1
|
Wu Z, Zhang F, Chen Z, Wang X, Liu X, Yang G, Wang S, Huang S, Luo HB, Huang YY, Wu D. Discovery and optimization of 4-(imidazo[1,2-a]pyrimidin-3-yl)thiazol-2-amine derivatives as novel phosphodiesterase 4 inhibitors. Mol Divers 2025; 29:2283-2291. [PMID: 39313709 DOI: 10.1007/s11030-024-10991-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 09/09/2024] [Indexed: 09/25/2024]
Abstract
Phosphodiesterases (PDEs) are important intracellular enzymes that hydrolyze the second messengers cAMP and/or cGMP. Now several studies have shown that PDE4 received particular attention due to which it represents the most prominent cAMP-metabolizing enzyme involved in many diseases. In this study, we performed prescreening of our internal compound library and discovered the compound (PTC-209) with moderate PDE4 inhibitory activity (IC50 of 4.78 ± 0.08 μM). And a series of 4-(imidazo[1,2-a]pyrimidin-3-yl)thiazol-2-amine derivatives as novel PDE4 inhibitors starting from PTC-209 were successfully designed and synthesized using a structure-based discovery strategy. L19, the most potent inhibitor, exhibited good inhibitory activity (IC50 of 0.48 ± 0.02 μM) and remarkable metabolic stability in rat liver microsomes. Our study presents an example of discovery novel PDE4 inhibitors, which would be helpful for design and optimization of novel inhibitors in future.
Collapse
Affiliation(s)
- Zongmin Wu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China
- School of Life and Health Sciences, Hainan University, Haikou, 570228, China
| | - Furong Zhang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China
- School of Life and Health Sciences, Hainan University, Haikou, 570228, China
| | - Zhexin Chen
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China
| | - Xue Wang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China
| | - Xingfu Liu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China
| | - Guofeng Yang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China
| | - Sen Wang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China
| | - Shuheng Huang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China
| | - Hai-Bin Luo
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China
| | - Yi-You Huang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China.
| | - Deyan Wu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, China.
| |
Collapse
|
2
|
Lin H, Bei Y, Shen Z, Wei T, Ge Y, Yu L, Xu H, He W, Dai Y, Yao D, Dai H. HDAC9 Deficiency Upregulates cGMP-dependent Kinase II to Mitigate Neuronal Apoptosis in Ischemic Stroke. Transl Stroke Res 2025; 16:868-881. [PMID: 38940872 DOI: 10.1007/s12975-024-01272-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/05/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024]
Abstract
Histone deacetylase 9 (HDAC9) is implicated in ischemic stroke by genome-wide association studies. We conducted a series of experiments using a mouse model of ischemic stroke (middle cerebral artery occlusion followed by reperfusion) to examine the potential role of HDAC9. Briefly, HDAC9 was upregulated in the penumbra. Deletion of HDAC9 from neurons reduced infarction volume, inhibited neuronal apoptosis in the penumbra, and improved neurological outcomes. HDAC9 knockout from neurons in the penumbra upregulated cGMP-dependent kinase II (cGK II), blocking which abrogated the protective effects of HDAC9 deletion. Mechanistically, HDAC9 interacts with the transcription factor MEF2, thereby inhibiting MEF2's binding to the promoter region of the cGK II gene, which results in the suppression of cGK II expression. Inhibiting the interaction between HDAC9 and MEF2 by BML210 upregulated cGK II and attenuated ischemic injury in mice. These results encourage targeting the HDAC9-MEF2 interaction in developing novel therapy against ischemic stroke.
Collapse
Affiliation(s)
- Haoran Lin
- Department of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, China
| | - Yun Bei
- Department of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, China
| | - Zexu Shen
- Department of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, China
| | - Taofeng Wei
- Department of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, China
| | - Yuyang Ge
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Lingyan Yu
- Department of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, China
| | - Huimin Xu
- Department of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, China
| | - Wei He
- Department of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, China
| | - Yunjian Dai
- Department of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, China
| | - Difei Yao
- Department of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, China
| | - Haibin Dai
- Department of Pharmacy, the Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, 310009, China.
- Clinical Pharmacy Research Center, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
3
|
Banerjee A, Thekkekkara D, Manjula SN, Nair SP, Lalitha MS. Correlation of autophagy and Alzheimer's disease with special emphasis on the role of phosphodiesterase-4. 3 Biotech 2025; 15:139. [PMID: 40292249 PMCID: PMC12018668 DOI: 10.1007/s13205-025-04306-5] [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: 02/14/2025] [Accepted: 04/02/2025] [Indexed: 04/30/2025] Open
Abstract
Autophagy disruption is important in Alzheimer's disease (AD) as it prevents misfolded proteins from being removed, which leads to the accumulation of amyloid plaques and neurofibrillary tangles (NFTs). Restoring autophagy improves neuronal survival and cognitive function, according to experimental models. In AD models, mTOR inhibition and AMPK activation enhance synaptic plasticity and lessen learning deficits. Inhibitors of phosphodiesterase-4 (PDE4) improve cognition and reduce neuroinflammation via altering cyclic adenosine monophosphate (cAMP) transmission. Furthermore, autophagic-lysosomal clearance is encouraged by upregulating transcription factor EB (TFEB), which lessens the pathogenic damage linked to AD. These results point to autophagy modification as a promising therapeutic approach, with the mTOR, AMPK, cAMP, and TFEB pathways being possible targets for drugs. Though much evidence is based on animal studies, these findings provide valuable insights into autophagy's role in AD pathology, offering promising directions for future research and drug development.
Collapse
Affiliation(s)
- Aniruddha Banerjee
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - Dithu Thekkekkara
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - S. N. Manjula
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - Salini P. Nair
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| | - Mankala Sree Lalitha
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysore, Karnataka 570 015 India
| |
Collapse
|
4
|
Sainimnuan S, Chimprasit A, Hannongbua S, Saparpakorn P. Role of interaction mode of phenanthrene derivatives as selective PDE5 inhibitors using molecular dynamics simulations and quantum chemical calculations. Mol Divers 2025; 29:1683-1696. [PMID: 39080154 DOI: 10.1007/s11030-024-10944-3] [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: 06/05/2024] [Accepted: 07/22/2024] [Indexed: 03/15/2025]
Abstract
Phosphodiesterase type 5 (PDE5) inhibitors play a crucial role in blocking PDE5 to improve erectile dysfunction (ED). However, most PDE5 drugs revealed side effects including the loss of vision due to the PDE6 inhibition. Phenanthrene derivatives isolated from E. macrobulbon were previously reported as PDE5 inhibitors. Two phenanthrene derivatives (cpds 1-2) revealed better inhibition to PDE5 than PDE6 and cpd 1 is more selective to PDE5 than cpd 2. To elucidate why the phenanthrene derivatives could inhibit PDE5 and PDE6, their binding modes were investigated using molecular dynamics simulations and quantum chemical calculations, as compared to the PDE5 drugs. From the results, all four drugs and phenanthrene derivatives revealed similar π-π interactions to Phe820 in PDE5. Additional H-bond interaction to Gln817 in PDE5 resulted in better PDE5 inhibition of vardenafil and tadalafil. Moreover, cpds 1-2 were able to form the H-bond interaction with Asp764 in PDE5. In the case of the PDE6, the loss of π-π interaction to Phe776 and H-bond interaction to Gln773 indicated the important points for losing the PDE6 inhibition. In conclusion, to develop the new potent PDE5 inhibitors, not only the important interaction with PDE5 but also the interaction with PDE6 should be considered. In phenanthrene derivatives, the middle ring was significant to form π-π interactions to Phe820 in PDE5 and hydroxyl substituent was also the key part to form the H-bond interaction with Asp764 in PDE5. Principal component analysis (PCA) and free energy landscape (FEL) analysis indicated the stability of the system. The bioavailability, drug-likeness, and pharmacokinetics of phenanthrene derivatives were also predicted. These derivatives revealed good drug-likeness and GI absorption. The obtained results showed that phenanthrene derivatives could be interesting for the development of PDE5 inhibitors in the future.
Collapse
Affiliation(s)
- Supawadee Sainimnuan
- Department of Chemistry, Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand
- Center for Advanced Studies in Nanotechnology for Chemical, Food and Agricultural Industries, KU Institute for Advanced Studies, Kasetsart University, Bangkok, 10900, Thailand
| | - Aunlika Chimprasit
- Department of Chemistry, Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand
- Center for Advanced Studies in Nanotechnology for Chemical, Food and Agricultural Industries, KU Institute for Advanced Studies, Kasetsart University, Bangkok, 10900, Thailand
| | - Supa Hannongbua
- Department of Chemistry, Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand
- Center for Advanced Studies in Nanotechnology for Chemical, Food and Agricultural Industries, KU Institute for Advanced Studies, Kasetsart University, Bangkok, 10900, Thailand
| | - Patchreenart Saparpakorn
- Department of Chemistry, Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand.
- Center for Advanced Studies in Nanotechnology for Chemical, Food and Agricultural Industries, KU Institute for Advanced Studies, Kasetsart University, Bangkok, 10900, Thailand.
| |
Collapse
|
5
|
Figueiredo IAD, Martins AMDO, Cavalcanti AMT, Fernandes JM, Gomes LEDS, Vieira MM, de Oliveira GNM, Felício IM, de Oliveira LN, Ramalho IGDS, de Sousa NF, Scotti L, Scotti MT, Alves JLDB, Diniz MDFFM, Ximenes DIJ, Vasconcelos LHC, Cavalcante FDA. Repeated-Dose Toxicity of Lauric Acid and Its Preventive Effect Against Tracheal Hyper-Responsiveness in Wistar Rats with Possible In Silico Molecular Targets. Pharmaceuticals (Basel) 2025; 18:221. [PMID: 40006035 PMCID: PMC11859213 DOI: 10.3390/ph18020221] [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: 12/03/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Lauric acid (LA), a medium-chain fatty acid, is a promising drug for asthma treatment. This study evaluated the toxicity of repeated doses and the effect of LA on pulmonary ventilation and tracheal reactivity in asthmatic Wistar rats and identified possible molecular targets of LA action in silico. METHODS The rats were divided into control (CG) and LA-treated groups at 100 mg/kg (AL100G) for toxicity analysis. Pulmonary ventilation and tracheal reactivity were assessed in the control (CG), asthmatic (AG), asthmatic treated with LA at 25, 50, or 100 mg/kg (AAL25G, AAL50G, and AAL100G), and dexamethasone-treated groups (ADEXAG). RESULTS The results showed that LA at a dose of 100 mg/kg did not cause death or toxicity. A pulmonary ventilation analysis indicated that AG had reduced minute volume, which was prevented in AAL25G. LA at all doses prevented carbachol-induced tracheal hyper-responsiveness and reduced the relaxing effect of aminophylline, as observed in AG. An in silico analysis revealed that LA had a good affinity for nine proteins (β2-adrenergic receptor, CaV, BKCa, KATP, adenylyl cyclase, PKG, eNOS, iNOS, and COX-2). CONCLUSIONS LA at 100 mg/kg has low toxicity, prevents hyper-responsiveness in an asthma model in rats, and acts as a multitarget compound with a good affinity for proteins related to airway hyper-responsiveness.
Collapse
Affiliation(s)
- Indyra Alencar Duarte Figueiredo
- Laboratório de Farmacologia Funcional Prof. George Thomas, Instituto de Pesquisa em Fármacos e Medicamentos, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil; (A.M.d.O.M.); (A.M.T.C.); (J.M.F.); (L.E.d.S.G.); (M.M.V.); (G.N.M.d.O.); (I.M.F.); (L.N.d.O.); (I.G.d.S.R.); (N.F.d.S.); (F.d.A.C.)
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil; (L.S.); (M.T.S.); (M.d.F.F.M.D.)
| | - Alissa Maria de Oliveira Martins
- Laboratório de Farmacologia Funcional Prof. George Thomas, Instituto de Pesquisa em Fármacos e Medicamentos, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil; (A.M.d.O.M.); (A.M.T.C.); (J.M.F.); (L.E.d.S.G.); (M.M.V.); (G.N.M.d.O.); (I.M.F.); (L.N.d.O.); (I.G.d.S.R.); (N.F.d.S.); (F.d.A.C.)
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil; (L.S.); (M.T.S.); (M.d.F.F.M.D.)
| | - Alexya Mikelle Teixeira Cavalcanti
- Laboratório de Farmacologia Funcional Prof. George Thomas, Instituto de Pesquisa em Fármacos e Medicamentos, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil; (A.M.d.O.M.); (A.M.T.C.); (J.M.F.); (L.E.d.S.G.); (M.M.V.); (G.N.M.d.O.); (I.M.F.); (L.N.d.O.); (I.G.d.S.R.); (N.F.d.S.); (F.d.A.C.)
| | - Jayne Muniz Fernandes
- Laboratório de Farmacologia Funcional Prof. George Thomas, Instituto de Pesquisa em Fármacos e Medicamentos, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil; (A.M.d.O.M.); (A.M.T.C.); (J.M.F.); (L.E.d.S.G.); (M.M.V.); (G.N.M.d.O.); (I.M.F.); (L.N.d.O.); (I.G.d.S.R.); (N.F.d.S.); (F.d.A.C.)
| | - Ludmila Emilly da Silva Gomes
- Laboratório de Farmacologia Funcional Prof. George Thomas, Instituto de Pesquisa em Fármacos e Medicamentos, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil; (A.M.d.O.M.); (A.M.T.C.); (J.M.F.); (L.E.d.S.G.); (M.M.V.); (G.N.M.d.O.); (I.M.F.); (L.N.d.O.); (I.G.d.S.R.); (N.F.d.S.); (F.d.A.C.)
| | - Mateus Mendes Vieira
- Laboratório de Farmacologia Funcional Prof. George Thomas, Instituto de Pesquisa em Fármacos e Medicamentos, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil; (A.M.d.O.M.); (A.M.T.C.); (J.M.F.); (L.E.d.S.G.); (M.M.V.); (G.N.M.d.O.); (I.M.F.); (L.N.d.O.); (I.G.d.S.R.); (N.F.d.S.); (F.d.A.C.)
| | - Gabriel Nunes Machado de Oliveira
- Laboratório de Farmacologia Funcional Prof. George Thomas, Instituto de Pesquisa em Fármacos e Medicamentos, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil; (A.M.d.O.M.); (A.M.T.C.); (J.M.F.); (L.E.d.S.G.); (M.M.V.); (G.N.M.d.O.); (I.M.F.); (L.N.d.O.); (I.G.d.S.R.); (N.F.d.S.); (F.d.A.C.)
| | - Isabela Motta Felício
- Laboratório de Farmacologia Funcional Prof. George Thomas, Instituto de Pesquisa em Fármacos e Medicamentos, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil; (A.M.d.O.M.); (A.M.T.C.); (J.M.F.); (L.E.d.S.G.); (M.M.V.); (G.N.M.d.O.); (I.M.F.); (L.N.d.O.); (I.G.d.S.R.); (N.F.d.S.); (F.d.A.C.)
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil; (L.S.); (M.T.S.); (M.d.F.F.M.D.)
| | - Lucas Nóbrega de Oliveira
- Laboratório de Farmacologia Funcional Prof. George Thomas, Instituto de Pesquisa em Fármacos e Medicamentos, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil; (A.M.d.O.M.); (A.M.T.C.); (J.M.F.); (L.E.d.S.G.); (M.M.V.); (G.N.M.d.O.); (I.M.F.); (L.N.d.O.); (I.G.d.S.R.); (N.F.d.S.); (F.d.A.C.)
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil; (L.S.); (M.T.S.); (M.d.F.F.M.D.)
| | - Igor Gabriel da Silva Ramalho
- Laboratório de Farmacologia Funcional Prof. George Thomas, Instituto de Pesquisa em Fármacos e Medicamentos, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil; (A.M.d.O.M.); (A.M.T.C.); (J.M.F.); (L.E.d.S.G.); (M.M.V.); (G.N.M.d.O.); (I.M.F.); (L.N.d.O.); (I.G.d.S.R.); (N.F.d.S.); (F.d.A.C.)
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil; (L.S.); (M.T.S.); (M.d.F.F.M.D.)
| | - Natália Ferreira de Sousa
- Laboratório de Farmacologia Funcional Prof. George Thomas, Instituto de Pesquisa em Fármacos e Medicamentos, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil; (A.M.d.O.M.); (A.M.T.C.); (J.M.F.); (L.E.d.S.G.); (M.M.V.); (G.N.M.d.O.); (I.M.F.); (L.N.d.O.); (I.G.d.S.R.); (N.F.d.S.); (F.d.A.C.)
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil; (L.S.); (M.T.S.); (M.d.F.F.M.D.)
| | - Luciana Scotti
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil; (L.S.); (M.T.S.); (M.d.F.F.M.D.)
| | - Marcus Tullius Scotti
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil; (L.S.); (M.T.S.); (M.d.F.F.M.D.)
- Departamento de Química, Centro de Ciências Exatas e da Natureza, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil
| | - José Luiz de Brito Alves
- Departamento de Nutrição, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil;
| | - Margareth de Fátima Formiga Melo Diniz
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil; (L.S.); (M.T.S.); (M.d.F.F.M.D.)
- Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal da Paraíba, Cidade Universitária, João Pessoa 58051-900, PB, Brazil;
| | - Daniele Idalino Janebro Ximenes
- Departamento de Ciências Farmacêuticas, Centro de Ciências da Saúde, Universidade Federal da Paraíba, Cidade Universitária, João Pessoa 58051-900, PB, Brazil;
| | - Luiz Henrique César Vasconcelos
- Laboratório de Farmacologia Funcional Prof. George Thomas, Instituto de Pesquisa em Fármacos e Medicamentos, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil; (A.M.d.O.M.); (A.M.T.C.); (J.M.F.); (L.E.d.S.G.); (M.M.V.); (G.N.M.d.O.); (I.M.F.); (L.N.d.O.); (I.G.d.S.R.); (N.F.d.S.); (F.d.A.C.)
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil; (L.S.); (M.T.S.); (M.d.F.F.M.D.)
- Departamento de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal da Paraíba, Cidade Universitária, João Pessoa 58051-900, PB, Brazil
| | - Fabiana de Andrade Cavalcante
- Laboratório de Farmacologia Funcional Prof. George Thomas, Instituto de Pesquisa em Fármacos e Medicamentos, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil; (A.M.d.O.M.); (A.M.T.C.); (J.M.F.); (L.E.d.S.G.); (M.M.V.); (G.N.M.d.O.); (I.M.F.); (L.N.d.O.); (I.G.d.S.R.); (N.F.d.S.); (F.d.A.C.)
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Centro de Ciências da Saúde, Universidade Federal da Paraíba, João Pessoa 58051-900, PB, Brazil; (L.S.); (M.T.S.); (M.d.F.F.M.D.)
- Departamento de Ciências Biomédicas, Centro de Ciências da Saúde, Universidade Federal da Paraíba, Cidade Universitária, João Pessoa 58051-900, PB, Brazil
| |
Collapse
|
6
|
Kelly MP, Nikolaev VO, Gobejishvili L, Lugnier C, Hesslinger C, Nickolaus P, Kass DA, Pereira de Vasconcelos W, Fischmeister R, Brocke S, Epstein PM, Piazza GA, Keeton AB, Zhou G, Abdel-Halim M, Abadi AH, Baillie GS, Giembycz MA, Bolger G, Snyder G, Tasken K, Saidu NEB, Schmidt M, Zaccolo M, Schermuly RT, Ke H, Cote RH, Mohammadi Jouabadi S, Roks AJM. Cyclic nucleotide phosphodiesterases as drug targets. Pharmacol Rev 2025; 77:100042. [PMID: 40081105 DOI: 10.1016/j.pharmr.2025.100042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 01/13/2025] [Indexed: 03/15/2025] Open
Abstract
Cyclic nucleotides are synthesized by adenylyl and/or guanylyl cyclase, and downstream of this synthesis, the cyclic nucleotide phosphodiesterase families (PDEs) specifically hydrolyze cyclic nucleotides. PDEs control cyclic adenosine-3',5'monophosphate (cAMP) and cyclic guanosine-3',5'-monophosphate (cGMP) intracellular levels by mediating their quick return to the basal steady state levels. This often takes place in subcellular nanodomains. Thus, PDEs govern short-term protein phosphorylation, long-term protein expression, and even epigenetic mechanisms by modulating cyclic nucleotide levels. Consequently, their involvement in both health and disease is extensively investigated. PDE inhibition has emerged as a promising clinical intervention method, with ongoing developments aiming to enhance its efficacy and applicability. In this comprehensive review, we extensively look into the intricate landscape of PDEs biochemistry, exploring their diverse roles in various tissues. Furthermore, we outline the underlying mechanisms of PDEs in different pathophysiological conditions. Additionally, we review the application of PDE inhibition in related diseases, shedding light on current advancements and future prospects for clinical intervention. SIGNIFICANCE STATEMENT: Regulating PDEs is a critical checkpoint for numerous (patho)physiological conditions. However, despite the development of several PDE inhibitors aimed at controlling overactivated PDEs, their applicability in clinical settings poses challenges. In this context, our focus is on pharmacodynamics and the structure activity of PDEs, aiming to illustrate how selectivity and efficacy can be optimized. Additionally, this review points to current preclinical and clinical evidence that depicts various optimization efforts and indications.
Collapse
Affiliation(s)
- Michy P Kelly
- Department of Neurobiology, Center for Research on Aging, University of Maryland School of Medicine, Baltimore, Maryland
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Leila Gobejishvili
- Department of Physiology, School of Medicine, University of Louisville, Kentucky, Louisville
| | - Claire Lugnier
- Translational CardioVascular Medicine, CRBS, UR 3074, Strasbourg, France
| | | | - Peter Nickolaus
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Rodolphe Fischmeister
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Orsay, France
| | - Stefan Brocke
- Department of Immunology, UConn Health, Farmington, Connecticut
| | - Paul M Epstein
- Department of Cell Biology, UConn Health, Farmington, Connecticut
| | - Gary A Piazza
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Adam B Keeton
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Gang Zhou
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - George S Baillie
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Mark A Giembycz
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Gretchen Snyder
- Molecular Neuropharmacology, Intra-Cellular Therapies Inc (ITI), New York, New York
| | - Kjetil Tasken
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nathaniel E B Saidu
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics and National Institute for Health and Care Research Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Ralph T Schermuly
- Department of internal Medicine, Justus Liebig University of Giessen, Giessen, Germany
| | - Hengming Ke
- Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina
| | - Rick H Cote
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, New Hampshire
| | - Soroush Mohammadi Jouabadi
- Section of Vascular and Metabolic Disease, Department of Internal Medicine, Erasmus MC University Medical Center, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Anton J M Roks
- Section of Vascular and Metabolic Disease, Department of Internal Medicine, Erasmus MC University Medical Center, Erasmus University Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
7
|
Nemr MTM, Abdelaziz MA, Teleb M, Elmasry AE, Elshaier YAAM. An overview on pharmaceutical applications of phosphodiesterase enzyme 5 (PDE5) inhibitors. Mol Divers 2024:10.1007/s11030-024-11016-2. [PMID: 39592536 DOI: 10.1007/s11030-024-11016-2] [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: 08/24/2024] [Accepted: 10/12/2024] [Indexed: 11/28/2024]
Abstract
Phosphodiesterase enzyme 5 (PDE5) inhibitors have emerged as one of the leading molecules for the treatment of erectile dysfunction (ED). PDE5 inhibitors are categorized structurally into several classes. PDE5 inhibitors have been a multidisciplinary endeavor that attracts the attention of researchers because of their multiple pharmaceutical applications. Beyond their action on ED, PDE5 inhibitors are widely used in treatment of benign prostatic hypertrophy (BPH), Eisenmenger's syndrome, Raynaud's Disease, Intrauterine growth retardation (IUGR), Mountain sickness, Bladder pain syndrome/interstitial cystitis (BPS/IC), pulmonary arterial hypertension and type II diabetes (insulin resistance). In addition, PDE5 inhibitors also show promising antiproliferative activity, anti-Alzheimer and COX-1/COX-2 inhibitory activity (anti-inflammatory). Pharmacokinetics, Pharmacogenetics and toxicity of PDE5 inhibitors were finally explored. The diverse therapeutic applications, the high feasibility of structural modification and the appropriate pharmacokinetic properties of PDE5 inhibitors have motivated researchers to develop new scaffolds that have been either under clinical trials or approved by FDA and utilize them to overcome some recent global concerns, such as COVID-19.
Collapse
Affiliation(s)
- Mohamed T M Nemr
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Eini Street 11562, Cairo, Egypt.
| | | | - Mohamed Teleb
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt
- Faculty of Pharmacy, Alamein International University (AIU), Alamein City, Alamein City, 5060310, Egypt
| | - Ahmed E Elmasry
- Organic & Medicinal Chemistry Department, Faculty of Pharmacy, University of Sadat City, Menoufia, Egypt
| | - Yaseen A A M Elshaier
- Organic & Medicinal Chemistry Department, Faculty of Pharmacy, University of Sadat City, Menoufia, Egypt.
| |
Collapse
|
8
|
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
|
9
|
Wang S, Yang G, Zhang K, Chen Z, Qiu M, Hou S, Zheng T, Wu Z, Ma Q, Zhang F, Gao G, Huang YY, Zhou Q, Luo HB, Wu D. Structural optimization of Moracin M as novel selective phosphodiesterase 4 inhibitors for the treatment of idiopathic pulmonary fibrosis. Bioorg Chem 2024; 149:107474. [PMID: 38805909 DOI: 10.1016/j.bioorg.2024.107474] [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: 04/05/2024] [Revised: 05/06/2024] [Accepted: 05/16/2024] [Indexed: 05/30/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and high mortality lung disease. Although the antifibrotic drugs pirfenidone and nintedanib could slow the rate of lung function decline, the usual course of the condition is inexorably to respiratory failure and death. Therefore, new approaches and novel therapeutic drugs for the treatment of IPF are urgently needed. And the selective PDE4 inhibitor has in vivo and in vitro anti-fibrotic effects in IPF models. But the clinical application of most PDE4 inhibitors are limited by their unexpected and severe side effects such as nausea, vomiting, and diarrhea. Herein, structure-based optimizations of the natural product Moracin M resulted in a novel a novel series of 2-arylbenzofurans as potent PDE4 inhibitors. The most potent inhibitor L13 has an IC50 of 36 ± 7 nM with remarkable selectivity across the PDE families and administration of L13·citrate (10.0 mg/kg) exhibited comparable anti-pulmonary fibrosis effects to pirfenidone (300 mg/kg) in a bleomycin-induced IPF mice model, indicate that L13 is a potential lead for the treatment of IPF.
Collapse
Affiliation(s)
- Sen Wang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Guofeng Yang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Kai Zhang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Zhexin Chen
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Meiying Qiu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Siyu Hou
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Tiansheng Zheng
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Zongmin Wu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Qinjiang Ma
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Furong Zhang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China; School of Life and Health Sciences, Hainan University, Haikou 570228, China
| | - Ge Gao
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Yi-You Huang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Qian Zhou
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China.
| | - Hai-Bin Luo
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China.
| | - Deyan Wu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China.
| |
Collapse
|
10
|
Chaiyabutr N, Noiprom J, Promruangreang K, Vasaruchapong T, Laoungbua P, Khow O, Chanhome L, Sitprija V. Acute phase reactions in Daboia siamensis venom and fraction-induced acute kidney injury: the role of oxidative stress and inflammatory pathways in in vivo rabbit and ex vivo rabbit kidney models. J Venom Anim Toxins Incl Trop Dis 2024; 30:e20230070. [PMID: 38808074 PMCID: PMC11131233 DOI: 10.1590/1678-9199-jvatitd-2023-0070] [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: 09/08/2023] [Accepted: 03/25/2024] [Indexed: 05/30/2024] Open
Abstract
Background This study examines the direct nephrotoxic effects of Daboia siamensis venom (RVV) and venom fractions in in vivo and isolated perfused kidneys (IPK) to understand the role of inflammation pathways and susceptibility to oxidative stress in venom or fraction-induced acute renal failure. Methods We administered RVV and its venom fractions (PLA2, MP, LAAO, and PDE) to rabbits in vivo and in the IPK model. We measured oxidative stress biomarkers (SOD, CAT, GSH, and MDA) in kidney tissue, as well as inflammatory cytokines (TNF-α, IL-1β, IFN-γ, IL-4, IL-5, and IL-10), MDA and GSH levels in plasma and urine. We also calculated fractional excretion (FE) for pro-/anti-inflammatory cytokines and oxidative stress biomarkers, including the ratios of pro-/anti-inflammatory cytokines in urine after envenomation. Results In both kidney models, significant increases in MDA, SOD, CAT, and GSH levels were observed in kidney tissues, along with elevated concentrations of MDA and GSH in plasma and urine after injecting RVV and venom fractions. Moreover, RVV injections led to progressive increases in FEMDA and decreases in FEGSH. The concentrations of IL-4, IL-5, IL-10, IFN-γ, and TNF-α in plasma increased in vivo, as well as in the urine of the IPK model, but not for IL-1β in both plasma and urine after RVV administrations. Urinary fractional excretion of TNF-α, IL-1β, IFN-γ, IL-4, IL-5, and IL-10 tended to decrease in vivo but showed elevated levels in the IPK model. A single RVV injection in vivo disrupted the balance of urinary cytokines, significantly reducing either the TNF-α/IL-10 ratio or the IFN-γ/IL-10 ratio. Conclusion RVV induces renal tubular toxicity by increasing oxidative stress production and elevating inflammatory cytokines in urine. During the acute phase of acute kidney injury, the balance of urine cytokines shifts toward anti-inflammatory dominance within the first two hours post-RVV and venom fractions.
Collapse
Affiliation(s)
- Narongsak Chaiyabutr
- Queen Saovabha Memorial Institute, The Thai Red Cross Society,
Pathumwan, Bangkok, Thailand
| | - Jureeporn Noiprom
- Department of Research and Development, Queen Saovabha Memorial
Institute, The Thai Red Cross Society, Bangkok, Thailand
| | - Kanyanat Promruangreang
- Department of Research and Development, Queen Saovabha Memorial
Institute, The Thai Red Cross Society, Bangkok, Thailand
| | - Taksa Vasaruchapong
- Snake Farm, Queen Saovabha Memorial Institute, The Thai Red Cross
Society, Bangkok, Thailand
| | - Panithi Laoungbua
- Snake Farm, Queen Saovabha Memorial Institute, The Thai Red Cross
Society, Bangkok, Thailand
| | - Orawan Khow
- Department of Research and Development, Queen Saovabha Memorial
Institute, The Thai Red Cross Society, Bangkok, Thailand
| | - Lawan Chanhome
- Snake Farm, Queen Saovabha Memorial Institute, The Thai Red Cross
Society, Bangkok, Thailand
| | - Visith Sitprija
- Queen Saovabha Memorial Institute, The Thai Red Cross Society,
Pathumwan, Bangkok, Thailand
| |
Collapse
|
11
|
Kokot M, Dehghannasiri R, Baharav T, Salzman J, Deorowicz S. SPLASH2 provides ultra-efficient, scalable, and unsupervised discovery on raw sequencing reads. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.17.533189. [PMID: 36993432 PMCID: PMC10055302 DOI: 10.1101/2023.03.17.533189] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
SPLASH is an unsupervised, reference-free, and unifying algorithm that discovers regulated sequence variation through statistical analysis of k-mer composition, subsuming many application-specific methods. Here, we introduce SPLASH2, a fast, scalable implementation of SPLASH based on an efficient k-mer counting approach. SPLASH2 enables rapid analysis of massive datasets from a wide range of sequencing technologies and biological contexts, delivering unparalleled scale and speed. The SPLASH2 algorithm unveils new biology (without tuning) in single-cell RNA-sequencing data from human muscle cells, as well as bulk RNA-seq from the entire Cancer Cell Line Encyclopedia (CCLE), including substantial unannotated alternative splicing in cancer transcriptome. The same untuned SPLASH2 algorithm recovers the BCR-ABL gene fusion, and detects circRNA sensitively and specifically, underscoring SPLASH2's unmatched precision and scalability across diverse RNA-seq detection tasks.
Collapse
Affiliation(s)
- Marek Kokot
- Department of Algorithmics and Software, Silesian University of Technology, Gliwice, Poland
| | - Roozbeh Dehghannasiri
- Department of Biomedical Data Science, Stanford University, Stanford, 94305, USA
- Department of Biochemistry, Stanford University, Stanford, 94305, USA
| | - Tavor Baharav
- Department of Electrical Engineering, Stanford University, Stanford, 94305, USA
| | - Julia Salzman
- Department of Biomedical Data Science, Stanford University, Stanford, 94305, USA
- Department of Biochemistry, Stanford University, Stanford, 94305, USA
- Department of Statistics (by courtesy), Stanford University, Stanford, 94305, USA
| | - Sebastian Deorowicz
- Department of Algorithmics and Software, Silesian University of Technology, Gliwice, Poland
| |
Collapse
|
12
|
Zhang H, Liu Y, Liu J, Chen J, Wang J, Hua H, Jiang Y. cAMP-PKA/EPAC signaling and cancer: the interplay in tumor microenvironment. J Hematol Oncol 2024; 17:5. [PMID: 38233872 PMCID: PMC10792844 DOI: 10.1186/s13045-024-01524-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/02/2024] [Indexed: 01/19/2024] Open
Abstract
Cancer is a complex disease resulting from abnormal cell growth that is induced by a number of genetic and environmental factors. The tumor microenvironment (TME), which involves extracellular matrix, cancer-associated fibroblasts (CAF), tumor-infiltrating immune cells and angiogenesis, plays a critical role in tumor progression. Cyclic adenosine monophosphate (cAMP) is a second messenger that has pleiotropic effects on the TME. The downstream effectors of cAMP include cAMP-dependent protein kinase (PKA), exchange protein activated by cAMP (EPAC) and ion channels. While cAMP can activate PKA or EPAC and promote cancer cell growth, it can also inhibit cell proliferation and survival in context- and cancer type-dependent manner. Tumor-associated stromal cells, such as CAF and immune cells, can release cytokines and growth factors that either stimulate or inhibit cAMP production within the TME. Recent studies have shown that targeting cAMP signaling in the TME has therapeutic benefits in cancer. Small-molecule agents that inhibit adenylate cyclase and PKA have been shown to inhibit tumor growth. In addition, cAMP-elevating agents, such as forskolin, can not only induce cancer cell death, but also directly inhibit cell proliferation in some cancer types. In this review, we summarize current understanding of cAMP signaling in cancer biology and immunology and discuss the basis for its context-dependent dual role in oncogenesis. Understanding the precise mechanisms by which cAMP and the TME interact in cancer will be critical for the development of effective therapies. Future studies aimed at investigating the cAMP-cancer axis and its regulation in the TME may provide new insights into the underlying mechanisms of tumorigenesis and lead to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Hongying Zhang
- Cancer Center, Laboratory of Oncogene, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yongliang Liu
- Cancer Center, Laboratory of Oncogene, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jieya Liu
- Cancer Center, Laboratory of Oncogene, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jinzhu Chen
- Cancer Center, Laboratory of Oncogene, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiao Wang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Hui Hua
- Laboratory of Stem Cell Biology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yangfu Jiang
- Cancer Center, Laboratory of Oncogene, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
13
|
Zhu Z, Tang W, Qiu X, Xin X, Zhang J. Advances in targeting Phosphodiesterase 1: From mechanisms to potential therapeutics. Eur J Med Chem 2024; 263:115967. [PMID: 38000211 DOI: 10.1016/j.ejmech.2023.115967] [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: 10/04/2023] [Revised: 11/07/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023]
Abstract
Phosphodiesterase 1 (PDE1) is an enzyme entrusted with the hydrolysis of the second messengers cAMP and cGMP, thereby governing a plethora of metabolic processes, encompassing ion channel modulation and cellular apoptosis. Recent advancements in the realm of small molecule structural variations have greatly facilitated the exploration of innovative applications for PDE1. Remarkably, a recent series of PDE1 inhibitors (PDE1i) have been meticulously formulated and devised, showcasing enhanced selectivity and potency. Among them, ITI-214 has entered Phase II clinical trials, holding promise for the treatment of Parkinson's disease and heart failure. Nevertheless, the majority of current PDE1 inhibitors have encountered substantial side effects in clinical trials attributable to their limited selectivity, this predicament presents a formidable obstacle in the development of specific small molecule inhibitors targeting PDE1. This Perspective endeavors to illuminate the potential design approaches, structure-activity relationships, and biological activities of current PDE1i, aiming to offer support and insights for clinical practice and the development of novel PDE1i.
Collapse
Affiliation(s)
- Ziyu Zhu
- Department of Neurology, Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Wentao Tang
- Department of Neurology, Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xuemei Qiu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xin Xin
- State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Jifa Zhang
- Department of Neurology, Joint Research Institution of Altitude Health and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; State Key Laboratory of Biotherapy and Cancer Center, Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
14
|
Kochoian BA, Bure C, Papa SM. Targeting Striatal Glutamate and Phosphodiesterases to Control L-DOPA-Induced Dyskinesia. Cells 2023; 12:2754. [PMID: 38067182 PMCID: PMC10706484 DOI: 10.3390/cells12232754] [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: 11/01/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
A large body of work during the past several decades has been focused on therapeutic strategies to control L-DOPA-induced dyskinesias (LIDs), common motor complications of long-term L-DOPA therapy in Parkinson's disease (PD). Yet, LIDs remain a clinical challenge for the management of patients with advanced disease. Glutamatergic dysregulation of striatal projection neurons (SPNs) appears to be a key contributor to altered motor responses to L-DOPA. Targeting striatal hyperactivity at the glutamatergic neurotransmission level led to significant preclinical and clinical trials of a variety of antiglutamatergic agents. In fact, the only FDA-approved treatment for LIDs is amantadine, a drug with NMDAR antagonistic actions. Still, novel agents with improved pharmacological profiles are needed for LID therapy. Recently other therapeutic targets to reduce dysregulated SPN activity at the signal transduction level have emerged. In particular, mechanisms regulating the levels of cyclic nucleotides play a major role in the transduction of dopamine signals in SPNs. The phosphodiesterases (PDEs), a large family of enzymes that degrade cyclic nucleotides in a specific manner, are of special interest. We will review the research for antiglutamatergic and PDE inhibition strategies in view of the future development of novel LID therapies.
Collapse
Affiliation(s)
- Brik A. Kochoian
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; (B.A.K.); (C.B.)
| | - Cassandra Bure
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; (B.A.K.); (C.B.)
| | - Stella M. Papa
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; (B.A.K.); (C.B.)
- Department of Neurology, Emory University School of Medicine, Atlanta, GA 30329, USA
| |
Collapse
|
15
|
Michel MC, Cardozo L, Chermansky CJ, Cruz F, Igawa Y, Lee KS, Sahai A, Wein AJ, Andersson KE. Current and Emerging Pharmacological Targets and Treatments of Urinary Incontinence and Related Disorders. Pharmacol Rev 2023; 75:554-674. [PMID: 36918261 DOI: 10.1124/pharmrev.121.000523] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 03/16/2023] Open
Abstract
Overactive bladder syndrome with and without urinary incontinence and related conditions, signs, and disorders such as detrusor overactivity, neurogenic lower urinary tract dysfunction, underactive bladder, stress urinary incontinence, and nocturia are common in the general population and have a major impact on the quality of life of the affected patients and their partners. Based on the deliberations of the subcommittee on pharmacological treatments of the 7th International Consultation on Incontinence, we present a comprehensive review of established drug targets in the treatment of overactive bladder syndrome and the aforementioned related conditions and the approved drugs used in its treatment. Investigational drug targets and compounds are also reviewed. We conclude that, despite a range of available medical treatment options, a considerable medical need continues to exist. This is largely because the existing treatments are symptomatic and have limited efficacy and/or tolerability, which leads to poor long-term adherence. SIGNIFICANCE STATEMENT: Urinary incontinence and related disorders are prevalent in the general population. While many treatments have been approved, few patients stay on long-term treatment despite none of them being curative. This paper provides a comprehensive discussion of existing and emerging treatment options for various types of incontinence and related disorders.
Collapse
Affiliation(s)
- Martin C Michel
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany (M.C.M.); Department of Urogynaecology, King's College Hospital, London, UK (L.C.); Department of Urology, Magee Women's Hospital, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania (C.J.C.); Department of Urology, Faculty of Medicine of University of Porto, Hospital São João and i3S Institute for Innovation and Investigation in Health, Porto, Portugal (F.C.); Department of Urology, Nagano Prefectural Shinshu Medical Center, Suzaka, Japan (Y.I.); Department of Urology Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (K-S.L.); Guy's Hospital and King's College London, London, UK (A.S.); Dept. of Urology, Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.J.W.); Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.J.W.); and Institute for Laboratory Medicine, Lund University, Lund, Sweden (K-E.A.)
| | - Linda Cardozo
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany (M.C.M.); Department of Urogynaecology, King's College Hospital, London, UK (L.C.); Department of Urology, Magee Women's Hospital, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania (C.J.C.); Department of Urology, Faculty of Medicine of University of Porto, Hospital São João and i3S Institute for Innovation and Investigation in Health, Porto, Portugal (F.C.); Department of Urology, Nagano Prefectural Shinshu Medical Center, Suzaka, Japan (Y.I.); Department of Urology Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (K-S.L.); Guy's Hospital and King's College London, London, UK (A.S.); Dept. of Urology, Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.J.W.); Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.J.W.); and Institute for Laboratory Medicine, Lund University, Lund, Sweden (K-E.A.)
| | - Christopher J Chermansky
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany (M.C.M.); Department of Urogynaecology, King's College Hospital, London, UK (L.C.); Department of Urology, Magee Women's Hospital, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania (C.J.C.); Department of Urology, Faculty of Medicine of University of Porto, Hospital São João and i3S Institute for Innovation and Investigation in Health, Porto, Portugal (F.C.); Department of Urology, Nagano Prefectural Shinshu Medical Center, Suzaka, Japan (Y.I.); Department of Urology Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (K-S.L.); Guy's Hospital and King's College London, London, UK (A.S.); Dept. of Urology, Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.J.W.); Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.J.W.); and Institute for Laboratory Medicine, Lund University, Lund, Sweden (K-E.A.)
| | - Francisco Cruz
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany (M.C.M.); Department of Urogynaecology, King's College Hospital, London, UK (L.C.); Department of Urology, Magee Women's Hospital, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania (C.J.C.); Department of Urology, Faculty of Medicine of University of Porto, Hospital São João and i3S Institute for Innovation and Investigation in Health, Porto, Portugal (F.C.); Department of Urology, Nagano Prefectural Shinshu Medical Center, Suzaka, Japan (Y.I.); Department of Urology Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (K-S.L.); Guy's Hospital and King's College London, London, UK (A.S.); Dept. of Urology, Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.J.W.); Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.J.W.); and Institute for Laboratory Medicine, Lund University, Lund, Sweden (K-E.A.)
| | - Yasuhiko Igawa
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany (M.C.M.); Department of Urogynaecology, King's College Hospital, London, UK (L.C.); Department of Urology, Magee Women's Hospital, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania (C.J.C.); Department of Urology, Faculty of Medicine of University of Porto, Hospital São João and i3S Institute for Innovation and Investigation in Health, Porto, Portugal (F.C.); Department of Urology, Nagano Prefectural Shinshu Medical Center, Suzaka, Japan (Y.I.); Department of Urology Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (K-S.L.); Guy's Hospital and King's College London, London, UK (A.S.); Dept. of Urology, Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.J.W.); Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.J.W.); and Institute for Laboratory Medicine, Lund University, Lund, Sweden (K-E.A.)
| | - Kyu-Sung Lee
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany (M.C.M.); Department of Urogynaecology, King's College Hospital, London, UK (L.C.); Department of Urology, Magee Women's Hospital, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania (C.J.C.); Department of Urology, Faculty of Medicine of University of Porto, Hospital São João and i3S Institute for Innovation and Investigation in Health, Porto, Portugal (F.C.); Department of Urology, Nagano Prefectural Shinshu Medical Center, Suzaka, Japan (Y.I.); Department of Urology Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (K-S.L.); Guy's Hospital and King's College London, London, UK (A.S.); Dept. of Urology, Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.J.W.); Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.J.W.); and Institute for Laboratory Medicine, Lund University, Lund, Sweden (K-E.A.)
| | - Arun Sahai
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany (M.C.M.); Department of Urogynaecology, King's College Hospital, London, UK (L.C.); Department of Urology, Magee Women's Hospital, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania (C.J.C.); Department of Urology, Faculty of Medicine of University of Porto, Hospital São João and i3S Institute for Innovation and Investigation in Health, Porto, Portugal (F.C.); Department of Urology, Nagano Prefectural Shinshu Medical Center, Suzaka, Japan (Y.I.); Department of Urology Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (K-S.L.); Guy's Hospital and King's College London, London, UK (A.S.); Dept. of Urology, Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.J.W.); Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.J.W.); and Institute for Laboratory Medicine, Lund University, Lund, Sweden (K-E.A.)
| | - Alan J Wein
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany (M.C.M.); Department of Urogynaecology, King's College Hospital, London, UK (L.C.); Department of Urology, Magee Women's Hospital, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania (C.J.C.); Department of Urology, Faculty of Medicine of University of Porto, Hospital São João and i3S Institute for Innovation and Investigation in Health, Porto, Portugal (F.C.); Department of Urology, Nagano Prefectural Shinshu Medical Center, Suzaka, Japan (Y.I.); Department of Urology Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (K-S.L.); Guy's Hospital and King's College London, London, UK (A.S.); Dept. of Urology, Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.J.W.); Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.J.W.); and Institute for Laboratory Medicine, Lund University, Lund, Sweden (K-E.A.)
| | - Karl-Erik Andersson
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University, Mainz, Germany (M.C.M.); Department of Urogynaecology, King's College Hospital, London, UK (L.C.); Department of Urology, Magee Women's Hospital, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania (C.J.C.); Department of Urology, Faculty of Medicine of University of Porto, Hospital São João and i3S Institute for Innovation and Investigation in Health, Porto, Portugal (F.C.); Department of Urology, Nagano Prefectural Shinshu Medical Center, Suzaka, Japan (Y.I.); Department of Urology Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea (K-S.L.); Guy's Hospital and King's College London, London, UK (A.S.); Dept. of Urology, Perlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.J.W.); Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina (A.J.W.); and Institute for Laboratory Medicine, Lund University, Lund, Sweden (K-E.A.)
| |
Collapse
|
16
|
Tsuda N, Tian Y, Fujimoto M, Kuramoto J, Makiuchi S, Ojima H, Gotoh M, Hiraoka N, Yoshida T, Kanai Y, Arai E. DNA methylation status of the SPHK1 and LTB genes underlies the clinicopathological diversity of non-alcoholic steatohepatitis-related hepatocellular carcinomas. J Cancer Res Clin Oncol 2023; 149:5109-5125. [PMID: 36348017 PMCID: PMC10349775 DOI: 10.1007/s00432-022-04445-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/19/2022] [Indexed: 11/09/2022]
Abstract
PURPOSE This study was performed to identify the DNA methylation profiles underlying the clinicopathological diversity of non-alcoholic steatohepatitis (NASH)-related hepatocellular carcinomas (HCCs). METHODS: Genome-wide DNA methylation analysis of 88 liver tissue samples was performed using the Infinium assay. RESULTS Principal component analysis revealed that distinct DNA methylation profiles differing from such profiles in normal control liver tissue had already been established in non-cancerous liver tissue showing NASH, which is considered to be a precancerous condition. Hierarchical clustering separated 26 NASH-related HCCs into Cluster I (n = 8) and Cluster II (n = 18). Such epigenetic clustering was significantly correlated with histopathological diversity, i.e. poorer tumor differentiation, tumor steatosis and development of a scirrhous HCC component. Significant differences in DNA methylation levels between the two clusters were accumulated in molecular pathways participating in cell adhesion and cytoskeletal remodeling, as well as cell proliferation and apoptosis. Among tumor-related genes characterizing Clusters I and II, differences in the levels of DNA methylation and mRNA expression for the SPHK1, INHBA, LTB and PDE3B genes were correlated with poorer tumor differentiation. 5-Aza-2'-deoxycytidine treatment of HCC cells revealed epigenetic regulation of the SPHK1 and LTB genes. Knockdown experiments showed that SPHK1 promotes cell proliferation, represses apoptosis and enhances migration, whereas LTB enhances migration of HCC cells. DNA hypomethylation resulting in increased expression of SPHK1 and LTB in poorly differentiated HCCs may underlie the aggressive phenotype of such HCCs. CONCLUSION These data indicate that DNA methylation profiles may determine the clinicopathological heterogeneity of NASH-related HCCs via alterations of tumor-related gene expression.
Collapse
Affiliation(s)
- Noboru Tsuda
- Department of Pathology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Ying Tian
- Department of Pathology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Mao Fujimoto
- Department of Pathology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Junko Kuramoto
- Department of Pathology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Satomi Makiuchi
- Department of Pathology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Hidenori Ojima
- Department of Pathology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan
| | - Masahiro Gotoh
- Fundamental Innovative Oncology Core Center, National Cancer Center Research Institute, Tokyo, 104-0045, Japan
| | - Nobuyoshi Hiraoka
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, 104-0045, Japan
| | - Teruhiko Yoshida
- Fundamental Innovative Oncology Core Center, National Cancer Center Research Institute, Tokyo, 104-0045, Japan
| | - Yae Kanai
- Department of Pathology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan.
| | - Eri Arai
- Department of Pathology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-Ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
17
|
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: 4] [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
|
18
|
Azargoonjahromi A. Dual role of nitric oxide in Alzheimer's Disease. Nitric Oxide 2023; 134-135:23-37. [PMID: 37019299 DOI: 10.1016/j.niox.2023.03.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/02/2023] [Accepted: 03/31/2023] [Indexed: 04/05/2023]
Abstract
Nitric oxide (NO), an enzymatic product of nitric oxide synthase (NOS), has been associated with a variety of neurological diseases such as Alzheimer's disease (AD). NO has long been thought to contribute to neurotoxic insults caused by neuroinflammation in AD. This perception shifts as more attention is paid to the early stages before cognitive problems manifest. However, it has revealed a compensatory neuroprotective role for NO that protects synapses by increasing neuronal excitability. NO can positively affect neurons by inducing neuroplasticity, neuroprotection, and myelination, as well as having cytolytic activity to reduce inflammation. NO can also induce long-term potentiation (LTP), a process by which synaptic connections among neurons become more potent. Not to mention that such functions give rise to AD protection. Notably, it is unquestionably necessary to conduct more research to clarify NO pathways in neurodegenerative dementias because doing so could help us better understand their pathophysiology and develop more effective treatment options. All these findings bring us to the prevailing notion that NO can be used either as a therapeutic agent in patients afflicted with AD and other memory impairment disorders or as a contributor to the neurotoxic and aggressive factor in AD. In this review, after presenting a general background on AD and NO, various factors that have a pivotal role in both protecting and exacerbating AD and their correlation with NO will be elucidated. Following this, both the neuroprotective and neurotoxic effects of NO on neurons and glial cells among AD cases will be discussed in detail.
Collapse
|
19
|
Neuronal lack of PDE7a disrupted working memory, spatial learning, and memory but facilitated cued fear memory in mice. Prog Neuropsychopharmacol Biol Psychiatry 2023; 120:110655. [PMID: 36220621 DOI: 10.1016/j.pnpbp.2022.110655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/01/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND PDEs regulate cAMP levels which is critical for PKA activity-dependent activation of CREB-mediated transcription in learning and memory. Inhibitors of PDEs like PDE4 and Pde7 improve learning and memory in rodents. However, the role of PDE7 in cognition or learning and memory has not been reported yet. METHODS Therefore, we aimed to explore the cognitive effects of a PDE7 subtype, PDE7a, using combined pharmacological and genetic approaches. RESULTS PDE7a-nko mice showed deficient working memory, impaired novel object recognition, deficient spatial learning & memory, and contextual fear memory, contrary to enhanced cued fear memory, highlighting the potential opposite role of PDE7a in the hippocampal neurons. Further, pharmacological inhibition of PDE7 by AGF2.20 selectively strengthens cued fear memory in C57BL/6 J mice, decreasing its extinction but did not affect cognitive processes assessed in other behavioral tests. The further biochemical analysis detected deficient cAMP in neural cell culture with genetic excision of the PDE7a gene, as well as in the hippocampus of PDE7a-nko mice in vivo. Importantly, we found overexpression of PKA-R and the reduced level of pPKA-C in the hippocampus of PDE7a-nko mice, suggesting a novel mechanism of the cAMP regulation by PDE7a. Consequently, the decreased phosphorylation of CREB, CAMKII, eif2a, ERK, and AMPK, and reduced total level of NR2A have been found in the brain of PDE7a-nko animals. Notably, genetic excision of PDE7a in neurons was not able to change the expression of NR2B, BDNF, synapsin1, synaptophysin, or snap25. CONCLUSION Altogether, our current findings demonstrated, for the first time, the role of PDE7a in cognitive processes. Future studies will untangle PDE7a-dependent neurobiological and molecular-cellular mechanisms related to cAMP-associated disorders.
Collapse
|
20
|
Wu J, Zhao X, Xiao C, Xiong G, Ye X, Li L, Fang Y, Chen H, Yang W, Du X. The role of lung macrophages in chronic obstructive pulmonary disease. Respir Med 2022; 205:107035. [PMID: 36343504 DOI: 10.1016/j.rmed.2022.107035] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/17/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) as a common, preventable and treatable chronic respiratory disease in clinic, gets continuous deterioration and we can't take effective intervention at present. Lung macrophages (LMs) are closely related to the occurrence and development of COPD, but the specific mechanism is not completely clear. In this review we will focus on the role of LMs and potential avenues for therapeutic targeting for LMs in COPD.
Collapse
Affiliation(s)
- Jianli Wu
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Xia Zhao
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Chuang Xiao
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, China
| | - Guosheng Xiong
- Thoracic Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Xiulin Ye
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Lin Li
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Yan Fang
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Hong Chen
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Weimin Yang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, China.
| | - Xiaohua Du
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China.
| |
Collapse
|
21
|
Endothelial and Vascular Smooth Muscle Dysfunction in Hypertension. Biochem Pharmacol 2022; 205:115263. [PMID: 36174768 DOI: 10.1016/j.bcp.2022.115263] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 12/11/2022]
Abstract
The development of essential hypertension involves several factors. Vascular dysfunction, characterized by endothelial dysfunction, low-grade inflammation and structural remodeling, plays an important role in the initiation and maintenance of essential hypertension. Although the mechanistic pathways by which essential hypertension develops are poorly understood, several pharmacological classes available on the clinical settings improve blood pressure by interfering in the cardiac output and/or vascular function. This review is divided in two major sections. The first section depicts the major molecular pathways as renin angiotensin aldosterone system (RAAS), endothelin, nitric oxide signalling pathway and oxidative stress in the development of vascular dysfunction. The second section describes the role of some pharmacological classes such as i) RAAS inhibitors, ii) dual angiotensin receptor-neprilysin inhibitors, iii) endothelin-1 receptor antagonists, iv) soluble guanylate cyclase modulators, v) phosphodiesterase type 5 inhibitors and vi) sodium-glucose cotransporter 2 inhibitors in the context of hypertension. Some classes are already approved in the treatment of hypertension, but others are not yet approved. However, due to their potential benefits these classes were included.
Collapse
|
22
|
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
|
23
|
Wei H, Wei J, Zhang S, Dong S, Li G, Ran W, Dong C, Zhang W, Che C, Luo W, Xu H, Dong Z, Wang J, Wang L. Easily automated radiosynthesis of [18F]P10A-1910 and its clinical translation to quantify phosphodiesterase 10A in human brain. Front Bioeng Biotechnol 2022; 10:983488. [PMID: 36147528 PMCID: PMC9486304 DOI: 10.3389/fbioe.2022.983488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Our previous work showed that [18F]P10A-1910 was a potential radioligand for use in imaging phosphodiesterase 10A (PDE10A). Specifically, it had high brain penetration and specific binding that was demonstrated in both rodents and non-human primates. Here, we present the first automatic cGMP-level production of [18F]P10A-1910 and translational PET/MRI study in living human brains. Successful one-step radiolabeling of [18F]P10A-1910 on a GE TRACERlab FX2N synthesis module was realized via two different methods. First, formulated [18F]P10A-1910 was derived from heating spirocyclic iodonium ylide in a tetra-n-butyl ammonium methanesulfonate solution. At the end of synthesis, it was obtained in non-decay corrected radiochemical yields (n.d.c. RCYs) of 12.4 ± 1.3%, with molar activities (MAs) of 90.3 ± 12.6 μmol (n = 7) (Method I). The boronic pinacol ester combined with copper and oxygen also delivered the radioligand with 16.8 ± 1.0% n. d.c. RCYs and 77.3 ± 20.7 GBq/μmol (n = 7) MAs after formulation (Method II). The radiochemical purity, radionuclidic purity, solvent residue, sterility, endotoxin content and other parameters were all validated for human use. Consistent with the distribution of PDE10A in the brain, escalating uptake of [18F]P10A-1910 was observed in the order of cerebellum (reference region), substantial nigra, caudate and putamen. The non-displaceable binding potential (BPND) was estimated by simplified reference-tissue model (SRTM); linear regressions demonstrated that BPND was well correlated with the most widely used semiquantitative parameter SUV. The strongest correlation was observed with SUV(50–60 min) (R2 = 0.966, p < 0.01). Collectively, these results indicated that a static scan protocol could be easily performed for PET imaging of PDE10A. Most importantly, that [18F]P10A-1910 is a promising radioligand to clinically quantify PDE10A.
Collapse
Affiliation(s)
- Huiyi Wei
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Junjie Wei
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Shaojuan Zhang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Shiliang Dong
- Center of Bariatric Surgery, Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Guocong Li
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Wenqing Ran
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Chenchen Dong
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Weibin Zhang
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Chao Che
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Wenzhao Luo
- Institute of Analysis, Guangdong Academy of Sciences (China National Analytical Center), Guangzhou, China
| | - Hao Xu
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zhiyong Dong
- Center of Bariatric Surgery, Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital of Jinan University, Guangzhou, China
- *Correspondence: Lu Wang, ; Jinghao Wang, ; Zhiyong Dong,
| | - Jinghao Wang
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Department of Pharmacy, The First Affiliated Hospital of Jinan University, Guangzhou, China
- *Correspondence: Lu Wang, ; Jinghao Wang, ; Zhiyong Dong,
| | - Lu Wang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital of Jinan University, Guangzhou, China
- *Correspondence: Lu Wang, ; Jinghao Wang, ; Zhiyong Dong,
| |
Collapse
|
24
|
Sapuleni J, Szymanska M, Meidan R. Diverse actions of sirtuin-1 on ovulatory genes and cell death pathways in human granulosa cells. Reprod Biol Endocrinol 2022; 20:104. [PMID: 35840944 PMCID: PMC9284863 DOI: 10.1186/s12958-022-00970-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 06/22/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Human granulosa-lutein cells (hGLCs) amply express sirtuin-1 (SIRT1), a NAD + -dependent deacetylase that is associated with various cellular functions. SIRT1 was shown to elevate cAMP on its own and additively with human chorionic gonadotropin (hCG), it is therefore interesting to examine if SIRT1 affects other essential hGLC functions. METHODS Primary hGLCs, obtained from the follicular aspirates of women undergoing IVF and SV40-transfected, immortalized hGLCs (SVOG cells), were used. Primary cells were treated with SIRT1 specific activator SRT2104, as well as hCG or their combination. Additionally, siRNA-targeting SIRT1 construct was used to silence endogenous SIRT1 in SVOG cells. PTGS2, EREG, VEGFA and FGF2 expression was determined using quantitative polymerase chain reaction (qPCR). Apoptotic and necroptotic proteins were determined by specific antibodies in western blotting. Cell viability/apoptosis was determined by the XTT and flow cytometry analyses. Data were analyzed using student t-test or Mann-Whitney U test or one-way ANOVA followed by Tukey HSD post hoc test. RESULTS In primary and immortalized hGLCs, SRT2104 significantly upregulated key ovulatory and angiogenic genes: PTGS2, EREG, FGF2 and VEGFA, these effects tended to be further augmented in the presence of hCG. Additionally, SRT2104 dose and time-dependently decreased viable cell numbers. Flow cytometry of Annexin V stained cells confirmed that SIRT1 reduced live cell numbers and increased late apoptotic and necrotic cells. Moreover, we found that SIRT1 markedly reduced anti-apoptotic BCL-XL and MCL1 protein levels and increased cleaved forms of pro-apoptotic proteins caspase-3 and PARP. SIRT1 also significantly induced necroptotic proteins RIPK1 and MLKL. RIPK1 inhibitor, necrostatin-1 mitigated SIRT1 actions on RIPK1 and MLKL but also on cleaved caspase-3 and PARP and in accordance on live and apoptotic cells, implying a role for RIPK1 in SIRT1-induced cell death. SIRT1 silencing produced inverse effects on sorted cell populations, anti-apoptotic, pro-apoptotic and necroptotic proteins, corroborating SIRT1 activation. CONCLUSIONS These findings reveal that in hGLCs, SIRT1 enhances the expression of ovulatory and angiogenic genes while eventually advancing cell death pathways. Interestingly, these seemingly contradictory events may have occurred in a cAMP-dependent manner.
Collapse
Affiliation(s)
- Jackson Sapuleni
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, 761001, Rehovot, Israel
| | - Magdalena Szymanska
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, 761001, Rehovot, Israel
- Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Rina Meidan
- Department of Animal Sciences, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, 761001, Rehovot, Israel.
| |
Collapse
|
25
|
Yuan F, Ren H, Tan W, Wang Y, Luo H. Effect of phosphodiesterase-4 inhibitor rolipram on colonic hypermotility in water avoidance stress rat model. Neurogastroenterol Motil 2022; 34:e14317. [PMID: 35037375 PMCID: PMC9286810 DOI: 10.1111/nmo.14317] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 11/01/2021] [Accepted: 12/20/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Phosphodiesterase (PDE) inhibition has been reported to play a role in regulating gut motility, but the evidence is insufficient, and the mechanism remains unknown. The aim of this study was to investigate the possible role of phosphodiesterase-4 (PDE4) inhibitor rolipram in water avoidance stress-induced colonic hypermotility. METHODS A rat model of irritable bowel syndrome (IBS) with diarrhea (IBS-D) was established by water avoidance stress (WAS). Intestinal motility was assessed by fecal pellets expulsion per hour. The cyclic adenosine monophosphate (cAMP) and nitric oxide (NO) level in colon tissue were detected using ELISA assay and the Griess test, respectively. Western blotting was performed to assess the protein level of PDE, PKA/p-CREB, and neuronal nitric oxide synthase (nNOS) in the colon. To determine the role of rolipram in gut motility, the rats of the WAS + Rolipram and Rolipram group were injected with rolipram intraperitoneally. The colonic contractile activity was recorded with a RM6240 multichannel physiological signal system. KEY RESULTS WAS-induced gastrointestinal hypermotility and increased defecation in rats. After repeated stress, protein levels of PDE4 in the colon were promoted while PKA/p-CREB and nNOS were highly decreased. cAMP content in colon tissue did not change significantly. However, NO content decreased after WAS, and rolipram partly enhanced NO in WAS-exposed rats. In addition, intraperitoneal injection of rolipram partly inhibited the colonic motility in vivo. Meanwhile, we observed rolipram inhibited the contraction of colonic smooth muscle strips, and this inhibitory effect was abolished by Nω-Nitro-L-arginine (L-NNA), a nitric oxide synthase (NOS) inhibitor, tetrodotoxin (TTX), a blocker of neuronal voltage-dependent Na+ channels, Rp-Adenosine 3',5'-cyclic monophosphorothioate triethylammonium salt hydrate (Rp-cAMPS), an antagonist of cAMP. CONCLUSIONS AND INFERENCES Rolipram could relieve stress-induced gastrointestinal hypermotility. This effect may be partly through the cAMP-PKA-p-CREB pathway and NO pathway.
Collapse
Affiliation(s)
- FangTing Yuan
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanHubei ProvinceChina
| | - HaiXia Ren
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanHubei ProvinceChina
| | - Wei Tan
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanHubei ProvinceChina
| | - Ying Wang
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanHubei ProvinceChina,Key Laboratory of Hubei Province for Digestive System DiseasesRenmin Hospital of Wuhan UniversityWuhanHubei ProvinceChina
| | - HeSheng Luo
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanHubei ProvinceChina
| |
Collapse
|
26
|
Huang MX, Tian YJ, Han C, Liu RD, Xie X, Yuan Y, Yang YY, Li Z, Chen J, Luo HB, Wu Y. Structural Modifications of Nimodipine Lead to Novel PDE1 Inhibitors with Anti-pulmonary Fibrosis Effects. J Med Chem 2022; 65:8444-8455. [PMID: 35666471 DOI: 10.1021/acs.jmedchem.2c00458] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Our previous research demonstrated that phosphodiesterase-1 (PDE1) could work as a potential target against idiopathic pulmonary fibrosis. Nimodipine, a calcium antagonist commonly used to improve hypertension, was reported to have inhibition against PDE1. Herein, a series of nimodipine analogues were discovered as novel selective and potent PDE1 inhibitors after structural modifications. Compound 2g exhibited excellent inhibitory activity against PDE1C (IC50 = 10 nM), high selectivity over other PDEs except for PDE4, and weak calcium channel antagonistic activity. Administration of compound 2g exhibited remarkable therapeutic effects in a rat model of pulmonary fibrosis induced by bleomycin and prevented myofibroblast differentiation induced by TGF-β1. The expressions of PDE1B and PDE1C were found to be increased and concentrated in the focus of fibrosis. Compound 2g increased the levels of 3',5'-cyclic adenosine monophosphate (cAMP) and 3',5'-cyclic guanosine monophosphate (cGMP) in the lungs of rats with pulmonary fibrosis, supporting the fact that the anti-fibrosis effects of 2g were through the regulation of cAMP and cGMP.
Collapse
Affiliation(s)
- Meng-Xing Huang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yi-Jing Tian
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Chuan Han
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Run-Duo Liu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xi Xie
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan 570228, China
| | - Yijun Yuan
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan 570228, China
| | - Yi-Yi Yang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhe Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jianwen Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Hai-Bin Luo
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.,Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Life and Pharmaceutical Sciences, Hainan University, Haikou, Hainan 570228, China.,Song Li' Academician Workstation of Hainan University (School of Pharmaceutical Sciences), Yazhou Bay, Sanya 572000, China
| | - Yinuo Wu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
27
|
Gomes DA, Joubert AM, Visagie MH. In Vitro Effects of Papaverine on Cell Migration and Vascular Endothelial Growth Factor in Cancer Cell Lines. Int J Mol Sci 2022; 23:4654. [PMID: 35563045 PMCID: PMC9104338 DOI: 10.3390/ijms23094654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 04/21/2022] [Accepted: 04/21/2022] [Indexed: 02/04/2023] Open
Abstract
Papaverine (PPV) is a benzylisoquinoline alkaloid isolated from Papaver somniferum that exerts antiproliferative activity. However, several questions remain regarding the biochemical pathways affected by PPV in tumourigenic cells. In this study, the influence of PPV on cell migration (light microscopy), expression of vascular endothelial growth factor (VEGF) B, VEGF R1, VEGF R2, and phosphorylated focal adhesion kinase (pFAK) were investigated using spectrophotometry in MDA-MB-231-, A549- and DU145 cell lines. The migration assay revealed that, after 48 h, PPV (100 µM) reduced cell migration to 81%, 91%, and 71% in MDA-MB-231-, A549-, and DU145 cells, respectively. VEGF B expression was reduced to 0.79-, 0.71-, and 0.73-fold after 48 h of exposure to PPV in MDA-MB-231-, A549- and DU145 cells, while PPV exposure of 48 h increased VEGF R1 expression in MDA-MB-231- and DU145 cells to 1.38 and 1.46. A fold decrease in VEGF R1 expression was observed in A549 cells to 0.90 after exposure to 150 µM. No statistically significant effects were observed on VEGF R2- and FAK expression after exposure to PPV. This study contributes to the understanding of the effects of a phytomedicinal alkaloid compound in cancer cells and may provide novel approaches to the application of non-addictive alkaloids.
Collapse
Affiliation(s)
| | | | - Michelle Helen Visagie
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Gezina, Pretoria 0031, South Africa; (D.A.G.); (A.M.J.)
| |
Collapse
|
28
|
Xiao Z, Wei H, Xu Y, Haider A, Wei J, Yuan S, Rong J, Zhao C, Li G, Zhang W, Chen H, Li Y, Zhang L, Sun J, Zhang S, Luo HB, Yan S, Cai Q, Hou L, Che C, Liang SH, Wang L. Discovery of a highly specific 18F-labeled PET ligand for phosphodiesterase 10A enabled by novel spirocyclic iodonium ylide radiofluorination. Acta Pharm Sin B 2022; 12:1963-1975. [PMID: 35847497 PMCID: PMC9279629 DOI: 10.1016/j.apsb.2021.11.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/30/2021] [Accepted: 10/20/2021] [Indexed: 12/14/2022] Open
Abstract
As a member of cyclic nucleotide phosphodiesterase (PDE) enzyme family, PDE10A is in charge of the degradation of cyclic adenosine (cAMP) and guanosine monophosphates (cGMP). While PDE10A is primarily expressed in the medium spiny neurons of the striatum, it has been implicated in a variety of neurological disorders. Indeed, inhibition of PDE10A has proven to be of potential use for the treatment of central nervous system (CNS) pathologies caused by dysfunction of the basal ganglia–of which the striatum constitutes the largest component. A PDE10A-targeted positron emission tomography (PET) radioligand would enable a better assessment of the pathophysiologic role of PDE10A, as well as confirm the relationship between target occupancy and administrated dose of a given drug candidate, thus accelerating the development of effective PDE10A inhibitors. In this study, we designed and synthesized a novel 18F-aryl PDE10A PET radioligand, codenamed [18F]P10A-1910 ([18F]9), in high radiochemical yield and molar activity via spirocyclic iodonium ylide-mediated radiofluorination. [18F]9 possessed good in vitro binding affinity (IC50 = 2.1 nmol/L) and selectivity towards PDE10A. Further, [18F]9 exhibited reasonable lipophilicity (logD = 3.50) and brain permeability (Papp > 10 × 10−6 cm/s in MDCK-MDR1 cells). PET imaging studies of [18F]9 revealed high striatal uptake and excellent in vivo specificity with reversible tracer kinetics. Preclinical studies in rodents revealed an improved plasma and brain stability of [18F]9 when compared to the current reference standard for PDE10A-targeted PET, [18F]MNI659. Further, dose–response experiments with a series of escalating doses of PDE10A inhibitor 1 in rhesus monkey brains confirmed the utility of [18F]9 for evaluating target occupancy in vivo in higher species. In conclusion, our results indicated that [18F]9 is a promising PDE10A PET radioligand for clinical translation.
Collapse
Affiliation(s)
- Zhiwei Xiao
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| | - Huiyi Wei
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Yi Xu
- Department of Cardiology, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Ahmed Haider
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| | - Junjie Wei
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Shiyu Yuan
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Jian Rong
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| | - Chunyu Zhao
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| | - Guocong Li
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Weibin Zhang
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Huangcan Chen
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Yuefeng Li
- Guangdong Landau Biotechnology Co. Ltd., Guangzhou 510555, China
| | - Lingling Zhang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Jiyun Sun
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| | - Shaojuan Zhang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Hai-Bin Luo
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Sen Yan
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou 510632, China
| | - Qijun Cai
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Lu Hou
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Chao Che
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Corresponding authors. Tel./fax: +86 755 26032530 (Chao Che), +1 617 7266165 (Steven H. Liang), +86 20 38688692 (Lu Wang).
| | - Steven H. Liang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
- Corresponding authors. Tel./fax: +86 755 26032530 (Chao Che), +1 617 7266165 (Steven H. Liang), +86 20 38688692 (Lu Wang).
| | - Lu Wang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
- Corresponding authors. Tel./fax: +86 755 26032530 (Chao Che), +1 617 7266165 (Steven H. Liang), +86 20 38688692 (Lu Wang).
| |
Collapse
|
29
|
Ostrom KF, LaVigne JE, Brust TF, Seifert R, Dessauer CW, Watts VJ, Ostrom RS. Physiological roles of mammalian transmembrane adenylyl cyclase isoforms. Physiol Rev 2022; 102:815-857. [PMID: 34698552 PMCID: PMC8759965 DOI: 10.1152/physrev.00013.2021] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 09/20/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022] Open
Abstract
Adenylyl cyclases (ACs) catalyze the conversion of ATP to the ubiquitous second messenger cAMP. Mammals possess nine isoforms of transmembrane ACs, dubbed AC1-9, that serve as major effector enzymes of G protein-coupled receptors (GPCRs). The transmembrane ACs display varying expression patterns across tissues, giving the potential for them to have a wide array of physiological roles. Cells express multiple AC isoforms, implying that ACs have redundant functions. Furthermore, all transmembrane ACs are activated by Gαs, so it was long assumed that all ACs are activated by Gαs-coupled GPCRs. AC isoforms partition to different microdomains of the plasma membrane and form prearranged signaling complexes with specific GPCRs that contribute to cAMP signaling compartments. This compartmentation allows for a diversity of cellular and physiological responses by enabling unique signaling events to be triggered by different pools of cAMP. Isoform-specific pharmacological activators or inhibitors are lacking for most ACs, making knockdown and overexpression the primary tools for examining the physiological roles of a given isoform. Much progress has been made in understanding the physiological effects mediated through individual transmembrane ACs. GPCR-AC-cAMP signaling pathways play significant roles in regulating functions of every cell and tissue, so understanding each AC isoform's role holds potential for uncovering new approaches for treating a vast array of pathophysiological conditions.
Collapse
Affiliation(s)
| | - Justin E LaVigne
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana
| | - Tarsis F Brust
- Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, Palm Beach Atlantic University, West Palm Beach, Florida
| | - Roland Seifert
- Institute of Pharmacology, Hannover Medical School, Hannover, Germany
| | - Carmen W Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas
| | - Val J Watts
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana
- Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, Indiana
| | - Rennolds S Ostrom
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California
| |
Collapse
|
30
|
Structure-based discovery of orally efficient inhibitors via unique interactions with H-pocket of PDE8 for the treatment of vascular dementia. Acta Pharm Sin B 2022; 12:3103-3112. [PMID: 35865094 PMCID: PMC9293670 DOI: 10.1016/j.apsb.2022.02.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/20/2022] [Accepted: 02/12/2022] [Indexed: 11/23/2022] Open
Abstract
Our previous study demonstrated that phosphodiesterase 8 (PDE8) could work as a potential target for vascular dementia (VaD) using a chemical probe 3a. However, compound 3a is a chiral compound which was obtained by chiral resolution on HPLC, restricting its usage in clinic. Herein, a series of non-chiral 9-benzyl-2-chloro-adenine derivatives were discovered as novel PDE8 inhibitors. Lead 15 exhibited potent inhibitory activity against PDE8A (IC50 = 11 nmol/L), high selectivity over other PDEs, and remarkable drug-like properties (worthy to mention is that its bioavailability was up to 100%). Oral administration of 15 significantly improved the cAMP level of the right brain and exhibited dose-dependent effects on cognitive improvement in a VaD mouse model. Notably, the X-ray crystal structure of the PDE8A–15 complex showed that the potent affinity and high selectivity of 15 might come from the distinctive interactions with H-pocket including T-shaped π–π interactions with Phe785 as well as a unique H-bond network, which have never been observed in other PDE−inhibitor complex before, providing new strategies for the further rational design of novel selective inhibitors against PDE8.
Collapse
|
31
|
Yan B, Ding Z, Zhang W, Cai G, Han H, Ma Y, Cao Y, Wang J, Chen S, Ai Y. Multiple PDE3A modulators act as molecular glues promoting PDE3A-SLFN12 interaction and induce SLFN12 dephosphorylation and cell death. Cell Chem Biol 2022; 29:958-969.e5. [PMID: 35104454 DOI: 10.1016/j.chembiol.2022.01.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 10/30/2021] [Accepted: 01/06/2022] [Indexed: 12/20/2022]
Abstract
The canonical function of phosphodiesterase 3A (PDE3A) is to hydrolyze the phosphodiester bonds in second messenger molecules, such as cyclic AMP (cAMP) and cyclic guanosine monophosphate (cGMP). Recently, a phosphodiesterase-activity-independent role for PDE3A was reported. In this noncanonical function, PDE3A physically interacts with Schlafen 12 (SLFN12) upon treatment of cells with cytotoxic PDE3A modulators. Here, we confirmed that the cytotoxic PDE3A modulators act as molecular glues to initiate the association of PDE3A and SLFN12. The PDE3A-SLFN12 interaction increases the protein stability of SLFN12 located in the cytoplasm, while at the same time also inducing SLFN12 dephosphorylation (including serines 368 and 573). Mutational analysis demonstrates that dephosphorylation is required for cell death induced by cytotoxic PDE3A modulators. Finally, we found that dephosphorylation promoted the rRNA RNase activity of SLFN12 and show that this nucleolytic activity is essential for SLFN12's cell-death-inducing function. Thus, our study deepens the understanding of the biochemical mechanisms underlying SLFN12-mediated cell death.
Collapse
Affiliation(s)
- Bo Yan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China; National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, People's Republic of China
| | - Zhangcheng Ding
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, People's Republic of China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 100871, People's Republic of China
| | - Wenbin Zhang
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, People's Republic of China; School of Life Sciences, Peking University, Beijing 100871, People's Republic of China
| | - Gaihong Cai
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, People's Republic of China
| | - Hui Han
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, People's Republic of China
| | - Yan Ma
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, People's Republic of China
| | - Yang Cao
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, People's Republic of China
| | - Jiawen Wang
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, People's Republic of China
| | - She Chen
- National Institute of Biological Sciences, 7 Science Park Road, Zhongguancun Life Science Park, Beijing 102206, People's Republic of China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 100871, People's Republic of China
| | - Youwei Ai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, People's Republic of China.
| |
Collapse
|
32
|
Fan T, Hou Y, Ge W, Fan T, Feng X, Guo W, Song X, Gao R, Wang J. Phosphodiesterase 4D promotes angiotensin II-induced hypertension in mice via smooth muscle cell contraction. Commun Biol 2022; 5:81. [PMID: 35058564 PMCID: PMC8776755 DOI: 10.1038/s42003-022-03029-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 12/23/2021] [Indexed: 11/09/2022] Open
Abstract
AbstractHypertension is a common chronic disease, which leads to cardio-cerebrovascular diseases, and its prevalence is increasing. The cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) pathway participates in multiple cardiovascular diseases. Phosphodiesterase (PDE) 4 has been shown to regulate PKA activity via cAMP specific hydrolysis. However, whether PDE4-cAMP-PKA pathway influences hypertension remains unknown. Herein, we reveal that PDE4D (one of PDE4 isoforms) expression is upregulated in the aortas of experimental hypertension induced by angiotensin II (Ang II). Furthermore, knockout of Pde4d in mouse smooth muscle cells (SMCs) attenuates Ang II-induced hypertension, arterial wall media thickening, vascular fibrosis and vasocontraction. Additionally, we find that PDE4D deficiency activates PKA-AMP-activated protein kinase (AMPK) signaling pathway to inhibit myosin phosphatase targeting subunit 1 (MYPT1)-myosin light chain (MLC) phosphorylation, relieving Ang II-induced SMC contraction in vitro and in vivo. Our results also indicate that rolipram, a PDE4 inhibitor, may be a potential drug for hypertension therapy.
Collapse
|
33
|
Le ML, Jiang MY, Han C, Yang YY, Wu Y. PDE1 inhibitors: a review of the recent patent literature (2008-present). Expert Opin Ther Pat 2022; 32:423-439. [PMID: 35016587 DOI: 10.1080/13543776.2022.2027910] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
INTRODUCTION : PDE1 has been demonstrated to be a potential drug target for a variety of diseases, such as Alzheimer's disease and cardiovascular disease. In the past decades, numerous PDE1 inhibitors with structural diversities have been developed and patented by pharmaceutical companies, providing drug candidates for exploring novel disease indications of PDE1. AREA COVERED : This review aims to provide an overview of PDE1 inhibitors reported in patents from 2008 to present. EXPERT OPINION : Among current PDE1 inhibitors, only a few of them showed high selectivity over other PDEs, which might cause severe side effects in clinic. The development of highly selective PDE1 inhibitors is still the "top priority" in the following research. The selective recognition mechanism of PDE1 with inhibitors should be further elucidated by X-ray crystallography in order to provide evidences for the rational design of selective PDE1 inhibitors. In addition, PDE1 inhibitors should be applied in the different clinical indications beyond CNS diseases.
Collapse
Affiliation(s)
- Mei-Ling Le
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Mei-Yan Jiang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Chuan Han
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Yi-Yi Yang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Yinuo Wu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, P. R. China
| |
Collapse
|
34
|
PDE-Mediated Cyclic Nucleotide Compartmentation in Vascular Smooth Muscle Cells: From Basic to a Clinical Perspective. J Cardiovasc Dev Dis 2021; 9:jcdd9010004. [PMID: 35050214 PMCID: PMC8777754 DOI: 10.3390/jcdd9010004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular diseases are important causes of mortality and morbidity worldwide. Vascular smooth muscle cells (SMCs) are major components of blood vessels and are involved in physiologic and pathophysiologic conditions. In healthy vessels, vascular SMCs contribute to vasotone and regulate blood flow by cyclic nucleotide intracellular pathways. However, vascular SMCs lose their contractile phenotype under pathological conditions and alter contractility or signalling mechanisms, including cyclic nucleotide compartmentation. In the present review, we focus on compartmentalized signaling of cyclic nucleotides in vascular smooth muscle. A deeper understanding of these mechanisms clarifies the most relevant axes for the regulation of vascular tone. Furthermore, this allows the detection of possible changes associated with pathological processes, which may be of help for the discovery of novel drugs.
Collapse
|
35
|
Giesen J, Mergia E, Koesling D, Russwurm M. Hippocampal AMPA- and NMDA-induced cGMP signals are mainly generated by NO-GC2 and are under tight control by PDEs 1 and 2. Eur J Neurosci 2021; 55:18-31. [PMID: 34902209 DOI: 10.1111/ejn.15564] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 11/29/2021] [Accepted: 12/07/2021] [Indexed: 11/30/2022]
Abstract
In the central nervous system, the nitric oxide (NO)/cyclic guanosine monophosphate (cGMP) signalling cascade has an established role in fine-tuning of synaptic transmission. In the present study, we asked which isoform of NO-sensitive guanylyl cyclase, NO-GC1 or NO-GC2, is responsible for generation of N-methyl-d-aspartate (NMDA)- and AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid)-induced cGMP signals and which of the phosphodiesterases (PDEs) is responsible for degradation. To this end, we performed live cell fluorescence measurements of primary hippocampal neurons isolated from NO-GC isoform-deficient mice. Although both isoforms contributed to the NMDA- and AMPA-induced cGMP signals, NO-GC2 clearly played the predominant role. Whereas under PDE-inhibiting conditions the cGMP levels elicited by both glutamatergic ligands were comparable, NMDA-induced cGMP signals were clearly higher than the AMPA-induced ones in the absence of PDE inhibitors. Thus, AMPA-induced cGMP signals are more tightly controlled by PDE-mediated degradation than NMDA-induced signals. In addition, these findings are compatible with the existence of at least two different pools of cGMP in both of which PDE1 and PDE2-known to be highly expressed in the hippocampus-are mainly responsible for cGMP degradation. The finding that distinct pools of cGMP are equipped with different amounts of PDEs highlights the importance of PDEs for the shape of NO-induced cGMP signals in the central nervous system.
Collapse
Affiliation(s)
- Jan Giesen
- Institute of Pharmacology and Toxicology, Ruhr-University Bochum, Bochum, Germany
| | - Evanthia Mergia
- Institute of Pharmacology and Toxicology, Ruhr-University Bochum, Bochum, Germany
| | - Doris Koesling
- Institute of Pharmacology and Toxicology, Ruhr-University Bochum, Bochum, Germany
| | - Michael Russwurm
- Institute of Pharmacology and Toxicology, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
36
|
Staicu FD, Martínez-Soto JC, Canovas S, Matás C. Nitric oxide-targeted protein phosphorylation during human sperm capacitation. Sci Rep 2021; 11:20979. [PMID: 34697378 PMCID: PMC8546126 DOI: 10.1038/s41598-021-00494-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 10/07/2021] [Indexed: 12/25/2022] Open
Abstract
Among many other molecules, nitric oxide insures the correct progress of sperm capacitation by mediating phosphorylation events. For a more comprehensive understanding of how this happens, we capacitated human spermatozoa from healthy men in the presence/absence of S-Nitrosoglutathione, a nitric oxide donor, two nitric oxide synthase inhibitors, NG-Nitro-l-arginine Methyl Ester Hydrochloride and Aminoguanidine Hemisulfate salt and, finally, with/without l-Arginine, the substrate for nitric oxide synthesis, and/or human follicular fluid. When analyzing the phosphorylation of protein kinase A substrates and tyrosine residues, we particularly observed how the inhibition of nitric oxide synthesis affects certain protein bands (~ 110, ~ 87, ~ 75 and ~ 62 kD) by lowering their phosphorylation degree, even when spermatozoa were incubated with l-Arginine and/or follicular fluid. Mass spectrometry analysis identified 29 proteins in these species, related to: spermatogenesis, binding to the zona pellucida, energy and metabolism, stress response, motility and structural organization, signaling and protein turnover. Significant changes in the phosphorylation degree of specific proteins could impair their biological activity and result in severe fertility-related phenotypes. These findings provide a deeper understanding of nitric oxide’s role in the capacitation process, and consequently, future studies in infertile patients should determine how nitric oxide mediates phosphorylation events in the species here described.
Collapse
Affiliation(s)
- Florentin-Daniel Staicu
- Department of Physiology, Veterinary Faculty, University of Murcia, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), Calle Campus Universitario, 11, 30100, Murcia, Spain.,Institute for Biomedical Research of Murcia (IMIB), Murcia, Spain
| | | | - Sebastian Canovas
- Institute for Biomedical Research of Murcia (IMIB), Murcia, Spain.,Department of Physiology, Nursery Faculty, University of Murcia, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), Murcia, Spain
| | - Carmen Matás
- Department of Physiology, Veterinary Faculty, University of Murcia, International Excellence Campus for Higher Education and Research (Campus Mare Nostrum), Calle Campus Universitario, 11, 30100, Murcia, Spain. .,Institute for Biomedical Research of Murcia (IMIB), Murcia, Spain.
| |
Collapse
|
37
|
Zhao C, Mo L, Lei T, Yan Y, Han S, Miao J, Gao Y, Wang X, Zhao W, Huang C. miR-5701 promoted apoptosis of clear cell renal cell carcinoma cells by targeting phosphodiesterase-1B. Anticancer Drugs 2021; 32:855-863. [PMID: 33929990 DOI: 10.1097/cad.0000000000001078] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Increasing evidence has demonstrated that microRNAs play critical roles in malignant biological behaviors, including cancerogenesis, cancer progression and metastasis, through the regulation of target genes expression. As miR-5701 has recently been identified to play roles as tumor suppressor miRNA in the development of some kinds of cancers, in this study we sought to investigate the role of miR-5701 in clear cell renal cell carcinoma (ccRCC). Colony formation, cell apoptosis and proliferation assays were employed, and the results showed that miR-5701 inhibited proliferation and promoted apoptosis of ccRCC cells. Western blotting and dual-luciferase reporter assays were used to confirm that PDE1B is a new direct target of miR-5701. Furthermore, overexpression of PDE1B attenuated the effects of miR-5701, indicating that miR-5701 inhibited proliferation and promoted apoptosis of ccRCC cells via targeting PDE1B. Taken together, the data presented here indicate that t miR-5701 is a tumor suppressor in ccRCC and PDE1B is a new target of miR-5701.
Collapse
Affiliation(s)
- Changan Zhao
- Department of Pathology, School of Basic Medical Sciences
- Institute of Genetics and Developmental Biology, Xi'an Jiaotong University Health Science Center
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University
| | - Liping Mo
- Department of Pathology, School of Basic Medical Sciences
| | - Ting Lei
- Department of Pathology, School of Basic Medical Sciences
| | - Yan Yan
- Department of Pathology, The First Hospital of Xi'an
| | - Shuiping Han
- Department of Pathology, School of Basic Medical Sciences
| | - Jiyu Miao
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University
| | - Yi Gao
- Department of Cell Biology and Genetics, Medical College of Yan'an University, Yan'an
| | - Xiaofei Wang
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University
| | - Wenbao Zhao
- Department of Pathology, School of Basic Medical Sciences
| | - Chen Huang
- Institute of Genetics and Developmental Biology, Xi'an Jiaotong University Health Science Center
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an Jiaotong University
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| |
Collapse
|
38
|
Tan JS, Liu NN, Guo TT, Hu S, Hua L. Genetic predisposition to COVID-19 may increase the risk of hypertension disorders in pregnancy: A two-sample Mendelian randomization study. Pregnancy Hypertens 2021; 26:17-23. [PMID: 34428710 DOI: 10.1016/j.preghy.2021.08.112] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/23/2021] [Accepted: 08/12/2021] [Indexed: 11/16/2022]
Abstract
AIMS The aim of this study was to apply the Mendelian randomization (MR) design to explore the potential causal association between COVID-19 and the risk of hypertension disorders in pregnancy. METHODS Our primary genetic instrument comprised 8 single-nucleotide polymorphisms (SNPs) associated with COVID-19 at genome-wide significance. Data on the associations between the SNPs and the risk of hypertension disorders in pregnancy were obtained from study based on a very large cohort of European population. The random-effects inverse-variance weighted method was conducted for the main analyses, with a complementary analysis of the weighted median and MR-Egger approaches. RESULTS Using IVW, we found that genetically predicted COVID-19 was significantly positively associated with hypertension disorders in pregnancy, with an odds ratio (OR) of 1.111 [95% confidence interval (CI) 1.042-1.184; P = 0.001]. Weighted median regression also showed directionally similar estimates [OR 1.098 (95% CI, 1.013-1.190), P = 0.023]. Both funnel plots and MR-Egger intercepts suggest no directional pleiotropic effects observed. CONCLUSIONS Our findings provide direct evidence that there is a shared genetic predisposition so that patients infected with COVID-19 may be causally associated with increased risk of hypertension disorders in pregnancy.
Collapse
Affiliation(s)
- Jiang-Shan Tan
- Thrombosis Center, National Clinical Research Center of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Ning-Ning Liu
- Peking University Sixth Hospital/Institute of Mental Health, Beijing 100191, China; NHC Key Laboratory of Mental Health (Peking University), National Clinical Research Center for Mental Disorders (Peking University Sixth Hospital), Beijing 100191, China
| | - Ting-Ting Guo
- Thrombosis Center, National Clinical Research Center of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Song Hu
- Thrombosis Center, National Clinical Research Center of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China
| | - Lu Hua
- Thrombosis Center, National Clinical Research Center of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100037, China.
| |
Collapse
|
39
|
Hanisak J, Soriano A, Adam GC, Basso A, Bauman D, Bell D, Frank E, O’Donnell G, Tawa P, Verras A, Yu Y, Zhang L, Seganish WM. Discovery of the First Non-cGMP Mimetic Small Molecule Activators of cGMP-Dependent Protein Kinase 1 α (PKG1α). ACS Med Chem Lett 2021; 12:1275-1282. [PMID: 34413956 DOI: 10.1021/acsmedchemlett.1c00264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 07/08/2021] [Indexed: 12/21/2022] Open
Abstract
PKG1α is a central node in cGMP signaling. Current therapeutics that look to activate this pathway rely on elevation of cGMP levels and subsequent activation of PKG1α. Direct activation of PKG1α could potentially drive additional efficacy without associated side effects of blanket cGMP elevation. We undertook a high-throughput screen to identify novel activators. After triaging through numerous false positive hits, attributed to compound mediated oxidation and activation of PKG1α, a piperidine series of compounds was validated. The hit 1 was a weak activator with EC50 = 47 μM. The activity could be improved to single digit micromolar, as seen in compounds 21 and 25 (7.0 and 3.7 μM, respectively). Several compounds were tested in a pVASP cell-based assay, and for compounds with moderate permeability, good agreement was observed between the biochemical and functional assays. These compounds will function as efficient tools to further interrogate PKG1α biology.
Collapse
Affiliation(s)
- Jennifer Hanisak
- Discovery Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Aileen Soriano
- Mass Spectrometry and Biophysics, Computational and Structural Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Gregory C. Adam
- Quantitative Biosciences, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Andrea Basso
- Mass Spectrometry and Biophysics, Computational and Structural Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - David Bauman
- Discovery Biology, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - David Bell
- Mass Spectrometry and Biophysics, Computational and Structural Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Emily Frank
- Mass Spectrometry and Biophysics, Computational and Structural Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Gregory O’Donnell
- Quantitative Biosciences, Merck & Co., Inc, West Point, Pennsylvania 19486, United States
| | - Paul Tawa
- Mass Spectrometry and Biophysics, Computational and Structural Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Andreas Verras
- Computational and Structural Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Yang Yu
- Discovery Chemistry, Merck & Co., Inc., Kenilworth, New Jersey 07033, United States
| | - Lei Zhang
- Biologics Analytical Research and Development, Merck & Co., Inc., Kenilworth, New Jersey, 07033 United States
| | - W. Michael Seganish
- Discovery Chemistry, Merck & Co., Inc., South San Francisco, California 94080, United States
| |
Collapse
|
40
|
Phosphodiesterase 7(PDE7): A unique drug target for central nervous system diseases. Neuropharmacology 2021; 196:108694. [PMID: 34245775 DOI: 10.1016/j.neuropharm.2021.108694] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/11/2021] [Accepted: 06/29/2021] [Indexed: 12/19/2022]
Abstract
Phosphodiesterase 7 (PDE7), one of the 11 phosphodiesterase (PDE) families, specifically hydrolyzes cyclic 3', 5'-adenosine monophosphate (cAMP). PDE7 is involved in many important functional processes in physiology and pathology by regulating intracellular cAMP signaling. Studies have demonstrated that PDE7 is widely expressed in the central nervous system (CNS) and potentially related to pathogenesis of many CNS diseases. Here, we summarized the classification and distribution of PDE7 in the brain and its functional roles in the mediation of CNS diseases such as Parkinson's disease (PD), Alzheimer's disease (AD), multiple sclerosis (MS), and schizophrenia. It is expected that the findings collected here will not only lead to a better understanding of the mechanisms by which PDE7 mediates CNS function and diseases, but also aid in the development of novel drugs targeting PDE7 for treatment of CNS diseases.
Collapse
|
41
|
Paes D, Schepers M, Rombaut B, van den Hove D, Vanmierlo T, Prickaerts J. The Molecular Biology of Phosphodiesterase 4 Enzymes as Pharmacological Targets: An Interplay of Isoforms, Conformational States, and Inhibitors. Pharmacol Rev 2021; 73:1016-1049. [PMID: 34233947 DOI: 10.1124/pharmrev.120.000273] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The phosphodiesterase 4 (PDE4) enzyme family plays a pivotal role in regulating levels of the second messenger cAMP. Consequently, PDE4 inhibitors have been investigated as a therapeutic strategy to enhance cAMP signaling in a broad range of diseases, including several types of cancers, as well as in various neurologic, dermatological, and inflammatory diseases. Despite their widespread therapeutic potential, the progression of PDE4 inhibitors into the clinic has been hampered because of their related relatively small therapeutic window, which increases the chance of producing adverse side effects. Interestingly, the PDE4 enzyme family consists of several subtypes and isoforms that can be modified post-translationally or can engage in specific protein-protein interactions to yield a variety of conformational states. Inhibition of specific PDE4 subtypes, isoforms, or conformational states may lead to more precise effects and hence improve the safety profile of PDE4 inhibition. In this review, we provide an overview of the variety of PDE4 isoforms and how their activity and inhibition is influenced by post-translational modifications and interactions with partner proteins. Furthermore, we describe the importance of screening potential PDE4 inhibitors in view of different PDE4 subtypes, isoforms, and conformational states rather than testing compounds directed toward a specific PDE4 catalytic domain. Lastly, potential mechanisms underlying PDE4-mediated adverse effects are outlined. In this review, we illustrate that PDE4 inhibitors retain their therapeutic potential in myriad diseases, but target identification should be more precise to establish selective inhibition of disease-affected PDE4 isoforms while avoiding isoforms involved in adverse effects. SIGNIFICANCE STATEMENT: Although the PDE4 enzyme family is a therapeutic target in an extensive range of disorders, clinical use of PDE4 inhibitors has been hindered because of the adverse side effects. This review elaborately shows that safer and more effective PDE4 targeting is possible by characterizing 1) which PDE4 subtypes and isoforms exist, 2) how PDE4 isoforms can adopt specific conformations upon post-translational modifications and protein-protein interactions, and 3) which PDE4 inhibitors can selectively bind specific PDE4 subtypes, isoforms, and/or conformations.
Collapse
Affiliation(s)
- Dean Paes
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, EURON, Maastricht University, Maastricht, The Netherlands (D.P, M.S., B.R., D.v.d.H., T.V., J.P.); Department of Neuroscience, Neuro-Immune Connect and Repair laboratory, Biomedical Research Institute, Hasselt University, Hasselt, Belgium (D.P., M.S., B.R., T.V.); and Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany (D.v.d.H.)
| | - Melissa Schepers
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, EURON, Maastricht University, Maastricht, The Netherlands (D.P, M.S., B.R., D.v.d.H., T.V., J.P.); Department of Neuroscience, Neuro-Immune Connect and Repair laboratory, Biomedical Research Institute, Hasselt University, Hasselt, Belgium (D.P., M.S., B.R., T.V.); and Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany (D.v.d.H.)
| | - Ben Rombaut
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, EURON, Maastricht University, Maastricht, The Netherlands (D.P, M.S., B.R., D.v.d.H., T.V., J.P.); Department of Neuroscience, Neuro-Immune Connect and Repair laboratory, Biomedical Research Institute, Hasselt University, Hasselt, Belgium (D.P., M.S., B.R., T.V.); and Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany (D.v.d.H.)
| | - Daniel van den Hove
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, EURON, Maastricht University, Maastricht, The Netherlands (D.P, M.S., B.R., D.v.d.H., T.V., J.P.); Department of Neuroscience, Neuro-Immune Connect and Repair laboratory, Biomedical Research Institute, Hasselt University, Hasselt, Belgium (D.P., M.S., B.R., T.V.); and Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany (D.v.d.H.)
| | - Tim Vanmierlo
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, EURON, Maastricht University, Maastricht, The Netherlands (D.P, M.S., B.R., D.v.d.H., T.V., J.P.); Department of Neuroscience, Neuro-Immune Connect and Repair laboratory, Biomedical Research Institute, Hasselt University, Hasselt, Belgium (D.P., M.S., B.R., T.V.); and Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany (D.v.d.H.)
| | - Jos Prickaerts
- Department of Psychiatry & Neuropsychology, School for Mental Health and Neuroscience, EURON, Maastricht University, Maastricht, The Netherlands (D.P, M.S., B.R., D.v.d.H., T.V., J.P.); Department of Neuroscience, Neuro-Immune Connect and Repair laboratory, Biomedical Research Institute, Hasselt University, Hasselt, Belgium (D.P., M.S., B.R., T.V.); and Department of Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany (D.v.d.H.)
| |
Collapse
|
42
|
Mirone V, Napolitano L, D'Emmanuele di Villa Bianca R, Mitidieri E, Sorrentino R, Vanelli A, Vanacore D, Turnaturi C, La Rocca R, Celentano G, Arcaniolo D, Cirino G. A new original nutraceutical formulation ameliorates the effect of Tadalafil on clinical score and cGMP accumulation. ACTA ACUST UNITED AC 2021; 93:221-226. [PMID: 34286560 DOI: 10.4081/aiua.2021.2.221] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/05/2021] [Indexed: 11/23/2022]
Abstract
OBJECTIVE To assess the efficacy of the combination of Tadalafil 5 mg and nutritional supplements composed by Panax ginseng, Moringa Oleifera and Rutin on erectile function in men with mild and moderate vasculogenic ED. METHODS we prospectively enrolled 86 patients divided into two groups A (45), B (33) in this multicenter randomized, doubleblind, placebo-controlled trial . Drop out was 8 patients (3 patients in group A and 5 in Group B). At screening visit patients underwent clinical examination, blood test (hormonal and metabolic profile) and filled out the IIEF-5 questionnaire and the SEP-2, SEP-3. Patients were randomized by a computergenerated list to receive either Tadalafil 5 mg once daily plus nutritional supplement once daily (group A) or Tadalafil 5 mg plus placebo with the same administration schedule (group B) for 3 months. Blood samples, IIEF-5, SEP-2 and SEP-3 have been collected again after 3 months. cGMP was measured in platelets of 38 patients at baseline and after one months. RESULTS Mean age was 59.98 ± 6.90 (range 38-69), mean IIEF-5 score at baseline was 13.59 ± 3.90. After three months of treatment, IIEF-5 score significantly improved in both groups compared to baseline (13.18 ± 3.75 vs 20.48 ± 2.24, p < 0.0001; 14.15 ± 4.09 vs 19.06 ± 4.36, p < 0.0001, in group A and group B respectively). Patients treated with Tadalafil plus nutritional supplement showed a significantly higher increase in IIEF-5 score compared to those who received placebo (7.27 ± 2.20 and 4.9 ± 2.79, respectively; p < 0.0001;). No hormonal differences and metabolic effects were found. According cGMP result, nutritional supplements ameliorates and extends the activity of the chronic treatment. CONCLUSIONS IIEF-5 significant increase in group B, can be ascribed to the nutritional supplement properties and antioxidant effects of moringa oleifera, ginseng and rutin and this can enhance the endothelial NO and cGMP production.
Collapse
Affiliation(s)
- Vincenzo Mirone
- Department of Neurosciences, Sciences of Reproduction, and Odontostomatology, University of Naples Federico II, Naples.
| | - Luigi Napolitano
- Department of Neurosciences, Sciences of Reproduction, and Odontostomatology, University of Naples Federico II, Naples.
| | | | - Emma Mitidieri
- Department of Pharmacy, School of Medicine and Surgery, University of Naples, Federico II, Naples.
| | - Raffaella Sorrentino
- Department of Pharmacy, School of Medicine and Surgery, University of Naples, Federico II, Naples.
| | | | - Domenico Vanacore
- Department of Pharmacy, School of Medicine and Surgery, University of Naples, Federico II, Naples.
| | - Carlotta Turnaturi
- Department of Pharmacy, School of Medicine and Surgery, University of Naples, Federico II, Naples.
| | - Roberto La Rocca
- Department of Neurosciences, Sciences of Reproduction, and Odontostomatology, University of Naples Federico II, Naples.
| | - Giuseppe Celentano
- Department of Neurosciences, Sciences of Reproduction, and Odontostomatology, University of Naples Federico II, Naples.
| | - Davide Arcaniolo
- Urology Unit, Department of Woman Child and of General and Specialist Surgery, University of Campania "Luigi Vanvitelli", Naples.
| | - Giuseppe Cirino
- Department of Pharmacy, School of Medicine and Surgery, University of Naples, Federico II, Naples.
| |
Collapse
|
43
|
Arora S, Surakiatchanukul T, Arora T, Cagini C, Lupidi M, Chhablani J. Sildenafil in ophthalmology: An update. Surv Ophthalmol 2021; 67:463-487. [PMID: 34175342 DOI: 10.1016/j.survophthal.2021.06.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 12/14/2022]
Abstract
Sildenafil citrate, a selective oral phosphodiesterase 5 inhibitor, is a widely used drug for erectile dysfunction that acts by elevating cGMP levels and causing smooth muscle relaxation. It also has 10% activity against PDE6, a key enzyme in phototransduction cascade in the retina. Recent ocular imaging developments have further revealed the influence of sildenafil on ocular hemodynamics, particularly choroidal perfusion. Choroidal thickness is increased, and choroidal perfusion is also enhanced by autoregulatory mechanisms that are further dependent on age and microvascular abnormalities. Studies demonstrating high intraocular pressure via a "parallel pathway" from increased choroidal volume and blood flow to the ciliary body have challenged previous concepts. Another new observation is the effect of sildenafil on bipolar cells and cyclic-nucleotide gated channels. We discuss potential deleterious effects (central serous chorioretinopathy, glaucoma, ischemic optic neuropathy, and risks to recessive carriers of retinitis pigmentosa), potential beneficial effects (ameliorate choroidal ischemia, prevent thickening of Bruch membrane, and promote recovery of the ellipsoid zone) in macular degeneration, as well as potential drug interactions of sildenafil.
Collapse
Affiliation(s)
- Supriya Arora
- Bahamas Vision Centre and Princess Margaret Hospital, Nassau NP, Bahamas.
| | - Thamolwan Surakiatchanukul
- Department of Ophthalmology, Jamaica Hospital Medical Center, New York Medical College, Jamaica, NY, USA
| | - Tarun Arora
- Bahamas Vision Centre and Princess Margaret Hospital, Nassau NP, Bahamas.
| | - Carlo Cagini
- Department of Biochemical and Surgical Sciences, Section of ophthalmology, University of Perugia, Perugia, Italy
| | - Marco Lupidi
- Department of Biochemical and Surgical Sciences, Section of ophthalmology, University of Perugia, Perugia, Italy
| | - Jay Chhablani
- University of Pittsburgh, UPMC Eye Center, Pittsburgh, PA, USA.
| |
Collapse
|
44
|
Jankowska A, Pawłowski M, Chłoń-Rzepa G. Diabetic Theory in Anti-Alzheimer's Drug Research and Development. Part 2: Therapeutic Potential of cAMP-Specific Phosphodiesterase Inhibitors. Curr Med Chem 2021; 28:3535-3553. [PMID: 32940168 DOI: 10.2174/0929867327666200917125857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/21/2020] [Accepted: 08/25/2020] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is one of the most prevalent age-related neurodegenerative disease that affects the cognition, behavior, and daily activities of individuals. Studies indicate that this disease is characterized by several pathological mechanisms, including the accumulation of amyloid-beta peptide, hyperphosphorylation of tau protein, impairment of cholinergic neurotransmission, and increase in inflammatory responses within the central nervous system. Chronic neuroinflammation associated with AD is closely related to disturbances in metabolic processes, including insulin release and glucose metabolism. As AD is also called type III diabetes, diverse compounds having antidiabetic effects have been investigated as potential drugs for its symptomatic and disease-modifying treatment. In addition to insulin and oral antidiabetic drugs, scientific attention has been paid to cyclic-3',5'-adenosine monophosphate (cAMP)-specific phosphodiesterase (PDE) inhibitors that can modulate the concentration of glucose and related hormones and exert beneficial effects on memory, mood, and emotional processing. In this review, we present the most recent reports focusing on the involvement of cAMP-specific PDE4, PDE7, and PDE8 in glycemic and inflammatory response controls as well as the potential utility of the PDE inhibitors in the treatment of AD. Besides the results of in vitro and in vivo studies, the review also presents recent reports from clinical trials.
Collapse
Affiliation(s)
- Agnieszka Jankowska
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Medicinal Chemistry, 9 Medyczna Street, Krakow 30-688, Poland
| | - Maciej Pawłowski
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Medicinal Chemistry, 9 Medyczna Street, Krakow 30-688, Poland
| | - Grażyna Chłoń-Rzepa
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Medicinal Chemistry, 9 Medyczna Street, Krakow 30-688, Poland
| |
Collapse
|
45
|
The Potential Role of Sildenafil in Cancer Management through EPR Augmentation. J Pers Med 2021; 11:jpm11060585. [PMID: 34205602 PMCID: PMC8234771 DOI: 10.3390/jpm11060585] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/16/2021] [Accepted: 06/18/2021] [Indexed: 12/25/2022] Open
Abstract
Enhanced permeation retention (EPR) was a significant milestone discovery by Maeda et al. paving the path for the emerging field of nanomedicine to become a powerful tool in the fight against cancer. Sildenafil is a potent inhibitor of phosphodiesterase 5 (PDE-5) used for the treatment of erectile dysfunction (ED) through the relaxation of smooth muscles and the modulation of vascular endothelial permeability. Overexpression of PDE-5 has been reported in lung, colon, metastatic breast cancers, and bladder squamous carcinoma. Moreover, sildenafil has been reported to increase the sensitivity of tumor cells of different origins to the cytotoxic effect of chemotherapeutic agents with augmented apoptosis mediated through inducing the downregulation of Bcl-xL and FAP-1 expression, enhancing reactive oxygen species (ROS) generation, phosphorylating BAD and Bcl-2, upregulating caspase-3,8,9 activities, and blocking cells at G0/G1 cell cycle phase. Sildenafil has also demonstrated inhibitory effects on the efflux activity of ATP-binding cassette (ABC) transporters such as ABCC4, ABCC5, ABCB1, and ABCG2, ultimately reversing multidrug resistance. Accordingly, there has been a growing interest in using sildenafil as monotherapy or chemoadjuvant in EPR augmentation and management of different types of cancer. In this review, we critically examine the basic molecular mechanism of sildenafil related to cancer biology and discuss the overall potential of sildenafil in enhancing EPR-based anticancer drug delivery, pointing to the outcomes of the most important related preclinical and clinical studies.
Collapse
|
46
|
Meador KJ, Leeman-Markowski B, Medina AE, Illamola SM, Seliger J, Novak G, Lin C, Ivanisevic M, Razavi B, Marino S, Boyd A, Loring DW. Vinpocetine, cognition, and epilepsy. Epilepsy Behav 2021; 119:107988. [PMID: 33957389 DOI: 10.1016/j.yebeh.2021.107988] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 04/07/2021] [Accepted: 04/07/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Vinpocetine has been shown to enhance memory in animal models, with possible cognitive benefit in humans. The present study sought to demonstrate if vinpocetine can enhance cognition in healthy volunteers or patients with epilepsy. In addition, we compare blood levels of vinpocetine and its active metabolite (apovincaminic acid; AVA) in humans and animals to further characterize factors related to possible therapeutic benefit. METHODS The cognitive effects of vinpocetine were assessed in healthy adult volunteers (n = 8) using a double-blind, randomized, crossover design at single doses (placebo, 10, 20, and 60 mg oral). Cognitive effects of vinpocetine in patients with focal epilepsy (n = 8) were tested using a double-blind, randomized, crossover design at single doses (placebo, 20 mg oral) followed by one-month open label at 20 mg oral three times a day. The neuropsychological battery included both computerized and non-computerized tests. Levels of vinpocetine and AVA in the human studies were compared to levels in 45 mice across time dosed at 5-20 mg/kg intraperitoneal of vinpocetine. RESULTS No significant cognitive benefits were seen in healthy volunteers or patients with epilepsy. No appreciable side effects occurred. Vinpocetine and AVA levels were lower in humans than animals. CONCLUSIONS Vinpocetine was well tolerated, but was not associated with positive cognitive effects. However, blood levels obtained in humans were substantially less than levels in animals obtained from dosages known to be effective in one model. This suggests that higher dosages are needed in humans to assess vinpocetine's cognitive efficacy.
Collapse
Affiliation(s)
- Kimford J Meador
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA.
| | - Beth Leeman-Markowski
- Department of Neurology, New York University and VA New York Harbor Healthcare System, NY, NY USA.
| | | | - Sílvia M Illamola
- Department of Experimental & Clinical Pharmacology, University of Minnesota, Minneapolis, MN, USA.
| | - Jordan Seliger
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA.
| | - Gloria Novak
- Department of Neurology, Emory University, Atlanta, GA, USA.
| | - Christine Lin
- School of Medicine, University of California, San Diego, CA, USA.
| | | | - Babak Razavi
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA.
| | - Susan Marino
- Department of Experimental & Clinical Pharmacology, University of Minnesota, Minneapolis, MN, USA.
| | | | - David W Loring
- Department of Neurology, Emory University, Atlanta, GA, USA.
| |
Collapse
|
47
|
Mikami S. Discovery of Clinical Candidate TAK-915, a Highly Potent, Selective, and Brain Penetrating Novel Phosphodiesterase 2A Inhibitor. J SYN ORG CHEM JPN 2021. [DOI: 10.5059/yukigoseikyokaishi.79.581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
48
|
Gulati S, Palczewski K. New focus on regulation of the rod photoreceptor phosphodiesterase. Curr Opin Struct Biol 2021; 69:99-107. [PMID: 33945959 DOI: 10.1016/j.sbi.2021.03.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/18/2021] [Accepted: 03/25/2021] [Indexed: 02/03/2023]
Abstract
Rod photoreceptor phosphodiesterase (PDE6) is the key catalytic enzyme of visual phototransduction. PDE6 is the only member of the phosphodiesterase family that consists of a heterodimeric catalytic core composed of PDE6α and PDE6β subunits and two inhibitory PDE6γ subunits. Both PDE6α and PDE6β contain two regulatory GAF domains and one catalytic domain. GAF domains and the tightly bound PDE6γ subunits allosterically regulate the activity of the catalytic domain in association with the GTP-bound transducin alpha subunit (Gtα-GTP). Recent cryo-electron microscopy structures of the PDE6αγβγ and PDE6αγβγ-(Gtα-GTP)2 complexes have provided valuable knowledge shedding additional light on the allosteric activation of PDE6 by Gtα-GTP. Here we discuss recent developments in our understanding of the mechanism of PDE6 activation.
Collapse
Affiliation(s)
| | - Krzysztof Palczewski
- Gavin Herbert Eye Institute and the Department of Ophthalmology, Center for Translational Vision Research, University of California, 829 Health Sciences Road, Irvine, CA 92617, USA; The Department of Physiology & Biophysics, University of California, Irvine, CA 92697, USA; The Department of Chemistry, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
49
|
Khan MI, Nikoui V, Naveed A, Mumtaz F, Zaman H, Haider A, Aman W, Wahab A, Khan SN, Ullah N, Dehpour AR. Antidepressant-like effect of ethanol in mice forced swimming test is mediated via inhibition of NMDA/nitric oxide/cGMP signaling pathway. Alcohol 2021; 92:53-63. [PMID: 33581263 DOI: 10.1016/j.alcohol.2021.01.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 01/04/2021] [Accepted: 01/14/2021] [Indexed: 12/25/2022]
Abstract
There is evidence for a dramatic relationship between depression and alcohol consumption. Depressed patients may abuse ethanol because this agent reduces the symptoms of depression. In the current study, we aimed to investigate the NMDA/nitric oxide/cGMP pathway in the antidepressant-like effect of ethanol in an animal model of behavioral despair. Animals were subjected to locomotor activity in an open-field test separately, followed by a forced swimming test. During the forced swimming test (FST), ethanol (2 and 2.5 g/kg) significantly decreased the immobility time without altering the locomotor activity of animals. The antidepressant-like effect of ethanol (2.5 g/kg) was reversed by co-administration of N-methyl-D-aspartate (NMDA, 75 mg/kg), L-arginine (750 mg/kg), or sildenafil (5 mg/kg). In contrast, co-administration of MK-801 (0.05 mg/kg), ketamine (1 mg/kg), and ifenprodil (0.5 mg/kg) as antagonists of NMDAR, and NG-nitro-L-arginine methyl ester (L-NAME, 10 mg/kg), 7-nitroindazole (7-NI, 30 mg/kg), and methylene blue (10 mg/kg) as inhibitors of nitric oxide synthase (NOS), or 1H-[1,2,4]oxadiazole[4,3-a]quinoxalin-1-one (ODQ) (20 mg/kg), a nitric oxide/cyclic-guanosine monophosphate (NO-cGMP) inhibitor, with a subeffective dose of ethanol (1.5 g/kg), significantly decreased the immobility time in the FST. Furthermore, injection of ethanol 2.5 g/kg alone or 1.5 g/kg with a 7-NI subeffective dose, significantly decreased the nitrite levels in the hippocampus and prefrontal cortex. Hence, it is concluded that blockade of NMDA receptors and the nitric oxide/cyclic-guanosine monophosphate (NO-cGMP) pathway might be involved in the antidepressant-like effect of ethanol in mice.
Collapse
Affiliation(s)
- Muhammad Imran Khan
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, G7-Islamabad, Pakistan; Department of Pharmacy, Kohat University of Science and Technology, Kohat, 26000, KPK, Pakistan.
| | - Vahid Nikoui
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Aamir Naveed
- Department of Psychiatry, PIMS, Islamabad, Pakistan
| | - Faiza Mumtaz
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Zaman
- Department of Pharmacy, Kohat University of Science and Technology, Kohat, 26000, KPK, Pakistan
| | - Adnan Haider
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Punjab, 46000, Pakistan
| | - Waqar Aman
- Faculty of Pharmacy, University of Central Punjab, Lahore, Punjab, Pakistan
| | - Abdul Wahab
- Department of Pharmacy, Kohat University of Science and Technology, Kohat, 26000, KPK, Pakistan
| | - Shahid Niaz Khan
- Department of Zoology, Kohat University of Science & Technology, Kohat, Pakistan
| | - Najeeb Ullah
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, KPK, Pakistan
| | - Ahmad Reza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
50
|
Anatomy, Pathophysiology, Molecular Mechanisms, and Clinical Management of Erectile Dysfunction in Patients Affected by Coronary Artery Disease: A Review. Biomedicines 2021; 9:biomedicines9040432. [PMID: 33923709 PMCID: PMC8074129 DOI: 10.3390/biomedicines9040432] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/06/2021] [Accepted: 04/13/2021] [Indexed: 12/17/2022] Open
Abstract
Erectile dysfunction (ED) has been defined as the inability to attain or maintain penile erection sufficient for successful sexual intercourse. ED carries a notable influence on life quality, with significant implications for family and social relationships. Because atherosclerosis of penile arteries represents one of the most frequent ED causes, patients presenting with it should always be investigated for potential coexistent coronary or peripheral disease. Up to 75% of ED patients have a stenosis of the iliac-pudendal-penile arteries, supplying the male genital organ’s perfusion. Recently, pathophysiology and molecular basis of male erection have been elucidated, giving the ground to pharmacological and mechanical revascularization treatment of this condition. This review will focus on the normal anatomy and physiology of erection, the pathophysiology of ED, the relation between ED and cardiovascular diseases, and, lastly, on the molecular basis of erectile dysfunction.
Collapse
|