1
|
Luque GM, Jabloñski M, Schiavi-Ehrenhaus LJ, Del Prado RC, Balbach M, Romarowski A, Martin-Hidalgo D, Visconti PE, Krapf D, Darszon A, Krapf D, Buck J, Levin LR, Buffone MG. Bovine serum albumin-induced calcium influx triggers soluble adenylyl cyclase activation and cyclic AMP signalling pathways in mouse sperm capacitation. J Physiol 2025; 603:2633-2653. [PMID: 40320899 DOI: 10.1113/jp288389] [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: 02/04/2025] [Accepted: 04/08/2025] [Indexed: 05/14/2025] Open
Abstract
Sperm capacitation involves a series of biochemical and physiological changes essential for fertilization. A critical regulator of capacitation, the soluble adenylyl cyclase (sAC; ADCY10)-dependent production of the second messenger cyclic AMP (cAMP), drives key downstream events such as protein kinase A (PKA) substrate phosphorylation. sAC activity is directly stimulated by bicarbonate (HCO3 -) and calcium (Ca2+). CatSper, a sperm-specific Ca2+ channel, is considered the primary pathway for Ca2+ influx during capacitation; however, emerging evidence suggests additional pathways exist. This study reveals that bovine serum albumin (BSA) influences the dynamics of intracellular Ca2+ concentration ([Ca2+]i) in CatSper1 knockout (KO) sperm and plays a novel role in sAC activation. Using single-cell live imaging and flow cytometry, we observed a rapid [Ca2+]i rise in the head of CatSper1 KO sperm under capacitating conditions, indicating an alternative Ca2+ entry mechanism. BSA alone, in the absence of HCO3 -, triggered a significant [Ca2+]i rise. Removal of extracellular Ca2+ abolished this [Ca2+]i rise, confirming the necessity of Ca2+ influx. This BSA-induced [Ca2+]i rise was upstream of sAC activation, since it was not affected by sAC inhibitors and led to increased cAMP production and PKA substrate phosphorylation. Our findings provide new insights into the regulatory mechanisms of sAC, highlighting the existence of a CatSper-independent Ca2+ entry pathway activated by BSA during sperm capacitation. This rapid [Ca2+]i rise is initiated in the sperm head and propagates throughout the cell, and is sufficient to activate sAC and stimulate cAMP synthesis independently of HCO3 -. KEY POINTS: Sperm capacitation, essential for fertilization, is regulated by sAC, which produces cAMP in response to HCO3 - and Ca2+, driving key events like protein kinase A substrate phosphorylation. We demonstrate the existence of a CatSper-independent Ca2+ entry pathway that initiates in the sperm head and propagates throughout the cell, occurring rapidly after sperm encounters albumin, a critical component of the capacitation medium used in in vitro fertilization procedures in mammals. This albumin-induced Ca2+ influx is sufficient to activate sAC and stimulate cAMP synthesis independently of HCO3 -. We further reveal a novel role for albumin, beyond its well-established function as a cholesterol acceptor, in triggering this rapid Ca2+ influx and downstream signalling events essential for sperm capacitation. By demonstrating a CatSper-independent regulatory pathway, we expand the current paradigm of Ca2+ signalling in sperm physiology.
Collapse
Affiliation(s)
- Guillermina M Luque
- Instituto de Biología y Medicina Experimental (IBYME), Ciudad Autónoma de Buenos Aires, Argentina
| | - Martina Jabloñski
- Instituto de Biología y Medicina Experimental (IBYME), Ciudad Autónoma de Buenos Aires, Argentina
| | - Liza J Schiavi-Ehrenhaus
- Instituto de Biología y Medicina Experimental (IBYME), Ciudad Autónoma de Buenos Aires, Argentina
| | - Rita C Del Prado
- Instituto de Biología y Medicina Experimental (IBYME), Ciudad Autónoma de Buenos Aires, Argentina
| | | | - Ana Romarowski
- Instituto de Biología y Medicina Experimental (IBYME), Ciudad Autónoma de Buenos Aires, Argentina
- University of Massachusetts Amherst (UMass Amherst), Amherst, MA, USA
| | | | - Pablo E Visconti
- University of Massachusetts Amherst (UMass Amherst), Amherst, MA, USA
| | - Dario Krapf
- Instituto de Biología Molecular y Celular de Rosario (IBR), Rosario, Santa Fe, Argentina
| | - Alberto Darszon
- Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos, Mexico
| | - Diego Krapf
- Colorado State University, Fort Collins, CO, USA
| | | | | | - Mariano G Buffone
- Instituto de Biología y Medicina Experimental (IBYME), Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
2
|
Yang M, Hussain HMJ, Khan M, Muhammad Z, Zhou J, Ma A, Huang X, Ye J, Chen M, Zhi A, Liu T, Khan R, Asim A, Shah W, Zeb A, Ahmad N, Zhang H, Xu B, Ma H, Shi Q, Shi B. Deficiency in DNAH12 causes male infertility by impairing DNAH1 and DNALI1 recruitment in humans and mice. eLife 2025; 13:RP100350. [PMID: 40146200 PMCID: PMC11949491 DOI: 10.7554/elife.100350] [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] [Indexed: 03/28/2025] Open
Abstract
Asthenoteratozoospermia, a prevalent cause of male infertility, lacks a well-defined etiology. DNAH12 is a special dynein featured by the absence of a microtubule-binding domain, however, its functions in spermatogenesis remain largely unknown. Through comprehensive genetic analyses involving whole-exome sequencing and subsequent Sanger sequencing on infertile patients and fertile controls from six distinct families, we unveiled six biallelic mutations in DNAH12 that co-segregate recessively with male infertility in the studied families. Transmission electron microscopy (TEM) revealed pronounced axonemal abnormalities, including inner dynein arms (IDAs) impairment and central pair (CP) loss in sperm flagella of the patients. Mouse models (Dnah12-/- and Dnah12mut/mut) were generated and recapitulated the reproductive defects in the patients. Noteworthy, DNAH12 deficiency did not show effects on cilium organization and function. Mechanistically, DNAH12 was confirmed to interact with two other IDA components DNALI1 and DNAH1, while disruption of DNAH12 leads to failed recruitment of DNALI1 and DNAH1 to IDAs and compromised sperm development. Furthermore, DNAH12 also interacts with radial spoke head proteins RSPH1, RSPH9, and DNAJB13 to regulate CP stability. Moreover, the infertility of Dnah12-/- mice could be overcome by intracytoplasmic sperm injection (ICSI) treatment. Collectively, DNAH12 plays a crucial role in the proper organization of axoneme in sperm flagella, but not cilia, by recruiting DNAH1 and DNALI1 in both humans and mice. These findings expand our comprehension of dynein component assembly in flagella and cilia and provide a valuable marker for genetic counseling and diagnosis of asthenoteratozoospermia in clinical practice.
Collapse
Affiliation(s)
- Menglei Yang
- Center for Reproduction and Genetics, Department of Obstetrics and Gynecology, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Hafiz Muhammad Jafar Hussain
- Center for Reproduction and Genetics, Department of Obstetrics and Gynecology, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Manan Khan
- Center for Reproduction and Genetics, Department of Obstetrics and Gynecology, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Zubair Muhammad
- Center for Reproduction and Genetics, Department of Obstetrics and Gynecology, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Jianteng Zhou
- Center for Reproduction and Genetics, Department of Obstetrics and Gynecology, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Ao Ma
- Center for Reproduction and Genetics, Department of Obstetrics and Gynecology, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Xiongheng Huang
- Center for Reproduction and Genetics, Department of Obstetrics and Gynecology, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Jingwei Ye
- Center for Reproduction and Genetics, Department of Obstetrics and Gynecology, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Min Chen
- Center for Reproduction and Genetics, Department of Obstetrics and Gynecology, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Aoran Zhi
- Center for Reproduction and Genetics, Department of Obstetrics and Gynecology, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Tao Liu
- Center for Reproduction and Genetics, Department of Obstetrics and Gynecology, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Ranjha Khan
- Center for Reproduction and Genetics, Department of Obstetrics and Gynecology, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Ali Asim
- Center for Reproduction and Genetics, Department of Obstetrics and Gynecology, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Wasim Shah
- Center for Reproduction and Genetics, Department of Obstetrics and Gynecology, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Aurang Zeb
- Center for Reproduction and Genetics, Department of Obstetrics and Gynecology, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Nisar Ahmad
- Center for Reproduction and Genetics, Department of Obstetrics and Gynecology, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Huan Zhang
- Center for Reproduction and Genetics, Department of Obstetrics and Gynecology, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Bo Xu
- Center for Reproduction and Genetics, Department of Obstetrics and Gynecology, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Hui Ma
- Center for Reproduction and Genetics, Department of Obstetrics and Gynecology, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Qinghua Shi
- Center for Reproduction and Genetics, Department of Obstetrics and Gynecology, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| | - Baolu Shi
- Center for Reproduction and Genetics, Department of Obstetrics and Gynecology, First Affiliated Hospital of USTC, Hefei National Research Center for Physical Sciences at the Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Biomedical Sciences and Health Laboratory of Anhui Province, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Division of Life Sciences and Medicine, University of Science and Technology of ChinaHefeiChina
| |
Collapse
|
3
|
Ayoub S, Rivera Sanchez NDR, Fischoeder J, Balbach M, Levin LR, Buck J, Ritagliati C. Cyclic AMP Rescue of Motility in Sperm Devoid of Soluble Adenylyl Cyclase. Int J Mol Sci 2025; 26:1489. [PMID: 40003956 PMCID: PMC11855772 DOI: 10.3390/ijms26041489] [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: 01/10/2025] [Revised: 02/04/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
The second messenger cAMP plays multiple critical roles in the control of sperm functions essential for male fertility, including motility. The enzyme soluble adenylyl cyclase (sAC; ADCY10) was shown genetically and pharmacologically to be the essential source of cAMP mediating many of these functions. Male mice and men with genetic deletions of sAC are infertile, and their sperm are progressively immotile. Pharmacologically, delivery of potent and specific sAC inhibitors to male mice renders them temporarily infertile, and their sperm are similarly immotile. Here, we show that males from a second, independently derived mouse sAC knockout line are also infertile with progressively immotile sperm. We use these mouse models to determine optimal conditions for pharmacologically elevating intracellular cAMP to rescue the sAC null motility defect. We show that cell-permeable cAMP analogs, but not forskolin, rescue the motility defects of sAC deficient sperm, and we demonstrate that 8Br-cAMP is an efficient cAMP analog to rescue motility.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Carla Ritagliati
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA (L.R.L.)
| |
Collapse
|
4
|
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
|
5
|
Wang WL, Lian H, Liang Y, Ye Y, Tam PKH, Chen Y. Molecular Mechanisms of Fibrosis in Cholestatic Liver Diseases and Regenerative Medicine-Based Therapies. Cells 2024; 13:1997. [PMID: 39682745 PMCID: PMC11640075 DOI: 10.3390/cells13231997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/18/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
The aim of this review is to explore the potential of new regenerative medicine approaches in the treatment of cholestatic liver fibrosis. Cholestatic liver diseases, such as primary biliary cholangitis (PBC), primary sclerosing cholangitis (PSC), and biliary atresia (BA), due to the accumulation of bile, often progress to liver fibrosis, cirrhosis, and liver failure. When the disease becomes severe enough to require liver transplantation. Deeply understanding the disease's progression and fibrosis formation is crucial for better diagnosis and treatment. Current liver fibrosis treatments mainly target the root causes and no direct treatment method in fibrosis itself. Recent advances in regenerative medicine offer a potential approach that may help find the ways to target fibrosis directly, offering hope for improved outcomes. We also summarize, analyze, and discuss the current state and benefits of regenerative medicine therapies such as mesenchymal stem cell (MSC) therapy, induced pluripotent stem cells (iPSCs), and organoid technology, which may help the treatment of cholestatic liver diseases. Focusing on the latest research may reveal new targets and enhance therapeutic efficacy, potentially leading to more effective management and even curative strategies for cholestatic liver diseases.
Collapse
Affiliation(s)
- Wei-Lu Wang
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China; (W.-L.W.); (H.L.); (Y.L.)
| | - Haoran Lian
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China; (W.-L.W.); (H.L.); (Y.L.)
| | - Yingyu Liang
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China; (W.-L.W.); (H.L.); (Y.L.)
| | - Yongqin Ye
- Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China;
| | - Paul Kwong Hang Tam
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China; (W.-L.W.); (H.L.); (Y.L.)
- Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China;
- Precision Regenerative Medicine Research Centre, Medical Sciences Division, Macau University of Science and Technology, Macao SAR, China
| | - Yan Chen
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China; (W.-L.W.); (H.L.); (Y.L.)
- Faculty of Medicine, Macau University of Science and Technology, Macao SAR, China;
- Precision Regenerative Medicine Research Centre, Medical Sciences Division, Macau University of Science and Technology, Macao SAR, China
| |
Collapse
|
6
|
Kumar M, Mehan S, Kumar A, Sharma T, Khan Z, Tiwari A, Das Gupta G, Narula AS. Therapeutic efficacy of Genistein in activation of neuronal AC/cAMP/CREB/PKA and mitochondrial ETC-Complex pathways in experimental model of autism: Evidence from CSF, blood plasma and brain analysis. Brain Res 2024; 1846:149251. [PMID: 39384128 DOI: 10.1016/j.brainres.2024.149251] [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: 07/16/2024] [Revised: 09/10/2024] [Accepted: 09/22/2024] [Indexed: 10/11/2024]
Abstract
Autism is a complex neurodevelopmental condition characterized by repetitive behaviors, impaired social communication, and various associated conditions such as depression and anxiety. Its multifactorial etiology includes genetic, environmental, dietary, and gastrointestinal contributions. Pathologically, Autism is linked to mitochondrial dysfunction, oxidative stress, neuroinflammation, and neurotransmitter imbalances involving GABA, glutamate, dopamine, and oxytocin. Propionic acid (PRPA) is a short-chain fatty acid produced by gut bacteria, influencing central nervous system functions. Elevated PRPA levels can exacerbate Autism-related symptoms by disrupting metabolic processes and crossing the blood-brain barrier. Our research investigates the neuroprotective potential of Genistein (GNT), an isoflavone compound with known benefits in neuropsychiatric and neurodegenerative disorders, through modulation of the AC/cAMP/CREB/PKA signaling pathway and mitochondrial ETC complex (I-IV) function. In silico analyses revealed GNT's high affinity for these targets. Subsequent in vitro and in vivo experiments using a PRPA-induced rat model of autism demonstrated that GNT (40 and 80 mg/kg., orally) significantly improves locomotion, neuromuscular coordination, and cognitive functions in PRPA-treated rodents. Behavioral assessments showed reduced immobility in the forced swim test, enhanced Morris water maze performance, and restored regular locomotor activity. On a molecular level, GNT restored levels of key signaling molecules (AC, cAMP, CREB, PKA) and mitochondrial complexes (I-V), disrupted by PRPA exposure. Additionally, GNT reduced neuroinflammation and apoptosis, normalized neurotransmitter levels, and improved the complete blood count profile. Histopathological analyses confirmed that GNT ameliorated PRPA-induced brain injuries, restored normal brain morphology, reduced demyelination, and promoted neurogenesis. The study supports GNT's potential in autism treatment by modulating neural pathways, reducing inflammation, and restoring neurotransmitter balance.
Collapse
Affiliation(s)
- Manjeet Kumar
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India; Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India; Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India.
| | - Aakash Kumar
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India; Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Tarun Sharma
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India; Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India; Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Aarti Tiwari
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India; Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Ghanshyam Das Gupta
- Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India; Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| |
Collapse
|
7
|
Visconti PE, Levin LR, Buck J. David Garbers and the Birth of cAMP Biology in Mammalian Sperm. Mol Reprod Dev 2024; 91:e23773. [PMID: 39385557 PMCID: PMC11910748 DOI: 10.1002/mrd.23773] [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: 07/29/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024]
Abstract
Dr. David Garbers made many impactful contributions to science and vastly improved our understanding of sperm biology. In this review, we focus on his identification of a key role for the second messenger cAMP in mammalian sperm. As a graduate student David discovered that sperm motility, which is essential for sperm to fertilize the egg, is under the control of the (at the time) recently identified, prototypical second messenger cAMP. Fast-forwarding to the present, agents which turn off sperm's ability to generate cAMP and block sperm motility are being investigated as potential nonhormonal contraceptives for men and women. Should these efforts prove successful, Dave's discoveries will prove to be the spark which ignited a revolution in human health.
Collapse
Affiliation(s)
- Pablo E Visconti
- Department of Veterinary and Animal Science, University of Massachusetts, Amherst, Massachusetts, USA
| | - Lonny R Levin
- Department of Pharmacology, Weill Cornell Medicine, New York, New York, USA
| | - Jochen Buck
- Department of Pharmacology, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
8
|
Kaupp UB, Kendall O. David Garbers' Contributions to Chemotaxis Signaling in Sperm. Mol Reprod Dev 2024; 91:e23774. [PMID: 39445585 DOI: 10.1002/mrd.23774] [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: 07/15/2024] [Revised: 09/12/2024] [Accepted: 09/15/2024] [Indexed: 10/25/2024]
Abstract
This review focuses on the contribution of the late David Garbers to chemotaxis of sperm, in particular from sea urchin. We will describe his discovery of chemotactic peptides and their cognate receptors, his discovery of a sperm-specific, unique Na+/H+ exchanger that represents a chimera between a solute carrier (SLC) and an ion channel. Finally, we will discuss his contributions to the understanding of cAMP signaling in sperm via soluble adenylyl cyclase (sAC) and its control by Ca2+ ions.
Collapse
Affiliation(s)
- U B Kaupp
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Life & Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Olivia Kendall
- Life & Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| |
Collapse
|
9
|
Li HL, Verhoeven A, Elferink RO. The role of soluble adenylyl cyclase in sensing and regulating intracellular pH. Pflugers Arch 2024; 476:457-465. [PMID: 38581526 PMCID: PMC11006738 DOI: 10.1007/s00424-024-02952-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/08/2024]
Abstract
Soluble adenylyl cyclase (sAC) differs from transmembrane adenylyl cyclases (tmAC) in many aspects. In particular, the activity of sAC is not regulated by G-proteins but by the prevailing bicarbonate concentrations inside cells. Therefore, sAC serves as an exquisite intracellular pH sensor, with the capacity to translate pH changes into the regulation of localization and/or activity of cellular proteins involved in pH homeostasis. In this review, we provide an overview of literature describing the regulation of sAC activity by bicarbonate, pinpointing the importance of compartmentalization of intracellular cAMP signaling cascades. In addition, examples of processes involving proton and bicarbonate transport in different cell types, in which sAC plays an important regulatory role, were described in detail.
Collapse
Affiliation(s)
- Hang Lam Li
- Tytgat Institute for Liver and Intestinal Research, Research Institute AGEM, Amsterdam UMC, Meibergdreef 69-71, 1105BK, Amsterdam, the Netherlands
| | - Arthur Verhoeven
- Tytgat Institute for Liver and Intestinal Research, Research Institute AGEM, Amsterdam UMC, Meibergdreef 69-71, 1105BK, Amsterdam, the Netherlands
| | - Ronald Oude Elferink
- Tytgat Institute for Liver and Intestinal Research, Research Institute AGEM, Amsterdam UMC, Meibergdreef 69-71, 1105BK, Amsterdam, the Netherlands.
| |
Collapse
|
10
|
Li HL, Go S, Chang JC, Verhoeven A, Elferink RO. Soluble adenylyl cyclase, the cell-autonomous member of the family. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166936. [PMID: 37951509 DOI: 10.1016/j.bbadis.2023.166936] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/12/2023] [Accepted: 10/24/2023] [Indexed: 11/14/2023]
Abstract
Soluble adenylyl cyclase (sAC) is the evolutionarily most ancient of a set of 10 adenylyl cyclases (Adcys). While Adcy1 to Adcy9 are cAMP-producing enzymes that are activated by G-protein coupled receptors (GPCRs), Adcy10 (sAC) is an intracellular adenylyl cyclase. sAC plays a pivotal role in numerous cellular processes, ranging from basic physiological functions to complex signaling cascades. As a distinct member of the adenylyl cyclase family, sAC is not activated by GPCRs and stands apart due to its unique characteristics, regulation, and localization within cells. This minireview aims to honour Ulli Brandt, the outgoing Executive Editor of our journal, Biochimica Biophysica Acta (BBA), and longstanding Executive Editor of the BBA section Bioenergetics. We will therefore focus this review on bioenergetic aspects of sAC and, in addition, review some important recent general developments in the field of research on sAC.
Collapse
Affiliation(s)
- Hang Lam Li
- Tytgat Institute for Liver and Intestinal Research, Research Institute AGEM, Amsterdam UMC, the Netherlands
| | - Simei Go
- Tytgat Institute for Liver and Intestinal Research, Research Institute AGEM, Amsterdam UMC, the Netherlands
| | - Jung-Chin Chang
- Tytgat Institute for Liver and Intestinal Research, Research Institute AGEM, Amsterdam UMC, the Netherlands
| | - Arthur Verhoeven
- Tytgat Institute for Liver and Intestinal Research, Research Institute AGEM, Amsterdam UMC, the Netherlands
| | - Ronald Oude Elferink
- Tytgat Institute for Liver and Intestinal Research, Research Institute AGEM, Amsterdam UMC, the Netherlands.
| |
Collapse
|
11
|
Abstract
Primary biliary cholangitis (PBC) is the most common of the autoimmune liver diseases, in which there is chronic small bile duct inflammation. The pathophysiology of PBC is multifactorial, involving immune dysregulation and damage to biliary epithelial cells, with influences from genetic factors, epigenetics, the gut-liver axis, and environmental exposures.
Collapse
Affiliation(s)
- Inbal Houri
- Division of Gastroenterology and Hepatology, Toronto Centre for Liver Disease, University of Toronto, 9th Floor Eaton Building, North Wing 219-B, 200 Elizabeth Street, Toronto, Ontario M5G 2C4, Canada
| | - Gideon M Hirschfield
- Division of Gastroenterology and Hepatology, Toronto Centre for Liver Disease, University of Toronto, 9th Floor Eaton Building, North Wing 219-B, 200 Elizabeth Street, Toronto, Ontario M5G 2C4, Canada.
| |
Collapse
|
12
|
Gannon HG, Riaz-Bradley A, Cann MJ. A Non-Functional Carbon Dioxide-Mediated Post-Translational Modification on Nucleoside Diphosphate Kinase of Arabidopsis thaliana. Int J Mol Sci 2024; 25:898. [PMID: 38255974 PMCID: PMC10815852 DOI: 10.3390/ijms25020898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
The carbamate post-translational modification (PTM), formed by the nucleophilic attack of carbon dioxide by a dissociated lysine epsilon-amino group, is proposed as a widespread mechanism for sensing this biologically important bioactive gas. Here, we demonstrate the discovery and in vitro characterization of a carbamate PTM on K9 of Arabidopsis nucleoside diphosphate kinase (AtNDK1). We demonstrate that altered side chain reactivity at K9 is deleterious for AtNDK1 structure and catalytic function, but that CO2 does not impact catalysis. We show that nucleotide substrate removes CO2 from AtNDK1, and the carbamate PTM is functionless within the detection limits of our experiments. The AtNDK1 K9 PTM is the first demonstration of a functionless carbamate. In light of this finding, we speculate that non-functionality is a possible feature of the many newly identified carbamate PTMs.
Collapse
Affiliation(s)
- Harry G. Gannon
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK; (H.G.G.)
| | - Amber Riaz-Bradley
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK; (H.G.G.)
| | - Martin J. Cann
- Department of Biosciences, Durham University, South Road, Durham DH1 3LE, UK; (H.G.G.)
- Biophysical Sciences Institute, Durham University, South Road, Durham DH1 3LE, UK
| |
Collapse
|
13
|
Bizerra PFV, Gilglioni EH, Li HL, Go S, Oude Elferink RPJ, Verhoeven AJ, Chang JC. Opposite regulation of glycogen metabolism by cAMP produced in the cytosol and at the plasma membrane. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119585. [PMID: 37714306 DOI: 10.1016/j.bbamcr.2023.119585] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 09/17/2023]
Abstract
Cyclic AMP is produced in cells by two different types of adenylyl cyclases: at the plasma membrane by the transmembrane adenylyl cyclases (tmACs, ADCY1~ADCY9) and in the cytosol by the evolutionarily more conserved soluble adenylyl cyclase (sAC, ADCY10). By employing high-resolution extracellular flux analysis in HepG2 cells to study glycogen breakdown in real time, we showed that cAMP regulates glycogen metabolism in opposite directions depending on its location of synthesis within cells and the downstream cAMP effectors. While the canonical tmAC-cAMP-PKA signaling promotes glycogenolysis, we demonstrate here that the non-canonical sAC-cAMP-Epac1 signaling suppresses glycogenolysis. Mechanistically, suppression of sAC-cAMP-Epac1 leads to Ser-15 phosphorylation and thereby activation of the liver-form glycogen phosphorylase to promote glycogenolysis. Our findings highlight the importance of cAMP microdomain organization for distinct metabolic regulation and establish sAC as a novel regulator of glycogen metabolism.
Collapse
Affiliation(s)
- Paulo F V Bizerra
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; State University of Maringá, Paraná, Brazil
| | - Eduardo H Gilglioni
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles, Brussels, Belgium
| | - Hang Lam Li
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Simei Go
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Ronald P J Oude Elferink
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Arthur J Verhoeven
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Jung-Chin Chang
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Division of Cell Biology, Metabolism & Cancer, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
14
|
Grahn E, Kaufmann SV, Askarova M, Ninov M, Welp LM, Berger TK, Urlaub H, Kaupp UB. Control of intracellular pH and bicarbonate by CO 2 diffusion into human sperm. Nat Commun 2023; 14:5395. [PMID: 37669933 PMCID: PMC10480191 DOI: 10.1038/s41467-023-40855-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 08/14/2023] [Indexed: 09/07/2023] Open
Abstract
The reaction of CO2 with H2O to form bicarbonate (HCO3-) and H+ controls sperm motility and fertilization via HCO3--stimulated cAMP synthesis. A complex network of signaling proteins participates in this reaction. Here, we identify key players that regulate intracellular pH (pHi) and HCO3- in human sperm by quantitative mass spectrometry (MS) and kinetic patch-clamp fluorometry. The resting pHi is set by amiloride-sensitive Na+/H+ exchange. The sperm-specific putative Na+/H+ exchanger SLC9C1, unlike its sea urchin homologue, is not gated by voltage or cAMP. Transporters and channels implied in HCO3- transport are not detected, and may be present at copy numbers < 10 molecules/sperm cell. Instead, HCO3- is produced by diffusion of CO2 into cells and readjustment of the CO2/HCO3-/H+ equilibrium. The proton channel Hv1 may serve as a unidirectional valve that blunts the acidification ensuing from HCO3- synthesis. This work provides a new framework for the study of male infertility.
Collapse
Affiliation(s)
- Elena Grahn
- Max Planck Institute for Neurobiology of Behavior-caesar, Molecular Sensory Systems, Ludwig-Erhard-Allee 2, 53175, Bonn, Germany
| | - Svenja V Kaufmann
- Max Planck Institute for Multidisciplinary Sciences, Bioanalytical Mass Spectrometry, Am Fassberg 11, 37077, Göttingen, Germany
| | - Malika Askarova
- Max Planck Institute for Neurobiology of Behavior-caesar, Molecular Sensory Systems, Ludwig-Erhard-Allee 2, 53175, Bonn, Germany
| | - Momchil Ninov
- Max Planck Institute for Multidisciplinary Sciences, Bioanalytical Mass Spectrometry, Am Fassberg 11, 37077, Göttingen, Germany
- University Medical Center Göttingen, Institute of Clinical Chemistry, Bioanalytics, Robert-Koch-Strasse 40, 37075, Göttingen, Germany
| | - Luisa M Welp
- Max Planck Institute for Multidisciplinary Sciences, Bioanalytical Mass Spectrometry, Am Fassberg 11, 37077, Göttingen, Germany
- University Medical Center Göttingen, Institute of Clinical Chemistry, Bioanalytics, Robert-Koch-Strasse 40, 37075, Göttingen, Germany
| | - Thomas K Berger
- Max Planck Institute for Neurobiology of Behavior-caesar, Molecular Sensory Systems, Ludwig-Erhard-Allee 2, 53175, Bonn, Germany.
- Department of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps-University Marburg, Deutschhausstrasse 1-2, 35037, Marburg, Germany.
| | - Henning Urlaub
- Max Planck Institute for Multidisciplinary Sciences, Bioanalytical Mass Spectrometry, Am Fassberg 11, 37077, Göttingen, Germany.
- University Medical Center Göttingen, Institute of Clinical Chemistry, Bioanalytics, Robert-Koch-Strasse 40, 37075, Göttingen, Germany.
- Cluster of Excellence, Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells (MBExC), University of Göttingen, Göttingen, Germany.
| | - U Benjamin Kaupp
- Max Planck Institute for Neurobiology of Behavior-caesar, Molecular Sensory Systems, Ludwig-Erhard-Allee 2, 53175, Bonn, Germany.
- Life & Medical Sciences Institute (LIMES), University Bonn, Carl-Troll-Strasse 31, 53115, Bonn, Germany.
| |
Collapse
|
15
|
Liu W, Zhang S, Li Q, Wu Y, Jia X, Feng W, Li Z, Shi Y, Hou Q, Ma J, Liu Y, Gao P, Ganz T, Liu S. Lactate modulates iron metabolism by binding soluble adenylyl cyclase. Cell Metab 2023; 35:1597-1612.e6. [PMID: 37480842 DOI: 10.1016/j.cmet.2023.06.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 04/18/2023] [Accepted: 06/27/2023] [Indexed: 07/24/2023]
Abstract
Overproduction of lactate (LA) can occur during exercise and in many diseases such as cancers. Individuals with hyperlactatemia often display anemia, decreased serum iron, and elevated hepcidin, a key regulator of iron metabolism. However, it is unknown whether and how LA regulates hepcidin expression. Here, we show LA binds to soluble adenylyl cyclase (sAC) in normal hepatocytes and affects systemic iron homeostasis in mice by increasing hepcidin expression. Comprehensive in vitro, in vivo, and in silico experiments show that the LA-sAC interaction raises cyclic adenosine monophosphate (cAMP) levels, which activates the PKA-Smad1/5/8 signaling pathway to increase hepcidin transcription. We verified this regulatory axis in wild-type mice and in mice with disordered iron homeostasis. LA also regulates hepcidin in humans at rest and subjected to extensive exercise that produce elevated LA. Our study links hyperlactatemia to iron deficiency, offering a mechanistic explanation for anemias seen in athletes and patients with lactic acidosis.
Collapse
Affiliation(s)
- Wei Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China; State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Shuping Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Quanjin Li
- University of Chinese Academy of Sciences, Beijing 100049, China; National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yue Wu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuan Jia
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenya Feng
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhaolong Li
- University of Chinese Academy of Sciences, Beijing 100049, China; National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yali Shi
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qingzhi Hou
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Juan Ma
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yajun Liu
- National Center for Orthopaedics, Beijing Jishuitan Hospital, Beijing 100035, China; Beijing Research Institute of Traumatology and Orthopaedics, Beijing 100035, China
| | - Pu Gao
- University of Chinese Academy of Sciences, Beijing 100049, China; National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Tomas Ganz
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sijin Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China; State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
16
|
Mariani NAP, Silva JV, Fardilha M, Silva EJR. Advances in non-hormonal male contraception targeting sperm motility. Hum Reprod Update 2023; 29:545-569. [PMID: 37141450 DOI: 10.1093/humupd/dmad008] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 03/23/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND The high rates of unintended pregnancy and the ever-growing world population impose health, economic, social, and environmental threats to countries. Expanding contraceptive options, including male methods, are urgently needed to tackle these global challenges. Male contraception is limited to condoms and vasectomy, which are unsuitable for many couples. Thus, novel male contraceptive methods may reduce unintended pregnancies, meet the contraceptive needs of couples, and foster gender equality in carrying the contraceptive burden. In this regard, the spermatozoon emerges as a source of druggable targets for on-demand, non-hormonal male contraception based on disrupting sperm motility or fertilization. OBJECTIVE AND RATIONALE A better understanding of the molecules governing sperm motility can lead to innovative approaches toward safe and effective male contraceptives. This review discusses cutting-edge knowledge on sperm-specific targets for male contraception, focusing on those with crucial roles in sperm motility. We also highlight challenges and opportunities in male contraceptive drug development targeting spermatozoa. SEARCH METHODS We conducted a literature search in the PubMed database using the following keywords: 'spermatozoa', 'sperm motility', 'male contraception', and 'drug targets' in combination with other related terms to the field. Publications until January 2023 written in English were considered. OUTCOMES Efforts for developing non-hormonal strategies for male contraception resulted in the identification of candidates specifically expressed or enriched in spermatozoa, including enzymes (PP1γ2, GAPDHS, and sAC), ion channels (CatSper and KSper), transmembrane transporters (sNHE, SLC26A8, and ATP1A4), and surface proteins (EPPIN). These targets are usually located in the sperm flagellum. Their indispensable roles in sperm motility and male fertility were confirmed by genetic or immunological approaches using animal models and gene mutations associated with male infertility due to sperm defects in humans. Their druggability was demonstrated by the identification of drug-like small organic ligands displaying spermiostatic activity in preclinical trials. WIDER IMPLICATIONS A wide range of sperm-associated proteins has arisen as key regulators of sperm motility, providing compelling druggable candidates for male contraception. Nevertheless, no pharmacological agent has reached clinical developmental stages. One reason is the slow progress in translating the preclinical and drug discovery findings into a drug-like candidate adequate for clinical development. Thus, intense collaboration among academia, private sectors, governments, and regulatory agencies will be crucial to combine expertise for the development of male contraceptives targeting sperm function by (i) improving target structural characterization and the design of highly selective ligands, (ii) conducting long-term preclinical safety, efficacy, and reversibility evaluation, and (iii) establishing rigorous guidelines and endpoints for clinical trials and regulatory evaluation, thus allowing their testing in humans.
Collapse
Affiliation(s)
- Noemia A P Mariani
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu, São Paulo State University, Botucatu, Brazil
| | - Joana V Silva
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
- QOPNA & LAQV, Department of Chemistry, University of Aveiro, Aveiro, Portugal
- Multidisciplinary Research in Biomedicine (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Margarida Fardilha
- Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | - Erick J R Silva
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu, São Paulo State University, Botucatu, Brazil
| |
Collapse
|
17
|
Wiggins SV, Schreiner R, Ferreira J, Marmorstein AD, Levin LR, Buck J. Carbonic Anhydrase Inhibitor Modulation of Intraocular Pressure Is Independent of Soluble Adenylyl Cyclase. J Ocul Pharmacol Ther 2023; 39:317-323. [PMID: 37097314 PMCID: PMC10398745 DOI: 10.1089/jop.2022.0180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 03/20/2023] [Indexed: 04/26/2023] Open
Abstract
Purpose: We investigated whether a clinically used carbonic anhydrase inhibitor (CAIs) can modulate intraocular pressure (IOP) through soluble adenylyl cyclase (sAC) signaling. Methods: IOP was measured 1 h after topical treatment with brinzolamide, a topically applied and clinically used CAIs, using direct cannulation of the anterior chamber in sAC knockout (KO) mice or C57BL/6J mice in the presence or absence of the sAC inhibitor (TDI-10229). Results: Mice treated with the sAC inhibitor TDI-10229 had elevated IOP. CAIs treatment significantly decreased increased intraocular pressure (IOP) in wild-type, sAC KO mice, as well as TDI-10229-treated mice. Conclusions: Inhibiting carbonic anhydrase reduces IOP independently from sAC in mice. Our studies suggest that the signaling cascade by which brinzolamide regulates IOP does not involve sAC.
Collapse
Affiliation(s)
- Shakarr V. Wiggins
- Department of Pharmacology, Weill Cornell Medicine, New York, New York, USA
- Graduate Program in Neuroscience, Weill Cornell Medicine, New York, New York, USA
| | - Ryan Schreiner
- Division of Regenerative Medicine, Department of Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, New York, USA
| | - Jacob Ferreira
- Department of Pharmacology, Weill Cornell Medicine, New York, New York, USA
| | | | - Lonny R. Levin
- Department of Pharmacology, Weill Cornell Medicine, New York, New York, USA
| | - Jochen Buck
- Department of Pharmacology, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
18
|
Sun S, Fushimi M, Rossetti T, Kaur N, Ferreira J, Miller M, Quast J, van den Heuvel J, Steegborn C, Levin LR, Buck J, Myers RW, Kargman S, Liverton N, Meinke PT, Huggins DJ. Scaffold Hopping and Optimization of Small Molecule Soluble Adenyl Cyclase Inhibitors Led by Free Energy Perturbation. J Chem Inf Model 2023; 63:2828-2841. [PMID: 37060320 DOI: 10.1021/acs.jcim.2c01577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2023]
Abstract
Free energy perturbation is a computational technique that can be used to predict how small changes to an inhibitor structure will affect the binding free energy to its target. In this paper, we describe the utility of free energy perturbation with FEP+ in the hit-to-lead stage of a drug discovery project targeting soluble adenyl cyclase. The project was structurally enabled by X-ray crystallography throughout. We employed free energy perturbation to first scaffold hop to a preferable chemotype and then optimize the binding affinity to sub-nanomolar levels while retaining druglike properties. The results illustrate that effective use of free energy perturbation can enable a drug discovery campaign to progress rapidly from hit to lead, facilitating proof-of-concept studies that enable target validation.
Collapse
Affiliation(s)
- Shan Sun
- Tri-Institutional Therapeutics Discovery Institute, New York, New York 10021, United States
| | - Makoto Fushimi
- Tri-Institutional Therapeutics Discovery Institute, New York, New York 10021, United States
| | - Thomas Rossetti
- Department of Pharmacology, Weill Cornell Medicine, New York City, New York 10056, United States
| | - Navpreet Kaur
- Department of Pharmacology, Weill Cornell Medicine, New York City, New York 10056, United States
| | - Jacob Ferreira
- Department of Pharmacology, Weill Cornell Medicine, New York City, New York 10056, United States
| | - Michael Miller
- Tri-Institutional Therapeutics Discovery Institute, New York, New York 10021, United States
| | - Jonathan Quast
- Department of Biochemistry, University of Bayreuth, Bayreuth 95440, Germany
| | | | - Clemens Steegborn
- Department of Biochemistry, University of Bayreuth, Bayreuth 95440, Germany
| | - Lonny R Levin
- Department of Pharmacology, Weill Cornell Medicine, New York City, New York 10056, United States
| | - Jochen Buck
- Department of Pharmacology, Weill Cornell Medicine, New York City, New York 10056, United States
| | - Robert W Myers
- Tri-Institutional Therapeutics Discovery Institute, New York, New York 10021, United States
| | - Stacia Kargman
- Tri-Institutional Therapeutics Discovery Institute, New York, New York 10021, United States
| | - Nigel Liverton
- Tri-Institutional Therapeutics Discovery Institute, New York, New York 10021, United States
| | - Peter T Meinke
- Tri-Institutional Therapeutics Discovery Institute, New York, New York 10021, United States
- Department of Pharmacology, Weill Cornell Medicine, New York City, New York 10056, United States
| | - David J Huggins
- Tri-Institutional Therapeutics Discovery Institute, New York, New York 10021, United States
- Department of Physiology and Biophysics, Weill Cornell Medical College of Cornell University, New York, New York 10065, United States
| |
Collapse
|
19
|
Ritagliati C, Ayoub S, Balbach M, Buck J, Levin LR. In vivo characterization of sAC null sperm. Front Cell Dev Biol 2023; 11:1134051. [PMID: 37152282 PMCID: PMC10160483 DOI: 10.3389/fcell.2023.1134051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 04/12/2023] [Indexed: 05/09/2023] Open
Abstract
Targeted disruption of the soluble adenylyl cyclase (ADCY10; sAC) gene results in male-specific sterility without affecting spermatogenesis, mating behavior, or spermatozoa morphology and count; however, it dramatically impairs sperm motility and prevents capacitation. These phenotypes were identified in sperm from sAC null mice surgically extracted from the epididymis and studied in vitro. Epididymal sperm are dormant, and never exposed to physiological activators in semen or the female reproductive tract. To study sAC null sperm under conditions which more closely resemble natural fertilization, we explored phenotypes of ejaculated sAC null sperm in vivo post-coitally as well as ex vivo, collected from the female reproductive tract. Ex vivo ejaculated sAC null sperm behaved similarly to epididymal sAC null sperm, except with respect to the physiologically induced acrosome reaction. These studies suggest there is a sAC-independent regulation of acrosome responsiveness induced upon ejaculation or exposure to factors in the female reproductive tract. We also studied the behavior of sAC null sperm in vivo post-coitally by taking advantage of transgenes with fluorescently labelled sperm. Transgenes expressing GFP in the acrosome and DsRed2 in the mitochondria located in the midpiece of sperm (DsRed2/Acr3-EGFP) allow visualization of sperm migration through the female reproductive tract after copulation. As previously reported, sperm from wild type (WT) double transgenic mice migrated from the uterus through the uterotubular junction (UTJ) into the oviduct within an hour post-copulation. In contrast, sperm from sAC null double transgenic mice were only found in the uterus. There were no sAC null sperm in the oviduct, even 8 h after copulation. These results demonstrate that sAC KO males are infertile because their sperm do not migrate to the fertilization site.
Collapse
|
20
|
Miller M, Rossetti T, Ferreira J, Ghanem L, Balbach M, Kaur N, Levin LR, Buck J, Kehr M, Coquille S, van den Heuvel J, Steegborn C, Fushimi M, Finkin-Groner E, Myers RW, Kargman S, Liverton NJ, Huggins DJ, Meinke PT. Design, Synthesis, and Pharmacological Evaluation of Second-Generation Soluble Adenylyl Cyclase (sAC, ADCY10) Inhibitors with Slow Dissociation Rates. J Med Chem 2022; 65:15208-15226. [PMID: 36346696 PMCID: PMC9866367 DOI: 10.1021/acs.jmedchem.2c01133] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Soluble adenylyl cyclase (sAC: ADCY10) is an enzyme involved in intracellular signaling. Inhibition of sAC has potential therapeutic utility in a number of areas. For example, sAC is integral to successful male fertility: sAC activation is required for sperm motility and ability to undergo the acrosome reaction, two processes central to oocyte fertilization. Pharmacologic evaluation of existing sAC inhibitors for utility as on-demand, nonhormonal male contraceptives suggested that both high intrinsic potency, fast on and slow dissociation rates are essential design elements for successful male contraceptive applications. During the course of the medicinal chemistry campaign described here, we identified sAC inhibitors that fulfill these criteria and are suitable for in vivo evaluation of diverse sAC pharmacology.
Collapse
Affiliation(s)
- Michael Miller
- Tri-Institutional Therapeutics Discovery Institute, New York, New York 10021, United States
| | - Thomas Rossetti
- Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, United States
| | - Jacob Ferreira
- Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, United States
| | - Lubna Ghanem
- Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, United States
| | - Melanie Balbach
- Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, United States
| | - Navpreet Kaur
- Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, United States
| | - Lonny R. Levin
- Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, United States
| | - Jochen Buck
- Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, United States
| | - Maria Kehr
- Department of Biochemistry, University of Bayreuth, 95440 Bayreuth, Germany
| | - Sandrine Coquille
- Department of Biochemistry, University of Bayreuth, 95440 Bayreuth, Germany
| | - Joop van den Heuvel
- Helmholtz Centre for Infection Research, Recombinant Protein Expression, 38124 Braunschweig, Germany
| | - Clemens Steegborn
- Department of Biochemistry, University of Bayreuth, 95440 Bayreuth, Germany
| | - Makoto Fushimi
- Tri-Institutional Therapeutics Discovery Institute, New York, New York 10021, United States
| | - Efrat Finkin-Groner
- Tri-Institutional Therapeutics Discovery Institute, New York, New York 10021, United States
| | - Robert W. Myers
- Tri-Institutional Therapeutics Discovery Institute, New York, New York 10021, United States
| | - Stacia Kargman
- Tri-Institutional Therapeutics Discovery Institute, New York, New York 10021, United States
| | - Nigel J. Liverton
- Tri-Institutional Therapeutics Discovery Institute, New York, New York 10021, United States
| | - David J. Huggins
- Tri-Institutional Therapeutics Discovery Institute, New York, New York 10021, United States; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York 10021, United States
| | - Peter T. Meinke
- Tri-Institutional Therapeutics Discovery Institute, New York, New York 10021, United States; Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, United States
| |
Collapse
|
21
|
Woranush W, Moskopp ML, Noll T, Dieterich P. Quantifying and mathematical modelling of the influence of soluble adenylate cyclase on cell cycle in human endothelial cells with Bayesian inference. J Cell Mol Med 2022; 26:5887-5900. [DOI: 10.1111/jcmm.17611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 08/28/2022] [Accepted: 10/05/2022] [Indexed: 11/16/2022] Open
Affiliation(s)
- Warunya Woranush
- Institut für Physiologie, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden Dresden Germany
| | - Mats Leif Moskopp
- Institut für Physiologie, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden Dresden Germany
- Vivantes Klinikum im Friedrichshain, Charité Academic Teaching Hospital, Klinik für Neurochirurgie Berlin Germany
| | - Thomas Noll
- Institut für Physiologie, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden Dresden Germany
| | - Peter Dieterich
- Institut für Physiologie, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden Dresden Germany
| |
Collapse
|
22
|
Cordero-Martínez J, Jimenez-Gutierrez GE, Aguirre-Alvarado C, Alacántara-Farfán V, Chamorro-Cevallos G, Roa-Espitia AL, Hernández-González EO, Rodríguez-Páez L. Participation of signaling proteins in sperm hyperactivation. Syst Biol Reprod Med 2022; 68:315-330. [DOI: 10.1080/19396368.2022.2122761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Affiliation(s)
- Joaquín Cordero-Martínez
- Laboratorio de Bioquímica Farmacológica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | | | - Charmina Aguirre-Alvarado
- Laboratorio de Bioquímica Farmacológica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
- Unidad de Investigación Médica en Inmunología e Infectología Centro Médico Nacional La Raza, IMSS, Ciudad de México, Mexico
| | - Verónica Alacántara-Farfán
- Laboratorio de Bioquímica Farmacológica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Germán Chamorro-Cevallos
- Laboratorio de Toxicología Preclínica Departamento de Farmacia Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Ana L. Roa-Espitia
- Departamento de Biología Celular Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Instituto Politécnico Nacional, México City, Mexico
| | - Enrique O. Hernández-González
- Departamento de Biología Celular Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Instituto Politécnico Nacional, México City, Mexico
| | - Lorena Rodríguez-Páez
- Laboratorio de Bioquímica Farmacológica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| |
Collapse
|
23
|
Rossetti T, Ferreira J, Ghanem L, Buck H, Steegborn C, Myers RW, Meinke PT, Levin LR, Buck J. Assessing potency and binding kinetics of soluble adenylyl cyclase (sAC) inhibitors to maximize therapeutic potential. Front Physiol 2022; 13:1013845. [PMID: 36246105 PMCID: PMC9554468 DOI: 10.3389/fphys.2022.1013845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
In mammalian cells, 10 different adenylyl cyclases produce the ubiquitous second messenger, cyclic adenosine monophosphate (cAMP). Amongst these cAMP-generating enzymes, bicarbonate (HCO3 -)-regulated soluble adenylyl cyclase (sAC; ADCY10) is uniquely essential in sperm for reproduction. For this reason, sAC has been proposed as a potential therapeutic target for non-hormonal contraceptives for men. Here, we describe key sAC-focused in vitro assays to identify and characterize sAC inhibitors for therapeutic use. The affinity and binding kinetics of an inhibitor can greatly influence in vivo efficacy, therefore, we developed improved assays for assessing these efficacy defining features.
Collapse
Affiliation(s)
- Thomas Rossetti
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
| | - Jacob Ferreira
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
| | - Lubna Ghanem
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
| | - Hannes Buck
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
| | - Clemens Steegborn
- Department of Biochemistry, University of Bayreuth, Bayreuth, Germany
| | - Robert W. Myers
- Tri-Institutional Therapeutics Discovery Institute, New York, NY, United States
| | - Peter T. Meinke
- Tri-Institutional Therapeutics Discovery Institute, New York, NY, United States
| | - Lonny R. Levin
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
| | - Jochen Buck
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
24
|
Chen J, Ding Q, An L, Wang H. Ca2+-stimulated adenylyl cyclases as therapeutic targets for psychiatric and neurodevelopmental disorders. Front Pharmacol 2022; 13:949384. [PMID: 36188604 PMCID: PMC9523369 DOI: 10.3389/fphar.2022.949384] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
As the main secondary messengers, cyclic AMP (cAMP) and Ca2+ trigger intracellular signal transduction cascade and, in turn, regulate many aspects of cellular function in developing and mature neurons. The group I adenylyl cyclase (ADCY, also known as AC) isoforms, including ADCY1, 3, and 8 (also known as AC1, AC3, and AC8), are stimulated by Ca2+ and thus functionally positioned to integrate cAMP and Ca2+ signaling. Emerging lines of evidence have suggested the association of the Ca2+-stimulated ADCYs with bipolar disorder, schizophrenia, major depressive disorder, post-traumatic stress disorder, and autism. In this review, we discuss the molecular and cellular features as well as the physiological functions of ADCY1, 3, and 8. We further discuss the recent therapeutic development to target the Ca2+-stimulated ADCYs for potential treatments of psychiatric and neurodevelopmental disorders.
Collapse
|
25
|
Ferreira J, Levin LR, Buck J. Strategies to safely target widely expressed soluble adenylyl cyclase for contraception. Front Pharmacol 2022; 13:953903. [PMID: 36091839 PMCID: PMC9452739 DOI: 10.3389/fphar.2022.953903] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
In humans, the prototypical second messenger cyclic AMP is produced by 10 adenylyl cyclase isoforms, which are divided into two classes. Nine isoforms are G protein coupled transmembrane adenylyl cyclases (tmACs; ADCY1-9) and the 10th is the bicarbonate regulated soluble adenylyl cyclase (sAC; ADCY10). This review details why sAC is uniquely druggable and outlines ways to target sAC for novel forms of male and female contraception.
Collapse
|
26
|
Ahmed MB, Alghamdi AAA, Islam SU, Lee JS, Lee YS. cAMP Signaling in Cancer: A PKA-CREB and EPAC-Centric Approach. Cells 2022; 11:cells11132020. [PMID: 35805104 PMCID: PMC9266045 DOI: 10.3390/cells11132020] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/17/2022] [Accepted: 06/23/2022] [Indexed: 02/01/2023] Open
Abstract
Cancer is one of the most common causes of death globally. Despite extensive research and considerable advances in cancer therapy, the fundamentals of the disease remain unclear. Understanding the key signaling mechanisms that cause cancer cell malignancy may help to uncover new pharmaco-targets. Cyclic adenosine monophosphate (cAMP) regulates various biological functions, including those in malignant cells. Understanding intracellular second messenger pathways is crucial for identifying downstream proteins involved in cancer growth and development. cAMP regulates cell signaling and a variety of physiological and pathological activities. There may be an impact on gene transcription from protein kinase A (PKA) as well as its downstream effectors, such as cAMP response element-binding protein (CREB). The position of CREB downstream of numerous growth signaling pathways implies its oncogenic potential in tumor cells. Tumor growth is associated with increased CREB expression and activation. PKA can be used as both an onco-drug target and a biomarker to find, identify, and stage tumors. Exploring cAMP effectors and their downstream pathways in cancer has become easier using exchange protein directly activated by cAMP (EPAC) modulators. This signaling system may inhibit or accelerate tumor growth depending on the tumor and its environment. As cAMP and its effectors are critical for cancer development, targeting them may be a useful cancer treatment strategy. Moreover, by reviewing the material from a distinct viewpoint, this review aims to give a knowledge of the impact of the cAMP signaling pathway and the related effectors on cancer incidence and development. These innovative insights seek to encourage the development of novel treatment techniques and new approaches.
Collapse
Affiliation(s)
- Muhammad Bilal Ahmed
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea; (M.B.A.); (J.-S.L.)
| | | | - Salman Ul Islam
- Department of Pharmacy, Cecos University, Peshawar, Street 1, Sector F 5 Phase 6 Hayatabad, Peshawar 25000, Pakistan;
| | - Joon-Seok Lee
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea; (M.B.A.); (J.-S.L.)
| | - Young-Sup Lee
- BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea; (M.B.A.); (J.-S.L.)
- Correspondence: ; Tel.: +82-53-950-6353; Fax: +82-53-943-2762
| |
Collapse
|
27
|
Teixeira-Nunes M, Retailleau P, Comisso M, Deruelle V, Mechold U, Renault L. Bacterial Nucleotidyl Cyclases Activated by Calmodulin or Actin in Host Cells: Enzyme Specificities and Cytotoxicity Mechanisms Identified to Date. Int J Mol Sci 2022; 23:ijms23126743. [PMID: 35743184 PMCID: PMC9223806 DOI: 10.3390/ijms23126743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 02/06/2023] Open
Abstract
Many pathogens manipulate host cell cAMP signaling pathways to promote their survival and proliferation. Bacterial Exoenzyme Y (ExoY) toxins belong to a family of invasive, structurally-related bacterial nucleotidyl cyclases (NC). Inactive in bacteria, they use proteins that are uniquely and abundantly present in eukaryotic cells to become potent, unregulated NC enzymes in host cells. Other well-known members of the family include Bacillus anthracis Edema Factor (EF) and Bordetella pertussis CyaA. Once bound to their eukaryotic protein cofactor, they can catalyze supra-physiological levels of various cyclic nucleotide monophosphates in infected cells. Originally identified in Pseudomonas aeruginosa, ExoY-related NC toxins appear now to be more widely distributed among various γ- and β-proteobacteria. ExoY-like toxins represent atypical, poorly characterized members within the NC toxin family. While the NC catalytic domains of EF and CyaA toxins use both calmodulin as cofactor, their counterparts in ExoY-like members from pathogens of the genus Pseudomonas or Vibrio use actin as a potent cofactor, in either its monomeric or polymerized form. This is an original subversion of actin for cytoskeleton-targeting toxins. Here, we review recent advances on the different members of the NC toxin family to highlight their common and distinct functional characteristics at the molecular, cytotoxic and enzymatic levels, and important aspects that need further characterizations.
Collapse
Affiliation(s)
- Magda Teixeira-Nunes
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, 91198 Gif-sur-Yvette, France; (M.T.-N.); (M.C.)
| | - Pascal Retailleau
- Institut de Chimie des Substances Naturelles (ICSN), CNRS-UPR2301, Université Paris-Saclay, 1 Avenue de la Terrasse, 91198 Gif-sur-Yvette, France;
| | - Martine Comisso
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, 91198 Gif-sur-Yvette, France; (M.T.-N.); (M.C.)
| | - Vincent Deruelle
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, CNRS UMR 3528, Institut Pasteur, 75015 Paris, France; (V.D.); (U.M.)
| | - Undine Mechold
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, CNRS UMR 3528, Institut Pasteur, 75015 Paris, France; (V.D.); (U.M.)
| | - Louis Renault
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, 91198 Gif-sur-Yvette, France; (M.T.-N.); (M.C.)
- Correspondence:
| |
Collapse
|
28
|
Chen K, Wang S, Sun Z. In Vivo Cardiac-specific Expression of Adenylyl Cyclase 4 Gene Protects against Klotho Deficiency-induced Heart Failure. Transl Res 2022; 244:101-113. [PMID: 35114419 PMCID: PMC9119924 DOI: 10.1016/j.trsl.2022.01.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/10/2022] [Accepted: 01/26/2022] [Indexed: 01/10/2023]
Abstract
Klotho is an aging-suppressor gene. Klotho gene deficiency causes heart failure in Klotho-hypomorphic mutant (KL (-/-)) mice. RNA-seq and western blot analysis showed that adenylyl cyclase type IV (AC4) mRNA and protein expression was largely decreased in cardiomyocytes of KL (-/-) mice. The objective of this study was to investigate whether in vivo cardiac-specific expression of AC4 gene protects against Klotho deficiency-induced heart failure. Interestingly, in vivo AAV-based cardiac-specific AC4 gene expression increased left ventricular fractional shortening, ejection fraction, stroke volume, and left ventricular end-diastolic volume in KL (-/-) mice, suggesting that cardiac-specific AC4 gene expression improves Klotho deficiency-induced heart dysfunction. Cardiac-specific AC4 gene expression also decreased Klotho deficiency-induced cardiac hypertrophy. Cardiac-specific AC4 gene expression alleviated Klotho deficiency-induced cardiac fibrosis and calcification. Furthermore, cardiac-specific AC4 gene expression attenuated mitochondrial dysfunction, superoxide accumulation and cardiomyocyte apoptotic cell death. Thus, downregulation of AC4 may contribute to Klotho deficiency-induced heart failure. Mechanistically, AAV2/9-αMHC-AC4 increased cardiomyocytic cAMP levels and thus regulated the PKA-PLN-SERCA2 signal pathway, which is critical in modulating calcium flux and mitochondrial function. In conclusion, cardiac-specific AC4 gene expression protects against Klotho deficiency-induced heart failure through increasing cardiomyocytic cAMP levels, which alleviates cAMP-dependent mitochondrial dysfunction, superoxide accumulation and apoptotic cell death. AC4 regulates superoxide levels via the cAMP-PKA pathway. AC4 could be a potential therapeutic target for heart failure associated with Klotho deficiency. Heart failure is the major cause of mortality in patients with chronic kidney disease (CKD). A decrease in Klotho levels is linked to CKD.
Collapse
Affiliation(s)
- Kai Chen
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America; Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Shirley Wang
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America; Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Zhongjie Sun
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America; Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America.
| |
Collapse
|
29
|
Devasani K, Yao Y. Expression and functions of adenylyl cyclases in the CNS. Fluids Barriers CNS 2022; 19:23. [PMID: 35307032 PMCID: PMC8935726 DOI: 10.1186/s12987-022-00322-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/07/2022] [Indexed: 12/27/2022] Open
Abstract
Adenylyl cyclases (ADCYs), by generating second messenger cAMP, play important roles in various cellular processes. Their expression, regulation and functions in the CNS, however, remain largely unknown. In this review, we first introduce the classification and structure of ADCYs, followed by a discussion of the regulation of mammalian ADCYs (ADCY1-10). Next, the expression and function of each mammalian ADCY isoform are summarized in a region/cell-specific manner. Furthermore, the effects of GPCR-ADCY signaling on blood-brain barrier (BBB) integrity are reviewed. Last, current challenges and future directions are discussed. We aim to provide a succinct review on ADCYs to foster new research in the future.
Collapse
Affiliation(s)
- Karan Devasani
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd., MDC 8, Tampa, FL, 33612, USA
| | - Yao Yao
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd., MDC 8, Tampa, FL, 33612, USA.
| |
Collapse
|
30
|
Abstract
Adenylyl cyclase 9 (AC9) is a membrane-bound enzyme that converts ATP into cAMP. The enzyme is weakly activated by forskolin, fully activated by the G protein Gαs subunit and is autoinhibited by the AC9 C-terminus. Although our recent structural studies of the AC9-Gαs complex provided the framework for understanding AC9 autoinhibition, the conformational changes that AC9 undergoes in response to activator binding remains poorly understood. Here, we present the cryo-EM structures of AC9 in several distinct states: (i) AC9 bound to a nucleotide inhibitor MANT-GTP, (ii) bound to an artificial activator (DARPin C4) and MANT-GTP, (iii) bound to DARPin C4 and a nucleotide analogue ATPαS, (iv) bound to Gαs and MANT-GTP. The artificial activator DARPin C4 partially activates AC9 by binding at a site that overlaps with the Gαs binding site. Together with the previously observed occluded and forskolin-bound conformations, structural comparisons of AC9 in the four conformations described here show that secondary structure rearrangements in the region surrounding the forskolin binding site are essential for AC9 activation. Adenylyl cyclases (ACs) generate the second messenger cAMP and play an important role in cellular signaling. Here, the authors use cryo-EM to trace the conformational changes resulting from binding to partial and full activators to one of these enzymes, AC9.
Collapse
|
31
|
Zhou G, Wang Z, Han S, Chen X, Li Z, Hu X, Li Y, Gao J. Multifaceted Roles of cAMP Signaling in the Repair Process of Spinal Cord Injury and Related Combination Treatments. Front Mol Neurosci 2022; 15:808510. [PMID: 35283731 PMCID: PMC8904388 DOI: 10.3389/fnmol.2022.808510] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/26/2022] [Indexed: 01/03/2023] Open
Abstract
Spinal cord injury (SCI) results in multiple pathophysiological processes, including blood–spinal cord barrier disruption, hemorrhage/ischemia, oxidative stress, neuroinflammation, scar formation, and demyelination. These responses eventually lead to severe tissue destruction and an inhibitory environment for neural regeneration.cAMP signaling is vital for neurite outgrowth and axonal guidance. Stimulating intracellular cAMP activity significantly promotes neuronal survival and axonal regrowth after SCI.However, neuronal cAMP levels in adult CNS are relatively low and will further decrease after injury. Targeting cAMP signaling has become a promising strategy for neural regeneration over the past two decades. Furthermore, studies have revealed that cAMP signaling is involved in the regulation of glial cell function in the microenvironment of SCI, including macrophages/microglia, reactive astrocytes, and oligodendrocytes. cAMP-elevating agents in the post-injury milieu increase the cAMP levels in both neurons and glial cells and facilitate injury repair through the interplay between neurons and glial cells and ultimately contribute to better morphological and functional outcomes. In recent years, combination treatments associated with cAMP signaling have been shown to exert synergistic effects on the recovery of SCI. Agents carried by nanoparticles exhibit increased water solubility and capacity to cross the blood–spinal cord barrier. Implanted bioscaffolds and injected hydrogels are potential carriers to release agents locally to avoid systemic side effects. Cell transplantation may provide permissive matrices to synergize with the cAMP-enhanced growth capacity of neurons. cAMP can also induce the oriented differentiation of transplanted neural stem/progenitor cells into neurons and increase the survival rate of cell grafts. Emerging progress focused on cAMP compartmentation provides researchers with new perspectives to understand the complexity of downstream signaling, which may facilitate the clinical translation of strategies targeting cAMP signaling for SCI repair.
Collapse
Affiliation(s)
- Gang Zhou
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhiyan Wang
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shiyuan Han
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiaokun Chen
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhimin Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xianghui Hu
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yongning Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Department of International Medical Service, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jun Gao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- *Correspondence: Jun Gao
| |
Collapse
|
32
|
Targeting GPCRs and Their Signaling as a Therapeutic Option in Melanoma. Cancers (Basel) 2022; 14:cancers14030706. [PMID: 35158973 PMCID: PMC8833576 DOI: 10.3390/cancers14030706] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/27/2022] [Accepted: 01/27/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Sixteen G-protein-coupled receptors (GPCRs) have been involved in melanogenesis or melanomagenesis. Here, we review these GPCRs, their associated signaling, and therapies. Abstract G-protein-coupled receptors (GPCRs) serve prominent roles in melanocyte lineage physiology, with an impact at all stages of development, as well as on mature melanocyte functions. GPCR ligands are present in the skin and regulate melanocyte homeostasis, including pigmentation. The role of GPCRs in the regulation of pigmentation and, consequently, protection against external aggression, such as ultraviolet radiation, has long been established. However, evidence of new functions of GPCRs directly in melanomagenesis has been highlighted in recent years. GPCRs are coupled, through their intracellular domains, to heterotrimeric G-proteins, which induce cellular signaling through various pathways. Such signaling modulates numerous essential cellular processes that occur during melanomagenesis, including proliferation and migration. GPCR-associated signaling in melanoma can be activated by the binding of paracrine factors to their receptors or directly by activating mutations. In this review, we present melanoma-associated alterations of GPCRs and their downstream signaling and discuss the various preclinical models used to evaluate new therapeutic approaches against GPCR activity in melanoma. Recent striking advances in our understanding of the structure, function, and regulation of GPCRs will undoubtedly broaden melanoma treatment options in the future.
Collapse
|
33
|
Bolger GB. The cAMP-signaling cancers: Clinically-divergent disorders with a common central pathway. Front Endocrinol (Lausanne) 2022; 13:1024423. [PMID: 36313756 PMCID: PMC9612118 DOI: 10.3389/fendo.2022.1024423] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Accepted: 09/27/2022] [Indexed: 12/01/2022] Open
Abstract
The cAMP-signaling cancers, which are defined by functionally-significant somatic mutations in one or more elements of the cAMP signaling pathway, have an unexpectedly wide range of cell origins, clinical manifestations, and potential therapeutic options. Mutations in at least 9 cAMP signaling pathway genes (TSHR, GPR101, GNAS, PDE8B, PDE11A, PRKARA1, PRKACA, PRKACB, and CREB) have been identified as driver mutations in human cancer. Although all cAMP-signaling pathway cancers are driven by mutation(s) that impinge on a single signaling pathway, the ultimate tumor phenotype reflects interactions between five critical variables: (1) the precise gene(s) that undergo mutation in each specific tumor type; (2) the effects of specific allele(s) in any given gene; (3) mutations in modifier genes (mutational "context"); (4) the tissue-specific expression of various cAMP signaling pathway elements in the tumor stem cell; and (5) and the precise biochemical regulation of the pathway components in tumor cells. These varying oncogenic mechanisms reveal novel and important targets for drug discovery. There is considerable diversity in the "druggability" of cAMP-signaling components, with some elements (GPCRs, cAMP-specific phosphodiesterases and kinases) appearing to be prime drug candidates, while other elements (transcription factors, protein-protein interactions) are currently refractory to robust drug-development efforts. Further refinement of the precise driver mutations in individual tumors will be essential for directing priorities in drug discovery efforts that target these mutations.
Collapse
|
34
|
Molecular mechanisms of sperm motility are conserved in an early-branching metazoan. Proc Natl Acad Sci U S A 2021; 118:2109993118. [PMID: 34810263 PMCID: PMC8640785 DOI: 10.1073/pnas.2109993118] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2021] [Indexed: 01/11/2023] Open
Abstract
Reef-building corals are the keystone species of the world’s most biodiverse yet threatened marine ecosystems. Coral reproduction, critical for reef resilience, requires that coral sperm swim through the water column to reach the egg. However, little is known about the mechanisms that regulate coral sperm motility. We found here that coral sperm motility is pH dependent and that activation of motility requires signaling via the pH-sensing enzyme soluble adenylyl cyclase. This study reveals the deep conservation of a sperm activation pathway from humans to corals, presenting the first comprehensive examination of the molecular mechanisms regulating sperm motility in an early-diverging animal. These results are critical for understanding the resilience of this sensitive life stage to a changing marine environment. Efficient and targeted sperm motility is essential for animal reproductive success. Sperm from mammals and echinoderms utilize a highly conserved signaling mechanism in which sperm motility is stimulated by pH-dependent activation of the cAMP-producing enzyme soluble adenylyl cyclase (sAC). However, the presence of this pathway in early-branching metazoans has remained unexplored. Here, we found that elevating cytoplasmic pH induced a rapid burst of cAMP signaling and triggered the onset of motility in sperm from the reef-building coral Montipora capitata in a sAC-dependent manner. Expression of sAC in the mitochondrial-rich midpiece and flagellum of coral sperm support a dual role for this molecular pH sensor in regulating mitochondrial respiration and flagellar beating and thus motility. In addition, we found that additional members of the homologous signaling pathway described in echinoderms, both upstream and downstream of sAC, are expressed in coral sperm. These include the Na+/H+ exchanger SLC9C1, protein kinase A, and the CatSper Ca2+ channel conserved even in mammalian sperm. Indeed, the onset of motility corresponded with increased protein kinase A activity. Our discovery of this pathway in an early-branching metazoan species highlights the ancient origin of the pH-sAC-cAMP signaling node in sperm physiology and suggests that it may be present in many other marine invertebrate taxa for which sperm motility mechanisms remain unexplored. These results emphasize the need to better understand the role of pH-dependent signaling in the reproductive success of marine animals, particularly as climate change stressors continue to alter the physiology of corals and other marine invertebrates.
Collapse
|
35
|
Bhagirath AY, Bhatia V, Medapati MR, Singh N, Hinton M, Chelikani P, Dakshinamurti S. Critical cysteines in the functional interaction of adenylyl cyclase isoform 6 with Gαs. FASEB Bioadv 2021; 4:180-196. [PMID: 35664968 PMCID: PMC9159366 DOI: 10.1096/fba.2021-00073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 10/11/2021] [Accepted: 11/08/2021] [Indexed: 11/17/2022] Open
Abstract
Activation of adenylyl cyclases (ACs) by G‐protein Gαs catalyzes the production of cyclic adenosine monophosphate (cAMP), a key second messenger that regulates diverse physiological responses. There are 10 AC isoforms present in humans, with AC5 and AC6 proposed to play vital roles in cardiac function. We have previously shown that under hypoxic conditions, AC6 is amenable to post‐translational modification by nitrosylation, resulting in decreased AC catalytic activity. Using a computational model of the AC6–Gαs complex, we predicted key nitrosylation‐amenable cysteine residues involved in the interaction of AC6 with Gαs and pursued a structure–function analysis of these cysteine residues in both AC6 and Gαs. Our results based on site‐directed mutagenesis of AC6 and Gαs, a constitutively active Gαs, AC activity, and live cell intracellular cAMP assays suggest that Cys1004 in AC6 (subunit C2) and Cys237 in Gαs are present at the AC–Gαs interface and are important for the activation of AC6 by Gαs. We further provide mechanistic evidence to show that mutating Cys 1004 in the second catalytic domain of AC6 makes it amenable to inhibition by Gαi, which may account for decreased functional activity of AC6 when this residue is unavailable.
Collapse
Affiliation(s)
- Anjali Y. Bhagirath
- Biology of Breathing Theme Children's Hospital Research Institute of Manitoba (CHRIM) Winnipeg Manitoba Canada
- Department of Oral Biology University of Manitoba Winnipeg Manitoba Canada
| | - Vikram Bhatia
- Biology of Breathing Theme Children's Hospital Research Institute of Manitoba (CHRIM) Winnipeg Manitoba Canada
- Department of Oral Biology University of Manitoba Winnipeg Manitoba Canada
| | | | - Nisha Singh
- Department of Oral Biology University of Manitoba Winnipeg Manitoba Canada
| | - Martha Hinton
- Biology of Breathing Theme Children's Hospital Research Institute of Manitoba (CHRIM) Winnipeg Manitoba Canada
- Department of Physiology and Pathophysiology University of Manitoba Winnipeg Manitoba Canada
| | - Prashen Chelikani
- Biology of Breathing Theme Children's Hospital Research Institute of Manitoba (CHRIM) Winnipeg Manitoba Canada
- Department of Oral Biology University of Manitoba Winnipeg Manitoba Canada
| | - Shyamala Dakshinamurti
- Biology of Breathing Theme Children's Hospital Research Institute of Manitoba (CHRIM) Winnipeg Manitoba Canada
- Department of Physiology and Pathophysiology University of Manitoba Winnipeg Manitoba Canada
- Department of Pediatrics University of Manitoba Winnipeg Manitoba Canada
| |
Collapse
|
36
|
Gonzaga de França Lopes L, Gouveia Júnior FS, Karine Medeiros Holanda A, Maria Moreira de Carvalho I, Longhinotti E, Paulo TF, Abreu DS, Bernhardt PV, Gilles-Gonzalez MA, Cirino Nogueira Diógenes I, Henrique Silva Sousa E. Bioinorganic systems responsive to the diatomic gases O2, NO, and CO: From biological sensors to therapy. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
37
|
Balbach M, Fushimi M, Huggins DJ, Steegborn C, Meinke PT, Levin LR, Buck J. Optimization of lead compounds into on-demand, nonhormonal contraceptives: leveraging a public-private drug discovery institute collaboration†. Biol Reprod 2021; 103:176-182. [PMID: 32307523 PMCID: PMC7401349 DOI: 10.1093/biolre/ioaa052] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/08/2020] [Accepted: 04/15/2020] [Indexed: 12/20/2022] Open
Abstract
Efforts to develop new male or female nonhormonal, orally available contraceptives assume that to be effective and safe, targets must be (1) essential for fertility; (2) amenable to targeting by small-molecule inhibitors; and (3) restricted to the germline. In this perspective, we question the third assumption and propose that despite its wide expression, soluble adenylyl cyclase (sAC: ADCY10), which is essential for male fertility, is a valid target. We hypothesize that an acute-acting sAC inhibitor may provide orally available, on-demand, nonhormonal contraception for men without adverse, mechanism-based effects. To test this concept, we describe a collaboration between academia and the unique capabilities of a public-private drug discovery institute.
Collapse
Affiliation(s)
- Melanie Balbach
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Makoto Fushimi
- Tri-Institutional Therapeutics Discovery Institute, New York, NY, USA
| | - David J Huggins
- Tri-Institutional Therapeutics Discovery Institute, New York, NY, USA.,Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Clemens Steegborn
- Department of Biochemistry, University of Bayreuth, Bayreuth, Germany
| | - Peter T Meinke
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA.,Tri-Institutional Therapeutics Discovery Institute, New York, NY, USA
| | - Lonny R Levin
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Jochen Buck
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
38
|
Jakobsen E, Andersen JV, Christensen SK, Siamka O, Larsen MR, Waagepetersen HS, Aldana BI, Bak LK. Pharmacological inhibition of mitochondrial soluble adenylyl cyclase in astrocytes causes activation of AMP-activated protein kinase and induces breakdown of glycogen. Glia 2021; 69:2828-2844. [PMID: 34378239 DOI: 10.1002/glia.24072] [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: 04/13/2021] [Revised: 06/28/2021] [Accepted: 07/30/2021] [Indexed: 12/17/2022]
Abstract
Mobilization of astrocyte glycogen is key for processes such as synaptic plasticity and memory formation but the link between neuronal activity and glycogen breakdown is not fully known. Activation of cytosolic soluble adenylyl cyclase (sAC) in astrocytes has been suggested to link neuronal depolarization and glycogen breakdown partly based on experiments employing pharmacological inhibition of sAC. However, several studies have revealed that sAC located within mitochondria is a central regulator of respiration and oxidative phosphorylation. Thus, pharmacological sAC inhibition is likely to affect both cytosolic and mitochondrial sAC and if bioenergetic readouts are studied, the observed effects are likely to stem from inhibition of mitochondrial rather than cytosolic sAC. Here, we report that a pharmacologically induced inhibition of sAC activity lowers mitochondrial respiration, induces phosphorylation of the metabolic master switch AMP-activated protein kinase (AMPK), and decreases glycogen stores in cultured primary murine astrocytes. From these data and our discussion of the literature, mitochondrial sAC emerges as a key regulator of astrocyte bioenergetics. Lastly, we discuss the challenges of investigating the functional and metabolic roles of cytosolic versus mitochondrial sAC in astrocytes employing the currently available pharmacological tool compounds.
Collapse
Affiliation(s)
- Emil Jakobsen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens V Andersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sofie K Christensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Olga Siamka
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Denmark
| | - Helle S Waagepetersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Blanca I Aldana
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lasse K Bak
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
39
|
Aldana A, Carneiro J, Martínez-Mekler G, Darszon A. Discrete Dynamic Model of the Mammalian Sperm Acrosome Reaction: The Influence of Acrosomal pH and Physiological Heterogeneity. Front Physiol 2021; 12:682790. [PMID: 34349664 PMCID: PMC8328089 DOI: 10.3389/fphys.2021.682790] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/28/2021] [Indexed: 01/31/2023] Open
Abstract
The acrosome reaction (AR) is an exocytotic process essential for mammalian fertilization. It involves diverse physiological changes (biochemical, biophysical, and morphological) that culminate in the release of the acrosomal content to the extracellular medium as well as a reorganization of the plasma membrane (PM) that allows sperm to interact and fuse with the egg. In spite of many efforts, there are still important pending questions regarding the molecular mechanism regulating the AR. Particularly, the contribution of acrosomal alkalinization to AR triggering physiological conditions is not well understood. Also, the dependence of the proportion of sperm capable of undergoing AR on the physiological heterogeneity within a sperm population has not been studied. Here, we present a discrete mathematical model for the human sperm AR based on the physiological interactions among some of the main components of this complex exocytotic process. We show that this model can qualitatively reproduce diverse experimental results, and that it can be used to analyze how acrosomal pH (pH a ) and cell heterogeneity regulate AR. Our results confirm that a pH a increase can on its own trigger AR in a subpopulation of sperm, and furthermore, it indicates that this is a necessary step to trigger acrosomal exocytosis through progesterone, a known natural inducer of AR. Most importantly, we show that the proportion of sperm undergoing AR is directly related to the detailed structure of the population physiological heterogeneity.
Collapse
Affiliation(s)
- Andrés Aldana
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jorge Carneiro
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova, Oeiras, Portugal
| | - Gustavo Martínez-Mekler
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Instituto de Ciencias Físicas, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Alberto Darszon
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| |
Collapse
|
40
|
Baiocchi L, Lenci I, Milana M, Kennedy L, Sato K, Zhang W, Ekser B, Ceci L, Meadows V, Glaser S, Alpini G, Francis H. Cyclic AMP Signaling in Biliary Proliferation: A Possible Target for Cholangiocarcinoma Treatment? Cells 2021; 10:1692. [PMID: 34359861 PMCID: PMC8303798 DOI: 10.3390/cells10071692] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/30/2021] [Accepted: 06/30/2021] [Indexed: 12/12/2022] Open
Abstract
Cholangiocarcinoma is a lethal disease with scarce response to current systemic therapy. The rare occurrence and large heterogeneity of this cancer, together with poor knowledge of its molecular mechanisms, are elements contributing to the difficulties in finding an appropriate cure. Cholangiocytes (and their cellular precursors) are considered the liver component giving rise to cholangiocarcinoma. These cells respond to several hormones, neuropeptides and molecular stimuli employing the cAMP/PKA system for the translation of messages in the intracellular space. For instance, in physiological conditions, stimulation of the secretin receptor determines an increase of intracellular levels of cAMP, thus activating a series of molecular events, finally determining in bicarbonate-enriched choleresis. However, activation of the same receptor during cholangiocytes' injury promotes cellular growth again, using cAMP as the second messenger. Since several scientific pieces of evidence link cAMP signaling system to cholangiocytes' proliferation, the possible changes of this pathway during cancer growth also seem relevant. In this review, we summarize the current findings regarding the cAMP pathway and its role in biliary normal and neoplastic cell proliferation. Perspectives for targeting the cAMP machinery in cholangiocarcinoma therapy are also discussed.
Collapse
Affiliation(s)
- Leonardo Baiocchi
- Hepatology Unit, University of Tor Vergata, 00133 Rome, Italy; (L.B.); (I.L.); (M.M.)
| | - Ilaria Lenci
- Hepatology Unit, University of Tor Vergata, 00133 Rome, Italy; (L.B.); (I.L.); (M.M.)
| | - Martina Milana
- Hepatology Unit, University of Tor Vergata, 00133 Rome, Italy; (L.B.); (I.L.); (M.M.)
| | - Lindsey Kennedy
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, IN 46202, USA; (L.K.); (K.S.); (L.C.); (V.M.)
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| | - Keisaku Sato
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, IN 46202, USA; (L.K.); (K.S.); (L.C.); (V.M.)
| | - Wenjun Zhang
- Division of Transplant Surgery, Department of Surgery, Indiana University, Indianapolis, IN 46202, USA; (W.Z.); (B.E.)
| | - Burcin Ekser
- Division of Transplant Surgery, Department of Surgery, Indiana University, Indianapolis, IN 46202, USA; (W.Z.); (B.E.)
| | - Ludovica Ceci
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, IN 46202, USA; (L.K.); (K.S.); (L.C.); (V.M.)
| | - Vik Meadows
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, IN 46202, USA; (L.K.); (K.S.); (L.C.); (V.M.)
| | - Shannon Glaser
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX 77807, USA;
| | - Gianfranco Alpini
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, IN 46202, USA; (L.K.); (K.S.); (L.C.); (V.M.)
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| | - Heather Francis
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, IN 46202, USA; (L.K.); (K.S.); (L.C.); (V.M.)
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| |
Collapse
|
41
|
Cross-Talk Between the Adenylyl Cyclase/cAMP Pathway and Ca 2+ Homeostasis. Rev Physiol Biochem Pharmacol 2021; 179:73-116. [PMID: 33398503 DOI: 10.1007/112_2020_55] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cyclic AMP and Ca2+ are the first second or intracellular messengers identified, unveiling the cellular mechanisms activated by a plethora of extracellular signals, including hormones. Cyclic AMP generation is catalyzed by adenylyl cyclases (ACs), which convert ATP into cAMP and pyrophosphate. By the way, Ca2+, as energy, can neither be created nor be destroyed; Ca2+ can only be transported, from one compartment to another, or chelated by a variety of Ca2+-binding molecules. The fine regulation of cytosolic concentrations of cAMP and free Ca2+ is crucial in cell function and there is an intimate cross-talk between both messengers to fine-tune the cellular responses. Cancer is a multifactorial disease resulting from a combination of genetic and environmental factors. Frequent cases of cAMP and/or Ca2+ homeostasis remodeling have been described in cancer cells. In those tumoral cells, cAMP and Ca2+ signaling plays a crucial role in the development of hallmarks of cancer, including enhanced proliferation and migration, invasion, apoptosis resistance, or angiogenesis. This review summarizes the cross-talk between the ACs/cAMP and Ca2+ intracellular pathways with special attention to the functional and reciprocal regulation between Orai1 and AC8 in normal and cancer cells.
Collapse
|
42
|
Li H, Guan Y, Han C, Zhang Y, Liu Q, Wei W, Ma Y. The pathogenesis, models and therapeutic advances of primary biliary cholangitis. Biomed Pharmacother 2021; 140:111754. [PMID: 34044277 DOI: 10.1016/j.biopha.2021.111754] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/30/2022] Open
Abstract
Primary biliary cholangitis (PBC) is an autoimmune disease characterized by the destruction of intrahepatic small bile ducts and the presence of antimitochondrial antibody (AMA), eventually progresses to liver fibrosis and cirrhosis. Genetic predisposition and environmental factors are involved in the occurrence of PBC, and the epitopes exposure and the imbalance of autoimmune tolerance are the last straw. The apoptosis of biliary epithelial cell (BEC) leads to the release of autoantigen epitopes, which activate the immune system, and the disorder of innate and adaptive immunity eventually leads to the start of disease. Animal models have unique advantages in investigating the pathogenesis and drug exploitation of PBC. Multiple models have been reported, and spontaneous model and induced model have been widely used in relevant research of PBC in recent years. Currently, the only drugs licensed for PBC are ursodesoxycholic acid (UDCA) and obeticholic acid (OCA). In the last few years, as the learned more about the pathogenesis of PBC, more and more targets have been discovered, and multiple targeted drugs are being in developed. In this review, the pathogenesis, murine models and treatment strategies of PBC were summarized, and the current research status was discussed to provide insights for the further study of PBC.
Collapse
Affiliation(s)
- Hao Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Yanling Guan
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Chenchen Han
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Yu Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Qian Liu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China.
| | - Yang Ma
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, China.
| |
Collapse
|
43
|
Vega Hissi EG, De Costa Guardamagna AB, Garro AD, Falcon CR, Anderluh M, Tomašič T, Kikelj D, Yaneff A, Davio CA, Enriz RD, Zurita AR. A Potent N-(piperidin-4-yl)-1H-pyrrole-2-carboxamide Inhibitor of Adenylyl Cyclase of G. lamblia: Biological Evaluation and Molecular Modelling Studies. ChemMedChem 2021; 16:2094-2105. [PMID: 33783977 DOI: 10.1002/cmdc.202100037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/26/2021] [Indexed: 11/06/2022]
Abstract
In this work, we report a derivative of N-(piperidin-4-yl)-1H-pyrrole-2-carboxamide as a new inhibitor for adenylyl cyclase of Giardia lamblia which was obtained from a study using structural data of the nucleotidyl cyclase 1 (gNC1) of this parasite. For such a study, we developed a model for this specific enzyme by using homology techniques, which is the first model reported for gNC1 of G. lamblia. Our studies show that the new inhibitor has a competitive mechanism of action against this enzyme. 2-Hydroxyestradiol was used as the reference compound for comparative studies. Results in this work are important from two points of view. on the one hand, an experimentally corroborated model for gNC1 of G. lamblia obtained by molecular modelling is presented; on the other hand, the new inhibitor obtained is an undoubtedly excellent starting structure for the development of new metabolic inhibitors for G. lamblia.
Collapse
Affiliation(s)
- Esteban G Vega Hissi
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700, San Luis, Argentina
| | - Antonella B De Costa Guardamagna
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700, San Luis, Argentina
| | - Adriana D Garro
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700, San Luis, Argentina
| | - Cristian R Falcon
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700, San Luis, Argentina
| | - Marko Anderluh
- Department of Medicinal Chemistry, University of Ljubljana, Faculty of Pharmacy Askerceva, cesta 7, 1000, Ljubljana, Slovenia
| | - Tihomir Tomašič
- Department of Medicinal Chemistry, University of Ljubljana, Faculty of Pharmacy Askerceva, cesta 7, 1000, Ljubljana, Slovenia
| | - Danijel Kikelj
- Department of Medicinal Chemistry, University of Ljubljana, Faculty of Pharmacy Askerceva, cesta 7, 1000, Ljubljana, Slovenia
| | - Agustín Yaneff
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, C1113, AAD, Buenos Aires, Argentina
| | - Carlos A Davio
- Instituto de Investigaciones Farmacológicas (ININFA-UBA-CONICET), Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, C1113, AAD, Buenos Aires, Argentina
| | - Ricardo D Enriz
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700, San Luis, Argentina
| | - Adolfo R Zurita
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700, San Luis, Argentina
| |
Collapse
|
44
|
Effect of Soluble Adenylyl Cyclase (ADCY10) Inhibitors on the LH-Stimulated cAMP Synthesis in Mltc-1 Leydig Cell Line. Int J Mol Sci 2021; 22:ijms22094641. [PMID: 33924969 PMCID: PMC8125623 DOI: 10.3390/ijms22094641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/20/2021] [Accepted: 04/26/2021] [Indexed: 11/25/2022] Open
Abstract
In contrast to all transmembrane adenylyl cyclases except ADCY9, the cytosolic soluble adenylyl cyclase (ADCY10) is insensitive to forskolin stimulation and is uniquely modulated by calcium and bicarbonate ions. In the present paper, we focus on ADCY10 localization and a kinetic analysis of intracellular cAMP accumulation in response to human LH in the absence or presence of four different ADCY10 inhibitors (KH7, LRE1, 2-CE and 4-CE) in MTLC-1 cells. ADCY10 was immuno-detected in the cytoplasm of MLTC-1 cells and all four inhibitors were found to inhibit LH-stimulated cAMP accumulation and progesterone level in MLTC-1 and testosterone level primary Leydig cells. Interestingly, similar inhibitions were also evidenced in mouse testicular Leydig cells. In contrast, the tmAC-specific inhibitors ddAdo3′ and ddAdo5′, even at high concentration, exerted weak or no inhibition on cAMP accumulation, suggesting an important role of ADCY10 relative to tmACs in the MLTC-1 response to LH. The strong synergistic effect of HCO3− under LH stimulation further supports the involvement of ADCY10 in the response to LH.
Collapse
|
45
|
Salmerón C, Harter TS, Kwan GT, Roa JN, Blair SD, Rummer JL, Shiels HA, Goss GG, Wilson RW, Tresguerres M. Molecular and biochemical characterization of the bicarbonate-sensing soluble adenylyl cyclase from a bony fish, the rainbow trout Oncorhynchus mykiss. Interface Focus 2021; 11:20200026. [PMID: 33633829 DOI: 10.1098/rsfs.2020.0026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2020] [Indexed: 12/14/2022] Open
Abstract
Soluble adenylyl cyclase (sAC) is a HC O 3 - -stimulated enzyme that produces the ubiquitous signalling molecule cAMP, and deemed an evolutionarily conserved acid-base sensor. However, its presence is not yet confirmed in bony fishes, the most abundant and diverse of vertebrates. Here, we identified sAC genes in various cartilaginous, ray-finned and lobe-finned fish species. Next, we focused on rainbow trout sAC (rtsAC) and identified 20 potential alternative spliced mRNAs coding for protein isoforms ranging in size from 28 to 186 kDa. Biochemical and kinetic analyses on purified recombinant rtsAC protein determined stimulation by HC O 3 - at physiologically relevant levels for fish internal fluids (EC50 ∼ 7 mM). rtsAC activity was sensitive to KH7, LRE1, and DIDS (established inhibitors of sAC from other organisms), and insensitive to forskolin and 2,5-dideoxyadenosine (modulators of transmembrane adenylyl cyclases). Western blot and immunocytochemistry revealed high rtsAC expression in gill ion-transporting cells, hepatocytes, red blood cells, myocytes and cardiomyocytes. Analyses in the cell line RTgill-W1 suggested that some of the longer rtsAC isoforms may be preferentially localized in the nucleus, the Golgi apparatus and podosomes. These results indicate that sAC is poised to mediate multiple acid-base homeostatic responses in bony fishes, and provide cues about potential novel functions in mammals.
Collapse
Affiliation(s)
- Cristina Salmerón
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, USA.,Department of Pharmacology, University of California San Diego, San Diego, CA, USA
| | - Till S Harter
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, USA
| | - Garfield T Kwan
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, USA
| | - Jinae N Roa
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, USA
| | - Salvatore D Blair
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada.,Department of Biology, Winthrop University, Rock Hill, SC, USA
| | - Jodie L Rummer
- Australian Research Council Centre of Excellence for Coral Reef Studies, James Cook University, Townsville, Queensland, Australia
| | - Holly A Shiels
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK
| | - Greg G Goss
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Rod W Wilson
- Department of Biosciences, University of Exeter, Exeter, UK
| | - Martin Tresguerres
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
46
|
Rossetti T, Jackvony S, Buck J, Levin LR. Bicarbonate, carbon dioxide and pH sensing via mammalian bicarbonate-regulated soluble adenylyl cyclase. Interface Focus 2021; 11:20200034. [PMID: 33633833 PMCID: PMC7898154 DOI: 10.1098/rsfs.2020.0034] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2020] [Indexed: 12/11/2022] Open
Abstract
Soluble adenylyl cyclase (sAC; ADCY10) is a bicarbonate (HCO3 -)-regulated enzyme responsible for the generation of cyclic adenosine monophosphate (cAMP). sAC is distributed throughout the cell and within organelles and, as such, plays a role in numerous cellular signalling pathways. Carbonic anhydrases (CAs) nearly instantaneously equilibrate HCO3 -, protons and carbon dioxide (CO2); because of the ubiquitous presence of CAs within cells, HCO3 --regulated sAC can respond to changes in any of these factors. Thus, sAC can function as a physiological HCO3 -/CO2/pH sensor. Here, we outline examples where we have shown that sAC responds to changes in HCO3 -, CO2 or pH to regulate diverse physiological functions.
Collapse
Affiliation(s)
- Tom Rossetti
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
- Graduate Program in Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Stephanie Jackvony
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
- Graduate Program in Neuroscience, Weill Cornell Medicine, New York, NY 10065, USA
| | - Jochen Buck
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lonny R. Levin
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
47
|
Zhang X, Yu K, Ma L, Qian Z, Tian X, Miao Y, Niu Y, Xu X, Guo S, Yang Y, Wang Z, Xue X, Gu C, Fang W, Sun J, Yu Y, Wang J. Endogenous glutamate determines ferroptosis sensitivity via ADCY10-dependent YAP suppression in lung adenocarcinoma. Am J Cancer Res 2021; 11:5650-5674. [PMID: 33897873 PMCID: PMC8058707 DOI: 10.7150/thno.55482] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 03/03/2021] [Indexed: 12/22/2022] Open
Abstract
Rationale: Ferroptosis, a newly identified form of regulated cell death, can be induced following the inhibition of cystine-glutamate antiporter system XC- because of the impaired uptake of cystine. However, the outcome following the accumulation of endogenous glutamate in lung adenocarcinoma (LUAD) has not yet been determined. Yes-associated protein (YAP) is sustained by the hexosamine biosynthesis pathway (HBP)-dependent O-linked beta-N-acetylglucosaminylation (O-GlcNAcylation), and glutamine-fructose-6-phosphate transaminase (GFPT1), the rate-limiting enzyme of the HBP, can be phosphorylated and inhibited by adenylyl cyclase (ADCY)-mediated activation of protein kinase A (PKA). However, whether accumulated endogenous glutamate determines ferroptosis sensitivity by influencing the ADCY/PKA/HBP/YAP axis in LUAD cells is not understood. Methods: Cell viability, cell death and the generation of lipid reactive oxygen species (ROS) and malondialdehyde (MDA) were measured to evaluate the responses to the induction of ferroptosis following the inhibition of system XC-. Tandem mass tags (TMTs) were employed to explore potential factors critical for the ferroptosis sensitivity of LUAD cells. Immunoblotting (IB) and quantitative RT-PCR (qPCR) were used to analyze protein and mRNA expression. Co-immunoprecipitation (co-IP) assays were performed to identify protein-protein interactions and posttranslational modifications. Metabolite levels were measured using the appropriate kits. Transcriptional regulation was evaluated using a luciferase reporter assay, chromatin immunoprecipitation (ChIP), and electrophoretic mobility shift assay (EMSA). Drug administration and limiting dilution cell transplantation were performed with cell-derived xenograft (CDX) and patient-derived xenograft (PDX) mouse models. The associations among clinical outcome, drug efficacy and ADCY10 expression were determined based on data from patients who underwent curative surgery and evaluated with patient-derived primary LUAD cells and tissues. Results: The accumulation of endogenous glutamate following system XC- inhibition has been shown to determine ferroptosis sensitivity by suppressing YAP in LUAD cells. YAP O-GlcNAcylation and expression cannot be sustained in LUAD cells upon impairment of GFPT1. Thus, Hippo pathway-like phosphorylation and ubiquitination of YAP are enhanced. ADCY10 acts as a key downstream target and diversifies the effects of glutamate on the PKA-dependent suppression of GFPT1. We also discovered that the protumorigenic and proferroptotic effects of ADCY10 are mediated separately. Advanced-stage LUADs with high ADCY10 expression are sensitive to ferroptosis. Moreover, LUAD cells with acquired therapy resistance are also prone to higher ADCY10 expression and are more likely to respond to ferroptosis. Finally, a varying degree of secondary labile iron increase is caused by the failure to sustain YAP-stimulated transcriptional compensation for ferritin at later stages further explains why ferroptosis sensitivity varies among LUAD cells. Conclusions: Endogenous glutamate is critical for ferroptosis sensitivity following the inhibition of system XC- in LUAD cells, and ferroptosis-based treatment is a good choice for LUAD patients with later-stage and/or therapy-resistant tumors.
Collapse
|
48
|
Chang HW, Frey G, Liu H, Xing C, Steinman L, Boyle WJ, Short JM. Generating tumor-selective conditionally active biologic anti-CTLA4 antibodies via protein-associated chemical switches. Proc Natl Acad Sci U S A 2021; 118:e2020606118. [PMID: 33627407 PMCID: PMC7936328 DOI: 10.1073/pnas.2020606118] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Anticytotoxic T lymphocyte-associated protein 4 (CTLA4) antibodies have shown potent antitumor activity, but systemic immune activation leads to severe immune-related adverse events, limiting clinical usage. We developed novel, conditionally active biologic (CAB) anti-CTLA4 antibodies that are active only in the acidic tumor microenvironment. In healthy tissue, this binding is reversibly inhibited by a novel mechanism using physiological chemicals as protein-associated chemical switches (PaCS). No enzymes or potentially immunogenic covalent modifications to the antibody are required for activation in the tumor. The novel anti-CTLA4 antibodies show similar efficacy in animal models compared to an analog of a marketed anti-CTLA4 biologic, but have markedly reduced toxicity in nonhuman primates (in combination with an anti-PD1 checkpoint inhibitor), indicating a widened therapeutic index (TI). The PaCS encompass mechanisms that are applicable to a wide array of antibody formats (e.g., ADC, bispecifics) and antigens. Examples shown here include antibodies to EpCAM, Her2, Nectin4, CD73, and CD3. Existing antibodies can be engineered readily to be made sensitive to PaCS, and the inhibitory activity can be optimized for each antigen's varying expression level and tissue distribution. PaCS can modulate diverse physiological molecular interactions and are applicable to various pathologic conditions, enabling differential CAB antibody activities in normal versus disease microenvironments.
Collapse
MESH Headings
- 5'-Nucleotidase/antagonists & inhibitors
- 5'-Nucleotidase/genetics
- 5'-Nucleotidase/immunology
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal, Humanized/chemistry
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Neoplasm/chemistry
- Antibodies, Neoplasm/pharmacology
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/genetics
- B7-H1 Antigen/immunology
- Bicarbonates/chemistry
- CD3 Complex/antagonists & inhibitors
- CD3 Complex/genetics
- CD3 Complex/immunology
- CTLA-4 Antigen/antagonists & inhibitors
- CTLA-4 Antigen/genetics
- CTLA-4 Antigen/immunology
- Cell Adhesion Molecules/antagonists & inhibitors
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/immunology
- Colonic Neoplasms/genetics
- Colonic Neoplasms/immunology
- Colonic Neoplasms/pathology
- Colonic Neoplasms/therapy
- Epithelial Cell Adhesion Molecule/antagonists & inhibitors
- Epithelial Cell Adhesion Molecule/genetics
- Epithelial Cell Adhesion Molecule/immunology
- GPI-Linked Proteins/antagonists & inhibitors
- GPI-Linked Proteins/genetics
- GPI-Linked Proteins/immunology
- Gene Expression
- Humans
- Hydrogen Sulfide/chemistry
- Hydrogen-Ion Concentration
- Immunotherapy/methods
- Macaca fascicularis
- Mice
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/genetics
- Neoplasm Proteins/immunology
- Protein Engineering/methods
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/pathology
- Tumor Burden/drug effects
- Tumor Microenvironment/drug effects
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
| | | | | | | | - Lawrence Steinman
- Stanford University School of Medicine, Stanford University, Stanford, CA 94305
| | | | | |
Collapse
|
49
|
Soluble adenylyl cyclase regulates the cytosolic NADH/NAD + redox state and the bioenergetic switch between glycolysis and oxidative phosphorylation. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2021; 1862:148367. [PMID: 33412125 DOI: 10.1016/j.bbabio.2020.148367] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 12/11/2020] [Accepted: 12/19/2020] [Indexed: 12/22/2022]
Abstract
The evolutionarily conserved soluble adenylyl cyclase (sAC, ADCY10) mediates cAMP signaling exclusively in intracellular compartments. Because sAC activity is sensitive to local concentrations of ATP, bicarbonate, and free Ca2+, sAC is potentially an important metabolic sensor. Nonetheless, little is known about how sAC regulates energy metabolism in intact cells. In this study, we demonstrated that both pharmacological and genetic suppression of sAC resulted in increased lactate secretion and decreased pyruvate secretion in multiple cell lines and primary cultures of mouse hepatocytes and cholangiocytes. The increased extracellular lactate-to-pyruvate ratio upon sAC suppression reflected an increased cytosolic free [NADH]/[NAD+] ratio, which was corroborated by using the NADH/NAD+ redox biosensor Peredox-mCherry. Mechanistic studies in permeabilized HepG2 cells showed that sAC inhibition specifically suppressed complex I of the mitochondrial respiratory chain. A survey of cAMP effectors revealed that only selective inhibition of exchange protein activated by cAMP 1 (Epac1), but not protein kinase A (PKA) or Epac2, suppressed complex I-dependent respiration and significantly increased the cytosolic NADH/NAD+ redox state. Analysis of the ATP production rate and the adenylate energy charge showed that inhibiting sAC reciprocally affects ATP production by glycolysis and oxidative phosphorylation while maintaining cellular energy homeostasis. In conclusion, our study shows that, via the regulation of complex I-dependent mitochondrial respiration, sAC-Epac1 signaling regulates the cytosolic NADH/NAD+ redox state, and coordinates oxidative phosphorylation and glycolysis to maintain cellular energy homeostasis. As such, sAC is effectively a bioenergetic switch between aerobic glycolysis and oxidative phosphorylation at the post-translational level.
Collapse
|
50
|
Zhang H, Kong Q, Wang J, Jiang Y, Hua H. Complex roles of cAMP-PKA-CREB signaling in cancer. Exp Hematol Oncol 2020; 9:32. [PMID: 33292604 PMCID: PMC7684908 DOI: 10.1186/s40164-020-00191-1] [Citation(s) in RCA: 273] [Impact Index Per Article: 54.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 11/19/2020] [Indexed: 02/08/2023] Open
Abstract
Cyclic adenosine monophosphate (cAMP) is the first discovered second messenger, which plays pivotal roles in cell signaling, and regulates many physiological and pathological processes. cAMP can regulate the transcription of various target genes, mainly through protein kinase A (PKA) and its downstream effectors such as cAMP-responsive element binding protein (CREB). In addition, PKA can phosphorylate many kinases such as Raf, GSK3 and FAK. Aberrant cAMP-PKA signaling is involved in various types of human tumors. Especially, cAMP signaling may have both tumor-suppressive and tumor-promoting roles depending on the tumor types and context. cAMP-PKA signaling can regulate cancer cell growth, migration, invasion and metabolism. This review highlights the important roles of cAMP-PKA-CREB signaling in tumorigenesis. The potential strategies to target this pathway for cancer therapy are also discussed.
Collapse
Affiliation(s)
- Hongying Zhang
- Laboratory of Oncogene, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Qingbin Kong
- Laboratory of Oncogene, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jiao Wang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yangfu Jiang
- Laboratory of Oncogene, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Hui Hua
- Laboratory of Stem Cell Biology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|