1
|
Coelho FC, Cerchiaro G, Araújo SES, Daher JPL, Cardoso SA, Coelho GF, Guimarães AG. Is There a Connection between the Metabolism of Copper, Sulfur, and Molybdenum in Alzheimer’s Disease? New Insights on Disease Etiology. Int J Mol Sci 2022; 23:ijms23147935. [PMID: 35887282 PMCID: PMC9324259 DOI: 10.3390/ijms23147935] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/06/2022] [Accepted: 07/11/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer’s disease (AD) and other forms of dementia was ranked 3rd in both the Americas and Europe in 2019 in a World Health Organization (WHO) publication listing the leading causes of death and disability worldwide. Copper (Cu) imbalance has been reported in AD and increasing evidence suggests metal imbalance, including molybdenum (Mo), as a potential link with AD occurrence.We conducted an extensive literature review of the last 60 years of research on AD and its relationship with Cu, sulfur (S), and Mo at out of range levels.Weanalyzed the interactions among metallic elements’ metabolisms;Cu and Mo are biological antagonists, Mo is a sulfite oxidase and xanthine oxidase co-factor, and their low activities impair S metabolism and reduce uric acid, respectively. We found significant evidence in the literature of a new potential mechanism linking Cu imbalance to Mo and S abnormalities in AD etiology: under certain circumstances, the accumulation of Cu not bound to ceruloplasmin might affect the transport of Mo outside the blood vessels, causing a mild Mo deficiency that might lowerthe activity of Mo and S enzymes essential for neuronal activity. The current review provides an updated discussion of the plausible mechanisms combining Cu, S, and Mo alterations in AD.
Collapse
Affiliation(s)
- Fábio Cunha Coelho
- Laboratório de Fitotecnia (LFIT), Universidade Estadual do Norte Fluminense Darcy Ribeiro—UENF, Campos dos Goytacazes 28013-602, Brazil
- Correspondence: ; Tel.: +55-22-998509469
| | - Giselle Cerchiaro
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Av. dos Estados, 5001, Bl. B, Santo André 09210-170, Brazil;
| | - Sheila Espírito Santo Araújo
- Laboratório de Biologia Celular e Tecidual (LBCT), Universidade Estadual do Norte Fluminense Darcy Ribeiro—UENF, Campos dos Goytacazes 28013-602, Brazil; (S.E.S.A.); (A.G.G.)
| | - João Paulo Lima Daher
- Departamento de Patologia, Hospital Universitário Antônio Pedro, Universidade Federal Fluminense, Niterói 24210-350, Brazil;
| | - Silvia Almeida Cardoso
- Departamento de Medicina e Enfermagem (DEM), Universidade Federal de Viçosa, Viçosa 36579-900, Brazil;
| | - Gustavo Fialho Coelho
- Instituto de Ciências Médicas, Universidade Federal do Rio de Janeiro, Macaé 27930-560, Brazil;
| | - Arthur Giraldi Guimarães
- Laboratório de Biologia Celular e Tecidual (LBCT), Universidade Estadual do Norte Fluminense Darcy Ribeiro—UENF, Campos dos Goytacazes 28013-602, Brazil; (S.E.S.A.); (A.G.G.)
| |
Collapse
|
2
|
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: 28] [Impact Index Per Article: 9.3] [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
|
3
|
Di Benedetto G, Iannucci LF, Surdo NC, Zanin S, Conca F, Grisan F, Gerbino A, Lefkimmiatis K. Compartmentalized Signaling in Aging and Neurodegeneration. Cells 2021; 10:464. [PMID: 33671541 PMCID: PMC7926881 DOI: 10.3390/cells10020464] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 12/12/2022] Open
Abstract
The cyclic AMP (cAMP) signalling cascade is necessary for cell homeostasis and plays important roles in many processes. This is particularly relevant during ageing and age-related diseases, where drastic changes, generally decreases, in cAMP levels have been associated with the progressive decline in overall cell function and, eventually, the loss of cellular integrity. The functional relevance of reduced cAMP is clearly supported by the finding that increases in cAMP levels can reverse some of the effects of ageing. Nevertheless, despite these observations, the molecular mechanisms underlying the dysregulation of cAMP signalling in ageing are not well understood. Compartmentalization is widely accepted as the modality through which cAMP achieves its functional specificity; therefore, it is important to understand whether and how this mechanism is affected during ageing and to define which is its contribution to this process. Several animal models demonstrate the importance of specific cAMP signalling components in ageing, however, how age-related changes in each of these elements affect the compartmentalization of the cAMP pathway is largely unknown. In this review, we explore the connection of single components of the cAMP signalling cascade to ageing and age-related diseases whilst elaborating the literature in the context of cAMP signalling compartmentalization.
Collapse
Affiliation(s)
- Giulietta Di Benedetto
- Neuroscience Institute, National Research Council of Italy (CNR), 35121 Padova, Italy;
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
| | - Liliana F. Iannucci
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Nicoletta C. Surdo
- Neuroscience Institute, National Research Council of Italy (CNR), 35121 Padova, Italy;
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
| | - Sofia Zanin
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Filippo Conca
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Biology, University of Padova, 35122 Padova, Italy
| | - Francesca Grisan
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Biology, University of Padova, 35122 Padova, Italy
| | - Andrea Gerbino
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, 70121 Bari, Italy;
| | - Konstantinos Lefkimmiatis
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
4
|
Gorny N, Kelly MP. Alterations in cyclic nucleotide signaling are implicated in healthy aging and age-related pathologies of the brain. VITAMINS AND HORMONES 2021; 115:265-316. [PMID: 33706951 DOI: 10.1016/bs.vh.2020.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
It is not only important to consider how hormones may change with age, but also how downstream signaling pathways that couple to hormone receptors may change. Among these hormone-coupled signaling pathways are the 3',5'-cyclic guanosine monophosphate (cGMP) and 3',5'-cyclic adenosine monophosphate (cAMP) intracellular second messenger cascades. Here, we test the hypothesis that dysfunction of cAMP and/or cGMP synthesis, execution, and/or degradation occurs in the brain during healthy and pathological diseases such as Alzheimer's disease, Parkinson's disease, and Huntington's disease. Although most studies report lower cyclic nucleotide signaling in the aged brain, with further reductions noted in the context of age-related diseases, there are select examples where cAMP signaling may be elevated in select tissues. Thus, therapeutics would need to target cAMP/cGMP in a tissue-specific manner if efficacy for select symptoms is to be achieved without worsening others.
Collapse
Affiliation(s)
- Nicole Gorny
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Michy P Kelly
- Department of Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
5
|
Sanders O, Rajagopal L. Phosphodiesterase Inhibitors for Alzheimer's Disease: A Systematic Review of Clinical Trials and Epidemiology with a Mechanistic Rationale. J Alzheimers Dis Rep 2020; 4:185-215. [PMID: 32715279 PMCID: PMC7369141 DOI: 10.3233/adr-200191] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Preclinical studies, clinical trials, and reviews suggest increasing 3',5'-cyclic adenosine monophosphate (cAMP) and 3',5'-cyclic guanosine monophosphate (cGMP) with phosphodiesterase inhibitors is disease-modifying in Alzheimer's disease (AD). cAMP/protein kinase A (PKA) and cGMP/protein kinase G (PKG) signaling are disrupted in AD. cAMP/PKA and cGMP/PKG activate cAMP response element binding protein (CREB). CREB binds mitochondrial and nuclear DNA, inducing synaptogenesis, memory, and neuronal survival gene (e.g., brain-derived neurotrophic factor) and peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1α). cAMP/PKA and cGMP/PKG activate Sirtuin-1, which activates PGC1α. PGC1α induces mitochondrial biogenesis and antioxidant genes (e.g.,Nrf2) and represses BACE1. cAMP and cGMP inhibit BACE1-inducing NFκB and tau-phosphorylating GSK3β. OBJECTIVE AND METHODS We review efficacy-testing clinical trials, epidemiology, and meta-analyses to critically investigate whether phosphodiesteraseinhibitors prevent or treat AD. RESULTS Caffeine and cilostazol may lower AD risk. Denbufylline and sildenafil clinical trials are promising but preliminary and inconclusive. PF-04447943 and BI 409,306 are ineffective. Vinpocetine, cilostazol, and nicergoline trials are mixed. Deprenyl/selegiline trials show only short-term benefits. Broad-spectrum phosphodiesterase inhibitor propentofylline has been shown in five phase III trials to improve cognition, dementia severity, activities of daily living, and global assessment in mild-to-moderate AD patients on multiple scales, including the ADAS-Cogand the CIBIC-Plus in an 18-month phase III clinical trial. However, two books claimed based on a MedScape article an 18-month phase III trial failed, so propentofylline was discontinued. Now, propentofylline is used to treat canine cognitive dysfunction, which, like AD, involves age-associated wild-type Aβ deposition. CONCLUSION Phosphodiesterase inhibitors may prevent and treat AD.
Collapse
|
6
|
Feng H, Wang C, He W, Wu X, Li S, Zeng Z, Wei M, He B. Roflumilast ameliorates cognitive impairment in APP/PS1 mice via cAMP/CREB/BDNF signaling and anti-neuroinflammatory effects. Metab Brain Dis 2019; 34:583-591. [PMID: 30610438 DOI: 10.1007/s11011-018-0374-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 12/19/2018] [Indexed: 12/20/2022]
Abstract
Phosphodiesterase type 4 (PDE4) inhibitors can prevent the breakdown of the second messenger cyclic adenosine monophosphate (cAMP) and improve cognitive performances in several animal models of cognition. However, the clinical development of PDE4 inhibitors has been seriously hampered by severe side effects, such as vomiting and nausea. In this study, we investigated the effect and mechanism of roflumilast, an FDA-approved PDE4 inhibitor for treatment of chronic obstructive pulmonary disease (COPD), on learning and memory abilities in the APP/PS1 mouse model of Alzheimer's disease (AD). APP/PS1 transgenic mice received 3 intragastric doses of roflumilast (0.1, 0.2 and 0.4 mg/kg) daily for 3 weeks followed by behavioral tests. Chronic administration of roflumilast significantly improved the learning and memory abilities of APP/PS1 transgenic mice in the novel object recognition task, Morris water maze, and the step-down passive avoidance task. In addition, roflumilast increased the cAMP, phosphorylated cAMP response-element binding protein (p-CREB) and brain-derived neurotrophic factor (BDNF) levels, and reduced the nuclear translocation of nuclear factor-kappa B (NF-κB) p65, and proinflammatory cytokine (IL-6, TNF-a and IL-1β) levels in the hippocampus of APP/PS1 transgenic mice. In conclusion, these findings suggest that roflumilast can enhance cognitive function in APP/PS1 transgenic mice, which may be related to its stimulation of the cAMP/CREB/BDNF pathway and anti-neuroinflammatory effects.
Collapse
Affiliation(s)
- Huancun Feng
- Department of Pharmacy, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Canmao Wang
- Department of Pharmacy, Shenzhen University General Hospital, Shenzhen, 518000, Guangdong, China
| | - Wei He
- Department of Pharmacy, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Xinjun Wu
- Department of Biology, East Carolina University, Greenville, NC, 27858, USA
| | - Shujie Li
- Department of Pharmacy, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Zhenkun Zeng
- Department of Pharmacy, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Meidan Wei
- Department of Pharmacy, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, China
| | - Binghong He
- Department of Pharmacy, The Third Affiliated Hospital, Southern Medical University, Guangzhou, 510000, Guangdong, China.
| |
Collapse
|
7
|
You M, Pan Y, Liu Y, Chen Y, Wu Y, Si J, Wang K, Hu F. Royal Jelly Alleviates Cognitive Deficits and β-Amyloid Accumulation in APP/PS1 Mouse Model Via Activation of the cAMP/PKA/CREB/BDNF Pathway and Inhibition of Neuronal Apoptosis. Front Aging Neurosci 2019; 10:428. [PMID: 30687079 PMCID: PMC6338040 DOI: 10.3389/fnagi.2018.00428] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 12/11/2018] [Indexed: 12/06/2022] Open
Abstract
Alzheimer’s disease (AD) is characterized clinically by progressive cognitive decline and pathologically by the accumulation of amyloid-β (Aβ) in the brain. Royal jelly (RJ), a secretion of honeybee hypopharyngeal and mandibular glands, has previously been shown to have anti-aging and neuromodulatory activities. In this study, we discovered that 3 months of RJ treatment substantially ameliorated behavioral deficits of APP/PS1 mice in the Morris Water Maze (MWM) test and step-down passive avoidance test. Our data also showed that RJ significantly diminished amyloid plaque pathology in APP/PS1 mice. Furthermore, RJ alleviated c-Jun N-terminal kinase (JNK) phosphorylation-induced neuronal apoptosis by suppressing oxidative stress. Importantly, hippocampal cyclic adenosine monophosphate (cAMP), p-PKA, p-CREB and BDNF levels were significantly increased in the APP/PS1 mice after RJ treatment, indicating that the cAMP/PKA/CREB/BDNF pathway might be related to the ameliorative effect of RJ on cognitive decline. Collectively, these results provide a scientific basis for using RJ as a functional food for targeting AD pathology.
Collapse
Affiliation(s)
- Mengmeng You
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Yongming Pan
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Yichen Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Yifan Chen
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Yuqi Wu
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Juanjuan Si
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Kai Wang
- Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Fuliang Hu
- College of Animal Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
8
|
A Role for Phosphodiesterase 11A (PDE11A) in the Formation of Social Memories and the Stabilization of Mood. ADVANCES IN NEUROBIOLOGY 2018; 17:201-230. [PMID: 28956334 DOI: 10.1007/978-3-319-58811-7_8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The most recently discovered 3',5'-cyclic nucleotide phosphodiesterase family is the Phosphodiesterase 11 (PDE11) family, which is encoded by a single gene PDE11A. PDE11A is a dual-specific PDE, breaking down both cAMP and cGMP. There are four PDE11A splice variants (PDE11A1-4) with distinct tissue expression profiles and unique N-terminal regulatory regions, suggesting that each isoform could be individually targeted with a small molecule or biologic. PDE11A4 is the PDE11A isoform expressed in brain and is found in the hippocampal formation of humans and rodents. Studies in rodents show that PDE11A4 mRNA expression in brain is, in fact, restricted to the hippocampal formation (CA1, possibly CA2, subiculum, and the adjacently connected amygdalohippocampal area). Within the hippocampal formation of rodents, PDE11A4 protein is expressed in neurons but not astrocytes, with a distribution across nuclear, cytoplasmic, and membrane compartments. This subcellular localization of PDE11A4 is altered in response to social experience in mouse, and in vitro studies show the compartmentalization of PDE11A4 is controlled, at least in part, by homodimerization and N-terminal phosphorylation. PDE11A4 expression dramatically increases in the hippocampus with age in the rodent hippocampus, from early postnatal life to late aging, suggesting PDE11A4 function may evolve across the lifespan. Interestingly, PDE11A4 protein shows a three to tenfold enrichment in the rodent ventral hippocampal formation (VHIPP; a.k.a. anterior in primates) versus dorsal hippocampal formation (DHIPP). Consistent with this enrichment in VHIPP, studies in knockout mice show that PDE11A regulates the formation of social memories and the stabilization of mood and is a critical mechanism by which social experience feeds back to modify the brain and subsequent social behaviors. PDE11A4 likely controls behavior by regulating hippocampal glutamatergic, oxytocin, and cytokine signaling, as well as protein translation. Given its unique tissue distribution and relatively selective effects on behavior, PDE11A may represent a novel therapeutic target for neuropsychiatric, neurodevelopmental, or age-related disorders. Therapeutically targeting PDE11A4 may be a way to selectively restore aberrant cyclic nucleotide signaling in the hippocampal formation while leaving the rest of the brain and periphery untouched, thus, relieving deficits while avoiding unwanted side effects.
Collapse
|
9
|
Hansen RT, Zhang HT. The Past, Present, and Future of Phosphodiesterase-4 Modulation for Age-Induced Memory Loss. ADVANCES IN NEUROBIOLOGY 2018; 17:169-199. [PMID: 28956333 DOI: 10.1007/978-3-319-58811-7_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The purpose of this chapter is to highlight the state of progress for phosphodiesterase-4 (PDE4) modulation as a potential therapeutic for psychiatric illness, and to draw attention to particular hurdles and obstacles that must be overcome in future studies to develop PDE4-mediated therapeutics. Pathological and non-pathological related memory loss will be the focus of the chapter; however, we will at times also touch upon other psychiatric illnesses like anxiety and depression. First, we will provide a brief background of PDE4, and the rationale for its extensive study in cognition. Second, we will explore fundamental differences in individual PDE4 subtypes, and then begin to address differences between pathological and non-pathological aging. Alterations of cAMP/PDE4 signaling that occur within normal vs. pathological aging, and the potential for PDE4 modulation to combat these alterations within each context will be described. Finally, we will finish the chapter with obstacles that have hindered the field, and future studies and alternative viewpoints that need to be addressed. Overall, we hope this chapter will demonstrate the incredible complexity of PDE4 signaling in the brain, and will be useful in forming a strategy to develop future PDE4-mediated therapeutics for psychiatric illnesses.
Collapse
Affiliation(s)
- Rolf T Hansen
- Departments of Behavioral Medicine & Psychiatry and Physiology & Pharmacology, West Virginia University Health Sciences Center, 1 Medical Center Drive, Morgantown, WV, 26506-9137, USA
| | - Han-Ting Zhang
- Department of Behavioral Medicine and Psychiatry, West Virginia University Health Sciences Center, 1 Medical Center Drive, Morgantown, WV, 26506, USA. .,Department of Physiology and Pharmacology, West Virginia University Health Sciences Center, 1 Medical Center Drive, Morgantown, WV, 26506, USA. .,Institute of Pharmacology, Taishan Medical University, Taian, 271016, China.
| |
Collapse
|
10
|
Kelly MP. Cyclic nucleotide signaling changes associated with normal aging and age-related diseases of the brain. Cell Signal 2018; 42:281-291. [PMID: 29175000 PMCID: PMC5732030 DOI: 10.1016/j.cellsig.2017.11.004] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 11/21/2017] [Indexed: 01/23/2023]
Abstract
Deficits in brain function that are associated with aging and age-related diseases benefit very little from currently available therapies, suggesting a better understanding of the underlying molecular mechanisms is needed to develop improved drugs. Here, we review the literature to test the hypothesis that a break down in cyclic nucleotide signaling at the level of synthesis, execution, and/or degradation may contribute to these deficits. A number of findings have been reported in both the human and animal model literature that point to brain region-specific changes in Galphas (a.k.a. Gαs or Gsα), adenylyl cyclase, 3',5'-adenosine monophosphate (cAMP) levels, protein kinase A (PKA), cAMP response element binding protein (CREB), exchange protein activated by cAMP (Epac), hyperpolarization-activated cyclic nucleotide-gated ion channels (HCNs), atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), soluble and particulate guanylyl cyclase, 3',5'-guanosine monophosphate (cGMP), protein kinase G (PKG) and phosphodiesterases (PDEs). Among the most reproducible findings are 1) elevated circulating ANP and BNP levels being associated with cognitive dysfunction or dementia independent of cardiovascular effects, 2) reduced basal and/or NMDA-stimulated cGMP levels in brain with aging or Alzheimer's disease (AD), 3) reduced adenylyl cyclase activity in hippocampus and specific cortical regions with aging or AD, 4) reduced expression/activity of PKA in temporal cortex and hippocampus with AD, 5) reduced phosphorylation of CREB in hippocampus with aging or AD, 6) reduced expression/activity of the PDE4 family in brain with aging, 7) reduced expression of PDE10A in the striatum with Huntington's disease (HD) or Parkinson's disease, and 8) beneficial effects of select PDE inhibitors, particularly PDE10 inhibitors in HD models and PDE4 and PDE5 inhibitors in aging and AD models. Although these findings generally point to a reduction in cyclic nucleotide signaling being associated with aging and age-related diseases, there are exceptions. In particular, there is evidence for increased cAMP signaling specifically in aged prefrontal cortex, AD cerebral vessels, and PD hippocampus. Thus, if cyclic nucleotide signaling is going to be targeted effectively for therapeutic gain, it will have to be manipulated in a brain region-specific manner.
Collapse
Affiliation(s)
- Michy P Kelly
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, 6439 Garners Ferry Road, VA Bldg 1, 3rd Floor, D-12, Columbia, SC 29209, United States.
| |
Collapse
|
11
|
Abstract
High levels of amyloid-β peptide (Aβ) have been related to Alzheimer's disease pathogenesis. However, in the healthy brain, low physiologically relevant concentrations of Aβ are necessary for long-term potentiation (LTP) and memory. Because cGMP plays a key role in these processes, here we investigated whether the cyclic nucleotide cGMP influences Aβ levels and function during LTP and memory. We demonstrate that the increase of cGMP levels by the phosphodiesterase-5 inhibitors sildenafil and vardenafil induces a parallel release of Aβ due to a change in the approximation of amyloid precursor protein (APP) and the β-site APP cleaving enzyme 1. Moreover, electrophysiological and behavioral studies performed on animals of both sexes showed that blocking Aβ function, by using anti-murine Aβ antibodies or APP knock-out mice, prevents the cGMP-dependent enhancement of LTP and memory. Our data suggest that cGMP positively regulates Aβ levels in the healthy brain which, in turn, boosts synaptic plasticity and memory.SIGNIFICANCE STATEMENT Amyloid-β (Aβ) is a key pathogenetic factor in Alzheimer's disease. However, low concentrations of endogenous Aβ, mimicking levels of the peptide in the healthy brain, enhance hippocampal long-term potentiation (LTP) and memory. Because the second messenger cGMP exerts a central role in LTP mechanisms, here we studied whether cGMP affects Aβ levels and function during LTP. We show that cGMP enhances Aβ production by increasing the APP/BACE-1 convergence in endolysosomal compartments. Moreover, the cGMP-induced enhancement of LTP and memory was disrupted by blockade of Aβ, suggesting that the physiological effect of the cyclic nucleotide on LTP and memory is dependent upon Aβ.
Collapse
|
12
|
Dessauer CW, Watts VJ, Ostrom RS, Conti M, Dove S, Seifert R. International Union of Basic and Clinical Pharmacology. CI. Structures and Small Molecule Modulators of Mammalian Adenylyl Cyclases. Pharmacol Rev 2017; 69:93-139. [PMID: 28255005 PMCID: PMC5394921 DOI: 10.1124/pr.116.013078] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Adenylyl cyclases (ACs) generate the second messenger cAMP from ATP. Mammalian cells express nine transmembrane AC (mAC) isoforms (AC1-9) and a soluble AC (sAC, also referred to as AC10). This review will largely focus on mACs. mACs are activated by the G-protein Gαs and regulated by multiple mechanisms. mACs are differentially expressed in tissues and regulate numerous and diverse cell functions. mACs localize in distinct membrane compartments and form signaling complexes. sAC is activated by bicarbonate with physiologic roles first described in testis. Crystal structures of the catalytic core of a hybrid mAC and sAC are available. These structures provide detailed insights into the catalytic mechanism and constitute the basis for the development of isoform-selective activators and inhibitors. Although potent competitive and noncompetitive mAC inhibitors are available, it is challenging to obtain compounds with high isoform selectivity due to the conservation of the catalytic core. Accordingly, caution must be exerted with the interpretation of intact-cell studies. The development of isoform-selective activators, the plant diterpene forskolin being the starting compound, has been equally challenging. There is no known endogenous ligand for the forskolin binding site. Recently, development of selective sAC inhibitors was reported. An emerging field is the association of AC gene polymorphisms with human diseases. For example, mutations in the AC5 gene (ADCY5) cause hyperkinetic extrapyramidal motor disorders. Overall, in contrast to the guanylyl cyclase field, our understanding of the (patho)physiology of AC isoforms and the development of clinically useful drugs targeting ACs is still in its infancy.
Collapse
Affiliation(s)
- Carmen W Dessauer
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas (C.W.D.); Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (V.J.W.); Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (R.S.O.); Center for Reproductive Sciences, University of California San Francisco, San Francisco, California (M.C.); Institute of Pharmacy, University of Regensburg, Regensburg, Germany (S.D.); and Institute of Pharmacology, Hannover Medical School, Hannover, Germany (R.S.)
| | - Val J Watts
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas (C.W.D.); Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (V.J.W.); Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (R.S.O.); Center for Reproductive Sciences, University of California San Francisco, San Francisco, California (M.C.); Institute of Pharmacy, University of Regensburg, Regensburg, Germany (S.D.); and Institute of Pharmacology, Hannover Medical School, Hannover, Germany (R.S.)
| | - Rennolds S Ostrom
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas (C.W.D.); Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (V.J.W.); Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (R.S.O.); Center for Reproductive Sciences, University of California San Francisco, San Francisco, California (M.C.); Institute of Pharmacy, University of Regensburg, Regensburg, Germany (S.D.); and Institute of Pharmacology, Hannover Medical School, Hannover, Germany (R.S.)
| | - Marco Conti
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas (C.W.D.); Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (V.J.W.); Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (R.S.O.); Center for Reproductive Sciences, University of California San Francisco, San Francisco, California (M.C.); Institute of Pharmacy, University of Regensburg, Regensburg, Germany (S.D.); and Institute of Pharmacology, Hannover Medical School, Hannover, Germany (R.S.)
| | - Stefan Dove
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas (C.W.D.); Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (V.J.W.); Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (R.S.O.); Center for Reproductive Sciences, University of California San Francisco, San Francisco, California (M.C.); Institute of Pharmacy, University of Regensburg, Regensburg, Germany (S.D.); and Institute of Pharmacology, Hannover Medical School, Hannover, Germany (R.S.)
| | - Roland Seifert
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, Texas (C.W.D.); Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana (V.J.W.); Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (R.S.O.); Center for Reproductive Sciences, University of California San Francisco, San Francisco, California (M.C.); Institute of Pharmacy, University of Regensburg, Regensburg, Germany (S.D.); and Institute of Pharmacology, Hannover Medical School, Hannover, Germany (R.S.)
| |
Collapse
|
13
|
Hesse R, Lausser L, Gummert P, Schmid F, Wahler A, Schnack C, Kroker KS, Otto M, Tumani H, Kestler HA, Rosenbrock H, von Arnim CAF. Reduced cGMP levels in CSF of AD patients correlate with severity of dementia and current depression. Alzheimers Res Ther 2017; 9:17. [PMID: 28274265 PMCID: PMC5343324 DOI: 10.1186/s13195-017-0245-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/13/2017] [Indexed: 01/21/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disorder, primarily affecting memory. That disorder is thought to be a consequence of neuronal network disturbances and synapse loss. Decline in cognitive function is associated with a high burden of neuropsychiatric symptoms (NPSs) such as depression. The cyclic nucleotides cyclic adenosine-3',5'-monophosphate (cAMP) and cyclic guanosine-3',5'-monophosphate (cGMP) are essential second messengers that play a crucial role in memory processing as well as synaptic plasticity and are potential therapeutic targets. Biomarkers that are able to monitor potential treatment effects and that reflect the underlying pathology are of crucial interest. METHODS In this study, we measured cGMP and cAMP in cerebrospinal fluid (CSF) in a cohort of 133 subjects including 68 AD patients and 65 control subjects. To address the association with disease progression we correlated cognitive status with cyclic nucleotide levels. Because a high burden of NPSs is associated with decrease in cognitive function, we performed an exhaustive evaluation of AD-relevant marker combinations in a depressive subgroup. RESULTS We show that cGMP, but not cAMP, levels in the CSF of AD patients are significantly reduced compared with the control group. Reduced cGMP levels in AD patients correlate with memory impairment based on Mini-Mental State Examination score (r = 0.17, p = 0.048) and tau as a marker of neurodegeneration (r = -0.28, p = 0.001). Moreover, we were able to show that AD patients suffering from current depression show reduced cGMP levels (p = 0.07) and exhibit a higher degree of cognitive impairment than non-depressed AD patients. CONCLUSION These results provide further evidence for an involvement of cGMP in AD pathogenesis and accompanying co-morbidities, and may contribute to elucidating synaptic plasticity alterations during disease progression.
Collapse
Affiliation(s)
- Raphael Hesse
- Department of Neurology, Ulm University, Oberer Eselsberg 45, 89081 Ulm, Germany
| | - Ludwig Lausser
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Pauline Gummert
- Department of Neurology, Ulm University, Oberer Eselsberg 45, 89081 Ulm, Germany
| | - Florian Schmid
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Anke Wahler
- Department of Neurology, Ulm University, Oberer Eselsberg 45, 89081 Ulm, Germany
| | - Cathrin Schnack
- Department of Neurology, Ulm University, Oberer Eselsberg 45, 89081 Ulm, Germany
| | - Katja S. Kroker
- Department of Drug Discovery Support, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
| | - Markus Otto
- Department of Neurology, Ulm University, Oberer Eselsberg 45, 89081 Ulm, Germany
| | - Hayrettin Tumani
- Department of Neurology, Ulm University, Oberer Eselsberg 45, 89081 Ulm, Germany
| | - Hans A. Kestler
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Holger Rosenbrock
- Department of CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co KG, Biberach, Germany
| | | |
Collapse
|
14
|
Guo H, Cheng Y, Wang C, Wu J, Zou Z, Niu B, Yu H, Wang H, Xu J. FFPM, a PDE4 inhibitor, reverses learning and memory deficits in APP/PS1 transgenic mice via cAMP/PKA/CREB signaling and anti-inflammatory effects. Neuropharmacology 2017; 116:260-269. [PMID: 28065587 DOI: 10.1016/j.neuropharm.2017.01.004] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 12/25/2016] [Accepted: 01/05/2017] [Indexed: 12/25/2022]
Abstract
Thus far, phosphodiesterase-4 (PDE4) inhibitors have not been approved for application in Alzheimer's disease (AD) in a clinical setting due to severe side effects, such as nausea and vomiting. In this study, we investigated the effect of FFPM, a novel PDE4 inhibitor, on learning and memory abilities, as well as the underlying mechanism in the APP/PS1 mouse model of AD. Pharmacokinetic studies have revealed that FFPM efficiently permeates into the brain, and reached peak values in plasma 2 h after orally dosing. A 3-week treatment with FFPM, at doses of 0.25 mg/kg and 0.5 mg/kg, significantly improved the learning and memory abilities of APP/PS1 transgenic mice in the Morris water maze and the Step-down passive avoidance task. Interestingly, we found that while rolipram (0.5 mg/kg) reduced the duration of the α2 adrenergic receptor-mediated anesthesia induced by xylazine/ketamine, FFPM (0.5 mg/kg) or the vehicle did not have an evident effect. FFPM increased the cAMP, PKA and CREB phosphorylation and BDNF levels, and reduced the NF-κB p65, iNOS, TNF-α and IL-1β levels in the hippocampi of APP/PS1 trangenic mice, as observed by ELISA and Western blot analysis. Taken together, our data demonstrated that the reversal effect of FFPM on cognitive deficits in APP/PS1 transgenic mice might be related to stimulation of the cAMP/PKA/CREB/BDNF pathway and anti-inflammatory effects. Moreover, FFPM appears to have potential as an effective PDE4 inhibitor in AD treatment with little emetic potential.
Collapse
Affiliation(s)
- Haibiao Guo
- Neuropharmacology and Drug Discovery Group, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; Modern Chinese Medicine Research Institute of Hutchison Whampoa Guangzhou Baiyunshan Chinese Medicine Co., Ltd., Guangzhou 510515, China
| | - Yufang Cheng
- Neuropharmacology and Drug Discovery Group, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Canmao Wang
- Department of Pharmacy, Shenzhen Hospital of Southern Medical University, Shenzhen, Guangdong, 518000, China
| | - Jingang Wu
- Neuropharmacology and Drug Discovery Group, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhengqiang Zou
- Neuropharmacology and Drug Discovery Group, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Bo Niu
- Neuropharmacology and Drug Discovery Group, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hui Yu
- Neuropharmacology and Drug Discovery Group, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Haitao Wang
- Neuropharmacology and Drug Discovery Group, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Jiangping Xu
- Neuropharmacology and Drug Discovery Group, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
15
|
Wilson NM, Titus DJ, Oliva AA, Furones C, Atkins CM. Traumatic Brain Injury Upregulates Phosphodiesterase Expression in the Hippocampus. Front Syst Neurosci 2016; 10:5. [PMID: 26903822 PMCID: PMC4742790 DOI: 10.3389/fnsys.2016.00005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 01/18/2016] [Indexed: 11/13/2022] Open
Abstract
Traumatic brain injury (TBI) results in significant impairments in hippocampal synaptic plasticity. A molecule critically involved in hippocampal synaptic plasticity, 3′,5′-cyclic adenosine monophosphate, is downregulated in the hippocampus after TBI, but the mechanism that underlies this decrease is unknown. To address this question, we determined whether phosphodiesterase (PDE) expression in the hippocampus is altered by TBI. Young adult male Sprague Dawley rats received sham surgery or moderate parasagittal fluid-percussion brain injury. Animals were analyzed by western blotting for changes in PDE expression levels in the hippocampus. We found that PDE1A levels were significantly increased at 30 min, 1 h and 6 h after TBI. PDE4B2 and 4D2 were also significantly increased at 1, 6, and 24 h after TBI. Additionally, phosphorylation of PDE4A was significantly increased at 6 and 24 h after TBI. No significant changes were observed in levels of PDE1B, 1C, 3A, 8A, or 8B between 30 min to 7 days after TBI. To determine the spatial profile of these increases, we used immunohistochemistry and flow cytometry at 24 h after TBI. PDE1A and phospho-PDE4A localized to neuronal cell bodies. PDE4B2 was expressed in neuronal dendrites, microglia and infiltrating CD11b+ immune cells. PDE4D was predominantly found in microglia and infiltrating CD11b+ immune cells. To determine if inhibition of PDE4 would improve hippocampal synaptic plasticity deficits after TBI, we treated hippocampal slices with rolipram, a pan-PDE4 inhibitor. Rolipram partially rescued the depression in basal synaptic transmission and converted a decaying form of long-term potentiation (LTP) into long-lasting LTP. Overall, these results identify several possible PDE targets for reducing hippocampal synaptic plasticity deficits and improving cognitive function acutely after TBI.
Collapse
Affiliation(s)
- Nicole M Wilson
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine Miami, FL, USA
| | - David J Titus
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine Miami, FL, USA
| | - Anthony A Oliva
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine Miami, FL, USA
| | - Concepcion Furones
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine Miami, FL, USA
| | - Coleen M Atkins
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine Miami, FL, USA
| |
Collapse
|
16
|
Dagda RK, Das Banerjee T. Role of protein kinase A in regulating mitochondrial function and neuronal development: implications to neurodegenerative diseases. Rev Neurosci 2015; 26:359-70. [PMID: 25741943 DOI: 10.1515/revneuro-2014-0085] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 01/08/2015] [Indexed: 11/15/2022]
Abstract
In neurons, enhanced protein kinase A (PKA) signaling elevates synaptic plasticity, promotes neuronal development, and increases dopamine synthesis. By contrast, a decline in PKA signaling contributes to the etiology of several brain degenerative diseases, including Alzheimer's disease and Parkinson's disease, suggesting that PKA predominantly plays a neuroprotective role. A-kinase anchoring proteins (AKAPs) are large multidomain scaffold proteins that target PKA and other signaling molecules to distinct subcellular sites to strategically localize PKA signaling at dendrites, dendritic spines, cytosol, and axons. PKA can be recruited to the outer mitochondrial membrane by associating with three different AKAPs to regulate mitochondrial dynamics, structure, mitochondrial respiration, trafficking, dendrite morphology, and neuronal survival. In this review, we survey the myriad of essential neuronal functions modulated by PKA but place a special emphasis on mitochondrially localized PKA. Finally, we offer an updated overview of how loss of PKA signaling contributes to the etiology of several brain degenerative diseases.
Collapse
|
17
|
Membranous adenylyl cyclase 1 activation is regulated by oxidation of N- and C-terminal methionine residues in calmodulin. Biochem Pharmacol 2014; 93:196-209. [PMID: 25462816 DOI: 10.1016/j.bcp.2014.11.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 11/15/2014] [Accepted: 11/17/2014] [Indexed: 12/19/2022]
Abstract
Membranous adenylyl cyclase 1 (AC1) is associated with memory and learning. AC1 is activated by the eukaryotic Ca(2+)-sensor calmodulin (CaM), which contains nine methionine residues (Met) important for CaM-target interactions. During ageing, Met residues are oxidized to (S)- and (R)-methionine sulfoxide (MetSO) by reactive oxygen species arising from an age-related oxidative stress. We examined how oxidation by H2O2 of Met in CaM regulates CaM activation of AC1. We employed a series of thirteen mutant CaM proteins never assessed before in a single study, where leucine is substituted for Met, in order to analyze the effects of oxidation of specific Met. CaM activation of AC1 is regulated by oxidation of all of the C-terminal Met in CaM, and by two N-terminal Met, M36 and M51. CaM with all Met oxidized is unable to activate AC1. Activity is fully restored by the combined catalytic activities of methionine sulfoxide reductases A and B (MsrA and B), which catalyze reduction of the (S)- and (R)-MetSO stereoisomers. A small change in secondary structure is observed in wild-type CaM upon oxidation of all nine Met, but no significant secondary structure changes occur in the mutant proteins when Met residues are oxidized by H2O2, suggesting that localized polarity, flexibility and structural changes promote the functional changes accompanying oxidation. The results signify that AC1 catalytic activity can be delicately adjusted by mediating CaM activation of AC1 by reversible Met oxidation in CaM. The results are important for memory, learning and possible therapeutic routes for regulating AC1.
Collapse
|
18
|
Kelly MP, Adamowicz W, Bove S, Hartman AJ, Mariga A, Pathak G, Reinhart V, Romegialli A, Kleiman RJ. Select 3',5'-cyclic nucleotide phosphodiesterases exhibit altered expression in the aged rodent brain. Cell Signal 2013; 26:383-97. [PMID: 24184653 DOI: 10.1016/j.cellsig.2013.10.007] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 10/13/2013] [Accepted: 10/24/2013] [Indexed: 12/21/2022]
Abstract
3',5'-cyclic nucleotide phosphodiesterases (PDEs) are the only known enzymes to compartmentalize cAMP and cGMP, yet little is known about how PDEs are dynamically regulated across the lifespan. We mapped mRNA expression of all 21 PDE isoforms in the adult rat and mouse central nervous system (CNS) using quantitative polymerase chain reaction (qPCR) and in situ hybridization to assess conservation across species. We also compared PDE mRNA and protein in the brains of old (26 months) versus young (5 months) Sprague-Dawley rats, with select experiments replicated in old (9 months) versus young (2 months) BALB/cJ mice. We show that each PDE isoform exhibits a unique expression pattern across the brain that is highly conserved between rats, mice, and humans. PDE1B, PDE1C, PDE2A, PDE4A, PDE4D, PDE5A, PDE7A, PDE8A, PDE8B, PDE10A, and PDE11A showed an age-related increase or decrease in mRNA expression in at least 1 of the 4 brain regions examined (hippocampus, cortex, striatum, and cerebellum). In contrast, mRNA expression of PDE1A, PDE3A, PDE3B, PDE4B, PDE7A, PDE7B, and PDE9A did not change with age. Age-related increases in PDE11A4, PDE8A3, PDE8A4/5, and PDE1C1 protein expression were confirmed in hippocampus of old versus young rodents, as were age-related increases in PDE8A3 protein expression in the striatum. Age-related changes in PDE expression appear to have functional consequences as, relative to young rats, the hippocampi of old rats demonstrated strikingly decreased phosphorylation of GluR1, CaMKIIα, and CaMKIIβ, decreased expression of the transmembrane AMPA regulatory proteins γ2 (a.k.a. stargazin) and γ8, and increased trimethylation of H3K27. Interestingly, expression of PDE11A4, PDE8A4/5, PDE8A3, and PDE1C1 correlate with these functional endpoints in young but not old rats, suggesting that aging is not only associated with a change in PDE expression but also a change in PDE compartmentalization.
Collapse
Affiliation(s)
- Michy P Kelly
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology & Neuroscience, 6439 Garners Ferry Rd, Columbia, SC 29209, USA.
| | - Wendy Adamowicz
- Pfizer Global Research and Development, Neuroscience Research Unit, Eastern point Road, Groton, CT 06340, USA.
| | - Susan Bove
- Pfizer Global Research and Development, Neuroscience Research Unit, Eastern point Road, Groton, CT 06340, USA.
| | - Alexander J Hartman
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology & Neuroscience, 6439 Garners Ferry Rd, Columbia, SC 29209, USA
| | - Abigail Mariga
- Pfizer Global Research and Development, Neuroscience Research Unit, Eastern point Road, Groton, CT 06340, USA.
| | - Geetanjali Pathak
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology & Neuroscience, 6439 Garners Ferry Rd, Columbia, SC 29209, USA
| | - Veronica Reinhart
- Pfizer Global Research and Development, Neuroscience Research Unit, Eastern point Road, Groton, CT 06340, USA
| | - Alison Romegialli
- Pfizer Global Research and Development, Neuroscience Research Unit, Eastern point Road, Groton, CT 06340, USA.
| | - Robin J Kleiman
- Pfizer Global Research and Development, Neuroscience Research Unit, Eastern point Road, Groton, CT 06340, USA.
| |
Collapse
|
19
|
cAMP level modulates scleral collagen remodeling, a critical step in the development of myopia. PLoS One 2013; 8:e71441. [PMID: 23951163 PMCID: PMC3741144 DOI: 10.1371/journal.pone.0071441] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 06/28/2013] [Indexed: 11/19/2022] Open
Abstract
The development of myopia is associated with decreased ocular scleral collagen synthesis in humans and animal models. Collagen synthesis is, in part, under the influence of cyclic adenosine monophosphate (cAMP). We investigated the associations between cAMP, myopia development in guinea pigs, and collagen synthesis by human scleral fibroblasts (HSFs). Form-deprived myopia (FDM) was induced by unilateral masking of guinea pig eyes. Scleral cAMP levels increased selectively in the FDM eyes and returned to normal levels after unmasking and recovery. Unilateral subconjunctival treatment with the adenylyl cyclase (AC) activator forskolin resulted in a myopic shift accompanied by reduced collagen mRNA levels, but it did not affect retinal electroretinograms. The AC inhibitor SQ22536 attenuated the progression of FDM. Moreover, forskolin inhibited collagen mRNA levels and collagen secretion by HSFs. The inhibition was reversed by SQ22536. These results demonstrate a critical role of cAMP in control of myopia development. Selective regulation of cAMP to control scleral collagen synthesis may be a novel therapeutic strategy for preventing and treating myopia.
Collapse
|
20
|
Sierksma ASR, Rutten K, Sydlik S, Rostamian S, Steinbusch HWM, van den Hove DLA, Prickaerts J. Chronic phosphodiesterase type 2 inhibition improves memory in the APPswe/PS1dE9 mouse model of Alzheimer's disease. Neuropharmacology 2012; 64:124-36. [PMID: 22771768 DOI: 10.1016/j.neuropharm.2012.06.048] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 06/22/2012] [Accepted: 06/24/2012] [Indexed: 01/08/2023]
Abstract
Alzheimer's disease (AD) is characterized by progressive cognitive deficits and synaptic dysfunction. Over the last decade phosphodiesterase inhibitors (PDEIs) have received increasing attention as putative cognition enhancers and have been suggested as a novel treatment strategy for AD. Given their ability to prevent hydrolysis of cAMP and/or cGMP, they can stimulate the cAMP/protein kinase A (PKA)/cAMP element-binding protein (CREB) and cGMP/PKG/CREB pathway to enhance synaptic transmission by increasing CREB phosphorylation (pCREB) and brain-derived neurotrophic factor (BDNF) transcription. Based on previous research, we hypothesized that chronic PDE2I treatment would improve AD-related cognitive deficits, by decreasing amyloid-β (Aβ) plaque load, enhancing pCREB and BDNF levels and increasing synaptic density in the hippocampus of 8-month-old APPswe/PS1dE9 mice. Results indicated that chronic PDE2I treatment could indeed improve memory performance in APPswe/PS1dE9 mice, without affecting anxiety, depressive-like behavior or hypothalamus-pituitary-adrenal axis regulation. However, no treatment effects were observed on Aβ plaque load, pCREB or BDNF concentrations, or presynaptic density in the hippocampus, suggesting that other signaling pathways and/or effector molecules might be responsible for its cognition-enhancing effects. Presynaptic density in the stratum lucidum of the CA3 subregion was significantly higher in APPswe/PS1dE9 mice compared to WT controls, possibly reflecting a compensatory mechanism. In conclusion, PDEs in general, and PDE2 specifically, could be considered as promising therapeutic targets for cognition enhancement in AD, although the underlying mechanism of action remains to be elucidated. This article is part of a Special Issue entitled 'Cognitive Enhancers'.
Collapse
Affiliation(s)
- Annerieke S R Sierksma
- Department of Psychiatry and Neuropsychology, Faculty of Health, Medicine and Life Science, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
21
|
Absence of Ca2+-stimulated adenylyl cyclases leads to reduced synaptic plasticity and impaired experience-dependent fear memory. Transl Psychiatry 2012; 2:e126. [PMID: 22832970 PMCID: PMC3365269 DOI: 10.1038/tp.2012.50] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Ca(2+)-stimulated adenylyl cyclase (AC) 1 and 8 are two genes that have been shown to play critical roles in fear memory. AC1 and AC8 couple neuronal activity and intracellular Ca(2+) increases to the production of cyclic adenosine monophosphate and are localized synaptically, suggesting that Ca(2+)-stimulated ACs may modulate synaptic plasticity. Here, we first established that Ca(2+)-stimulated ACs modulate protein markers of synaptic activity at baseline and after learning. Primary hippocampal cell cultures showed that AC1/AC8 double-knockout (DKO) mice have reduced SV2, a synaptic vesicle protein, abundance along their dendritic processes, and this reduction can be rescued through lentivirus delivery of AC8 to the DKO cells. Additionally, phospho-synapsin, a protein implicated in the regulation of neurotransmitter release at the synapse, is decreased in vivo 1 h after conditioned fear (CF) training in DKO mice. Importantly, additional experiments showed that long-term potentiation deficits present in DKO mice are rescued by acutely replacing AC8 in the forebrain, further supporting the idea that Ca(2+)-stimulated AC activity is a crucial modulator of synaptic plasticity. Previous studies have demonstrated that memory is continually modulated by gene-environment interactions. The last set of experiments evaluated the effects of knocking out AC1 and AC8 genes on experience-dependent changes in CF memory. We showed that the strength of CF memory in wild-type mice is determined by previous environment, minimal or enriched, whereas memory in DKO mice is unaffected. Thus, overall these results show that AC1 and AC8 modulate markers of synaptic activity and help integrate environmental information to modulate fear memory.
Collapse
|
22
|
Oh MM, Oliveira FA, Disterhoft JF. Learning and aging related changes in intrinsic neuronal excitability. Front Aging Neurosci 2010; 2:2. [PMID: 20552042 PMCID: PMC2874400 DOI: 10.3389/neuro.24.002.2010] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Accepted: 01/11/2010] [Indexed: 11/16/2022] Open
Abstract
A goal of many laboratories that study aging is to find a key cellular change(s) that can be manipulated and restored to a young-like state, and thus, reverse the age-related cognitive deficits. We have chosen to focus our efforts on the alteration of intrinsic excitability (as reflected by the postburst afterhyperpolarization, AHP) during the learning process in hippocampal pyramidal neurons. We have consistently found that the postburst AHP is significantly reduced in hippocampal pyramidal neurons from young adults that have successfully learned a hippocampus-dependent task. In the context of aging, the baseline intrinsic excitability of hippocampal neurons is decreased and therefore cognitive learning is impaired. In aging animals that are able to learn, neuron changes in excitability similar to those seen in young neurons during learning occur. Our challenge, then, is to understand how and why excitability changes occur in neurons from aging brains and cause age-associated learning impairments. After understanding the changes, we should be able to formulate strategies for reversing them, thus making old neurons function more as they did when they were young. Such a reversal should rescue the age-related cognitive deficits.
Collapse
Affiliation(s)
- M. Matthew Oh
- Department of Physiology, Feinberg School of Medicine, Northwestern UniversityChicago, IL, USA
| | - Fernando A. Oliveira
- Department of Physiology, Feinberg School of Medicine, Northwestern UniversityChicago, IL, USA
| | - John F. Disterhoft
- Department of Physiology, Feinberg School of Medicine, Northwestern UniversityChicago, IL, USA
| |
Collapse
|
23
|
Egger M, Maity P, Hübner M, Seifert R, König B. Synthesis and Pharmacological Properties of New Tetracyclic Forskolin Analogues. European J Org Chem 2009. [DOI: 10.1002/ejoc.200900260] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
24
|
Learning-related postburst afterhyperpolarization reduction in CA1 pyramidal neurons is mediated by protein kinase A. Proc Natl Acad Sci U S A 2009; 106:1620-5. [PMID: 19164584 DOI: 10.1073/pnas.0807708106] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Learning-related reductions of the postburst afterhyperpolarization (AHP) in hippocampal pyramidal neurons have been shown ex vivo, after trace eyeblink conditioning. The AHP is also reduced by many neuromodulators, such as norepinephrine, via activation of protein kinases. Trace eyeblink conditioning, like other hippocampus-dependent tasks, relies on protein synthesis for consolidating the learned memory. Protein kinase A (PKA) has been shown to be a key contributor for protein synthesis via the cAMP-response element-binding pathway. Here, we have explored a potential involvement of PKA and protein kinase C (PKC) in maintaining the learning-related postburst AHP reduction observed in CA1 pyramidal neurons. Bath application of isoproterenol (1 muM), a beta-adrenergic agonist that activates PKA, significantly reduced the AHP in CA1 neurons from control animals, but not from rats that learned. This occlusion suggests that PKA activity is involved in maintaining the AHP reduction measured ex vivo after successful learning. In contrast, bath application of the PKC activator, (-) indolactam V (0.2 muM), significantly reduced the AHP in CA1 neurons from both control and trained rats, indicating that PKC activity is not involved in maintaining the AHP reduction at this point after learning.
Collapse
|
25
|
Burgos-Ramos E, Hervás-Aguilar A, Aguado-Llera D, Puebla-Jiménez L, Hernández-Pinto AM, Barrios V, Arilla-Ferreiro E. Somatostatin and Alzheimer's disease. Mol Cell Endocrinol 2008; 286:104-11. [PMID: 18359553 DOI: 10.1016/j.mce.2008.01.014] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2007] [Revised: 01/23/2008] [Accepted: 01/24/2008] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is characterized by the cerebral deposition of senile plaques that are mainly composed of a set of peptides referred to as amyloid beta-peptides (Abeta). Among the numerous neuropeptides produced in intrinsic cortical and hippocampal neurons, somatostatin (SRIF) has been found to be the most consistently reduced in the brain and cerebrospinal fluid of AD patients. SRIF receptors (SSTR), which mediate the neuromodulatory signals of SRIF, are also markedly depleted in the AD brain, there being subtype-selective alterations in cortical areas. In the rat temporal cortex, we have shown that intracerebroventricular infusion of Abeta25-35 results in a decrease in SRIF-like immunoreactivity and in SRIF receptor subtype 2 (SSTR2) mRNA and protein levels, in correlation with a decrease in SSTR functionality. Insulin-like growth factor-I prevents the reduction in these parameters induced by Abeta25-35. Abeta has recently been demonstrated to be degraded primarily by a neutral endopeptidase, neprilysin, in the brain. SRIF regulates brain Abeta levels via modulation of neprilysin activity. Because SRIF expression in the brain declines upon aging in various mammals, including rodents, apes and humans, the aging-dependent reduction of SRIF has been hypothesized to trigger accumulation of Abeta in the brain by suppressing neprilysin action. Here we present an overview of recent advances on the role of SRIF in AD and its relationship with Abeta peptides.
Collapse
Affiliation(s)
- E Burgos-Ramos
- Unidad de Neurobioquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Alcalá, Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
26
|
Pinto C, Papa D, Hübner M, Mou TC, Lushington GH, Seifert R. Activation and inhibition of adenylyl cyclase isoforms by forskolin analogs. J Pharmacol Exp Ther 2008; 325:27-36. [PMID: 18184830 DOI: 10.1124/jpet.107.131904] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Adenylyl cyclase (AC) isoforms 1 to 9 are differentially expressed in tissues and constitute an interesting drug target. ACs 1 to 8 are activated by the diterpene, forskolin (FS). It is unfortunate that there is a paucity of AC isoform-selective activators. To develop such compounds, an understanding of the structure/activity relationships of diterpenes is necessary. Therefore, we examined the effects of FS and nine FS analogs on ACs 1, 2, and 5 expressed in Spodoptera frugiperda insect cells. Diterpenes showed the highest potencies at AC1 and the lowest potencies at AC2. We identified full agonists, partial agonists, antagonists, and inverse agonists, i.e., diterpenes that reduced basal AC activity. Each AC isoform exhibited a distinct pharmacological profile. AC2 showed the highest basal activity of all AC isoforms and highest sensitivity to inverse agonistic effects of 1-deoxy-forskolin, 7-deacetyl-1,9-dideoxy-forskolin, and, particularly, BODIPY-forskolin. In contrast, BODIPY-forskolin acted as partial agonist at the other ACs. 1-Deoxy-forskolin analogs were devoid of agonistic activity at ACs but antagonized the effects of FS in a mixed competitive/noncompetitive manner. At purified catalytic AC subunits, BODIPY-forskolin acted as weak partial agonist/strong partial antagonist. Molecular modeling revealed that the BODIPY group rotates promiscuously outside of the FS-binding site. Collectively, ACs are not uniformly activated and inhibited by FS and FS analogs, demonstrating the feasibility to design isoform-selective FS analogs. The two- and multiple-state models, originally developed to conceptualize ligand effects at G-protein-coupled receptors, can be applied to ACs to explain certain experimental data.
Collapse
Affiliation(s)
- Cibele Pinto
- Department of Pharmacology and Toxicology, University of Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| | | | | | | | | | | |
Collapse
|
27
|
Albasanz JL, Rodríguez A, Ferrer I, Martín M. Up-regulation of adenosine A1 receptors in frontal cortex from Pick's disease cases. Eur J Neurosci 2007; 26:3501-8. [DOI: 10.1111/j.1460-9568.2007.05965.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
28
|
Perez-Buira S, Barrachina M, Rodriguez A, Albasanz JL, Martín M, Ferrer I. Expression levels of adenosine receptors in hippocampus and frontal cortex in argyrophilic grain disease. Neurosci Lett 2007; 423:194-9. [PMID: 17707587 DOI: 10.1016/j.neulet.2007.06.049] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2007] [Revised: 06/24/2007] [Accepted: 06/25/2007] [Indexed: 12/24/2022]
Abstract
Expression of adenosine receptors of the A1, A2A and A2B type has been examined in the post-mortem frontal cortex and hippocampus in argyrophilic grain disease (AGD), a tauopathy affecting the hippocampus but usually not the frontal cortex, in an attempt to learn about the modulation of the adenosine pathway in this disorder. Significant increased levels of A1, but not of A2A and A2B, have been observed in AGD in the hippocampus but not in the frontal cortex, when compared with age-matched controls. This is accompanied by increased levels of adenylyl cyclase (AC), an effector of A1, and by increased (although not significant) percentage of inhibition of forskolin-stimulated AC by the A1 agonist cyclohexyladenosine in the hippocampus in AGD. These findings indicate sensitization of A1/AC in the hippocampus in AGD, and support a putative activation of the A1/AC pathway that may facilitate protection of this preferentially involved region in AGD.
Collapse
Affiliation(s)
- S Perez-Buira
- Institut de Neuropatologia, Servei Anatomia Patològica, IDIBELL-Hospital Universitari de Bellvitge, Facultad de Medicina, Universitat de Barcelona, carrer Feixa Llarga sn, 08907 Hospitalet de Llobregat, Spain
| | | | | | | | | | | |
Collapse
|
29
|
Burgos-Ramos E, Hervás-Aguilar A, Puebla-Jiménez L, Boyano-Adánez MC, Arilla-Ferreiro E. Chronic but not acute intracerebroventricular administration of amyloid beta-peptide(25-35) decreases somatostatin content, adenylate cyclase activity, somatostatin-induced inhibition of adenylate cyclase activity, and adenylate cyclase I levels in the rat hippocampus. J Neurosci Res 2007; 85:433-42. [PMID: 17086550 DOI: 10.1002/jnr.21115] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Although alterations in adenylate cyclase (AC) activity and somatostatin (SRIF) receptor density have been reported in Alzheimer's disease, the effects of amyloid beta-peptide (Abeta) on these parameters in the hippocampus are unknown. Our aim was to investigate whether the peptide fragment Abeta(25-35) can affect the somatostatinergic system in the rat hippocampus. Hence, Abeta(25-35) was injected intracerebroventricularly (i.c.v.) to Wistar rats in a single dose or infused via an osmotic minipump connected to a cannula implanted in the right lateral ventricle during 14 days. The animals were decapitated 7 or 14 days after the single injection and 14 days after chronic infusion of the peptide. Chronic i.c.v. infusion of Abeta(25-35) decreased SRIF-like immunoreactive content without modifying the SRIF receptor density, SRIF receptor expression, or the Gialpha(1), Gialpha(2), and Gialpha(3) protein levels in the hippocampus. This treatment, however, caused a decrease in basal and forskolin-stimulated AC activity as well as in the capacity of SRIF to inhibit AC activity. Furthermore, the protein levels of the neural-specific AC type I were significantly decreased in the hippocampus of the treated rats, whereas an increase in the levels of AC V/VI was found, with no alterations in type VIII AC. A single i.c.v. dose of Abeta(25-35) exerted no effect on SRIF content or SRIF receptors but induced a slight decrease in forskolin-stimulated AC activity and its inhibition by SRIF. Because chronic Abeta(25-35) infusion impairs learning and memory whereas SRIF facilitates these functions, the alterations described here might be physiologically important given the decreased cognitive behavior previously reported in Abeta-treated rats.
Collapse
Affiliation(s)
- E Burgos-Ramos
- Grupo de Neurobioquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
| | | | | | | | | |
Collapse
|
30
|
Rodríguez A, Martín M, Albasanz JL, Barrachina M, Espinosa JC, Torres JM, Ferrer I. Adenosine A1 Receptor Protein Levels and Activity Is Increased in the Cerebral Cortex in Creutzfeldt-Jakob Disease and in Bovine Spongiform Encephalopathy-Infected Bovine-PrP Mice. J Neuropathol Exp Neurol 2006; 65:964-75. [PMID: 17021401 DOI: 10.1097/01.jnen.0000235120.59935.f5] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Prion diseases are characterized by neuronal loss, astrocytic gliosis, spongiform change, and abnormal protease-resistant prion protein (PrP) deposition. Creutzfeldt-Jakob disease (CJD) is the most prevalent human prion disease, whereas scrapie and bovine spongiform encephalopathy (BSE) are the most common animal prion diseases. Several candidates have been proposed as mediators of degeneration in prion diseases, one of them glutamate. Recent studies have shown reduced metabotropic glutamate receptor/phospholipase C signaling in the cerebral cortex in CJD, suggesting that this important neuromodulator and neuroprotector pathway is attenuated in CJD. Adenosine is involved in the regulation of different metabolic processes under physiological and pathologic conditions. Adenosine function is mediated by adenosine receptors, which are categorized into 4 types: A1, A2A, A2B, and A3. A1Rs are G-protein-coupled receptors that induce the inhibition of adenylyl cyclase activity. The most dramatic inhibitory actions of adenosine receptors are on the glutamatergic system. For these reasons, we examined the levels of A1Rs in the frontal cortex of 12 patients with CJD and 6 age-matched controls and in BSE-infected bovine-PrP transgenic mice (BoPrP-Tg110 mice) at different postincubation times to address modifications in A1Rs with disease progression. A significant increase in the protein levels of A1Rs was found in the cerebral cortex in CJD and in the murine BSE model at advanced stages of the disease and coincidental with the appearance of PrP expression. In addition, the activity of A1Rs was analyzed by in vitro assays with isolated membranes of the frontal cortex in CJD. Increased activity of the receptor, as revealed by the decreased forskolin-stimulated cAMP production in response to the A1R agonists cyclohexyl adenosine and cyclopentyl adenosine, was observed in CJD cases when compared with controls. Finally, mRNA A1R levels were similar in CJD and control cases, thus suggesting abnormal A1R turnover or dysregulation of raft-associated signaling pathways in CJD. These results show, for the first time, sensitization of A1Rs in prion diseases.
Collapse
Affiliation(s)
- Agustín Rodríguez
- Institut de Neuropatologia, Servei Anatomia Patològica, IDIBELL-Hospital Universitari de Bellvitge, Universitat de Barcelona, 08907 Hospitalet de Llobregat, Spain
| | | | | | | | | | | | | |
Collapse
|
31
|
Hawes JJ, Narasimhaiah R, Picciotto MR. Galanin attenuates cyclic AMP regulatory element-binding protein (CREB) phosphorylation induced by chronic morphine and naloxone challenge in Cath.a cells and primary striatal cultures. J Neurochem 2006; 96:1160-8. [PMID: 16417577 DOI: 10.1111/j.1471-4159.2005.03613.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Repeated morphine administration leads to molecular alterations of the neural circuitry in the locus coeruleus and nucleus accumbens. These changes include increased activity of several components of the cAMP signaling pathway that are thought to be associated with psychological and somatic signs of opiate withdrawal. The neuropeptide galanin has been shown to attenuate cAMP signaling in multiple cell types. The current study demonstrates that acute galanin treatment blocks the consequences of increased cAMP signaling following chronic opiate administration and withdrawal in Cath.a cells and primary cultures of striatal neurons as measured by phosphorylation of the transcription factor cAMP regulatory element-binding protein (CREB). In addition, galanin-mediated attenuation of CREB phosphorylation is independent of galanin-induced extracellular signal-regulated kinase (ERK) 1/2 phosphorylation in Cath.a cells. These data suggest that galanin receptors may serve as an additional potential therapeutic target for the treatment of opiate withdrawal.
Collapse
Affiliation(s)
- Jessica J Hawes
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut 06508, USA
| | | | | |
Collapse
|
32
|
Dalfó E, Albasanz JL, Rodríguez A, Martín M, Ferrer I. Abnormal Group I Metabotropic Glutamate Receptor Expression and Signaling in the Frontal Cortex in Pick Disease. J Neuropathol Exp Neurol 2005; 64:638-47. [PMID: 16042316 DOI: 10.1097/01.jnen.0000171649.86718.f2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Group I metabotropic glutamate receptors (mGluR1) regulate synaptic transmission through the stimulation of phospholipase Cbeta1 (PLCbeta1) and then by the activation of protein kinase C (PKC). Considering these properties, it is conceivable that major cortical functional deficits may be attributed to abnormal mGluR processing and signaling. The present work examines mGluRI expression and signaling in the frontal cortex (area 8) of 3 cases with Pick disease (PiD), a neurodegenerative disease with abnormal phospho-tau accumulation, in comparison with 3 age-matched controls by means of glutamate binding assays, enzymatic activity, gel electrophoresis and Western blotting, solubility and immunoprecipitation assays, and confocal microscopy. Reduced expression levels of PLCbeta1 and reduced PLCbeta1 activity have been found in PiD. The expression levels of the nonrelated phospholipase PLCgamma, a substrate of tyrosine kinase, are also reduced in PiD. This is accompanied by a marked decrease in the expression of cPKCalpha and increased expression of the inner band (76 kDa) of the nPKCdelta doublet at the expense of a decrease of the phosphorylated (active) form (78 kDa). In contrast, L-[3H]glutamate-specific binding to mGluRs is augmented in PiD cases, mainly because of the higher mGluR1 and mGluRs expression levels detected. No modifications in PLCbeta1 solubility have been observed in PiD and no interactions between PLCbeta1 and tau have been demonstrated in diseased and control cases. Moreover, double-labeling immunofluorescence and confocal microscopy have shown no colocalization of phospho-tau (AT8 antibody) and PLCbeta1 in phospho-tau inclusions, including Pick bodies. These results demontrate for the first time abnormal mGluR signaling in the cerebral cortex in PiD and selective vulnerability of phospholipases and PKC to PiD.
Collapse
Affiliation(s)
- Esther Dalfó
- Institut de Neuropatologia, Servei Anatomia Patològica, IDIBELL-Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Spain
| | | | | | | | | |
Collapse
|
33
|
Mons N, Segu L, Nogues X, Buhot MC. Effects of age and spatial learning on adenylyl cyclase mRNA expression in the mouse hippocampus. Neurobiol Aging 2004; 25:1095-106. [PMID: 15212834 DOI: 10.1016/j.neurobiolaging.2003.10.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2003] [Revised: 10/16/2003] [Accepted: 10/22/2003] [Indexed: 11/30/2022]
Abstract
Adenylyl cyclase (AC) subtypes have been implicated in memory processes and synaptic plasticity. In the present study, the effects of aging and learning on Ca2+/calmodulin-stimulable AC1, Ca2+-insensitive AC2 and Ca2+/calcineurin-inhibited AC9 mRNA level were compared in the dorsal hippocampus of young-adult and aged C57BL/6 mice using in situ hybridization. Both AC1 and AC9 mRNA expression were downregulated in aged hippocampus, whereas AC2 mRNA remained unchanged, suggesting differential sensitivities to the aging process. We next examined AC mRNA expression in the hippocampus after spatial learning in the Morris water maze. Acquisition of the spatial task was associated with an increase of AC1 and AC9 mRNA levels in both young-adult and aged groups, suggesting that Ca2+-sensitive ACs are oppositely regulated by aging and learning. However, aged-trained mice had reduced AC1 and AC9, but greater AC2, mRNA levels relative to young-trained mice and age-related learning impairments were correlated with reduced AC1 expression in area CA1. We suggest that reduced levels of hippocampal AC1 mRNA may greatly contribute to age-related defects in spatial memory.
Collapse
Affiliation(s)
- N Mons
- Laboratoire de Neurosciences Cognitives, UMR 5106, Avenue des Facultés, 33405 Talence Cedex, France.
| | | | | | | |
Collapse
|
34
|
James R, Adams RR, Christie S, Buchanan SR, Porteous DJ, Millar JK. Disrupted in Schizophrenia 1 (DISC1) is a multicompartmentalized protein that predominantly localizes to mitochondria. Mol Cell Neurosci 2004; 26:112-22. [PMID: 15121183 DOI: 10.1016/j.mcn.2004.01.013] [Citation(s) in RCA: 115] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2003] [Revised: 01/19/2004] [Accepted: 01/21/2004] [Indexed: 02/08/2023] Open
Abstract
DISC1 is disrupted by a chromosomal translocation cosegregating with schizophrenia and recurrent major depression in a large Scottish family and has also been reported as a potential susceptibility locus in independent populations. We reveal a widespread and complex pattern of DISC1 expression, with at least five forms of Disrupted in Schizophrenia 1 DISC1 detectable. Mitochondria are the predominant site of DISC1 expression with additional nuclear, cytoplasmic, and actin-associated locations evident. Although the subcellular targeting of DISC1 is clearly complex, the association with mitochondria is of interest as many mitochondrial deficits have been reported in schizophrenia and other neuropsychiatric illnesses. Moreover, of the many cellular functions performed by mitochondria, their role in oxidative phosphorylation, calcium homeostasis, and apoptosis may hold particular relevance for the neuronal disturbances believed to be involved in the pathogenesis of schizophrenia.
Collapse
Affiliation(s)
- R James
- Medical Genetics Section, Department of Medical Sciences, University of Edinburgh, Molecular Medicine Centre, Western General Hospital, Edinburgh EH4 2XU, UK.
| | | | | | | | | | | |
Collapse
|
35
|
Wang H, Ferguson GD, Pineda VV, Cundiff PE, Storm DR. Overexpression of type-1 adenylyl cyclase in mouse forebrain enhances recognition memory and LTP. Nat Neurosci 2004; 7:635-42. [PMID: 15133516 DOI: 10.1038/nn1248] [Citation(s) in RCA: 170] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2004] [Accepted: 04/01/2004] [Indexed: 01/15/2023]
Abstract
Cyclic AMP is a positive regulator of synaptic plasticity and is required for several forms of hippocampus-dependent memory including recognition memory. The type I adenylyl cyclase, Adcy1 (also known as AC1), is crucial in memory formation because it couples Ca(2+) to cyclic AMP increases in the hippocampus. Because Adcy1 is neurospecific, it is a potential pharmacological target for increasing cAMP specifically in the brain and for improving memory. We have generated transgenic mice that overexpress Adcy1 in the forebrain using the Camk2a (also known as alpha-CaMKII) promoter. These mice showed elevated long-term potentiation (LTP), increased memory for object recognition and slower rates of extinction for contextual memory. The increase in recognition memory and lower rates of contextual memory extinction may be due to enhanced extracellular signal-related kinase (ERK)/mitogen-activated protein kinase (MAPK) signaling, which is elevated in mice that overexpress Adcy1.
Collapse
Affiliation(s)
- Hongbing Wang
- Department of Pharmacology, University of Washington, Box 357280, 1959 NE Pacific Street, Seattle, Washington 98195-7280, USA
| | | | | | | | | |
Collapse
|
36
|
García-Jiménez A, Fastbom J, Ohm TG, Cowburn RF. G-protein alpha-subunit levels in hippocampus and entorhinal cortex of brains staged for Alzheimer's disease neurofibrillary and amyloid pathologies. Neuroreport 2003; 14:1523-7. [PMID: 12960778 DOI: 10.1097/00001756-200308060-00025] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
G-protein alpha-subunits (Galphao, Galphai, Galphas, Galphaq) and adenylyl cyclase (AC) I and II isoforms were quantified in hippocampus and entorhinal cortex from 22 cases staged for Alzheimer's disease (AD) pathologies according to Braak and Braak. Hippocampal Galphai levels declined significantly with neurofibrillary staging, whereas AC I levels in this region increased. Significant amyloid stage-related reductions of Galphai were seen in both the hippocampus and entorhinal cortex. The hippocampus also showed a significant reduction of Galphao with amyloid staging. It is concluded that levels of inhibitory G-protein subunits Galphao, and in particular Galphai, decrease in parallel to the extent of AD pathology.
Collapse
Affiliation(s)
- Angela García-Jiménez
- Neurotec Department, Division of Experimental Geriatrics, Karolinska Institutet, Novum, KFC, S-141 86 Huddinge, Sweden
| | | | | | | |
Collapse
|
37
|
Chong YH, Shin YJ, Suh YH. Cyclic AMP inhibition of tumor necrosis factor alpha production induced by amyloidogenic C-terminal peptide of Alzheimer's amyloid precursor protein in macrophages: involvement of multiple intracellular pathways and cyclic AMP response element binding protein. Mol Pharmacol 2003; 63:690-8. [PMID: 12606779 DOI: 10.1124/mol.63.3.690] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
In the present study, we focused on the molecular events involved in tumor necrosis factor-alpha (TNF-alpha) production in response to the amyloidogenic 105-amino acid carboxyl-terminal fragment (CT105) of amyloid precursor protein, a candidate alternative toxic element in Alzheimer's disease pathology, and the mechanisms by which cyclic AMP regulates the relating inflammatory signal cascades. CT105 at nanomolar concentrations strongly activated multiple signaling pathways involving tyrosine kinase-dependent extracellular signal-regulated kinase and p38 mitogen-activated protein kinases. Moreover, phosphatidylinositol 3-kinase/Akt signal was required for excess TNF-alpha production in human macrophages derived from THP-1 cells. Interferon-gamma significantly potentiated the induction of the CT105-mediated signal cascade. These multiple signaling pathways in turn converged, at least in part, at the nuclear transcription factor known as cAMP response element binding protein (CREB), which acts on the TNF-alpha gene promoter through the cAMP response element. The cell-permeable cAMP analog dibutyryl cAMP partially and almost simultaneously suppressed all of these CT105-induced signaling pathways through excessive CREB phosphorylation, which led to decreased CREB DNA binding activity and reduced TNF-alpha expression. Furthermore, dibutyryl cAMP decreased the interaction of the p65 nuclear factor-kappa B with CREB binding protein, thus further inhibiting CT105-mediated TNF-alpha expression. Collectively, the detailed molecular mechanisms of amyloidogenic CT-induced TNF-alpha production as negatively regulated by cAMP may advance the possibility of targeted treatment in Alzheimer's disease.
Collapse
Affiliation(s)
- Young Hae Chong
- Department of Microbiology, College of Medicine, Division of Molecular Biology and Neuroscience, Medical Research Center, Ewha Womans University, Seoul, Korea.
| | | | | |
Collapse
|
38
|
Chong YH, Shin SA, Lee HJ, Kang JHL, Suh YH. Molecular mechanisms underlying cyclic AMP inhibition of macrophage dependent TNF-alpha production and neurotoxicity in response to amyloidogenic C-terminal fragment of Alzheimer's amyloid precursor protein. J Neuroimmunol 2002; 133:160-74. [PMID: 12446019 DOI: 10.1016/s0165-5728(02)00349-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
In the present study, we characterized the intracellular pathway involved in the macrophage production of tumor necrosis factor-alpha (TNF-alpha) and the molecular mechanisms by which cyclic AMP (cAMP) regulates the neurotoxic inflammatory signaling cascade in response to the 105 amino acid carboxyl-terminal fragment (CT105) of amyloid precursor protein, a candidate of alternative toxic elements in Alzheimer's disease (AD) pathology. CT105 in combination with interferon-gamma (IFN-gamma) elicited a robust and sustained increase of TNF-alpha production due to enhanced TNF-alpha mRNA transcription, mediated via increased nuclear factor-kappaB (NF-kappaB) in human macrophages derived from monocytic THP-1 cells. A mechanistic analysis revealed that the cAMP analog, dibutyryl cyclic AMP (dbcAMP), or the adenyl cyclase activator, forskolin, effectively suppressed the stimulant-induced TNF-alpha production by reducing the nuclear translocation and DNA binding activity of NF-kappaB. The inhibitory mechanisms manifested by dbcAMP included the decreased phosphorylation/degradation of NF-kappaB inhibitor (IkappaB) followed by its increased synthesis/stability. Importantly, this macrophage derived TNF-alpha appears to be a key pathological mediator of the resultant neurotoxicity, which was attenuated by increased cAMP levels during macrophage stimulation with CT105. These findings provide evidence, which supports an important role of CT105 as a potent macrophage stimulator eliciting NF-kappaB-mediated inflammatory signals for excess TNF-alpha production, which in turn ultimately leads to the neurotoxicity. In addition, the detailed inhibitory mechanism of cAMP action implies that an increased cAMP level could be benefit against AD progression.
Collapse
Affiliation(s)
- Young Hae Chong
- Department of Microbiology, College of Medicine, Division of Molecular Biology and Neuroscience, Medical Research Center, Ewha Womans University, 911-1, Mok-6-dong, Yangcheonku, 158-710, Seoul, South Korea.
| | | | | | | | | |
Collapse
|