1
|
Rácz K, Segal Y, Lénárt K, Fillér C, Tóth A, Szegeczki V, Gergely P, Zákány R, Reglődi D, Juhász T. Cartilage degradation is followed by PAC1 receptor reduction in articular cartilage of human knee joints. GeroScience 2025:10.1007/s11357-025-01689-4. [PMID: 40369255 DOI: 10.1007/s11357-025-01689-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Accepted: 04/29/2025] [Indexed: 05/16/2025] Open
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is a neuropeptide expressed in the nervous system and also in various peripheral tissues, including the musculoskeletal system. PACAP has an important function in the regulation of chondrogenesis and plays a protective role in cartilage oxidative and mechanical stress. PACAP knockout (KO) mice show early signs of aging and osteoarthritis in knee joint articular cartilage. Its specific, most potent receptor is the PAC1 receptor, the activation of which leads to enhanced Sox9 expression and subsequently, it increases the expression of collagen type II, glucosaminoglycans and aggrecan. In the present study, we investigated articular cartilage of human knee joints taken from cadavers of varying ages. Thickness and extracellular matrix content of articular cartilage of knee joints decreases with aging. The cartilage degeneration process most likely begins between the ages of 40 to 50. Expression of PAC1 receptor decreases in parallel with the reduction of cartilage thickness, leading to subsequent reduced Sox9 expression with cartilage specific matrix production. In summary, we found correlation in the reduction of cartilage thickness and quality together with PAC1 receptor expression and activity.
Collapse
Affiliation(s)
- Kálmán Rácz
- Department of Forensic Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98, 4032, Debrecen, Hungary
| | - Yonatan Segal
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98, 4032, Debrecen, Hungary
| | - Kinga Lénárt
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98, 4032, Debrecen, Hungary
| | - Csaba Fillér
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98, 4032, Debrecen, Hungary
| | - Anna Tóth
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98, 4032, Debrecen, Hungary
| | - Vince Szegeczki
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98, 4032, Debrecen, Hungary
| | - Péter Gergely
- Department of Forensic Medicine, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98, 4032, Debrecen, Hungary
| | - Róza Zákány
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98, 4032, Debrecen, Hungary
| | - Dóra Reglődi
- Department of Anatomy, Medical School, HUN-REN-PTE PACAP Research Team, University of Pécs, Szigeti Út 12, 7624, Pécs, Hungary
| | - Tamás Juhász
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98, 4032, Debrecen, Hungary.
| |
Collapse
|
2
|
Yavas E, Zhuravka I, Fanselow MS. PAC1 receptor modulation of freezing and flight behavior in periaqueductal gray. GENES, BRAIN, AND BEHAVIOR 2023; 22:e12873. [PMID: 37983568 PMCID: PMC10733566 DOI: 10.1111/gbb.12873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/05/2023] [Accepted: 11/05/2023] [Indexed: 11/22/2023]
Abstract
The midbrain periaqueductal gray (PAG) region is a critical anatomical regulator of fear-related species-specific defensive reactions (SSDRs). Pituitary adenylate-cyclase-activating polypeptide (PACAP), and its main receptor PAC1, play an important role in fear-related behavior and anxiety disorders. However, the function of the PACAP-PAC1 system within the PAG with regards to SSDRs has received little attention. To address this gap, we used transgenic PAC1flox/flox mice to examine both conditional and unconditional defensive reactions. We performed conditional PAC1 gene deletion within the ventrolateral(vl)PAG of PAC1flox/flox mice using an adeno-associated virus (AAV) coding for Cre recombinase. Following viral expression, we used a white noise fear conditioning preparation that produces both an unconditional activity burst to the onset of noise that is followed by conditional freezing. On Day 1, mice received five white noise foot-shock pairings, whereas on Day 2, they were exposed to white noise five times without shock and we scored the activity burst and freezing to the white noise. Following behavioral testing, histology for immunofluorescent analysis was conducted in order to identify PACAP positive cells and stress-induced c-fos activity respectively. We found that PAC1 deletion in vlPAG increased the unconditional activity burst response but disrupted conditional freezing. PAC1 deletion was accompanied by higher c-fos activity following the behavioral experiments. Furthermore, a significant portion of PACAP-EGFP positive cells showed overlapping expression with VGAT, indicating their association with inhibitory neurons. The findings suggested that intact PACAP-PAC1 mechanisms are essential for SSDRs in vlPAG. Therefore, midbrain PACAP contributes to the underlying molecular mechanisms regulating fear responses.
Collapse
Affiliation(s)
- Ersin Yavas
- Department of PsychologyBartın UniversityBartınTurkey
| | - Irina Zhuravka
- Staglin Center for Brain and Behavioral HealthDepartment of Psychology, UCLALos AngelesCaliforniaUSA
| | - Michael S. Fanselow
- Staglin Center for Brain and Behavioral HealthDepartment of Psychology, UCLALos AngelesCaliforniaUSA
- Department of Psychiatry and Biobehavioral SciencesUCLALos AngelesCaliforniaUSA
| |
Collapse
|
3
|
Wang DX, Dong ZJ, Deng SX, Tian YM, Xiao YJ, Li X, Ma XR, Li L, Li P, Chang HZ, Liu L, Wang F, Wu Y, Gao X, Zheng SS, Gu HM, Zhang YN, Wu JB, Wu F, Peng Y, Zhang XW, Zhan RY, Gao LX, Sun Q, Guo X, Zhao XD, Luo JH, Zhou R, Han L, Shu Y, Zhao JW. GDF11 slows excitatory neuronal senescence and brain ageing by repressing p21. Nat Commun 2023; 14:7476. [PMID: 37978295 PMCID: PMC10656444 DOI: 10.1038/s41467-023-43292-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 11/06/2023] [Indexed: 11/19/2023] Open
Abstract
As a major neuron type in the brain, the excitatory neuron (EN) regulates the lifespan in C. elegans. How the EN acquires senescence, however, is unknown. Here, we show that growth differentiation factor 11 (GDF11) is predominantly expressed in the EN in the adult mouse, marmoset and human brain. In mice, selective knock-out of GDF11 in the post-mitotic EN shapes the brain ageing-related transcriptional profile, induces EN senescence and hyperexcitability, prunes their dendrites, impedes their synaptic input, impairs object recognition memory and shortens the lifespan, establishing a functional link between GDF11, brain ageing and cognition. In vitro GDF11 deletion causes cellular senescence in Neuro-2a cells. Mechanistically, GDF11 deletion induces neuronal senescence via Smad2-induced transcription of the pro-senescence factor p21. This work indicates that endogenous GDF11 acts as a brake on EN senescence and brain ageing.
Collapse
Affiliation(s)
- Di-Xian Wang
- Department of Pathology of Sir Run Run Shaw Hospital, and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang, China
- Center of Cryo-Electron Microscopy, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Zhao-Jun Dong
- Department of Pathology of Sir Run Run Shaw Hospital, and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang, China
- Center of Cryo-Electron Microscopy, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Sui-Xin Deng
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, 201508, Shanghai, China
| | | | - Yu-Jie Xiao
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, 201508, Shanghai, China
| | - Xinran Li
- The Global Scientific and Technological Innovation Center and the MOE Key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Xiao-Ru Ma
- Department of Pathology of Sir Run Run Shaw Hospital, and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang, China
- Center of Cryo-Electron Microscopy, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Liang Li
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, 201508, Shanghai, China
| | - Pengxiao Li
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai; Center for Systems Biomedicine, Shanghai Jiao Tong University, 200240, Shanghai, China
| | | | | | - Fan Wang
- Department of Pathology of Sir Run Run Shaw Hospital, and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang, China
- Center of Cryo-Electron Microscopy, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Yang Wu
- Department of Pathology of Sir Run Run Shaw Hospital, and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang, China
- Center of Cryo-Electron Microscopy, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Xiang Gao
- Department of Pathology of Sir Run Run Shaw Hospital, and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang, China
- Center of Cryo-Electron Microscopy, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Shuang-Shuang Zheng
- Department of Pathology of Sir Run Run Shaw Hospital, and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang, China
- Center of Cryo-Electron Microscopy, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Hui-Min Gu
- Department of Pathology of Sir Run Run Shaw Hospital, and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang, China
- Center of Cryo-Electron Microscopy, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Ya-Nan Zhang
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Jian-Bin Wu
- Department of Pathology of Sir Run Run Shaw Hospital, and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang, China
- Center of Cryo-Electron Microscopy, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Fan Wu
- Department of Neurosurgery, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, 310003, Hangzhou, China
| | - Yonglin Peng
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai; Center for Systems Biomedicine, Shanghai Jiao Tong University, 200240, Shanghai, China
| | - Xiao-Wen Zhang
- Department of Pathology of Sir Run Run Shaw Hospital, and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang, China
- Center of Cryo-Electron Microscopy, Zhejiang University, 310058, Hangzhou, Zhejiang, China
| | - Ren-Ya Zhan
- Department of Neurosurgery, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, 310003, Hangzhou, China
| | - Li-Xia Gao
- Department of Neurology of the Second Affiliated Hospital, Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, 310020, Hangzhou, China
| | - Qiming Sun
- Department of Biochemistry, and Department of Cardiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xing Guo
- Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Xiao-Dong Zhao
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai; Center for Systems Biomedicine, Shanghai Jiao Tong University, 200240, Shanghai, China
| | - Jian-Hong Luo
- Department of Neurobiology and Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 310058, Hangzhou, Zhejiang, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Zhejiang, China
| | - Ruhong Zhou
- Institute of Quantitative Biology, College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Lei Han
- BGI Research, 310030, Hangzhou, China.
| | - Yousheng Shu
- Department of Neurosurgery, Jinshan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Fudan University, 201508, Shanghai, China.
| | - Jing-Wei Zhao
- Department of Pathology of Sir Run Run Shaw Hospital, and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, 310058, Hangzhou, Zhejiang, China.
- Center of Cryo-Electron Microscopy, Zhejiang University, 310058, Hangzhou, Zhejiang, China.
| |
Collapse
|
4
|
Elgendy SA, Soliman MM, Ghamry HI, Shukry M, Mohammed LA, Nasr HE, Alotaibi BS, Jafri I, Sayed S, Osman A, Elnoury HA. Exploration of Tilmicosin Cardiotoxicity in Rats and the Protecting Role of the Rhodiola rosea Extract: Potential Roles of Cytokines, Antioxidant, Apoptotic, and Anti-Fibrotic Pathways. TOXICS 2023; 11:857. [PMID: 37888707 PMCID: PMC10610616 DOI: 10.3390/toxics11100857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/18/2023] [Accepted: 09/20/2023] [Indexed: 10/28/2023]
Abstract
Tilmicosin (TIL) is a common macrolide antibiotic in veterinary medicine. High doses of TIL can have adverse cardiovascular effects. This study examined the effects of Rhodiola rosea (RHO) that have anti-inflammatory, antioxidant, and anti-fibrotic effects on tilmicosin (TIL)-induced cardiac injury targeting anti-inflammatory, antioxidant, apoptotic, and anti-apoptotic signaling pathways with anti-fibrotic outcomes. Thirty-six male Wistar albino rats were randomly divided into groups of six rats each. Rats received saline as a negative control, CARV 1 mL orally (10 mg/kg BW), and RHO 1 mL orally at 400 mg/kg BW daily for 12 consecutive days. The TIL group once received a single subcutaneous injection (SC) dose of TIL (75 mg/kg BW) on the sixth day of the experiment to induce cardiac damage. The standard group (CARV + TIL) received CARV daily for 12 consecutive days with a single TIL SC injection 1 h after CARV administration only on the sixth day of study and continued for another six successive days on CARV. The protective group (RHO + TIL) received RHO daily for the same period as in CARV + TIL-treated rats and with the dosage mentioned before. Serum was extracted at the time of the rat's scarification at 13 days of study and examined for biochemical assessments in serum lactate dehydrogenase (LDH), cardiac troponin I (cTI), and creatine phosphokinase (CK-MB). Protein carbonyl (PC) contents, malondialdehyde (MDA), and total antioxidant capacity (TAC) in cardiac homogenate were used to measure these oxidative stress markers. Quantitative RT-PCR was used to express interferon-gamma (INF-γ), cyclooxygenase-2 (COX-2), OGG1, BAX, caspase-3, B-cell lymphoma-2 (Bcl-2), and superoxide dismutase (SOD) genes in cardiac tissues, which are correlated with inflammation, antioxidants, and apoptosis. Alpha-smooth muscle actin (α-SMA), calmodulin (CaMKII), and other genes associated with Ca2+ hemostasis and fibrosis were examined using IHC analysis in cardiac cells (myocardium). TIL administration significantly increased the examined cardiac markers, LDH, cTI, and CK-MB. TIL administration also increased ROS, PC, and MDA while decreasing antioxidant activities (TAC and SOD mRNA) in cardiac tissues. Serum inflammatory cytokines and genes of inflammatory markers, DNA damage (INF-γ, COX-2), and apoptotic genes (caspase-3 and BAX) were upregulated with downregulation of the anti-apoptotic gene Bcl-2 as well as the DNA repair OGG1 in cardiac tissues. Furthermore, CaMKII and α-SMA genes were upregulated at cellular levels using cardiac tissue IHC analysis. On the contrary, pretreatment with RHO and CARV alone significantly decreased the cardiac injury markers induced by TIL, inflammatory and anti-inflammatory cytokines, and tissue oxidative-antioxidant parameters. INF-γ, COX-2, OGG1, BAX, and caspase-3 mRNA were downregulated, as observed by real-time PCR, while SOD and Bcl-2 mRNA were upregulated. Furthermore, the CaMKII and α-SMA genes' immune reactivities were significantly decreased in the RHO-pretreated rats.
Collapse
Affiliation(s)
- Salwa A. Elgendy
- Department of Pharmacology, Faculty of Medicine, Benha University, Benha 13511, Egypt
| | - Mohamed Mohamed Soliman
- Department of Clinical Laboratory Sciences, Turabah University College, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
- Biochemistry Department, Faculty of Veterinary Medicine, Benha University, Toukh 13736, Egypt
| | - Heba I. Ghamry
- Nutrition and Food Science, Department of Home Economics, Faculty of Home Economics, King Khalid University, P.O. Box 960, Abha 61421, Saudi Arabia;
| | - Mustafa Shukry
- Department of Physiology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Lina Abdelhady Mohammed
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha 13511, Egypt (H.E.N.)
| | - Hend Elsayed Nasr
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha 13511, Egypt (H.E.N.)
| | - Badriyah S. Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Ibrahim Jafri
- Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Samy Sayed
- Department of Economic Entomology and Pesticides, Faculty of Agriculture, Cairo University, Giza 12613, Egypt;
- Department of Science and Technology, University College-Ranyah, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Amira Osman
- Department of Basic Medical and Dental Sciences, Faculty of Dentistry, Zarqa University, Zarqa 13110, Jordan;
- Department of Histology and Cell Biology, Faculty of Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Heba A. Elnoury
- Department of Pharmacology, Faculty of Medicine, Benha University, Benha 13511, Egypt
| |
Collapse
|
5
|
Gilmartin MR, Ferrara NC. Pituitary Adenylate Cyclase-Activating Polypeptide in Learning and Memory. Front Cell Neurosci 2021; 15:663418. [PMID: 34239418 PMCID: PMC8258392 DOI: 10.3389/fncel.2021.663418] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 06/02/2021] [Indexed: 02/01/2023] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a highly conserved neuropeptide that regulates neuronal physiology and transcription through Gs/Gq-coupled receptors. Its actions within hypothalamic, limbic, and mnemonic systems underlie its roles in stress regulation, affective processing, neuroprotection, and cognition. Recently, elevated PACAP levels and genetic disruption of PAC1 receptor signaling in humans has been linked to maladaptive threat learning and pathological stress and fear in post-traumatic stress disorder (PTSD). PACAP is positioned to integrate stress and memory in PTSD for which memory of the traumatic experience is central to the disorder. However, PACAP's role in memory has received comparatively less attention than its role in stress. In this review, we consider the evidence for PACAP-PAC1 receptor signaling in learning and plasticity, discuss emerging data on sex differences in PACAP signaling, and raise key questions for further study toward elucidating the contribution of PACAP to adaptive and maladaptive fear learning.
Collapse
Affiliation(s)
| | - Nicole C Ferrara
- Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| |
Collapse
|
6
|
Huang X, Li J, Song S, Wang L, Lin Z, Ouyang Z, Yu R. Hormesis effect of hydrogen peroxide on the promoter activity of neuropeptide receptor PAC1-R. J Food Biochem 2019; 43:e12877. [PMID: 31353704 DOI: 10.1111/jfbc.12877] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 04/04/2019] [Accepted: 04/11/2019] [Indexed: 12/18/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) receptor 1 (PAC1-R) is the neuropeptide PACAP-preferring receptor-mediating neuroprotective activity. In order to clarify the biological mechanism of its expression, we cloned the 2,526 bp promoter fragment from -2,500 to +26 of the transcription initiation site of human ADCYAP1R1 gene and constructed the novel promotor reporter system named pYr-PromDetect-PAC1p. It was found in SH-SY5Y cells low concentration (<10 nM) of hydrogen peroxide (H2 O2 ) significantly promoted the activity of PAC1-R promoter in dose-dependent way, which was significantly inhibited by the transcription factor specificity protein 1 (SP1) inhibitor mithramycin A and was further confirmed in the deletion mutation of the predicted SP1 binding sites. Moreover, higher concentration of H2 O2 (>10 nM) inhibited the activity of PAC1-R in dose-dependent way. The hormesis effect of H2 O2 on PAC1-R promoter would help to further clarify the physiological effect of low-dose reactive oxygen on nervous system. PRACTICAL APPLICATIONS: PAC1-R mediates well-known neuroprotective, neurotrophic, and neurogenesis effects, which is an important drug target for neurodegenerative diseases. The hormesis effects of oxidative stress on PAC1-R expression not only help to explain the hormesis effects of oxidative stress on nerve system, but also offer a novel strategy to increase the expression of PAC1-R for the nerve protection or nerve generation. For example, taking advantage of low degree of oxidative stress to increases the expression of PAC1-R might help prevent subsequent surgical serious injury on the nervous system. The activation of PAC1-R promoter by low concentration of H2 O2 would help to further clarify the physiological effect of low-dose reactive oxygen on nervous system.
Collapse
Affiliation(s)
- Xiaoling Huang
- Institute of Biomedicine, School of Life Science and Technology, Jinan University, Guangzhou, China
| | - Junfeng Li
- Institute of Biomedicine, School of Life Science and Technology, Jinan University, Guangzhou, China
| | - Suqin Song
- Institute of Biomedicine, School of Life Science and Technology, Jinan University, Guangzhou, China
| | - Like Wang
- Institute of Biomedicine, School of Life Science and Technology, Jinan University, Guangzhou, China
| | - Zhuochao Lin
- Institute of Biomedicine, School of Life Science and Technology, Jinan University, Guangzhou, China
| | - Zehua Ouyang
- Institute of Biomedicine, School of Life Science and Technology, Jinan University, Guangzhou, China
| | - Rongjie Yu
- Institute of Biomedicine, School of Life Science and Technology, Jinan University, Guangzhou, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
7
|
Abstract
Dysregulation of neuropeptides may play an important role in aging-induced impairments. In the long list of neuropeptides, pituitary adenylate cyclase-activating polypeptide (PACAP) represents a highly effective cytoprotective peptide that provides an endogenous control against a variety of tissue-damaging stimuli. PACAP has neuro- and general cytoprotective effects due to anti-apoptotic, anti-inflammatory, and antioxidant actions. As PACAP is also a part of the endogenous protective machinery, it can be hypothesized that the decreased protective effects in lack of endogenous PACAP would accelerate age-related degeneration and PACAP knockout mice would display age-related degenerative signs earlier. Recent results support this hypothesis showing that PACAP deficiency mimics aspects of age-related pathophysiological changes including increased neuronal vulnerability and systemic degeneration accompanied by increased apoptosis, oxidative stress, and inflammation. Decrease in PACAP expression has been shown in different species from invertebrates to humans. PACAP-deficient mice display numerous pathological alterations mimicking early aging, such as retinal changes, corneal keratinization and blurring, and systemic amyloidosis. In the present review, we summarize these findings and propose that PACAP deficiency could be a good model of premature aging.
Collapse
|
8
|
Alteration of the PAC1 Receptor Expression in the Basal Ganglia of MPTP-Induced Parkinsonian Macaque Monkeys. Neurotox Res 2017; 33:702-715. [PMID: 29230633 DOI: 10.1007/s12640-017-9841-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 09/14/2017] [Accepted: 09/20/2017] [Indexed: 12/22/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a well-known neuropeptide with strong neurotrophic and neuroprotective effects. PACAP exerts its protective actions via three G protein-coupled receptors: the specific Pac1 receptor (Pac1R) and the Vpac1/Vpac2 receptors, the neuroprotective effects being mainly mediated by the Pac1R. The protective role of PACAP in models of Parkinson's disease and other neurodegenerative diseases is now well-established in both in vitro and in vivo studies. PACAP and its receptors occur in the mammalian brain, including regions associated with Parkinson's disease. PACAP receptor upregulation or downregulation has been reported in several injury models or human diseases, but no data are available on alterations of receptor expression in Parkinson's disease. The model closest to the human disease is the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced macaque model. Therefore, our present aim was to evaluate changes in Pac1R expression in basal ganglia related to Parkinson's disease in a macaque model. Monkeys were rendered parkinsonian with MPTP, and striatum, pallidum, and cortex were evaluated for Pac1R immunostaining. We found that Pac1R immunosignal was markedly reduced in the caudate nucleus, putamen, and internal and external parts of the globus pallidus, while the immunoreactivity remained unchanged in the cortex of MPTP-treated parkinsonian monkey brains. This decrease was attenuated in some brain areas in monkeys treated with L-DOPA. The strong, specific decrease of the PACAP receptor immunosignal in the basal ganglia of parkinsonian macaque monkey brains suggests that the PACAP/Pac1R system may play an important role in the development/progression of the disease.
Collapse
|
9
|
Ivic I, Solymar M, Fulop BD, Hashimoto H, Toth G, Tamas A, Juhasz T, Koller A, Reglodi D. Aging-Induced Modulation of Pituitary Adenylate Cyclase-Activating Peptide- and Vasoactive Intestinal Peptide-Induced Vasomotor Responses in the Arteries of Mice. J Vasc Res 2017; 54:359-366. [PMID: 29131060 DOI: 10.1159/000481781] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/23/2017] [Indexed: 12/17/2022] Open
Abstract
Pituitary adenylate cyclase-activating peptide (PACAP; 1-38 and 1-27) and vasoactive intestinal peptide (VIP) are related neuropeptides of the secretin/glucagon family. Overlapping signaling through G-protein-coupled receptors mediates their vasomotor activity. We previously showed that PACAP deficiency (PACAP-KO) shifts the mechanisms of vascular response and maintains arterial relaxation through the VIP backup mechanism and (mainly) its VPAC1R, but their age-dependent modulation is still unknown. We hypothesized that backup mechanisms exist, which maintain the vasomotor activity of these peptides also in older age. Thus, we investigated the effects of exogenous VIP and PACAP peptides in isolated carotid arteries of 2- and 15-month-old wild-type (WT) and PACAP-KO mice. All peptides induced relaxation in the arteries of young WT mice, whereas in young PACAP-KO mice PACAP1-27 and VIP, but not PACAP1-38, induced relaxation. Unlike VIP, PACAP-induced vasomotor responses were reduced in aging WT mice. However, in the arteries of aging PACAP-KO mice, PACAP1-27- and VIP-induced responses were reduced, but PACAP1-38 showed a greater vasomotor response compared to that of young PACAP-KO animals. There were no significant differences between the vasomotor responses of aging WT and PACAP-KO mice. Our data suggest that, in the absence of PACAP both in young and old ages, the vascular response is mediated through backup mechanisms, most likely VIP, maintaining proper vascular relaxation in aging-induced PACAP insufficiency.
Collapse
Affiliation(s)
- Ivan Ivic
- Department of Anatomy, MTA-PTE PACAP Research Group, Medical School, University of Pecs, Pecs, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Huang J, Waters K, Machaalani R. Hypoxia and nicotine effects on Pituitary adenylate cyclase activating polypeptide (PACAP) and its receptor 1 (PAC1) in the developing piglet brainstem. Neurotoxicology 2017; 62:30-38. [DOI: 10.1016/j.neuro.2017.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 05/11/2017] [Accepted: 05/11/2017] [Indexed: 11/27/2022]
|
11
|
Han P, Nielsen M, Song M, Yin J, Permenter MR, Vogt JA, Engle JR, Dugger BN, Beach TG, Barnes CA, Shi J. The Impact of Aging on Brain Pituitary Adenylate Cyclase Activating Polypeptide, Pathology and Cognition in Mice and Rhesus Macaques. Front Aging Neurosci 2017; 9:180. [PMID: 28659785 PMCID: PMC5467357 DOI: 10.3389/fnagi.2017.00180] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 05/23/2017] [Indexed: 01/30/2023] Open
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) is associated with Alzheimer's disease (AD), but its age-related effects are unknown. We chose the rhesus macaque due to its closeness to human anatomy and physiology. We examined four variables: aging, cognitive performance, amyloid plaques and PACAP. Delayed nonmatching-to-sample recognition memory scores declined with age and correlated with PACAP levels in the striatum, parietal and temporal lobes. Because amyloid plaques were the only AD pathology in the old rhesus macaque, we further studied human amyloid precursor protein (hAPP) transgenic mice. Aging was associated with decreased performance in the Morris Water Maze (MWM). In wild type (WT) C57BL/6 mice, the performance was decreased at age 24-26 month whereas in hAPP transgenic mice, it was decreased as early as 9-12 month. Neuritic plaques in adult hAPP mice clustered in hippocampus and adjacent cortical regions, but did not propagate further into the frontal cortex. Cerebral PACAP protein levels were reduced in hAPP mice compared to age-matched WT mice, but the genetic predisposition dominated cognitive decline. Taken together, these data suggest an association among PACAP levels, aging, cognitive function and amyloid load in nonhuman primates, with both similarities and differences from human AD brains. Our results suggest caution in choosing animal models and in extrapolating data to human AD studies.
Collapse
Affiliation(s)
- Pengcheng Han
- Department of Neurology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical CenterPhoenix, AZ, United States
- Department of Pathology and Laboratory Medicine Resident Program, Medical University of South CarolinaCharleston, SC, United States
| | - Megan Nielsen
- Department of Neurology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical CenterPhoenix, AZ, United States
| | - Melissa Song
- Department of Neurology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical CenterPhoenix, AZ, United States
| | - Junxiang Yin
- Department of Neurology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical CenterPhoenix, AZ, United States
| | - Michele R. Permenter
- California National Primate Research Center, University of California, DavisDavis, CA, United States
| | - Julie A. Vogt
- California National Primate Research Center, University of California, DavisDavis, CA, United States
| | - James R. Engle
- California National Primate Research Center, University of California, DavisDavis, CA, United States
- Evely F. McKnight Brain Institute, University of ArizonaTucson, AZ, United States
| | - Brittany N. Dugger
- Institute for Neurodegenerative Diseases, University of California, San FranciscoSan Francisco, CA, United States
- Civin Laboratory for Neuropathology, Banner Sun Health Research InstituteSun City, AZ, United States
| | - Thomas G. Beach
- Civin Laboratory for Neuropathology, Banner Sun Health Research InstituteSun City, AZ, United States
| | - Carol A. Barnes
- California National Primate Research Center, University of California, DavisDavis, CA, United States
- Evely F. McKnight Brain Institute, University of ArizonaTucson, AZ, United States
- Division of Neural Systems, Memory and Aging, University of ArizonaTucson, AZ, United States
- Departments of Psychology, Neurology, and Neuroscience, University of ArizonaTucson, AZ, United States
| | - Jiong Shi
- Department of Neurology, Barrow Neurological Institute, St. Joseph’s Hospital and Medical CenterPhoenix, AZ, United States
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General HospitalTianjin, China
| |
Collapse
|
12
|
Marzagalli R, Leggio GM, Bucolo C, Pricoco E, Keay KA, Cardile V, Castorina S, Salomone S, Drago F, Castorina A. Genetic blockade of the dopamine D3 receptor enhances hippocampal expression of PACAP and receptors and alters their cortical distribution. Neuroscience 2016; 316:279-295. [PMID: 26718601 DOI: 10.1016/j.neuroscience.2015.12.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/17/2015] [Accepted: 12/17/2015] [Indexed: 02/08/2023]
Abstract
Dopamine D3 receptors (D3Rs) are implicated in several aspects of cognition, but their role in aversive conditioning has only been marginally uncovered. Investigations have reported that blockade of D3Rs enhances the acquisition of fear memories, a phenomenon tightly linked to the neuropeptide pituitary adenylate cyclase-activating peptide (PACAP). However, the impact of D3R ablation on the PACAPergic system in regions critical for the formation of new memories remains unexplored. To address this issue, levels of PACAP and its receptors were compared in the hippocampus and cerebral cortex (CX) of mice devoid of functional D3Rs (D3R(-/-)) and wild-types (WTs) using a series of comparative immunohistochemical and biochemical analyses. Morphometric and stereological data revealed increased hippocampal area and volume in D3R(-/-) mice, and augmented neuronal density in CA1 and CA2/3 subfields. PACAP levels were increased in the hippocampus of D3R(-/-) mice. Expression of PACAP receptors was also heightened in mutant mice. In the CX, PACAP immunoreactivity (IR), was restricted to cortical layer V in WTs, but was distributed throughout layers IV-VI in D3R(-/-) mice, along with increased mRNAs, protein concentration and staining scores. Consistently, PAC1, VPAC1 and VPAC2 IRs were variably redistributed in CX, with a general upregulation in cortical layers II-IV in knockout animals. Our interpretation of these findings is that disturbed dopamine neurotransmission due to genetic D3R blockade may enhance the PACAP/PAC1-VPAC axis, a key endogenous system for the processing of fear memories. This could explain, at least in part, the facilitated acquisition and consolidation of aversive memories in D3R(-/-) mice.
Collapse
Affiliation(s)
- R Marzagalli
- Department of Biomedical and Biotechnological Sciences, Section of Human Anatomy and Histology, University of Catania, Catania 95123, Italy
| | - G M Leggio
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania 95123, Italy
| | - C Bucolo
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania 95123, Italy
| | - E Pricoco
- Department G.F. Ingrassia, Azienda Ospedaliero-Universitaria "Policlinico-Vittorio Emanuele", Section of Anatomic Pathology, University of Catania, Catania, Italy
| | - K A Keay
- Discipline of Anatomy and Histology, School of Medical Sciences, The University of Sydney, Sydney 2006, NSW, Australia
| | - V Cardile
- Department of Biomedical and Biotechnological Sciences, Section of Physiology, University of Catania, Catania 95123, Italy
| | - S Castorina
- Department of Biomedical and Biotechnological Sciences, Section of Human Anatomy and Histology, University of Catania, Catania 95123, Italy
| | - S Salomone
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania 95123, Italy
| | - F Drago
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania 95123, Italy
| | - A Castorina
- Department of Biomedical and Biotechnological Sciences, Section of Human Anatomy and Histology, University of Catania, Catania 95123, Italy; Discipline of Anatomy and Histology, School of Medical Sciences, The University of Sydney, Sydney 2006, NSW, Australia.
| |
Collapse
|
13
|
Lee JC, Choe SY. Region-specific changes in the distribution of transient receptor potential vanilloid 4 channel (TRPV4) in the central nervous system of Alzheimer’s disease model mice. Genes Genomics 2016. [DOI: 10.1007/s13258-016-0389-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
14
|
Yu R, Cui Z, Li M, Yang Y, Zhong J. Dimer-dependent intrinsic/basal activity of the class B G protein-coupled receptor PAC1 promotes cellular anti-apoptotic activity through Wnt/β-catenin pathways that are associated with dimer endocytosis. PLoS One 2014; 9:e113913. [PMID: 25426938 PMCID: PMC4245242 DOI: 10.1371/journal.pone.0113913] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Accepted: 11/02/2014] [Indexed: 12/23/2022] Open
Abstract
The high expression of PACAP (pituitary adenylate cyclase-activating polypeptide)-preferring receptor PAC1 is associated with nerve injury and tumors. Our previous report (Yu R, et al. PLoS One 2012; 7: e51811) confirmed the dimerization of PAC1 and found that the M-PAC1 mutation in the N-terminal first Cys/Ala lost the ability to form dimers. In this study, Chinese hamster ovary (CHO-K1) cells overexpressing wild-type PAC1 (PAC1-CHO) had significantly higher anti-apoptotic activities against serum withdrawal-induced apoptosis associated with a lower caspase 3 activity and a higher Bcl-2 level in a ligand-independent manner than those of CHO cells overexpressing the mutant M-PAC1 (M-PAC1-CHO). PAC1-CHO had significantly higher β-catenin, cyclin D1 and c-myc levels corresponding to the Wnt/β-catenin signal than did M-PAC1-CHO. In addition, the Wnt/β-catenin pathway inhibitor XAV939 significantly inhibited the anti-apoptotic activities of PAC1-CHO. Top-flash assays demonstrated that PAC1-CHO had a significantly stronger Wnt/β-catenin signal than did M-PAC1-CHO. Acetylcysteine (NAC) as an inhibitor of the dimerization of PAC1 inhibited the anti-apoptotic activities that were endowed by PAC1 and decreased the Wnt/β-catenin signal in Top-flash assays. In the PAC1 Tet (tetracycline)-on inducible gene expression system by doxycycline (Dox), higher expression levels of PAC1 resulted in higher anti-apoptotic activities that were associated with a stronger Wnt/β-catenin signal. A similar correlation was also found with the down-regulation of PAC1 in the Neuro2a neuroblastoma cell. BiFC combined with fluorescence confocal imaging indicated that during serum-withdrawal-induced apoptosis, PAC1 dimers displayed significant endocytosis. These findings indicate that PAC1 has ligand-independent and dimer-dependent intrinsic/basal activity, conferring cells with anti-apoptotic activities against serum withdrawal, which is involved in the Wnt/β-catenin signal and is associated with the endocytosis of PAC1 dimers. The discovery and study of the dimer-dependent basal activity of PAC1 not only help us understand the physiological and pathological role of PAC1 but also promote the development of drugs targeting PAC1.
Collapse
Affiliation(s)
- Rongjie Yu
- Institute of Biomedicine, Department of Cell Biology, Jinan University, Guangzhou, China
- * E-mail:
| | - Zekai Cui
- Institute of Biomedicine, Department of Cell Biology, Jinan University, Guangzhou, China
| | - Mei Li
- Institute of Biomedicine, Department of Cell Biology, Jinan University, Guangzhou, China
| | - Yanxu Yang
- Institute of Biomedicine, Department of Cell Biology, Jinan University, Guangzhou, China
| | - Jiaping Zhong
- Institute of Biomedicine, Department of Cell Biology, Jinan University, Guangzhou, China
| |
Collapse
|
15
|
Yu R, Yang Y, Cui Z, Zheng L, Zeng Z, Zhang H. Novel peptide VIP-TAT with higher affinity for PAC1 inhibited scopolamine induced amnesia. Peptides 2014; 60:41-50. [PMID: 25086267 DOI: 10.1016/j.peptides.2014.07.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Revised: 07/19/2014] [Accepted: 07/21/2014] [Indexed: 12/25/2022]
Abstract
A novel peptide VIP-TAT with a cell penetrating peptide TAT at the C-terminus of VIP was constructed and prepared using intein mediated purification with an affinity chitin-binding tag (IMPACT) system to enhance the brain uptake efficiency for the medical application in central nervous system. It was found by labeling VIP-TAT and VIP with fluorescein isothiocyanate (FITC) that the extension with TAT increased the brain uptake efficiency of VIP-TAT significantly. Then short-term and long-term treatment with scopolamine (Scop) was used to evaluate the effect of VIP-TAT or VIP on Scop induced amnesia. Both short-term and long-term administration of VIP-TAT inhibited the latent time reduction in step-through test induced by Scop significantly, but long-term administration of VIP aggravated the Scop induced amnesia. Long-term i.p. injection of VIP-TAT was shown to have positive effect by inhibiting the oxidative damage, apoptosis and the cholinergic system activity reduction that induced by Scop, while VIP exerted negative effect in brain opposite to that in periphery system. The in vitro data showed that VIP-TAT had not only protective but also proliferative effect on Neuro2a cells which was inhibited by PAC1 antagonist PACAP(6-38). Competition binding assay and cAMP assay confirmed that VIP-TAT had higher affinity and activation for PAC1 than VIP. So it was concluded that the significantly stronger protective effect of VIP-TAT against Scop induced amnesia than VIP was due to (1) the enhanced brain uptake efficiency of VIP-TAT and (2) the increased affinity and activation of VIP-TAT for receptor PAC1.
Collapse
Affiliation(s)
- Rongjie Yu
- Cell Biology Institute, Department of Cell Biology, Jinan University, Guangzhou 510632, China.
| | - Yanxu Yang
- Cell Biology Institute, Department of Cell Biology, Jinan University, Guangzhou 510632, China
| | - Zekai Cui
- Cell Biology Institute, Department of Cell Biology, Jinan University, Guangzhou 510632, China
| | - Lijun Zheng
- Cell Biology Institute, Department of Cell Biology, Jinan University, Guangzhou 510632, China
| | - Zhixing Zeng
- Cell Biology Institute, Department of Cell Biology, Jinan University, Guangzhou 510632, China
| | - Huahua Zhang
- Laboratory of Medical Genetics of Guangdong Medical College, Dongguan, Guangdong 523808, China
| |
Collapse
|
16
|
Age-related changes in the distribution of transient receptor potential vanilloid 4 channel (TRPV4) in the central nervous system of rats. J Mol Histol 2014; 45:497-505. [PMID: 24917364 DOI: 10.1007/s10735-014-9578-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 05/26/2014] [Indexed: 10/25/2022]
|
17
|
Pirger Z, Naskar S, László Z, Kemenes G, Reglődi D, Kemenes I. Reversal of age-related learning deficiency by the vertebrate PACAP and IGF-1 in a novel invertebrate model of aging: the pond snail (Lymnaea stagnalis). J Gerontol A Biol Sci Med Sci 2014; 69:1331-8. [PMID: 24846768 PMCID: PMC4197904 DOI: 10.1093/gerona/glu068] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
With the increase of life span, nonpathological age-related memory decline is affecting an increasing number of people. However, there is evidence that age-associated memory impairment only suspends, rather than irreversibly extinguishes, the intrinsic capacity of the aging nervous system for plasticity (1). Here, using a molluscan model system, we show that the age-related decline in memory performance can be reversed by administration of the pituitary adenylate cyclase activating polypeptide (PACAP). Our earlier findings showed that a homolog of the vertebrate PACAP38 and its receptors exist in the pond snail (Lymnaea stagnalis) brain (2), and it is both necessary and instructive for memory formation after reward conditioning in young animals (3). Here we show that exogenous PACAP38 boosts memory formation in aged Lymnaea, where endogenous PACAP38 levels are low in the brain. Treatment with insulin-like growth factor-1, which in vertebrates was shown to transactivate PACAP type I (PAC1) receptors (4) also boosts memory formation in aged pond snails. Due to the evolutionarily conserved nature of these polypeptides and their established role in memory and synaptic plasticity, there is a very high probability that they could also act as “memory rejuvenating” agents in humans.
Collapse
Affiliation(s)
- Zsolt Pirger
- Balaton Limnological Institute, Centre for Ecological Research, Hungarian Academy of Sciences, Tihany, Hungary. Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton. Department of Anatomy MTA-PTE, "Momentum" PACAP Team, University of Pécs, Hungary
| | - Souvik Naskar
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton
| | - Zita László
- Balaton Limnological Institute, Centre for Ecological Research, Hungarian Academy of Sciences, Tihany, Hungary. Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton
| | - György Kemenes
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton
| | - Dóra Reglődi
- Department of Anatomy MTA-PTE, "Momentum" PACAP Team, University of Pécs, Hungary
| | - Ildikó Kemenes
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton.
| |
Collapse
|
18
|
Yu R, Guo X, Zhong J, Li M, Zeng Z, Zhang H. The N-terminal HSDCIF motif is required for cell surface trafficking and dimerization of family B G protein coupled receptor PAC1. PLoS One 2012; 7:e51811. [PMID: 23284775 PMCID: PMC3528735 DOI: 10.1371/journal.pone.0051811] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 11/06/2012] [Indexed: 12/19/2022] Open
Abstract
PAC1 is PACAP (pituitary adenylate cyclase-activating polypeptide) preferring receptor belonging to class B G protein coupled receptor (GPCR) mediating the most effects of PACAP. The important role of G protein coupled receptor homo/heteromerization in receptor folding, maturation, trafficking, and cell surface expression has become increasingly evident. The bimolecular fluorescence complementation (BiFC) and bioluminescence resonance energy transfer (BRET) assay were used in this research to confirm the dimerization of PAC1 for the first time. The structure-activity relationship focused on the N-terminal HSDCIF motif, which locates behind the signal sequence and has high homology with PACAP (1–6), was assayed using a receptor mutant with the deletion of the HSDCIF motif. The fluorescence confocal microscope observation showed that the deletion of the HSDCIF motif impaired the cell delivery of PAC1. The results of BiFC, BRET and westernblot indicated that the deletion of HSDCIF motif and the replacement of the Cys residue with Ala in HSDCIF motif resulted in the disruption of receptor dimerization. And the exogenous chemically synthesized oligopeptide HSDCIF (100 nmol/L) not only down-regulated the dimerization of PAC1, induced the internalization of PAC1, but also inhibited the proliferation of CHO cells expressing PAC1 stably and decreased the activity of PACAP on the cell viability. All these data suggested that the N-terminal HSDCIF motif played key role in the trafficking and the dimerization of PAC1, and the exogenous oligopeptide HSDCIF had effects on the cell signaling, trafficking and the dimerization of PAC1.
Collapse
Affiliation(s)
- Rongjie Yu
- Cell Biology Institute, the Department of Cell Biology, Jinan University, Guangzhou, China.
| | | | | | | | | | | |
Collapse
|
19
|
Ding Y, Cheng H, Yu R, Tang C, Liu X, Chen J. Effects of cyclopeptide C*HSDGIC* from the cyclization of PACAP (1-5) on the proliferation and UVB-induced apoptosis of the retinal ganglion cell line RGC-5. Peptides 2012; 36:280-5. [PMID: 22706041 DOI: 10.1016/j.peptides.2012.06.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) is a neuropeptide that confers potent neurotrophic and neuroprotective effects. Cyclopeptide C*HSDGIC* (CHC), which results from the cyclization of PACAP (1-5) with disulfide, has been demonstrated to represent a potent agonist for the PACAP-specific receptor PAC1 which mediates the majority of PACAP's effects. In this study, the expression of PAC1 in a rat retinal ganglion cell line (RGC-5) was confirmed using a western blot analysis, and it was determined that CHC promoted the proliferation of RGC-5 cells using the cell counting kit-8 (CCK8) assay and flow cytometry. Furthermore, the treatment of CHC attenuated the decrease of cell viability in cells exposed to UVB irradiation. Flow cytometry and a JC-1 assay revealed that the CHC treatment protected the RGC-5 cells against UVB-induced apoptosis. In addition, similar to PACAP, the anti-apoptotic effect of CHC was related to the down-regulation of caspase-3. In summary, these results demonstrate for the first time that PAC1 is present in RGC-5 cells and that CHC, a cyclopeptide from PACAP, promotes RGC-5 cell proliferation and attenuates UVB-induced apoptosis.
Collapse
Affiliation(s)
- Yong Ding
- Department of Ophthalmology, Affiliated First Hospital of Jinan University, Guangzhou, China
| | | | | | | | | | | |
Collapse
|
20
|
Lee JC, Seong J, Kim SH, Lee SJ, Cho YJ, An J, Nam DH, Joo KM, Cha CI. Replacement of microglial cells using Clodronate liposome and bone marrow transplantation in the central nervous system of SOD1G93A transgenic mice as an in vivo model of amyotrophic lateral sclerosis. Biochem Biophys Res Commun 2012; 418:359-65. [DOI: 10.1016/j.bbrc.2012.01.026] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Accepted: 01/06/2012] [Indexed: 02/06/2023]
|
21
|
Lee JC, Shin JH, Park BW, Kim GS, Kim JC, Kang KS, Cha CI. Region-specific changes in the immunoreactivity of SIRT1 expression in the central nervous system of SOD1G93A transgenic mice as an in vivo model of amyotrophic lateral sclerosis. Brain Res 2012; 1433:20-8. [DOI: 10.1016/j.brainres.2011.11.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2011] [Revised: 11/04/2011] [Accepted: 11/07/2011] [Indexed: 12/28/2022]
|
22
|
Cho YJ, Lee JC, Kang BG, An J, Song HS, Son O, Nam DH, Cha CI, Joo KM. Immunohistochemical study on the expression of calcium binding proteins (calbindin-D28k, calretinin, and parvalbumin) in the cerebral cortex and in the hippocampal region of nNOS knock-out(-/-) mice. Anat Cell Biol 2011; 44:106-15. [PMID: 21829754 PMCID: PMC3145839 DOI: 10.5115/acb.2011.44.2.106] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Revised: 05/06/2011] [Accepted: 05/24/2011] [Indexed: 01/31/2023] Open
Abstract
Nitric oxide (NO) modulates the activities of various channels and receptors to participate in the regulation of neuronal intracellular Ca(2+) levels. Ca(2+) binding protein (CaBP) expression may also be altered by NO. Accordingly, we examined expression changes in calbindin-D28k, calretinin, and parvalbumin in the cerebral cortex and hippocampal region of neuronal NO synthase knockout(-/-) (nNOS(-/-)) mice using immunohistochemistry. For the first time, we demonstrate that the expression of CaBPs is specifically altered in the cerebral cortex and hippocampal region of nNOS(-/-) mice and that their expression changed according to neuronal type. As changes in CaBP expression can influence temporal and spatial intracellular Ca(2+) levels, it appears that NO may be involved in various functions, such as modulating neuronal Ca(2+) homeostasis, regulating synaptic transmission, and neuroprotection, by influencing the expression of CaBPs. Therefore, these results suggest another mechanism by which NO participates in the regulation of neuronal Ca(2+) homeostasis. However, the exact mechanisms of this regulation and its functional significance require further investigation.
Collapse
Affiliation(s)
- Yu Jin Cho
- Department of Anatomy, College of Medicine, Seoul National University, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|