotides (cAMP, cGMP) play a major role in normal and pathologic signaling. Beyond receptors, cyclic nucleotide phosphodiesterases; (PDEs) rapidly convert the cyclic nucleotide in its respective 5'-nucleotide to control intracellular cAMP and/or cGMP levels to maintain a normal physiological state. However, in many pathologies, dysregulations of various PDEs (PDE1-PDE11) contribute mainly to organs and tissue failures related to uncontrolled phosphorylation cascade. Among these, PDE4 represents the greatest family, since it is constituted by 4 genes with multiple variants differently distributed at tissue, cellular and subcellular levels, allowing different fine-tuned regulations. Since the 1980s, pharmaceutical companies have developed PDE4 inhibitors (PDE4-I) to overcome cardiovascular diseases. Since, they have encountered many undesired problems, (emesis), they focused their research on other PDEs. Today, increases in the knowledge of complex PDE4 regulations in various tissues and pathologies, and the evolution in drug design, resulted in a renewal of PDE4-I development. The present review describes the recent PDE4-I development targeting cardiovascular diseases, obesity, diabetes, ulcerative colitis, and Crohn's disease, malignancies, fatty liver disease, osteoporosis, depression, as well as COVID-19. Today, the direct therapeutic approach of PDE4 is extended by developing allosteric inhibitors and protein/protein interactions allowing to act on the PDE interactome.
Collapse
Affiliation(s)
- Claire Lugnier
- Section de Structures Biologiques, Pharmacologie et Enzymologie, CNRS/Unistra, CRBS, UR 3072, CEDEX, 67084 Strasbourg, France
| |
Collapse
|
43
|
Ajiboye BO, Oyinloye BE, Ojo OA, Lawal OE, Jokomba YA, Balogun BA, Adeoye AO, Ajuwon OR. Effect of Flavonoid-Rich Extract From Dalbergiella welwitschii Leaf on Redox, Cholinergic, Monoaminergic, and Purinergic Dysfunction in Oxidative Testicular Injury: Ex Vivo and In Silico Studies. Bioinform Biol Insights 2022; 16:11779322221115546. [PMID: 35966809 PMCID: PMC9373118 DOI: 10.1177/11779322221115546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
The antioxidant, cholinergic, monoaminergic, and purinergic activities of flavonoid-rich extract from Dalbergiella welwitschii leaf (FEDW) were investigated on oxidative testicular injury (ex vivo) due to the local report on the use of this plant as anti-testicular injury. Flavonoid extract was obtained from FEDW using a standard procedure. Five male albino rats were used, testes harvested and incubated with FeSO4 for accessing the cholinergic, monoaminergic, and purinergic activities of the FEDW (ex vivo). Testicular tissues incubated with FeSO4 demonstrated a significant decrease in antioxidant biomarkers, arginase, ATPase, ENTPDase, 5'-nucleotidase, and PDE-5 activities, as well as Zn and sialic acid levels with an upsurge in malondialdehyde (MDA), and NO levels, myeloperoxidase, cholinesterases, monoamine oxidase (MAO), and angiotensin-converting enzyme (ACE) activities. Treatment of testicular tissues incubated with FeSO4 via different concentrations of FEDW significantly increased the activities of antioxidant, arginase, ATPase, E-NTPDase, 5'-nucleotidase, phosphodiesterase-5 (PDE-5), as well as Zn and sialic acid levels with a significant decrease in MDA, nitric oxide (NO), myeloperoxidase, cholinesterases, MAO, and ACE levels. Molecular docking revealed the molecular interactions of cyclooxygenase-2 (COX-2) with ellagic acid, piperine, and caffeine with piperine and caffeine obeyed the druggability and pharmacokinetic. These findings point to FEDW as a possible potential for the treatment of oxidative testicular injury.
Collapse
Affiliation(s)
- Basiru Olaitan Ajiboye
- Phytomedicine and Molecular Toxicology Research Laboratory, Department of Biochemistry, Federal University Oye-Ekiti, Oye-Ekiti, Nigeria.,Institute of Drug Research and Development, SE Bogoro Center, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Babatunji Emmanuel Oyinloye
- Institute of Drug Research and Development, SE Bogoro Center, Afe Babalola University, Ado-Ekiti, Nigeria.,Phytomedicine, Biochemical Toxicology and Biotechnology Research Laboratories, Department of Biochemistry, College of Sciences, Afe Babalola University, Ado-Ekiti, Nigeria.,Biotechnology and Structural Biology (BSB) Group, Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa, South Africa
| | - Oluwafemi Adeleke Ojo
- Phytomedicine, Molecular Toxicology, and Computational Biochemistry Research Laboratory (PMTCB-RL), Department of Biochemistry, Bowen University, Iwo, Nigeria
| | - Olaolu Ebenezer Lawal
- Phytomedicine and Molecular Toxicology Research Laboratory, Department of Biochemistry, Federal University Oye-Ekiti, Oye-Ekiti, Nigeria
| | - Yesirat Abimbola Jokomba
- Phytomedicine and Molecular Toxicology Research Laboratory, Department of Biochemistry, Federal University Oye-Ekiti, Oye-Ekiti, Nigeria
| | - Basheer Ajibola Balogun
- Clinical Biochemistry Unit, Department of Biochemistry, University of Ilorin, Ilorin, Nigeria
| | - Akinwunmi Oluwaseun Adeoye
- Biomembrane, Phytomedicine, and Drug Development Unit, Department of Biochemistry, Federal University Oye-Ekiti, Oye-Ekiti, Nigeria
| | - Olawale Rasaq Ajuwon
- Redox Biology Research Unit, Department of Biochemistry, Federal University Oye-Ekiti, Oye-Ekiti, Nigeria
| |
Collapse
|
44
|
Crocetti L, Floresta G, Cilibrizzi A, Giovannoni MP. An Overview of PDE4 Inhibitors in Clinical Trials: 2010 to Early 2022. Molecules 2022; 27:4964. [PMID: 35956914 PMCID: PMC9370432 DOI: 10.3390/molecules27154964] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 11/16/2022] Open
Abstract
Since the early 1980s, phosphodiesterase 4 (PDE4) has been an attractive target for the treatment of inflammation-based diseases. Several scientific advancements, by both academia and pharmaceutical companies, have enabled the identification of many synthetic ligands for this target, along with the acquisition of precise information on biological requirements and linked therapeutic opportunities. The transition from pre-clinical to clinical phase was not easy for the majority of these compounds, mainly due to their significant side effects, and it took almost thirty years for a PDE4 inhibitor to become a drug i.e., Roflumilast, used in the clinics for the treatment of chronic obstructive pulmonary disease. Since then, three additional compounds have reached the market a few years later: Crisaborole for atopic dermatitis, Apremilast for psoriatic arthritis and Ibudilast for Krabbe disease. The aim of this review is to provide an overview of the compounds that have reached clinical trials in the last ten years, with a focus on those most recently developed for respiratory, skin and neurological disorders.
Collapse
Affiliation(s)
- Letizia Crocetti
- NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy
| | - Giuseppe Floresta
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria 6, 95125 Catania, Italy
| | - Agostino Cilibrizzi
- Institute of Pharmaceutical Science, King’s College London, Stamford Street, London SE1 9NH, UK
| | - Maria Paola Giovannoni
- NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Via Ugo Schiff 6, 50019 Sesto Fiorentino, Italy
| |
Collapse
|
45
|
Emerging molecular technologies for light-mediated modulation of pancreatic beta-cell function. Mol Metab 2022; 64:101552. [PMID: 35863638 PMCID: PMC9352964 DOI: 10.1016/j.molmet.2022.101552] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/13/2022] [Accepted: 07/13/2022] [Indexed: 11/22/2022] Open
Abstract
Background Optogenetic modalities as well as optochemical and photopharmacological strategies, collectively termed optical methods, have revolutionized the control of cellular functions via light with great spatiotemporal precision. In comparison to the major advances in the photomodulation of signaling activities noted in neuroscience, similar applications to endocrine cells of the pancreas, particularly insulin-producing β-cells, have been limited. The availability of tools allowing light-mediated changes in the trafficking of ions such as K+ and Ca2+ and signaling intermediates such as cyclic adenosine monophosphate (cAMP), renders β-cells and their glucose-stimulated insulin secretion (GSIS) amenable to optoengineering for drug-free control of blood sugar. Scope of review The molecular circuit of the GSIS in β-cells is described with emphasis on intermediates which are targetable for optical intervention. Various pharmacological agents modifying the release of insulin are reviewed along with their documented side effects. These are contrasted with optical approaches, which have already been employed for engineering β-cell function or are considered for future such applications. Principal obstacles are also discussed as the implementation of optogenetics is pondered for tissue engineering and biology applications of the pancreas. Major Conclusions Notable advances in optogenetic, optochemical and photopharmacological tools are rendering feasible the smart engineering of pancreatic cells and tissues with light-regulated function paving the way for novel solutions for addressing pancreatic pathologies including diabetes.
Collapse
|
46
|
Ameliorative Sexual Behavior and Phosphodiesterase-5 Inhibitory Effects of Spondias mangifera Fruit Extract in Rodents: In Silico, In Vitro, and In Vivo Study. J Clin Med 2022; 11:jcm11133732. [PMID: 35807028 PMCID: PMC9267661 DOI: 10.3390/jcm11133732] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/07/2022] [Accepted: 06/22/2022] [Indexed: 12/14/2022] Open
Abstract
The ethanolic extracts of Spondias mangifera fruit (SMFE) were evaluated for aphrodisiac activity. The in-vitro phosphodiesterase-5 (PDE-5) inhibition was assessed based on in-silico molecular docking and simulation studies. In addition, the in-vivo sexual behavior was analyzed in the form of mount (MF, ML), intromission (IF, IL), and ejaculation (EF, EL) frequencies and latencies to validate the in-vitro results. Some biochemical parameters, including PDE-5, nitric oxide, and testosterone, were also observed. The above extract constituted β-amyrin, β-sitosterol, and oleanolic acid and showed tremendous binding with phosphodiesterase-5 and sildenafil. Both the sildenafil and ethanolic extracts (200 and 400 mg/kg/d bodyweight) significantly (p < 0.1, p < 0.05) increased MF, IF, and EF, respectively. In contrast, ML and IL significantly (p < 0.1) decreased, and EL significantly (p < 0.1) increased compared with a normal group of animals. The ethanolic extracts (200 and 400 mg/kg/d bodyweight) and sildenafil further significantly (p < 0.05, p < 0.1) diminished PDE-5 activity significantly (p < 0.05, p < 0.1) and enhanced nitric oxide and testosterone levels, as compared with normal rodents. Therefore, the S. mangifera ethanolic extract might be a valuable alternate aphrodisiac for erectile dysfunction.
Collapse
|
47
|
Mani A. PDE4DIP in health and diseases. Cell Signal 2022; 94:110322. [PMID: 35346821 PMCID: PMC9618167 DOI: 10.1016/j.cellsig.2022.110322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 11/15/2022]
Abstract
Cyclic-AMP (cAMP), the first second messenger to be identified, is synthesized, and is universally utilized as a second messenger, and plays important roles in integrity, and function of organs, including heart. Through its coupling with other intracellular messengers, cAMP facilitates excitation-contraction coupling, increases heart rate and conduction velocity. It is degraded by a class of enzymes called cAMP-dependent phosphodiesterase (PDE), with PDE3 and PDE4 being the predominant isoforms in the heart. This highly diverse class of enzymes degrade cAMP and through anchoring proteins generates dynamic microdomains to target specific proteins and control specific cell functions in response to various stimuli. The impaired function of the anchoring protein either by inherited genetic mutations or acquired injuries results in altered intracellular targeting, and blunted responsiveness to stimulating pathways and contributes to pathological cardiac remodeling, cardiac arrhythmias and reduced cell survival. Recent genetic studies provide compelling evidence for an association between the variants in the anchoring protein PDE4DIP and atrial fibrillation, stroke, and heart failure.
Collapse
Affiliation(s)
- Arya Mani
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA; Department of Genetics, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
48
|
Bukhari SNA, Alamgeer, Saeed S, Asim MH, Irfan HM, Ejaz H, Elsherif MA, Junaid K. Antihypertensive and Vasorelaxant Effects of Citrus aurantifolia Linn. Fruit: Proposed Mechanisms. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:5871424. [PMID: 35646151 PMCID: PMC9142296 DOI: 10.1155/2022/5871424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 04/23/2022] [Accepted: 05/04/2022] [Indexed: 11/23/2022]
Abstract
Background Citrus aurantifolia Linn. fruit, a natural dietary item, has long been used traditionally to treat hypertension in Pakistan. The current research work aims to explore the effect on blood pressure and its mechanisms. Methods The aqueous methanol extract of plant fruit was used to evaluate hypotensive/antihypertensive, vasorelaxation, and safety profiles. Moreover, the in vitro inhibitory effect of AMECA on phosphodiesterase was also evaluated. Results In hypotensive studies, extracts of Citrus aurantifolia fruit exhibited a concentration-dependent reduction in SBP, DBP, MAP, and heart rate. A similar effect has been observed on anesthetized rats, but the effects exerted by the extract were not altered significantly in the presence of L-NAME, atropine, captopril, and propranolol. Moreover, in coronary arteries, the extract significantly potentiated relaxations induced by cGMP- and cAMP-dependent relaxing agonists. When exposed to PDEs, the extract concentration dependently subdued cGMP-hydrolyzing activity of different PDEs with IC50 values of 40-130 μg/mL. Conclusion It is conceivable that extracts obtained from Citrus aurantifolia fruit produced hypotensive and antihypertensive effects in rats. The extract elicited endothelium-independent vasorelaxation, possibly by acting directly on smooth muscles of the coronary artery and by increasing cGMP and cAMP via nonselective inhibition of vascular PDEs.
Collapse
Affiliation(s)
- Syed Nasir Abbas Bukhari
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Al Jouf 72388, Saudi Arabia
| | - Alamgeer
- Punjab University College of Pharmacy, University of the Punjab, 54000 Lahore, Pakistan
| | - Sumera Saeed
- College of Pharmacy, University of Sargodha, 40100 Sargodha, Pakistan
| | | | | | - Hasan Ejaz
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Sakaka, Jouf University, Al Jouf 72388, Saudi Arabia
| | - Mervat A. Elsherif
- Chemistry Department, College of Science, Jouf University, Al Jouf 72388, Sakaka, Saudi Arabia
| | - Kashaf Junaid
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Sakaka, Jouf University, Al Jouf 72388, Saudi Arabia
| |
Collapse
|
49
|
Donate-Correa J, Sanchez-Niño MD, González-Luis A, Ferri C, Martín-Olivera A, Martín-Núñez E, Fernandez-Fernandez B, Tagua VG, Mora-Fernández C, Ortiz A, Navarro-González JF. Repurposing drugs for highly prevalent diseases: pentoxifylline, an old drug and a new opportunity for diabetic kidney disease. Clin Kidney J 2022; 15:2200-2213. [PMID: 36381364 PMCID: PMC9664582 DOI: 10.1093/ckj/sfac143] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Indexed: 11/30/2022] Open
Abstract
Diabetic kidney disease is one of the most frequent complications in patients with diabetes and constitutes a major cause of end-stage kidney disease. The prevalence of diabetic kidney disease continues to increase as a result of the growing epidemic of diabetes and obesity. Therefore, there is mounting urgency to design and optimize novel strategies and drugs that delay the progression of this pathology and contain this trend. The new approaches should go beyond the current therapy focussed on the control of traditional risk factors such as hyperglycaemia and hypertension. In this scenario, drug repurposing constitutes an economic and feasible approach based on the discovery of useful activities for old drugs. Pentoxifylline is a nonselective phosphodiesterase inhibitor currently indicated for peripheral artery disease. Clinical trials and meta-analyses have shown renoprotection secondary to anti-inflammatory and antifibrotic effects in diabetic patients treated with this old known drug, which makes pentoxifylline a candidate for repurposing in diabetic kidney disease.
Collapse
Affiliation(s)
- Javier Donate-Correa
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- GEENDIAB (Grupo Español para el estudio de la Nefropatía Diabética), Sociedad Española de Nefrología, Santander, Spain
- RICORS2040 (RD21/0005/0013), Instituto de Salud Carlos III, Madrid, Spain
| | - María Dolores Sanchez-Niño
- Departamento de Nefrología e Hipertensión, IIS-Fundación Jiménez Díaz y Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Ainhoa González-Luis
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- Escuela de doctorado, Universidad de La Laguna
| | - Carla Ferri
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- Escuela de doctorado, Universidad de La Laguna
| | - Alberto Martín-Olivera
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- Escuela de doctorado, Universidad de La Laguna
| | - Ernesto Martín-Núñez
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- RICORS2040 (RD21/0005/0013), Instituto de Salud Carlos III, Madrid, Spain
| | - Beatriz Fernandez-Fernandez
- Departamento de Nefrología e Hipertensión, IIS-Fundación Jiménez Díaz y Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- RICORS2040 (RD21/0005/0001), Instituto de Salud Carlos III, Madrid, Spain
| | - Víctor G Tagua
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Carmen Mora-Fernández
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- GEENDIAB (Grupo Español para el estudio de la Nefropatía Diabética), Sociedad Española de Nefrología, Santander, Spain
- RICORS2040 (RD21/0005/0013), Instituto de Salud Carlos III, Madrid, Spain
| | - Alberto Ortiz
- Departamento de Nefrología e Hipertensión, IIS-Fundación Jiménez Díaz y Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- RICORS2040 (RD21/0005/0001), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan F Navarro-González
- Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- GEENDIAB (Grupo Español para el estudio de la Nefropatía Diabética), Sociedad Española de Nefrología, Santander, Spain
- RICORS2040 (RD21/0005/0013), Instituto de Salud Carlos III, Madrid, Spain
- Servicio de Nefrología, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
- Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| |
Collapse
|
50
|
Zhao YY, Zhang LJ, Liang XY, Zhang XC, Chang JR, Shi M, Liu H, Zhou Y, Sun Z, Zhao YF. Pyruvate Upregulates Hepatic FGF21 Expression by Activating PDE and Inhibiting cAMP–Epac–CREB Signaling Pathway. Int J Mol Sci 2022; 23:ijms23105490. [PMID: 35628302 PMCID: PMC9141208 DOI: 10.3390/ijms23105490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 02/01/2023] Open
Abstract
Fibroblast growth factor 21 (FGF21) functions as a polypeptide hormone to regulate glucose and lipid metabolism, and its expression is regulated by cellular metabolic stress. Pyruvate is an important intermediate metabolite that acts as a key hub for cellular fuel metabolism. However, the effect of pyruvate on hepatic FGF21 expression and secretion remains unknown. Herein, we examined the gene expression and protein levels of FGF21 in human hepatoma HepG2 cells and mouse AML12 hepatocytes in vitro, as well as in mice in vivo. In HepG2 and AML12 cells, pyruvate at concentrations above 0.1 mM significantly increased FGF21 expression and secretion. The increase in cellular cAMP levels by adenylyl cyclase activation, phosphodiesterase (PDE) inhibition and 8-Bromo-cAMP administration significantly restrained pyruvate-stimulated FGF21 expression. Pyruvate significantly increased PDE activities, reduced cAMP levels and decreased CREB phosphorylation. The inhibition of exchange protein directed activated by cAMP (Epac) and cAMP response element binding protein (CREB) upregulated FGF21 expression, upon which pyruvate no longer increased FGF21 expression. The increase in plasma pyruvate levels in mice induced by the intraperitoneal injection of pyruvate significantly increased FGF21 gene expression and PDE activity with a reduction in cAMP levels and CREB phosphorylation in the mouse liver compared with the control. In conclusion, pyruvate activates PDEs to reduce cAMP and then inhibits the cAMP–Epac–CREB signaling pathway to upregulate FGF21 expression in hepatocytes.
Collapse
|