1
|
Singhrao N, Flores-Tamez VA, Moustafa YA, Reddy GR, Burns AE, Pinkerton KE, Chen CY, Navedo MF, Nieves-Cintrón M. Nicotine Impairs Smooth Muscle cAMP Signaling and Vascular Reactivity. Microcirculation 2024; 31:e12871. [PMID: 38805589 PMCID: PMC11303104 DOI: 10.1111/micc.12871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/29/2024] [Accepted: 05/13/2024] [Indexed: 05/30/2024]
Abstract
OBJECTIVE This study aimed to determine nicotine's impact on receptor-mediated cyclic adenosine monophosphate (cAMP) synthesis in vascular smooth muscle (VSM). We hypothesize that nicotine impairs β adrenergic-mediated cAMP signaling in VSM, leading to altered vascular reactivity. METHODS The effects of nicotine on cAMP signaling and vascular function were systematically tested in aortic VSM cells and acutely isolated aortas from mice expressing the cAMP sensor TEpacVV (Camper), specifically in VSM (e.g., CamperSM). RESULTS Isoproterenol (ISO)-induced β-adrenergic production of cAMP in VSM was significantly reduced in cells from second-hand smoke (SHS)-exposed mice and cultured wild-type VSM treated with nicotine. The decrease in cAMP synthesis caused by nicotine was verified in freshly isolated arteries from a mouse that had cAMP sensor expression in VSM (e.g., CamperSM mouse). Functionally, the changes in cAMP signaling in response to nicotine hindered ISO-induced vasodilation, but this was reversed by immediate PDE3 inhibition. CONCLUSIONS These results imply that nicotine alters VSM β adrenergic-mediated cAMP signaling and vasodilation, which may contribute to the dysregulation of vascular reactivity and the development of vascular complications for nicotine-containing product users.
Collapse
Affiliation(s)
- Navid Singhrao
- Department of Pharmacology, University of California, Davis, USA
| | | | | | | | - Abby E. Burns
- Department of Pharmacology, University of California, Davis, USA
| | - Kent E. Pinkerton
- Center for Health and the Environment, University of California, Davis, California, USA
| | - Chao-Yin Chen
- Department of Pharmacology, University of California, Davis, USA
| | - Manuel F. Navedo
- Department of Pharmacology, University of California, Davis, USA
| | | |
Collapse
|
2
|
Marques-Almeida T, Lanceros-Mendez S, Ribeiro C. State of the Art and Current Challenges on Electroactive Biomaterials and Strategies for Neural Tissue Regeneration. Adv Healthc Mater 2024; 13:e2301494. [PMID: 37843074 DOI: 10.1002/adhm.202301494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 09/22/2023] [Indexed: 10/17/2023]
Abstract
The loss or failure of an organ/tissue stands as one of the healthcare system's most prevalent, devastating, and costly challenges. Strategies for neural tissue repair and regeneration have received significant attention due to their particularly strong impact on patients' well-being. Many research efforts are dedicated not only to control the disease symptoms but also to find solutions to repair the damaged tissues. Neural tissue engineering (TE) plays a key role in addressing this problem and significant efforts are being carried out to develop strategies for neural repair treatment. In the last years, active materials allowing to tune cell-materials interaction are being increasingly used, representing a recent paradigm in TE applications. Among the most important stimuli influencing cell behavior are the electrical and mechanical ones. In this way, materials with the ability to provide this kind of stimuli to the neural cells seem to be appropriate to support neural TE. In this scope, this review summarizes the different biomaterials types used for neural TE, highlighting the relevance of using active biomaterials and electrical stimulation. Furthermore, this review provides not only a compilation of the most relevant studies and results but also strategies for novel and more biomimetic approaches for neural TE.
Collapse
Affiliation(s)
- Teresa Marques-Almeida
- Physics Centre of Minho and Porto Universities (CF-UM-UP), University of Minho, Braga, 4710-057, Portugal
- LaPMET - Laboratory of Physics for Materials and Emergent Technologies, University of Minho, Braga, 4710-057, Portugal
| | - Senentxu Lanceros-Mendez
- Physics Centre of Minho and Porto Universities (CF-UM-UP), University of Minho, Braga, 4710-057, Portugal
- LaPMET - Laboratory of Physics for Materials and Emergent Technologies, University of Minho, Braga, 4710-057, Portugal
- BCMaterials, Basque Center for Materials, Applications and Nanostructures, UPV/EHU Science Park, Leioa, 48940, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, 48009, Spain
| | - Clarisse Ribeiro
- Physics Centre of Minho and Porto Universities (CF-UM-UP), University of Minho, Braga, 4710-057, Portugal
- LaPMET - Laboratory of Physics for Materials and Emergent Technologies, University of Minho, Braga, 4710-057, Portugal
| |
Collapse
|
3
|
Zhang Y, Su B, Tian Y, Yu Z, Wu X, Ding J, Wu C, Wei D, Yin H, Sun J, Fan H. Magnetic manipulation of Fe 3O 4@BaTiO 3 nanochains to regulate extracellular topographical and electrical cues. Acta Biomater 2023; 168:470-483. [PMID: 37495167 DOI: 10.1016/j.actbio.2023.07.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 07/14/2023] [Accepted: 07/19/2023] [Indexed: 07/28/2023]
Abstract
Magnetic fields play an essential role in material science and biomedical engineering. Magnetic-responsive materials can be arranged orderly in matrix to realize the construction of an aligned scaffold under magnetic induction. However, a single topological cue is insufficient to activate neural tissue regeneration, demanding more cues to promote regeneration synergistically, such as electrical stimulation and a biomimetic matrix. Herein, we propose one-dimensional (1D) magnetoelectric Fe3O4@BaTiO3 nanochains with controllable lengths under the regulation of a magnetic field. These nanochains can be oriented in the biomimetic hydrogel under magnetic guidance and induce the hydrogel microfiber to align along the direction of the nanochains, which is beneficial for cell-oriented outgrowth. This aligned hydrogel enabled wireless electrical stimulation mediated by magnetoelectric nanochains under magnetic stimulation, thereby activating the voltage-gated ion channel. Consequently, topological and electrical cues in this multifunctional biomimetic hydrogel synergistically enhanced the expression of neural functional proteins, facilitating synapse remodeling and neural regeneration. Predictably, the construction of multifunctional hydrogels based on low-cost and facile synthesis of magnetoelectric nanochains is an emerging patient-friendly and effective therapeutic strategy for neural or other tissue regeneration. STATEMENT OF SIGNIFICANCE: A facile and controllable magnetic strategy is established to manipulate 1D nanomaterial growth, matrix topography, and wireless electrical stimulation of cells. First, the magnetic-assisted interface co-assembly was used to control the length of Fe3O4@BaTiO3 nanochains with enhanced magnetoelectric effect. Then, the motion of the magnetic-induced nanochains guided the orientation of nanofibers in a 3D biomimetic hydrogel matrix. Finally, wireless electrical signals and topological cues in the biomimetic matrix synergistically promoted orderly aligned cell outgrowth and membrane depolarization by Ca2+ influx, thus enhancing nerve cell synaptic plasticity and functional expression. Consequently, this work provides a conceptual strategy from material design to extracellular matrix signal manipulation and synergistic induction of tissue regeneration.
Collapse
Affiliation(s)
- Yusheng Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Borui Su
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Yuan Tian
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Zhuoting Yu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Xiaoyang Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Jie Ding
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Chengheng Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China; Institute of Regulatory Science for Medical Devices, Sichuan University, Chengdu 610064, Sichuan, China
| | - Dan Wei
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
| | - Huabin Yin
- James Watt School of Engineering, University of Glasgow, Glasgow G12 8LT, UK
| | - Jin Sun
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Hongsong Fan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
| |
Collapse
|
4
|
Ma T, Ding Q, Liu C, Wu H. Electromagnetic fields regulate calcium-mediated cell fate of stem cells: osteogenesis, chondrogenesis and apoptosis. Stem Cell Res Ther 2023; 14:133. [PMID: 37194107 DOI: 10.1186/s13287-023-03303-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/28/2023] [Indexed: 05/18/2023] Open
Abstract
Electromagnetic fields (EMF) are increasing in popularity as a safe and non-invasive therapy. On the one hand, it is widely acknowledged that EMF can regulate the proliferation and differentiation of stem cells, promoting the undifferentiated cells capable of osteogenesis, angiogenesis, and chondroblast differentiation to achieve bone repair purpose. On the other hand, EMF can inhibit tumor stem cells proliferation and promote apoptosis to suppress tumor growth. As an essential second messenger, intracellular calcium plays a role in regulating cell cycle, such as proliferation, differentiation and apoptosis. There is increasing evidence that the modulation of intracellular calcium ion by EMF leads to differential outcomes in different stem cells. This review summarizes the regulation of channels, transporters, and ion pumps by EMF-induced calcium oscillations. It furtherly discusses the role of molecules and pathways activated by EMF-dependent calcium oscillations in promoting bone and cartilage repair and inhibiting tumor stem cells growth.
Collapse
Affiliation(s)
- Tian Ma
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Qing Ding
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Chaoxu Liu
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| | - Hua Wu
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
5
|
Xia G, Shi H, Su Y, Han B, Shen C, Gao S, Chen Z, Xu C. Photoactivated adenylyl cyclases attenuate sepsis-induced cardiomyopathy by suppressing macrophage-mediated inflammation. Front Immunol 2022; 13:1008702. [PMID: 36330522 PMCID: PMC9624221 DOI: 10.3389/fimmu.2022.1008702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/30/2022] [Indexed: 11/18/2022] Open
Abstract
Sepsis-induced myocardiopathy, characterized by innate immune cells infiltration and proinflammatory cytokines release, may lead to perfusion failure or even life-threatening cardiogenic shock. Macrophages-mediated inflammation has been shown to contribute to sepsis-induced myocardiopathy. In the current study, we introduced two photoactivated adenylyl cyclases (PACs), Beggiatoa sp. PAC (bPAC) and Beggiatoa sp. IS2 PAC (biPAC) into macrophages by transfection to detect the effects of light-induced regulation of macrophage pro-inflammatory response and LPS-induced sepsis-induced myocardiopathy. By this method, we uncovered that blue light-induced bPAC or biPAC activation considerably inhibited the production of pro-inflammatory cytokines IL-1 and TNF-α, both at mRNA and protein levels. Further, we assembled a GelMA-Macrophages-LED system, which consists of GelMA—a type of light crosslink hydrogel, gene modulated macrophages and wireless LED device, to allow light to regulate cardiac inflammation in situ with murine models of LPS-induced sepsis. Our results showed significant inhibition of leukocytes infiltration, especially macrophages and neutrophils, suppression of pro-inflammatory cytokines release, and alleviation of sepsis-induced cardiac dysfunction. Thus, our study may represent an emerging means to treat sepsis-induced myocardiopathy and other cardiovascular diseases by photo-activated regulating macrophage function.
Collapse
Affiliation(s)
- Guofang Xia
- Department of Cardiology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongyu Shi
- Wusong Central Hospital, Shanghai, China
| | - Yuanyuan Su
- Department of Cardiology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Beibei Han
- Department of Cardiology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengxing Shen
- Department of Cardiology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shiqiang Gao
- Institute of Physiology, Department of Neurophysiology, Julius-Maximilians-University of Wuerzburg, Wuerzburg, Germany
| | - Zhong Chen
- Department of Cardiology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Congfeng Xu, ; Zhong Chen,
| | - Congfeng Xu
- Department of Cardiology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Congfeng Xu, ; Zhong Chen,
| |
Collapse
|
6
|
Stojilkovic SS, Previde RM, Sherman AS, Fletcher PA. Pituitary corticotroph identity and receptor-mediated signaling: A transcriptomics perspective. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2022; 25. [PMID: 36177190 PMCID: PMC9514143 DOI: 10.1016/j.coemr.2022.100364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent single-cell RNA sequencing has offered an unprecedented view of pituitary cell transcriptomic profiles. In this review, these new data are briefly discussed and compared with the classical literature, focusing on pituitary corticotrophs. These cells are introduced by discussing their marker genes, followed by a review of G protein-coupled receptor gene expression, heterotrimeric G protein genes, and genes encoding signaling pathways downstream of G proteins: adenylate cyclases, phosphodiesterases, phospholipases, and protein kinases. The expression patterns of enzyme-linked plasma membrane and nuclear hormone receptor genes was also analyzed. The overview of these selected groups of genes sheds new light on corticotrophic receptors and their signaling pathways and provides guidance for further basic and clinical research by identifying genes that not been studied so far.
Collapse
Affiliation(s)
- Stanko S. Stojilkovic
- Section on Cellular Signaling, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
- Correspondence: Stanko S. Stojilkovic ()
| | - Rafael M. Previde
- Section on Cellular Signaling, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Arthur S. Sherman
- Laboratory of Biological Modeling, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Patrick A. Fletcher
- Laboratory of Biological Modeling, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| |
Collapse
|
7
|
Preferential Expression of Ca2+-Stimulable Adenylyl Cyclase III in the Supraventricular Area, Including Arrhythmogenic Pulmonary Vein of the Rat Heart. Biomolecules 2022; 12:biom12050724. [PMID: 35625651 PMCID: PMC9138642 DOI: 10.3390/biom12050724] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/16/2022] [Accepted: 05/19/2022] [Indexed: 12/10/2022] Open
Abstract
Ectopic excitability in pulmonary veins (PVs) is the major cause of atrial fibrillation. We previously reported that the inositol trisphosphate receptor in rat PV cardiomyocytes cooperates with the Na+-Ca2+ exchanger to provoke ectopic automaticity in response to norepinephrine. Here, we focused on adenylyl cyclase (AC) as another effector of norepinephrine stimulation. RT-PCR, immunohistochemistry, and Western blotting revealed that the abundant expression of Ca2+-stimulable AC3 was restricted to the supraventricular area, including the PVs. All the other AC isotypes hardly displayed any region-specific expressions. Immunostaining of isolated cardiomyocytes showed an enriched expression of AC3 along the t-tubules in PV myocytes. The cAMP-dependent response of L-type Ca2+ currents in the PV and LA cells is strengthened by the 0.1 mM intracellular Ca2+ condition, unlike in the ventricular cells. The norepinephrine-induced automaticity of PV cardiomyocytes was reversibly suppressed by 100 µM SQ22536, an adenine-like AC inhibitor. These findings suggest that the specific expression of AC3 along t-tubules may contribute to arrhythmogenic automaticity in rat PV cardiomyocytes.
Collapse
|
8
|
Aydın B, Gören MZ, Kanlı Z, Cabadak H. Cross-Talk of Cholinergic and β-Adrenergic Receptor Signalling in Chronic Myeloid Leukemia K562 Cells. Clin Exp Pharmacol Physiol 2022; 49:515-524. [PMID: 35088452 DOI: 10.1111/1440-1681.13627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/26/2021] [Accepted: 12/02/2021] [Indexed: 11/29/2022]
Abstract
In many studies on breast, skin, and intestinal cancers, beta-adrenergic receptor antagonists have been shown to inhibit cell proliferation and angiogenesis and increase apoptosis in cancers. Carbachol inhibits chronic myeloid leukemia K562 cell proliferation. Beta-blockers are known to inhibit cell progression. The aim of this study, explain the mechanism of action of beta-adrenergic receptors agonists and antagonists on apoptosis in chronic myeloid leukemia cells. We tried to determine the effect of combined treatment of beta-adrenergic and cholinergic drugs on Adrenergic β1 and β2 gene expression, cell proliferation and apoptosis in chronic myeloid leukemia K562 cells. Cell proliferation was evaluated by the BrdU incorporation kit. Caspase 3, 8, 9 activities were measured by the caspase-assay kit. Protein expression level detected by western blotting. We found that exposure to propranolol either by combination with carbachol facilitates additive effects on inhibition of caspase 3 and 8 expression in chronic myeloid leukemia K562 cells. But caspase 9 expression level was increased by propranolol alone or with propranolol and Carbachol combination. The combined therapy of cholinergic and adrenergic receptor drugs will decrease cell proliferation in K562 cells. This decrease in cell proliferation may be mediated by the mitochondrial dependent intrinsic apoptosis pathway.
Collapse
Affiliation(s)
- Banu Aydın
- Department of Biophysics, Marmara University, School of Medicine, Basic Medical Sciences Building, Maltepe, Istanbul, 34854, Turkey
| | - Mehmet Zafer Gören
- Department of Medical Pharmacology, Marmara University, School of Medicine, Basic Medical Sciences Building, Maltepe, Istanbul, 34854, Turkey
| | - Zehra Kanlı
- Department of Biophysics, Marmara University, School of Medicine, Basic Medical Sciences Building, Maltepe, Istanbul, 34854, Turkey
| | - Hülya Cabadak
- Department of Biophysics, Marmara University, School of Medicine, Basic Medical Sciences Building, Maltepe, Istanbul, 34854, Turkey
| |
Collapse
|
9
|
Bello SF, Xu H, Guo L, Li K, Zheng M, Xu Y, Zhang S, Bekele EJ, Bahareldin AA, Zhu W, Zhang D, Zhang X, Ji C, Nie Q. Hypothalamic and ovarian transcriptome profiling reveals potential candidate genes in low and high egg production of white Muscovy ducks (Cairina moschata). Poult Sci 2021; 100:101310. [PMID: 34298381 PMCID: PMC8322464 DOI: 10.1016/j.psj.2021.101310] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 05/24/2021] [Accepted: 06/01/2021] [Indexed: 01/16/2023] Open
Abstract
In China, the low egg production rate is a major challenge to Muscovy duck farmers. Hypothalamus and ovary play essential role in egg production of birds. However, there are little or no reports from these tissues to identify potential candidate genes responsible for egg production in White Muscovy ducks. A total of 1,537 laying ducks were raised; the egg production traits which include age at first egg (days), number of eggs at 300 d, and number of eggs at 59 wk were recorded. Moreover, 4 lowest (LP) and 4 highest producing (HP) were selected at 59 wk of age, respectively. To understand the mechanism of egg laying regulation, we sequenced the hypothalamus and ovary transcriptome profiles in LP and HP using RNA-Seq. The results showed that the number of eggs at 300 d and number of eggs at 59 wk in the HP were significantly more (P < 0.001) than the LP ducks. In total, 106.98G clean bases were generated from 16 libraries with an average of 6.68G clean bases for each library. Further analysis showed 569 and 2,259 differentially expressed genes (DEGs) were identified in the hypothalamus and ovary between LP and HP, respectively. The KEGG pathway enrichment analysis revealed 114 and 139 pathways in the hypothalamus and ovary, respectively which includes Calcium signaling pathway, ECM-receptor interaction, Focal adhesion, MAPK signaling pathway, Apoptosis and Apelin signaling pathways that are involved in egg production. Based on the GO terms and KEGG pathways results, 10 potential candidate genes (P2RX1, LPAR2, ADORA1, FN1, AKT3, ADCY5, ADCY8, MAP3K8, PXN, and PTTG1) were identified to be responsible for egg production. Further, protein-protein interaction was analyzed to show the relationship between these candidate genes. Therefore, this study provides useful information on transcriptome of hypothalamus and ovary of LP and HP Muscovy ducks.
Collapse
Affiliation(s)
- Semiu Folaniyi Bello
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, Guangdong, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, Guangdong, China
| | - Haiping Xu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, Guangdong, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, Guangdong, China
| | - Lijin Guo
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, Guangdong, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, Guangdong, China
| | - Kan Li
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, Guangdong, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, Guangdong, China
| | - Ming Zheng
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, Guangdong, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, Guangdong, China
| | - Yibin Xu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, Guangdong, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, Guangdong, China
| | - Siyu Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, Guangdong, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, Guangdong, China
| | - Endashaw Jebessa Bekele
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, Guangdong, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, Guangdong, China
| | - Ali Abdalla Bahareldin
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, Guangdong, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, Guangdong, China
| | - Weijian Zhu
- Wens Foodstuff Group Co. Ltd., Yunfu, 527400 Guangdong, China
| | - Dexiang Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, Guangdong, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, Guangdong, China; Wens Foodstuff Group Co. Ltd., Yunfu, 527400 Guangdong, China
| | - Xiquan Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, Guangdong, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, Guangdong, China
| | - Congliang Ji
- Wens Foodstuff Group Co. Ltd., Yunfu, 527400 Guangdong, China
| | - Qinghua Nie
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, South China Agricultural University, Guangzhou 510642, Guangdong, China; Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding and Key Lab of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou 510642, Guangdong, China; Wens Foodstuff Group Co. Ltd., Yunfu, 527400 Guangdong, China.
| |
Collapse
|
10
|
Przykaza Ł, Kozniewska E. Ligands of the Neuropeptide Y Y2 Receptors as a Potential Multitarget Therapeutic Approach for the Protection of the Neurovascular Unit Against Acute Ischemia/Reperfusion: View from the Perspective of the Laboratory Bench. Transl Stroke Res 2021; 13:12-24. [PMID: 34292517 PMCID: PMC8766383 DOI: 10.1007/s12975-021-00930-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 07/11/2021] [Accepted: 07/12/2021] [Indexed: 01/02/2023]
Abstract
Ischemic stroke is the third leading cause of death and disability worldwide, with no available satisfactory prevention or treatment approach. The current treatment is limited to the use of “reperfusion methods,” i.e., an intravenous or intra-arterial infusion of a fibrinolytic agent, mechanical removal of the clot by thrombectomy, or a combination of both methods. It should be stressed, however, that only approximately 5% of all acute strokes are eligible for fibrinolytic treatment and fewer than 10% for thrombectomy. Despite the tremendous progress in understanding of the pathomechanisms of cerebral ischemia, the promising results of basic research on neuroprotection are not currently transferable to human stroke. A possible explanation for this failure is that experiments on in vivo animal models involve healthy young animals, and the experimental protocols seldom consider the importance of protecting the whole neurovascular unit (NVU), which ensures intracranial homeostasis and is seriously damaged by ischemia/reperfusion. One of the endogenous protective systems activated during ischemia and in neurodegenerative diseases is represented by neuropeptide Y (NPY). It has been demonstrated that activation of NPY Y2 receptors (Y2R) by a specific ligand decreases the volume of the postischemic infarction and improves performance in functional tests of rats with arterial hypertension subjected to middle cerebral artery occlusion/reperfusion. This functional improvement suggests the protection of the NVU. In this review, we focus on NPY and discuss the potential, multidirectional protective effects of Y2R agonists against acute focal ischemia/reperfusion injury, with special reference to the NVU.
Collapse
Affiliation(s)
- Łukasz Przykaza
- Laboratory of Experimental and Clinical Neurosurgery, Mossakowski Medical Research Institute Polish Academy of Sciences, A. Pawińskiego Str. 5, 02-106, Warsaw, Poland
| | - Ewa Kozniewska
- Laboratory of Experimental and Clinical Neurosurgery, Mossakowski Medical Research Institute Polish Academy of Sciences, A. Pawińskiego Str. 5, 02-106, Warsaw, Poland.
| |
Collapse
|
11
|
Cross-Talk Between the Adenylyl Cyclase/cAMP Pathway and Ca 2+ Homeostasis. Rev Physiol Biochem Pharmacol 2021; 179:73-116. [PMID: 33398503 DOI: 10.1007/112_2020_55] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cyclic AMP and Ca2+ are the first second or intracellular messengers identified, unveiling the cellular mechanisms activated by a plethora of extracellular signals, including hormones. Cyclic AMP generation is catalyzed by adenylyl cyclases (ACs), which convert ATP into cAMP and pyrophosphate. By the way, Ca2+, as energy, can neither be created nor be destroyed; Ca2+ can only be transported, from one compartment to another, or chelated by a variety of Ca2+-binding molecules. The fine regulation of cytosolic concentrations of cAMP and free Ca2+ is crucial in cell function and there is an intimate cross-talk between both messengers to fine-tune the cellular responses. Cancer is a multifactorial disease resulting from a combination of genetic and environmental factors. Frequent cases of cAMP and/or Ca2+ homeostasis remodeling have been described in cancer cells. In those tumoral cells, cAMP and Ca2+ signaling plays a crucial role in the development of hallmarks of cancer, including enhanced proliferation and migration, invasion, apoptosis resistance, or angiogenesis. This review summarizes the cross-talk between the ACs/cAMP and Ca2+ intracellular pathways with special attention to the functional and reciprocal regulation between Orai1 and AC8 in normal and cancer cells.
Collapse
|
12
|
Patt J, Alenfelder J, Pfeil EM, Voss JH, Merten N, Eryilmaz F, Heycke N, Rick U, Inoue A, Kehraus S, Deupi X, Müller CE, König GM, Crüsemann M, Kostenis E. An experimental strategy to probe Gq contribution to signal transduction in living cells. J Biol Chem 2021; 296:100472. [PMID: 33639168 PMCID: PMC8024710 DOI: 10.1016/j.jbc.2021.100472] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/14/2022] Open
Abstract
Heterotrimeric G protein subunits Gαq and Gα11 are inhibited by two cyclic depsipeptides, FR900359 (FR) and YM-254890 (YM), both of which are being used widely to implicate Gq/11 proteins in the regulation of diverse biological processes. An emerging major research question therefore is whether the cellular effects of both inhibitors are on-target, that is, mediated via specific inhibition of Gq/11 proteins, or off-target, that is, the result of nonspecific interactions with other proteins. Here we introduce a versatile experimental strategy to discriminate between these possibilities. We developed a Gαq variant with preserved catalytic activity, but refractory to FR/YM inhibition. A minimum of two amino acid changes were required and sufficient to achieve complete inhibitor resistance. We characterized the novel mutant in HEK293 cells depleted by CRISPR–Cas9 of endogenous Gαq and Gα11 to ensure precise control over the Gα-dependent cellular signaling route. Using a battery of cellular outcomes with known and concealed Gq contribution, we found that FR/YM specifically inhibited cellular signals after Gαq introduction via transient transfection. Conversely, both inhibitors were inert across all assays in cells expressing the drug-resistant variant. These findings eliminate the possibility that inhibition of non-Gq proteins contributes to the cellular effects of the two depsipeptides. We conclude that combined application of FR or YM along with the drug-resistant Gαq variant is a powerful in vitro strategy to discern on-target Gq against off-target non-Gq action. Consequently, it should be of high value for uncovering Gq input to complex biological processes with high accuracy and the requisite specificity.
Collapse
Affiliation(s)
- Julian Patt
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Judith Alenfelder
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Eva Marie Pfeil
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Jan Hendrik Voss
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Nicole Merten
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Funda Eryilmaz
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Nina Heycke
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Uli Rick
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Stefan Kehraus
- Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Xavier Deupi
- Laboratory of Biomolecular Research and Condensed Matter Theory Group, Paul Scherrer Institute, Villigen, Switzerland
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Gabriele M König
- Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Max Crüsemann
- Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Evi Kostenis
- Molecular, Cellular and Pharmacobiology Section, Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany.
| |
Collapse
|
13
|
Li C, Qian T, He R, Wan C, Liu Y, Yu H. Endoplasmic Reticulum-Plasma Membrane Contact Sites: Regulators, Mechanisms, and Physiological Functions. Front Cell Dev Biol 2021; 9:627700. [PMID: 33614657 PMCID: PMC7889955 DOI: 10.3389/fcell.2021.627700] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/13/2021] [Indexed: 12/13/2022] Open
Abstract
The endoplasmic reticulum (ER) forms direct membrane contact sites with the plasma membrane (PM) in eukaryotic cells. These ER-PM contact sites play essential roles in lipid homeostasis, ion dynamics, and cell signaling, which are carried out by protein-protein or protein-lipid interactions. Distinct tethering factors dynamically control the architecture of ER-PM junctions in response to intracellular signals or external stimuli. The physiological roles of ER-PM contact sites are dependent on a variety of regulators that individually or cooperatively perform functions in diverse cellular processes. This review focuses on proteins functioning at ER-PM contact sites and highlights the recent progress in their mechanisms and physiological roles.
Collapse
Affiliation(s)
- Chenlu Li
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Tiantian Qian
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Ruyue He
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Chun Wan
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, United States
| | - Yinghui Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Haijia Yu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
14
|
McCabe KJ, Rangamani P. Computational modeling approaches to cAMP/PKA signaling in cardiomyocytes. J Mol Cell Cardiol 2021; 154:32-40. [PMID: 33548239 DOI: 10.1016/j.yjmcc.2021.01.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/11/2021] [Accepted: 01/15/2021] [Indexed: 12/12/2022]
Abstract
The cAMP/PKA pathway is a fundamental regulator of excitation-contraction coupling in cardiomyocytes. Activation of cAMP has a variety of downstream effects on cardiac function including enhanced contraction, accelerated relaxation, adaptive stress response, mitochondrial regulation, and gene transcription. Experimental advances have shed light on the compartmentation of cAMP and PKA, which allow for control over the varied targets of these second messengers and is disrupted in heart failure conditions. Computational modeling is an important tool for understanding the spatial and temporal complexities of this system. In this review article, we outline the advances in computational modeling that have allowed for deeper understanding of cAMP/PKA dynamics in the cardiomyocyte in health and disease, and explore new modeling frameworks that may bring us closer to a more complete understanding of this system. We outline various compartmental and spatial signaling models that have been used to understand how β-adrenergic signaling pathways function in a variety of simulation conditions. We also discuss newer subcellular models of cardiovascular function that may be used as templates for the next phase of computational study of cAMP and PKA in the heart, and outline open challenges which are important to consider in future models.
Collapse
Affiliation(s)
- Kimberly J McCabe
- Simula Research Laboratory, Department of Computational Physiology, PO Box 134, 1325 Lysaker, Norway.
| | - Padmini Rangamani
- University of California San Diego, Department of Mechanical and Aerospace Engineering, 9500 Gilman Drive MC 0411, La Jolla, CA 92093, United States of America
| |
Collapse
|
15
|
Burton RAB, Terrar DA. Emerging Evidence for cAMP-calcium Cross Talk in Heart Atrial Nanodomains Where IP 3-Evoked Calcium Release Stimulates Adenylyl Cyclases. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2021; 4:25152564211008341. [PMID: 37366374 PMCID: PMC10243587 DOI: 10.1177/25152564211008341] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/01/2021] [Accepted: 03/07/2021] [Indexed: 06/28/2023]
Abstract
Calcium handling is vital to normal physiological function in the heart. Human atrial arrhythmias, eg. atrial fibrillation, are a major morbidity and mortality burden, yet major gaps remain in our understanding of how calcium signaling pathways function and interact. Inositol trisphosphate (IP3) is a calcium-mobilizing second messenger and its agonist-induced effects have been observed in many tissue types. In the atria IP3 receptors (IR3Rs) residing on junctional sarcoplasmic reticulum augment cellular calcium transients and, when over-stimulated, lead to arrhythmogenesis. Recent studies have demonstrated that the predominant pathway for IP3 actions in atrial myocytes depends on stimulation of calcium-dependent forms of adenylyl cyclase (AC8 and AC1) by IP3-evoked calcium release from the sarcoplasmic reticulum. AC8 shows co-localisation with IP3Rs and AC1 appears to be nearby. These observations support crosstalk between calcium and cAMP pathways in nanodomains in atria. Similar mechanisms also appear to operate in the pacemaker region of the sinoatrial node. Here we discuss these significant advances in our understanding of atrial physiology and pathology, together with implications for the identification of potential novel targets and modulators for the treatment of atrial arrhythmias.
Collapse
Affiliation(s)
| | - Derek A. Terrar
- Department of Pharmacology, University of Oxford, Oxford, UK
| |
Collapse
|
16
|
Linghu C, Johnson SL, Valdes PA, Shemesh OA, Park WM, Park D, Piatkevich KD, Wassie AT, Liu Y, An B, Barnes SA, Celiker OT, Yao CC, Yu CCJ, Wang R, Adamala KP, Bear MF, Keating AE, Boyden ES. Spatial Multiplexing of Fluorescent Reporters for Imaging Signaling Network Dynamics. Cell 2020; 183:1682-1698.e24. [PMID: 33232692 DOI: 10.1016/j.cell.2020.10.035] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/05/2020] [Accepted: 10/21/2020] [Indexed: 12/23/2022]
Abstract
In order to analyze how a signal transduction network converts cellular inputs into cellular outputs, ideally one would measure the dynamics of many signals within the network simultaneously. We found that, by fusing a fluorescent reporter to a pair of self-assembling peptides, it could be stably clustered within cells at random points, distant enough to be resolved by a microscope but close enough to spatially sample the relevant biology. Because such clusters, which we call signaling reporter islands (SiRIs), can be modularly designed, they permit a set of fluorescent reporters to be efficiently adapted for simultaneous measurement of multiple nodes of a signal transduction network within single cells. We created SiRIs for indicators of second messengers and kinases and used them, in hippocampal neurons in culture and intact brain slices, to discover relationships between the speed of calcium signaling, and the amplitude of PKA signaling, upon receiving a cAMP-driving stimulus.
Collapse
Affiliation(s)
- Changyang Linghu
- Department of Media Arts and Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; McGovern Institute for Brain Research, MIT, Cambridge, MA 02139, USA; Center for Neurobiological Engineering, MIT, Cambridge, MA 02139, USA; Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA 02139, USA
| | - Shannon L Johnson
- Department of Media Arts and Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; McGovern Institute for Brain Research, MIT, Cambridge, MA 02139, USA; Center for Neurobiological Engineering, MIT, Cambridge, MA 02139, USA
| | - Pablo A Valdes
- Department of Media Arts and Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; Department of Neurosurgery, Harvard Medical School, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Or A Shemesh
- Department of Media Arts and Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; McGovern Institute for Brain Research, MIT, Cambridge, MA 02139, USA; Center for Neurobiological Engineering, MIT, Cambridge, MA 02139, USA; Department of Biological Engineering, MIT, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA; Department of Neurobiology and Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Won Min Park
- Tim Taylor Department of Chemical Engineering, Kansas State University, Manhattan, KS 66506, USA
| | - Demian Park
- Department of Media Arts and Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; McGovern Institute for Brain Research, MIT, Cambridge, MA 02139, USA; Center for Neurobiological Engineering, MIT, Cambridge, MA 02139, USA
| | - Kiryl D Piatkevich
- Department of Media Arts and Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; McGovern Institute for Brain Research, MIT, Cambridge, MA 02139, USA; Center for Neurobiological Engineering, MIT, Cambridge, MA 02139, USA; Department of Biological Engineering, MIT, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA; School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Asmamaw T Wassie
- Department of Media Arts and Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; McGovern Institute for Brain Research, MIT, Cambridge, MA 02139, USA; Center for Neurobiological Engineering, MIT, Cambridge, MA 02139, USA; Department of Biological Engineering, MIT, Cambridge, MA 02139, USA
| | - Yixi Liu
- Department of Media Arts and Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; McGovern Institute for Brain Research, MIT, Cambridge, MA 02139, USA; Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA 02139, USA; Center for Neurobiological Engineering, MIT, Cambridge, MA 02139, USA
| | - Bobae An
- Department of Media Arts and Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; McGovern Institute for Brain Research, MIT, Cambridge, MA 02139, USA; Center for Neurobiological Engineering, MIT, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| | - Stephanie A Barnes
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA; Picower Institute for Learning and Memory, MIT, Cambridge, MA 02139, USA
| | - Orhan T Celiker
- Department of Media Arts and Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; McGovern Institute for Brain Research, MIT, Cambridge, MA 02139, USA; Center for Neurobiological Engineering, MIT, Cambridge, MA 02139, USA; Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA 02139, USA
| | - Chun-Chen Yao
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| | - Chih-Chieh Jay Yu
- Department of Media Arts and Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; McGovern Institute for Brain Research, MIT, Cambridge, MA 02139, USA; Center for Neurobiological Engineering, MIT, Cambridge, MA 02139, USA; Department of Biological Engineering, MIT, Cambridge, MA 02139, USA
| | - Ru Wang
- Department of Media Arts and Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; McGovern Institute for Brain Research, MIT, Cambridge, MA 02139, USA; Center for Neurobiological Engineering, MIT, Cambridge, MA 02139, USA
| | - Katarzyna P Adamala
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Mark F Bear
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA; Picower Institute for Learning and Memory, MIT, Cambridge, MA 02139, USA
| | - Amy E Keating
- Department of Biological Engineering, MIT, Cambridge, MA 02139, USA; Department of Biology, MIT, Cambridge, MA 02139, USA; Koch Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Edward S Boyden
- Department of Media Arts and Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA 02139, USA; McGovern Institute for Brain Research, MIT, Cambridge, MA 02139, USA; Center for Neurobiological Engineering, MIT, Cambridge, MA 02139, USA; Department of Biological Engineering, MIT, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA; Koch Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Cambridge, MA 02139, USA.
| |
Collapse
|
17
|
Tenner B, Getz M, Ross B, Ohadi D, Bohrer CH, Greenwald E, Mehta S, Xiao J, Rangamani P, Zhang J. Spatially compartmentalized phase regulation of a Ca 2+-cAMP-PKA oscillatory circuit. eLife 2020; 9:e55013. [PMID: 33201801 PMCID: PMC7671691 DOI: 10.7554/elife.55013] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 10/07/2020] [Indexed: 01/31/2023] Open
Abstract
Signaling networks are spatiotemporally organized to sense diverse inputs, process information, and carry out specific cellular tasks. In β cells, Ca2+, cyclic adenosine monophosphate (cAMP), and Protein Kinase A (PKA) exist in an oscillatory circuit characterized by a high degree of feedback. Here, we describe a mode of regulation within this circuit involving a spatial dependence of the relative phase between cAMP, PKA, and Ca2+. We show that in mouse MIN6 β cells, nanodomain clustering of Ca2+-sensitive adenylyl cyclases (ACs) drives oscillations of local cAMP levels to be precisely in-phase with Ca2+ oscillations, whereas Ca2+-sensitive phosphodiesterases maintain out-of-phase oscillations outside of the nanodomain. Disruption of this precise phase relationship perturbs Ca2+ oscillations, suggesting the relative phase within an oscillatory circuit can encode specific functional information. This work unveils a novel mechanism of cAMP compartmentation utilized for localized tuning of an oscillatory circuit and has broad implications for the spatiotemporal regulation of signaling networks.
Collapse
Affiliation(s)
- Brian Tenner
- Department of Biophysics and Biophysical Chemistry, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Pharmacology, University of California, San DiegoLa JollaUnited States
| | - Michael Getz
- Chemical Engineering Graduate Program, University of California, San DiegoLa JollaUnited States
| | - Brian Ross
- Department of Pharmacology, University of California, San DiegoLa JollaUnited States
| | - Donya Ohadi
- Department of Mechanical and Aerospace Engineering, University of California, San DiegoLa JollaUnited States
| | - Christopher H Bohrer
- Department of Biophysics and Biophysical Chemistry, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Eric Greenwald
- Department of Pharmacology, University of California, San DiegoLa JollaUnited States
| | - Sohum Mehta
- Department of Pharmacology, University of California, San DiegoLa JollaUnited States
| | - Jie Xiao
- Department of Biophysics and Biophysical Chemistry, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Padmini Rangamani
- Chemical Engineering Graduate Program, University of California, San DiegoLa JollaUnited States
- Department of Mechanical and Aerospace Engineering, University of California, San DiegoLa JollaUnited States
| | - Jin Zhang
- Department of Pharmacology, University of California, San DiegoLa JollaUnited States
- Department of Chemistry and Biochemistry, University of California, San DiegoLa JollaUnited States
| |
Collapse
|
18
|
Annamdevula NS, Sweat R, Gunn H, Griswold JR, Britain AL, Rich TC, Leavesley SJ. Measurement of 3-Dimensional cAMP Distributions in Living Cells using 4-Dimensional (x, y, z, and λ) Hyperspectral FRET Imaging and Analysis. J Vis Exp 2020. [PMID: 33191928 DOI: 10.3791/61720] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Cyclic AMP is a second messenger that is involved in a wide range of cellular and physiological activities. Several studies suggest that cAMP signals are compartmentalized, and that compartmentalization contributes to signaling specificity within the cAMP signaling pathway. The development of Fӧrster resonance energy transfer (FRET) based biosensors has furthered the ability to measure and visualize cAMP signals in cells. However, these measurements are often confined to two spatial dimensions, which may result in misinterpretation of data. To date, there have been only very limited measurements of cAMP signals in three spatial dimensions (x, y, and z), due to the technical limitations in using FRET sensors that inherently exhibit low signal to noise ratio (SNR). In addition, traditional filter-based imaging approaches are often ineffective for accurate measurement of cAMP signals in localized subcellular regions due to a range of factors, including spectral crosstalk, limited signal strength, and autofluorescence. To overcome these limitations and allow FRET-based biosensors to be used with multiple fluorophores, we have developed hyperspectral FRET imaging and analysis approaches that provide spectral specificity for calculating FRET efficiencies and the ability to spectrally separate FRET signals from confounding autofluorescence and/or signals from additional fluorescent labels. Here, we present the methodology for implementing hyperspectral FRET imaging as well as the need to construct an appropriate spectral library that is neither undersampled nor oversampled to perform spectral unmixing. While we present this methodology for measurement of three-dimensional cAMP distributions in pulmonary microvascular endothelial cells (PMVECs), this methodology could be used to study spatial distributions of cAMP in a range of cell types.
Collapse
Affiliation(s)
- Naga S Annamdevula
- Department of Pharmacology, University of South Alabama; Center for Lung Biology, University of South Alabama
| | - Rachel Sweat
- Department of Chemical and Biomolecular Engineering, University of South Alabama
| | - Hayden Gunn
- Department of Pharmacology, University of South Alabama
| | - John R Griswold
- Department of Chemical and Biomolecular Engineering, University of South Alabama
| | - Andrea L Britain
- Department of Pharmacology, University of South Alabama; Center for Lung Biology, University of South Alabama
| | - Thomas C Rich
- Department of Pharmacology, University of South Alabama; Center for Lung Biology, University of South Alabama
| | - Silas J Leavesley
- Department of Pharmacology, University of South Alabama; Center for Lung Biology, University of South Alabama; Department of Chemical and Biomolecular Engineering, University of South Alabama;
| |
Collapse
|
19
|
Agolini E, Cherchi C, Bellacchio E, Martinelli D, Cocciadiferro D, Cutrera R, Chiarini Testa MB, Barone C, Bianca S, Novelli A. Expanding the clinical and molecular spectrum of lethal congenital contracture syndrome 8 associated with biallelic variants of ADCY6. Clin Genet 2020; 97:649-654. [PMID: 31846058 DOI: 10.1111/cge.13691] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/06/2019] [Accepted: 12/11/2019] [Indexed: 11/26/2022]
Abstract
Arthrogryposis multiplex congenita (AMC) is defined as congenital, non-progressive contractures in more than two joints and in multiple body areas, resulting from reduced fetal mobility. So far, more than 400 causative genes for AMC have been identified. Some isolated AMC phenotypes arise as a result of mutations in genes encoding components required for motor neuron structure, function, and myelination, as in the case of ADCY6 encoding the enzyme adenylyl cyclase type 6. ADCY6 inactivation, due to biallelic variants, have been previously associated with the lethal congenital contracture syndrome 8 (LCCS8). So far, only four LCCS8 patients, from two families, have been reported. Here, we describe a new patient affected by a severe form of AMC, harboring two novel compound heterozygous variants in ADCY6. Our findings expand the clinical and mutational spectrum of LCCS8, showing a possible correlation between the impact of the ADCY6 missense variants reported to date, predicted by molecular modeling, and the severity of the phenotype.
Collapse
Affiliation(s)
- Emanuele Agolini
- Laboratory of Medical Genetics Unit, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Claudio Cherchi
- Respiratory Unit, Academic Department of Pediatrics, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Emanuele Bellacchio
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Diego Martinelli
- Division of Metabolism, Department of Pediatric Specialties, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Dario Cocciadiferro
- Laboratory of Medical Genetics Unit, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Renato Cutrera
- Respiratory Unit, Academic Department of Pediatrics, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Maria B Chiarini Testa
- Respiratory Unit, Academic Department of Pediatrics, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| | - Chiara Barone
- Medical Genetics, Referral Centre for Rare Genetic Diseases, ARNAS Garibaldi, Catania, Italy
| | - Sebastiano Bianca
- Medical Genetics, Referral Centre for Rare Genetic Diseases, ARNAS Garibaldi, Catania, Italy
| | - Antonio Novelli
- Laboratory of Medical Genetics Unit, Ospedale Pediatrico Bambino Gesù, Rome, Italy
| |
Collapse
|
20
|
cAMP and Photoreceptor Cell Death in Retinal Degeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1185:301-304. [PMID: 31884628 DOI: 10.1007/978-3-030-27378-1_49] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Inherited retinal degenerations (IRDs) are a genetically heterogeneous group of disorders characterized by the progressive loss of photoreceptor cells. Despite this heterogeneity in the disease-causing mutation, common underlying mechanisms promoting photoreceptor cell death may be present. Dysregulation of photoreceptor cyclic nucleotide signaling may be one such common feature differentiating healthy from diseased photoreceptors. Here we review evidence that elevated retinal cAMP levels promote photoreceptor death and are a common feature of numerous animal models of IRDs. Improving our understanding of how cAMP levels become elevated and identifying downstream effectors may prove important for the development of therapeutics that will be applicable to multiple forms of the disease.
Collapse
|
21
|
Nazri MUIA, Idris I, Ross O, Ismail WIW. Neurological Disorder Brain Model: A Lesson from Marine Worms (Annelida: Polychaeta). Malays J Med Sci 2019; 26:5-18. [PMID: 31908583 PMCID: PMC6939724 DOI: 10.21315/mjms2019.26.6.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 07/09/2019] [Indexed: 12/25/2022] Open
Abstract
The incidence of neurodegenerative diseases is directly proportional to age. The prevalence of non-communicable diseases, for example, Alzheimer's and Parkinson's, is expected to rise in the coming years. Understanding the etiopathology of these diseases is a crucial step that needs to be taken to develop drugs for their treatment. Animal models are being increasingly used to expand the knowledge and understanding on neurodegenerative diseases. Marine worms, known as polychaetes (phylum Annelida), which are abundantly and frequently found in benthic environments, possess a simple yet complete nervous system (including a true brain that is centralised and specialised) compared to other annelids. Hence, polychaetes can potentially be the next candidate for a nerve disease model. The ability to activate the entire nervous system regeneration (NSR) is among the remarkable features of many polychaetes species. However, the information on NSR in polychaetes and how it can potentially model neurodegenerative diseases in humans is still lacking. By exploring such studies, we may eventually be able to circumvent the developmental constraints that limit NSR in the human nervous system. This article is intended to briefly review responsible mechanisms and signalling pathways of NSR in marine polychaetes and to make a comparison with other established models of neurodegenerative disease.
Collapse
Affiliation(s)
| | - Izwandy Idris
- Institute of Oceanography and Environment, Universiti Malaysia Terengganu, Terengganu, Malaysia
| | - Othman Ross
- Institute of Oceanography and Environment, Universiti Malaysia Terengganu, Terengganu, Malaysia
| | - Wan Iryani Wan Ismail
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Terengganu, Malaysia
| |
Collapse
|
22
|
Ohadi D, Rangamani P. Geometric Control of Frequency Modulation of cAMP Oscillations due to Calcium in Dendritic Spines. Biophys J 2019; 117:1981-1994. [PMID: 31668747 PMCID: PMC7018999 DOI: 10.1016/j.bpj.2019.10.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 07/30/2019] [Accepted: 10/02/2019] [Indexed: 12/12/2022] Open
Abstract
The spatiotemporal regulation of cyclic adenosine monophosphate (cAMP) and its dynamic interactions with other second messengers such as calcium are critical features of signaling specificity required for neuronal development and connectivity. cAMP is known to contribute to long-term potentiation and memory formation by controlling the formation and regulation of dendritic spines. Despite the recent advances in biosensing techniques for monitoring spatiotemporal cAMP dynamics, the underlying molecular mechanisms that attribute to the subcellular modulation of cAMP remain unknown. In this work, we model the spatiotemporal dynamics of calcium-induced cAMP signaling pathway in dendritic spines. Using a three-dimensional reaction-diffusion model, we investigate the effect of different spatial characteristics of cAMP dynamics that may be responsible for subcellular regulation of cAMP concentrations. Our model predicts that the volume/surface ratio of the spine, regulated through the spine head size, spine neck size, and the presence of physical barriers (spine apparatus), is an important regulator of cAMP dynamics. Furthermore, localization of the enzymes responsible for the synthesis and degradation of cAMP in different compartments also modulates the oscillatory patterns of cAMP through exponential relationships. Our findings shed light on the significance of complex geometric and localization relationships for cAMP dynamics in dendritic spines.
Collapse
Affiliation(s)
- Donya Ohadi
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, California
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, California.
| |
Collapse
|
23
|
Menthol Increases Bendiocarb Efficacy Through Activation of Octopamine Receptors and Protein Kinase A. Molecules 2019; 24:molecules24203775. [PMID: 31635151 PMCID: PMC6832705 DOI: 10.3390/molecules24203775] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/17/2019] [Accepted: 10/18/2019] [Indexed: 12/31/2022] Open
Abstract
Great effort is put into seeking a new and effective strategies to control insect pests. One of them is to combine natural products with chemical insecticides to increase their effectiveness. In the study presented, menthol which is an essential oil component was evaluated on its ability to increase the efficiency of bendiocarb, carbamate insecticide. A multi-approach study was conducted using biochemical method (to measure acetylcholinesterase enzyme activity), electrophysiological technique (microelectrode recordings in DUM neurons in situ), and confocal microscopy (for calcium imaging). In the electrophysiological experiments, menthol caused hyperpolarization, which was blocked by an octopamine receptor antagonist (phentolamine) and an inhibitor of protein kinase A (H-89). It also raised the intracellular calcium level. The effect of bendiocarb was potentiated by menthol and this phenomenon was abolished by phentolamine and H-89 but not by protein kinase C inhibitor (bisindolylmaleimide IX). The results indicate that menthol increases carbamate insecticide efficiency by acting on octopamine receptors and triggering protein kinase A phosphorylation pathway.
Collapse
|
24
|
Ohadi D, Schmitt DL, Calabrese B, Halpain S, Zhang J, Rangamani P. Computational Modeling Reveals Frequency Modulation of Calcium-cAMP/PKA Pathway in Dendritic Spines. Biophys J 2019; 117:1963-1980. [PMID: 31668749 PMCID: PMC7031750 DOI: 10.1016/j.bpj.2019.10.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 07/30/2019] [Accepted: 10/02/2019] [Indexed: 12/20/2022] Open
Abstract
Dendritic spines are the primary excitatory postsynaptic sites that act as subcompartments of signaling. Ca2+ is often the first and most rapid signal in spines. Downstream of calcium, the cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) pathway plays a critical role in the regulation of spine formation, morphological modifications, and ultimately, learning and memory. Although the dynamics of calcium are reasonably well-studied, calcium-induced cAMP/PKA dynamics, particularly with respect to frequency modulation, are not fully explored. In this study, we present a well-mixed model for the dynamics of calcium-induced cAMP/PKA dynamics in dendritic spines. The model is constrained using experimental observations in the literature. Further, we measured the calcium oscillation frequency in dendritic spines of cultured hippocampal CA1 neurons and used these dynamics as model inputs. Our model predicts that the various steps in this pathway act as frequency modulators for calcium, and the high frequency of calcium input is filtered by adenylyl cyclase 1 and phosphodiesterases in this pathway such that cAMP/PKA only responds to lower frequencies. This prediction has important implications for noise filtering and long-timescale signal transduction in dendritic spines. A companion manuscript presents a three-dimensional spatial model for the same pathway.
Collapse
Affiliation(s)
- Donya Ohadi
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California
| | - Danielle L Schmitt
- Department of Pharmacology, University of California San Diego, La Jolla, California
| | - Barbara Calabrese
- Division of Biological Sciences and Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Shelley Halpain
- Division of Biological Sciences and Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Jin Zhang
- Department of Pharmacology, University of California San Diego, La Jolla, California
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California.
| |
Collapse
|
25
|
Mertz C, Krarup S, Jensen CD, Lindholm SEH, Kjær C, Pinborg LH, Bak LK. Aspects of cAMP Signaling in Epileptogenesis and Seizures and Its Potential as Drug Target. Neurochem Res 2019; 45:1247-1255. [PMID: 31414342 DOI: 10.1007/s11064-019-02853-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/30/2019] [Accepted: 08/03/2019] [Indexed: 12/19/2022]
Abstract
Epilepsy is one of the most common chronic neurological conditions. Today, close to 30 different medications to prevent epileptic seizures are in use; yet, far from all patients become seizure free upon medical treatment. Thus, there is a need for new pharmacological approaches including novel drug targets for the management of epilepsy. Despite the fact that a role for cAMP signaling in epileptogenesis and seizures was first suggested some four decades ago, none of the current medications target the cAMP signaling system. The reasons for this are probably many including limited knowledge of the underlying biology and pathology as well as difficulties in designing selective drugs for the different components of the cAMP signaling system. This review explores selected aspects of cAMP signaling in the context of epileptogenesis and seizures including cAMP response element binding (CREB)-mediated transcriptional regulation. We discuss the therapeutic potential of targeting cAMP signaling in epilepsy and point to an increased knowledge of the A-kinase anchoring protein-based signaling hubs as being of seminal importance for future drug discovery within the field. Further, in terms of targeting CREB, we argue that targeting upstream cAMP signals might be more fruitful than targeting CREB itself. Finally, we point to astrocytes as cellular targets in epilepsy since cAMP signals may regulate astrocytic K+ clearance affecting neuronal excitability.
Collapse
Affiliation(s)
- Christoffer Mertz
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Sara Krarup
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Cecilie D Jensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Sandy E H Lindholm
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Christina Kjær
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100, Copenhagen, Denmark.,Department of Technology, Faculty of Health and Technology, University College Copenhagen, 2200, Copenhagen, Denmark
| | - Lars H Pinborg
- Epilepsy Clinic & Neurobiology Research Unit, Copenhagen University Hospital, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Lasse K Bak
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100, Copenhagen, Denmark.
| |
Collapse
|
26
|
Li Z, Liu T, Gilmore A, Gómez NM, Mitchell CH, Li YP, Oursler MJ, Yang S. Regulator of G Protein Signaling Protein 12 (Rgs12) Controls Mouse Osteoblast Differentiation via Calcium Channel/Oscillation and Gαi-ERK Signaling. J Bone Miner Res 2019; 34:752-764. [PMID: 30489658 PMCID: PMC7675783 DOI: 10.1002/jbmr.3645] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 11/13/2018] [Accepted: 11/17/2018] [Indexed: 12/11/2022]
Abstract
Bone homeostasis intimately relies on the balance between osteoblasts (OBs) and osteoclasts (OCs). Our previous studies have revealed that regulator of G protein signaling protein 12 (Rgs12), the largest protein in the Rgs super family, is essential for osteoclastogenesis from hematopoietic cells and OC precursors. However, how Rgs12 regulates OB differentiation and function is still unknown. To understand that, we generated an OB-targeted Rgs12 conditional knockout (CKO) mice model by crossing Rgs12fl/fl mice with Osterix (Osx)-Cre transgenic mice. We found that Rgs12 was highly expressed in both OB precursor cells (OPCs) and OBs of wild-type (WT) mice, and gradually increased during OB differentiation, whereas Rgs12-CKO mice (OsxCre/+ ; Rgs12fl/fl ) exhibited a dramatic decrease in both trabecular and cortical bone mass, with reduced numbers of OBs and increased apoptotic cell population. Loss of Rgs12 in OPCs in vitro significantly inhibited OB differentiation and the expression of OB marker genes, resulting in suppression of OB maturation and mineralization. Further mechanism study showed that deletion of Rgs12 in OPCs significantly inhibited guanosine triphosphatase (GTPase) activity and cyclic adenosine monophosphate (cAMP) level, and impaired Calcium (Ca2+ ) oscillations via restraints of major Ca2+ entry sources (extracellular Ca2+ influx and intracellular Ca2+ release from endoplasmic reticulum), partially contributed by the blockage of L-type Ca2+ channel mediated Ca2+ influx. Downstream mediator extracellular signal-related protein kinase (ERK) was found inactive in OBs of OsxCre/+ ; Rgs12fl/fl mice and in OPCs after Rgs12 deletion, whereas application of pertussis toxin (PTX) or overexpression of Rgs12 could rescue the defective OB differentiation via restoration of ERK phosphorylation. Our findings reveal that Rgs12 is an important regulator during osteogenesis and highlight Rgs12 as a potential therapeutic target for bone disorders. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Ziqing Li
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania Philadelphia, PA 19104, USA
| | - Tongjun Liu
- Department of Oral Biology, School of Dental Medicine, University of Buffalo, State University of New York, Buffalo, NY 14215, USA
- Department of Implantology, Shandong Provincial Key Laboratory of Oral Biomedicine, School of Stomatology, Shandong University
- Department of Stomatology, the Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong province 250000, China
| | - Alyssa Gilmore
- Department of Oral Biology, School of Dental Medicine, University of Buffalo, State University of New York, Buffalo, NY 14215, USA
| | - Néstor Más Gómez
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania Philadelphia, PA 19104, USA
| | - Claire H Mitchell
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania Philadelphia, PA 19104, USA
- Department of Physiology, School of Medicine, University of Pennsylvania Philadelphia, PA 19104, USA
| | - Yi-ping Li
- Department of Pathology, University of Alabama in Birmingham, Birmingham, AL 35294, USA
| | - Merry J Oursler
- Department of Medicine, Endocrine Research Unit, Mayo Clinic, Rochester, MN 55905, USA
| | - Shuying Yang
- Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania Philadelphia, PA 19104, USA
- The Penn Center for Musculoskeletal Disorders, University of Pennsylvania Philadelphia, PA 19104, USA
- Department of Oral Biology, School of Dental Medicine, University of Buffalo, State University of New York, Buffalo, NY 14215, USA
| |
Collapse
|
27
|
Yannopoulos D, Bartos JA, Aufderheide TP, Callaway CW, Deo R, Garcia S, Halperin HR, Kern KB, Kudenchuk PJ, Neumar RW, Raveendran G. The Evolving Role of the Cardiac Catheterization Laboratory in the Management of Patients With Out-of-Hospital Cardiac Arrest: A Scientific Statement From the American Heart Association. Circulation 2019; 139:e530-e552. [DOI: 10.1161/cir.0000000000000630] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Coronary artery disease is prevalent in different causes of out-of-hospital cardiac arrest (OHCA), especially in individuals presenting with shockable rhythms of ventricular fibrillation/pulseless ventricular tachycardia (VF/pVT). The purpose of this report is to review the known prevalence and potential importance of coronary artery disease in patients with OHCA and to describe the emerging paradigm of treatment with advanced perfusion/reperfusion techniques and their potential benefits on the basis of available evidence. Although randomized clinical trials are planned or ongoing, current scientific evidence rests principally on observational case series with their potential confounding selection bias. Among patients resuscitated from VF/pVT OHCA with ST-segment elevation on their postresuscitation ECG, the prevalence of coronary artery disease has been shown to be 70% to 85%. More than 90% of these patients have had successful percutaneous coronary intervention. Conversely, among patients resuscitated from VF/pVT OHCA without ST-segment elevation on their postresuscitation ECG, the prevalence of coronary artery disease has been shown to be 25% to 50%. For these patients, early access to the cardiac catheterization laboratory is associated with a 10% to 15% absolute higher functionally favorable survival rate compared with more conservative approaches of late or no access to the cardiac catheterization laboratory. In patients with VF/pVT OHCA refractory to standard treatment, a new treatment paradigm is also emerging that uses venoarterial extracorporeal membrane oxygenation to facilitate return of normal perfusion and to support further resuscitation efforts, including coronary angiography and percutaneous coronary intervention. The burden of coronary artery disease is high in this patient population, presumably causative in most patients. The strategy of venoarterial extracorporeal membrane oxygenation, coronary angiography, and percutaneous coronary intervention has resulted in functionally favorable survival rates ranging from 9% to 45% in observational studies in this patient population. Patients with VF/pVT should be considered at the highest severity in the continuum of acute coronary syndromes. These patients have a significant burden of coronary artery disease and acute coronary thrombotic events. Evidence from randomized trials will further define optimal clinical practice.
Collapse
|
28
|
Drelich A, Judy B, He X, Chang Q, Yu S, Li X, Lu F, Wakamiya M, Popov V, Zhou J, Ksiazek T, Gong B. Exchange Protein Directly Activated by cAMP Modulates Ebola Virus Uptake into Vascular Endothelial Cells. Viruses 2018; 10:v10100563. [PMID: 30332733 PMCID: PMC6213290 DOI: 10.3390/v10100563] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/13/2018] [Accepted: 10/13/2018] [Indexed: 12/16/2022] Open
Abstract
Members of the family Filoviridae, including Ebola virus (EBOV) and Marburg virus (MARV), cause severe hemorrhagic fever in humans and nonhuman primates. Given their high lethality, a comprehensive understanding of filoviral pathogenesis is urgently needed. In the present studies, we revealed that the exchange protein directly activated by cAMP 1 (EPAC1) gene deletion protects vasculature in ex vivo explants from EBOV infection. Importantly, pharmacological inhibition of EPAC1 using EPAC-specific inhibitors (ESIs) mimicked the EPAC1 knockout phenotype in the ex vivo model. ESI treatment dramatically decreased EBOV infectivity in both ex vivo vasculature and in vitro vascular endothelial cells (ECs). Furthermore, postexposure protection of ECs against EBOV infection was conferred using ESIs. Protective efficacy of ESIs in ECs was observed also in MARV infection. Additional studies using a vesicular stomatitis virus pseudotype that expresses EBOV glycoprotein (EGP-VSV) confirmed that ESIs reduced infection in ECs. Ultrastructural studies suggested that ESIs blocked EGP-VSV internalization via inhibition of macropinocytosis. The inactivation of EPAC1 affects the early stage of viral entry after viral binding to the cell surface, but before early endosome formation, in a phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)-dependent manner. Our study delineated a new critical role of EPAC1 during EBOV uptake into ECs.
Collapse
Affiliation(s)
- Aleksandra Drelich
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Barbara Judy
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Xi He
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Department of Cardiovascular Surgery, Changhai Institute of Cardiovascular Surgery, Shanghai 200433, China.
| | - Qing Chang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Shangyi Yu
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.
- Department of Cardiovascular Surgery, Changhai Institute of Cardiovascular Surgery, Shanghai 200433, China.
| | - Xiang Li
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Fanglin Lu
- Department of Cardiovascular Surgery, Changhai Institute of Cardiovascular Surgery, Shanghai 200433, China.
| | - Maki Wakamiya
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Vsevolod Popov
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Jia Zhou
- Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Thomas Ksiazek
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Bin Gong
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
29
|
Leslie SN, Nairn AC. cAMP regulation of protein phosphatases PP1 and PP2A in brain. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1866:64-73. [PMID: 30401536 DOI: 10.1016/j.bbamcr.2018.09.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 09/13/2018] [Indexed: 12/21/2022]
Abstract
Normal functioning of the brain is dependent upon a complex web of communication between numerous cell types. Within neuronal networks, the faithful transmission of information between neurons relies on an equally complex organization of inter- and intra-cellular signaling systems that act to modulate protein activity. In particular, post-translational modifications (PTMs) are responsible for regulating protein activity in response to neurochemical signaling. The key second messenger, cyclic adenosine 3',5'-monophosphate (cAMP), regulates one of the most ubiquitous and influential PTMs, phosphorylation. While cAMP is canonically viewed as regulating the addition of phosphate groups through its activation of cAMP-dependent protein kinases, it plays an equally critical role in regulating removal of phosphate through indirect control of protein phosphatase activity. This dichotomy of regulation by cAMP places it as one of the key regulators of protein activity in response to neuronal signal transduction throughout the brain. In this review we focus on the role of cAMP in regulation of the serine/threonine phosphatases protein phosphatase 1 (PP1) and protein phosphatase 2A (PP2A) and the relevance of control of PP1 and PP2A to regulation of brain function and behavior.
Collapse
Affiliation(s)
- Shannon N Leslie
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT, United States of America
| | - Angus C Nairn
- Department of Psychiatry, Yale University, New Haven, CT, United States of America
| |
Collapse
|
30
|
Johnstone TB, Agarwal SR, Harvey RD, Ostrom RS. cAMP Signaling Compartmentation: Adenylyl Cyclases as Anchors of Dynamic Signaling Complexes. Mol Pharmacol 2018; 93:270-276. [PMID: 29217670 PMCID: PMC5820540 DOI: 10.1124/mol.117.110825] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 12/04/2017] [Indexed: 11/22/2022] Open
Abstract
It is widely accepted that cAMP signaling is compartmentalized within cells. However, our knowledge of how receptors, cAMP signaling enzymes, effectors, and other key proteins form specific signaling complexes to regulate specific cell responses is limited. The multicomponent nature of these systems and the spatiotemporal dynamics involved as proteins interact and move within a cell make cAMP responses highly complex. Adenylyl cyclases, the enzymatic source of cAMP production, are key starting points for understanding cAMP compartments and defining the functional signaling complexes. Three basic elements are required to form a signaling compartment. First, a localized signal is generated by a G protein-coupled receptor paired to one or more of the nine different transmembrane adenylyl cyclase isoforms that generate the cAMP signal in the cytosol. The diffusion of cAMP is subsequently limited by several factors, including expression of any number of phosphodiesterases (of which there are 24 genes plus spice variants). Finally, signal response elements are differentially localized to respond to cAMP produced within each locale. A-kinase-anchoring proteins, of which there are 43 different isoforms, facilitate this by targeting protein kinase A to specific substrates. Thousands of potential combinations of these three elements are possible in any given cell type, making the characterization of cAMP signaling compartments daunting. This review will focus on what is known about how cells organize cAMP signaling components as well as identify the unknowns. We make an argument for adenylyl cyclases being central to the formation and maintenance of these signaling complexes.
Collapse
Affiliation(s)
- Timothy B Johnstone
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (T.B.J., R.S.O.); and Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno Nevada (S.R.A., R.D.H.)
| | - Shailesh R Agarwal
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (T.B.J., R.S.O.); and Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno Nevada (S.R.A., R.D.H.)
| | - Robert D Harvey
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (T.B.J., R.S.O.); and Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno Nevada (S.R.A., R.D.H.)
| | - Rennolds S Ostrom
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, California (T.B.J., R.S.O.); and Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno Nevada (S.R.A., R.D.H.)
| |
Collapse
|
31
|
Robichaux WG, Cheng X. Intracellular cAMP Sensor EPAC: Physiology, Pathophysiology, and Therapeutics Development. Physiol Rev 2018; 98:919-1053. [PMID: 29537337 PMCID: PMC6050347 DOI: 10.1152/physrev.00025.2017] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 09/05/2017] [Accepted: 09/06/2017] [Indexed: 12/13/2022] Open
Abstract
This review focuses on one family of the known cAMP receptors, the exchange proteins directly activated by cAMP (EPACs), also known as the cAMP-regulated guanine nucleotide exchange factors (cAMP-GEFs). Although EPAC proteins are fairly new additions to the growing list of cAMP effectors, and relatively "young" in the cAMP discovery timeline, the significance of an EPAC presence in different cell systems is extraordinary. The study of EPACs has considerably expanded the diversity and adaptive nature of cAMP signaling associated with numerous physiological and pathophysiological responses. This review comprehensively covers EPAC protein functions at the molecular, cellular, physiological, and pathophysiological levels; and in turn, the applications of employing EPAC-based biosensors as detection tools for dissecting cAMP signaling and the implications for targeting EPAC proteins for therapeutic development are also discussed.
Collapse
Affiliation(s)
- William G Robichaux
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center , Houston, Texas
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, The Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center , Houston, Texas
| |
Collapse
|
32
|
De novo transcriptomics reveal distinct phototransduction signaling components in the retina and skin of a color-changing vertebrate, the hogfish (Lachnolaimus maximus). J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2018; 204:475-485. [PMID: 29492668 DOI: 10.1007/s00359-018-1254-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/22/2018] [Accepted: 02/22/2018] [Indexed: 10/17/2022]
Abstract
Across diverse taxa, an increasing number of photoreceptive systems are being discovered in tissues outside of the eye, such as in the skin. Dermal photoreception is believed to serve a variety of functions, including rapid color change via specialized cells called chromatophores. In vitro studies of this system among color-changing fish have suggested the use of a phototransduction signaling cascade that fundamentally differs from that of the retina. Thus, the goal of this study was to identify phototransduction genes and compare their expression in the retina and skin of a color-changing fish, the hogfish Lachnolaimus maximus. De novo transcriptomics revealed the expression of genes that may underlie distinct, yet complete phototransduction cascades in L. maximus retina and skin. In contrast to the five visual opsin genes and cGMP-dependent phototransduction components expressed in the retina of L. maximus, only a single short-wavelength sensitive opsin (SWS1) and putative cAMP-dependent phototransduction components were expressed in the skin. These data suggest a separate evolutionary history of phototransduction in the retina and skin of certain vertebrates and, for the first time, indicate an expression repertoire of genes that underlie a non-retinal phototransduction pathway in the skin of a color-changing fish.
Collapse
|
33
|
Endogenous Gαq-Coupled Neuromodulator Receptors Activate Protein Kinase A. Neuron 2017; 96:1070-1083.e5. [PMID: 29154125 DOI: 10.1016/j.neuron.2017.10.023] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 09/11/2017] [Accepted: 10/16/2017] [Indexed: 01/09/2023]
Abstract
Protein kinase A (PKA) integrates inputs from G-protein-coupled neuromodulator receptors to modulate synaptic and cellular function. Gαs signaling stimulates PKA activity, whereas Gαi inhibits PKA activity. Gαq, on the other hand, signals through phospholipase C, and it remains unclear whether Gαq-coupled receptors signal to PKA in their native context. Here, using two independent optical reporters of PKA activity in acute mouse hippocampus slices, we show that endogenous Gαq-coupled muscarinic acetylcholine receptors activate PKA. Mechanistically, this effect is mediated by parallel signaling via either calcium or protein kinase C. Furthermore, multiple Gαq-coupled receptors modulate phosphorylation by PKA, a classical Gαs/Gαi effector. Thus, these results highlight PKA as a biochemical integrator of three major types of GPCRs and necessitate reconsideration of classic models used to predict neuronal signaling in response to the large family of Gαq-coupled receptors.
Collapse
|
34
|
Getz MC, Nirody JA, Rangamani P. Stability analysis in spatial modeling of cell signaling. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2017; 10. [PMID: 28787545 DOI: 10.1002/wsbm.1395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 06/06/2017] [Accepted: 06/11/2017] [Indexed: 11/08/2022]
Abstract
Advances in high-resolution microscopy and other techniques have emphasized the spatio-temporal nature of information transfer through signal transduction pathways. The compartmentalization of signaling molecules and the existence of microdomains are now widely acknowledged as key features in biochemical signaling. To complement experimental observations of spatio-temporal dynamics, mathematical modeling has emerged as a powerful tool. Using modeling, one can not only recapitulate experimentally observed dynamics of signaling molecules, but also gain an understanding of the underlying mechanisms in order to generate experimentally testable predictions. Reaction-diffusion systems are commonly used to this end; however, the analysis of coupled nonlinear systems of partial differential equations, generated by considering large reaction networks is often challenging. Here, we aim to provide an introductory tutorial for the application of reaction-diffusion models to the spatio-temporal dynamics of signaling pathways. In particular, we outline the steps for stability analysis of such models, with a focus on biochemical signal transduction. WIREs Syst Biol Med 2018, 10:e1395. doi: 10.1002/wsbm.1395 This article is categorized under: Biological Mechanisms > Cell Signaling Analytical and Computational Methods > Dynamical Methods Models of Systems Properties and Processes > Mechanistic Models.
Collapse
Affiliation(s)
- Michael C Getz
- Chemical Engineering Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Jasmine A Nirody
- Biophysics Graduate Program, University of California, Berkeley, CA, USA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
35
|
Abstract
Electrical and/or electromechanical stimulation has been shown to play a significant role in regenerating various functionalities in soft tissues, such as tendons, muscles, and nerves. In this work, we investigate the piezoelectric polymer polyvinylidene fluoride (PVDF) as a potential substrate for wireless neuronal differentiation. Piezoelectric PVDF enables generation of electrical charges on its surface upon acoustic stimulation, inducing neuritogenesis of PC12 cells. We demonstrate that the effect of pure piezoelectric stimulation on neurite generation in PC12 cells is comparable to the ones induced by neuronal growth factor (NGF). In inhibitor experiments, our results indicate that dynamic stimulation of PVDF by ultrasonic (US) waves activates calcium channels, thus inducing the generation of neurites via a cyclic adenosine monophosphate (cAMP)-dependent pathway. This mechanism is independent from the well-studied NGF induced mitogen-activated protein kinases/extracellular signal-regulated kinases (MAPK/ERK) pathway. The use of US, in combination with piezoelectric polymers, is advantageous since focused power transmission can occur deep into biological tissues, which holds great promise for the development of non-invasive neuroregenerative devices.
Collapse
|
36
|
Zhao J, Wang L, Li Y. Electroacupuncture Alleviates the Inflammatory Response via Effects on M1 and M2 Macrophages after Spinal Cord Injury. Acupunct Med 2017; 35:224-230. [PMID: 28077367 DOI: 10.1136/acupmed-2016-011107] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2016] [Indexed: 01/09/2023]
Abstract
Background Macrophages/microglia are important effector cells at the site of spinal cord injury (SCI). M1-type macrophages facilitate innate immunity to remove foreign microbes and wound debris from the injury site. M2-type macrophages exhibit tissue repair properties and attenuate production of pro-inflammatory cytokines. Regulation of the polarisation of M1/M2 macrophages may affect the inflammatory response in SCI and may be related to neurotrophin-3 (NT-3). Electroacupuncture (EA) at GV acupuncture points can be used as an adjuvant therapy for SCI. Aim To investigate the effects of EA on Basso, Beattie and Bresnahan (BBB) functional evaluation and inflammatory cytokines (tumour necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6 and IL-10), and on the proportions of M1/M2 macrophages, and to provide a greater understanding of the mechanisms underlying the potential clinical treatment of SCI. Methods A rat SCI model was induced by spinal segment transection at T10 in 16 Sprague-Dawley rats. A further eight rats were included as a Control group. Ten surviving SCI model rats were divided into two groups (n=5 each): an SCI group that remained untreated; and an SCI+EA group that received EA at GV6 and GV9. Results EA improved BBB scores, inhibited the proportion of M1 macrophages and TNF-α, IL-1β and IL-6 levels, and downregulated the M1 marker CD86. By contrast, EA enhanced IL-10, the proportion of M2 macrophages and upregulated the M2 marker CD206 and NT-3 expression. Conclusions EA had a positive impact on SCI model rats. This may be related to the neuroprotective effect of NT-3, which may increase the polarisation of M2 microglia/macrophages.
Collapse
Affiliation(s)
- Jiagui Zhao
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Likui Wang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Yuanhai Li
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| |
Collapse
|
37
|
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: 128] [Impact Index Per Article: 18.3] [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
|
38
|
Maus M, Cuk M, Patel B, Lian J, Ouimet M, Kaufmann U, Yang J, Horvath R, Hornig-Do HT, Chrzanowska-Lightowlers ZM, Moore KJ, Cuervo AM, Feske S. Store-Operated Ca 2+ Entry Controls Induction of Lipolysis and the Transcriptional Reprogramming to Lipid Metabolism. Cell Metab 2017; 25:698-712. [PMID: 28132808 PMCID: PMC5342942 DOI: 10.1016/j.cmet.2016.12.021] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 09/01/2016] [Accepted: 12/28/2016] [Indexed: 12/26/2022]
Abstract
Ca2+ signals were reported to control lipid homeostasis, but the Ca2+ channels and pathways involved are largely unknown. Store-operated Ca2+ entry (SOCE) is a ubiquitous Ca2+ influx pathway regulated by stromal interaction molecule 1 (STIM1), STIM2, and the Ca2+ channel ORAI1. We show that SOCE-deficient mice accumulate pathological amounts of lipid droplets in the liver, heart, and skeletal muscle. Cells from patients with loss-of-function mutations in STIM1 or ORAI1 show a similar phenotype, suggesting a cell-intrinsic role for SOCE in the regulation of lipid metabolism. SOCE is crucial to induce mobilization of fatty acids from lipid droplets, lipolysis, and mitochondrial fatty acid oxidation. SOCE regulates cyclic AMP production and the expression of neutral lipases as well as the transcriptional regulators of lipid metabolism, peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α), and peroxisome proliferator-activated receptor α (PPARα). SOCE-deficient cells upregulate lipophagy, which protects them from lipotoxicity. Our data provide evidence for an important role of SOCE in lipid metabolism.
Collapse
Affiliation(s)
- Mate Maus
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Mario Cuk
- Department of Pediatrics, Zagreb University Hospital Centre and School of Medicine, 10 000 Zagreb, Croatia
| | - Bindi Patel
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jayson Lian
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Mireille Ouimet
- Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Ulrike Kaufmann
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Jun Yang
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Rita Horvath
- Wellcome Trust Centre for Mitochondrial Research, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Hue-Tran Hornig-Do
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Zofia M Chrzanowska-Lightowlers
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Kathryn J Moore
- Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Stefan Feske
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
39
|
Bueno Bergantin L. Novel Challenges for the Therapeutics of Depression: Pharmacological Modulation of Interaction between the Intracellular Signaling Pathways Mediated by Ca2+ and cAMP. JOURNAL OF ADDICTION THERAPY AND RESEARCH 2017; 1:001-006. [DOI: 10.29328/journal.jatr.1001001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/03/2024]
|
40
|
LB B. The “Calcium Paradox” Due To Ca2+/Camp Interaction: New Insights for the Neuroscience Field. JOURNAL OF NEUROSCIENCE AND NEUROLOGICAL DISORDERS 2017; 1:012-015. [DOI: 10.29328/journal.jnnd.1001002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/03/2024]
|
41
|
Ramos-Espiritu L, Kleinboelting S, Navarrete FA, Alvau A, Visconti PE, Valsecchi F, Starkov A, Manfredi G, Buck H, Adura C, Zippin JH, van den Heuvel J, Glickman JF, Steegborn C, Levin LR, Buck J. Discovery of LRE1 as a specific and allosteric inhibitor of soluble adenylyl cyclase. Nat Chem Biol 2016; 12:838-44. [PMID: 27547922 PMCID: PMC5030147 DOI: 10.1038/nchembio.2151] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 05/23/2016] [Indexed: 12/22/2022]
Abstract
The prototypical second messenger cAMP regulates a wide variety of physiological processes. It can simultaneously mediate diverse functions by acting locally in independently regulated microdomains. In mammalian cells, two types of adenylyl cyclase generate cAMP: G-protein-regulated transmembrane adenylyl cyclases and bicarbonate-, calcium- and ATP-regulated soluble adenylyl cyclase (sAC). Because each type of cyclase regulates distinct microdomains, methods to distinguish between them are needed to understand cAMP signaling. We developed a mass-spectrometry-based adenylyl cyclase assay, which we used to identify a new sAC-specific inhibitor, LRE1. LRE1 bound to the bicarbonate activator binding site and inhibited sAC via a unique allosteric mechanism. LRE1 prevented sAC-dependent processes in cellular and physiological systems, and it will facilitate exploration of the therapeutic potential of sAC inhibition.
Collapse
Affiliation(s)
- Lavoisier Ramos-Espiritu
- Department of Pharmacology, Weill Cornell Medical College, New York, New York, USA
- The High-Throughput Screening and Spectroscopy Resource Center, The Rockefeller University, New York, New York, USA
| | | | - Felipe A Navarrete
- Department of Veterinary and Animal Science, University of Massachusetts, Amherst, Massachusetts, USA
| | - Antonio Alvau
- Department of Veterinary and Animal Science, University of Massachusetts, Amherst, Massachusetts, USA
| | - Pablo E Visconti
- Department of Veterinary and Animal Science, University of Massachusetts, Amherst, Massachusetts, USA
| | - Federica Valsecchi
- Brain and Mind Research Institute, Weill Cornell Medical College, New York, New York, USA
| | - Anatoly Starkov
- Brain and Mind Research Institute, Weill Cornell Medical College, New York, New York, USA
| | - Giovanni Manfredi
- Brain and Mind Research Institute, Weill Cornell Medical College, New York, New York, USA
| | - Hannes Buck
- Department of Pharmacology, Weill Cornell Medical College, New York, New York, USA
| | - Carolina Adura
- The High-Throughput Screening and Spectroscopy Resource Center, The Rockefeller University, New York, New York, USA
| | - Jonathan H Zippin
- Department of Dermatology, Weill Cornell Medical College, New York, New York, USA
| | | | - J Fraser Glickman
- The High-Throughput Screening and Spectroscopy Resource Center, The Rockefeller University, New York, New York, USA
| | - Clemens Steegborn
- Department of Biochemistry, University of Bayreuth, Bayreuth, Germany
| | - Lonny R Levin
- Department of Pharmacology, Weill Cornell Medical College, New York, New York, USA
| | - Jochen Buck
- Department of Pharmacology, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
42
|
Riedl A, Shamsi Z, Anderton M, Goldfarb P, Wiseman A. Differing features of proteins in membranes may result in antioxidant or prooxidant action: opposite effects on lipid peroxidation of alcohol dehydrogenase and albumin in liposomal systems. Redox Rep 2016; 2:35-40. [DOI: 10.1080/13510002.1996.11747024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
43
|
Li X, Wang L, Li Y, Zhao N, Zhen L, Fu J, Yang Q. Calcium regulates motility and protein phosphorylation by changing cAMP and ATP concentrations in boar sperm in vitro. Anim Reprod Sci 2016; 172:39-51. [PMID: 27423488 DOI: 10.1016/j.anireprosci.2016.07.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 06/29/2016] [Accepted: 07/01/2016] [Indexed: 12/15/2022]
Abstract
Considering the importance of calcium (Ca(2+)) in regulating sperm capacitation, hyperactivation and acrosome reaction, little is known about the molecular mechanism of action of this ion in this process. In the present study, assessment of the molecular mechanism from the perspective of energy metabolism occurred. Sperm motility variables were determined using computer-assisted sperm analysis (CASA) and the phosphorylation of PKA substrates, tyrosine residues and AMP-activated protein kinase (AMPK) were analyzed by Western blot. Moreover, intracellular sperm-specific glyceraldehyde 3-phosphatedehydrogenase (GAPDH) activity, 3'-5'-cyclic adenosine monophosphate (cAMP) and adenosine 5'-triphosphate (ATP) concentrations were assessed in boar sperm treated with Ca(2+). Results of the present study indicated that, under greater extracellular Ca(2+)concentrations (≥3.0mM), sperm motility and protein phosphorylation were inhibited. Interestingly, these changes were correlated with that of GAPDH activity, AMPK phosphorylation, cAMP and ATP concentrations. The negative effects of Ca(2+) on these intracellular processes were attenuated by addition of the calmodulin (CaM) inhibitor W7 and the inhibitor of calmodulin-dependent protein kinase (CaMK), KN-93. In the presence of greater extracellular Ca(2+), however, the phosphorylation pathway was suppressed by H-89. Taken together, these results suggested that Ca(2+) had a dual role in regulating boar sperm motility and protein phosphorylation due to the changes of cAMP and ATP concentrations, in response to cAMP-mediated signal transduction and the Ca(2+) signaling cascade. The present study provided some novel insights into the molecular mechanism underlying the effects of Ca(2+) on boar sperm as well as the involvement of energy metabolism in this mechanism.
Collapse
Affiliation(s)
- Xinhong Li
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Lirui Wang
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuhua Li
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Na Zhao
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Linqing Zhen
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jieli Fu
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qiangzhen Yang
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
44
|
Bergantin LB, Caricati-Neto A. Challenges for the pharmacological treatment of neurological and psychiatric disorders: Implications of the Ca(2+)/cAMP intracellular signalling interaction. Eur J Pharmacol 2016; 788:255-260. [PMID: 27349146 DOI: 10.1016/j.ejphar.2016.06.034] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 06/15/2016] [Accepted: 06/21/2016] [Indexed: 11/15/2022]
Abstract
In 2013, we discovered that the entitled "calcium paradox" phenomenon, which means a paradoxical sympathetic hyperactivity produced by l-type Ca(2+) channel blockers (CCBs), used in antihypertensive therapy, is due to interaction between the intracellular signalling pathways mediated by Ca(2+) and cAMP (Ca(2+)/cAMP interaction). In 2015, we proposed that the pharmacological manipulation of this interaction could be a new therapeutic strategy for increasing neurotransmission in psychiatric disorders, and producing neuroprotection in the neurodegenerative diseases. Besides the paradoxical sympathetic hyperactivity produced by CCBs, several clinical studies have been demonstrating pleiotropic effects of CCBs, including neuroprotective effects. CCBs genuinely exhibit cognitive-enhancing abilities and reduce the risk of dementia, including Alzheimer's, Parkinson´s disease and others. The molecular mechanisms involved in these pleiotropic effects remain under debate. Our recent discovery that the "calcium paradox" phenomenon is due to Ca(2+)/cAMP interaction may provide new insights for the pharmacological treatment of neurological and psychiatric disorders, including enhancement of current therapies mainly by reducing adverse effects, and improving effectiveness of modern medicines. Whether Ca(2+)/cAMP interaction is involved in CCBs pleiotropic effects also deserves special attention. Then, the pharmacological manipulation of the Ca(2+)/cAMP interaction could be a more efficient therapeutic strategy for increasing neurotransmission in psychiatric disorders, and producing neuroprotection in the neurodegenerative diseases. Thus, in this review we summarize the current knowledge of this field, making new directions and future perspectives.
Collapse
Affiliation(s)
- Leandro Bueno Bergantin
- Department of Pharmacology, Universidade Federal de São Paulo, Escola Paulista de Medicina, Laboratory of Autonomic and Cardiovascular Pharmacology, 55 11 5576-4973, Rua Pedro de Toledo, 669, Vila Clementino, São Paulo, SP CEP 04039-032, Brazil.
| | - Afonso Caricati-Neto
- Department of Pharmacology, Universidade Federal de São Paulo, Escola Paulista de Medicina, Laboratory of Autonomic and Cardiovascular Pharmacology, 55 11 5576-4973, Rua Pedro de Toledo, 669, Vila Clementino, São Paulo, SP CEP 04039-032, Brazil
| |
Collapse
|
45
|
Kreft M, Jorgačevski J, Vardjan N, Zorec R. Unproductive exocytosis. J Neurochem 2016; 137:880-9. [DOI: 10.1111/jnc.13561] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 01/21/2016] [Accepted: 01/25/2016] [Indexed: 01/04/2023]
Affiliation(s)
- Marko Kreft
- Laboratory of Neuroendocrinology-Molecular Cell Physiology; Faculty of Medicine; University of Ljubljana; Ljubljana Slovenia
- Celica BIOMEDICAL; Ljubljana Slovenia
- Department of Biology; Biotechnical Faculty; University of Ljubljana; Ljubljana Slovenia
| | - Jernej Jorgačevski
- Laboratory of Neuroendocrinology-Molecular Cell Physiology; Faculty of Medicine; University of Ljubljana; Ljubljana Slovenia
- Celica BIOMEDICAL; Ljubljana Slovenia
| | - Nina Vardjan
- Laboratory of Neuroendocrinology-Molecular Cell Physiology; Faculty of Medicine; University of Ljubljana; Ljubljana Slovenia
- Celica BIOMEDICAL; Ljubljana Slovenia
| | - Robert Zorec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology; Faculty of Medicine; University of Ljubljana; Ljubljana Slovenia
- Celica BIOMEDICAL; Ljubljana Slovenia
| |
Collapse
|
46
|
Assessing the relevance of light for fungi: Implications and insights into the network of signal transmission. ADVANCES IN APPLIED MICROBIOLOGY 2016; 76:27-78. [PMID: 21924971 DOI: 10.1016/b978-0-12-387048-3.00002-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Light represents an important environmental cue, which provides information enabling fungi to prepare and react to the different ambient conditions between day and night. This adaptation requires both anticipation of the changing conditions, which is accomplished by daily rhythmicity of gene expression brought about by the circadian clock, and reaction to sudden illumination. Besides perception of the light signal, also integration of this signal with other environmental cues, most importantly nutrient availability, necessitates light-dependent regulation of signal transduction pathways and metabolic pathways. An influence of light and/or the circadian clock is known for the cAMP pathway, heterotrimeric G-protein signaling, mitogen-activated protein kinases, two-component phosphorelays, and Ca(2+) signaling. Moreover, also the target of rapamycin signaling pathway and reactive oxygen species as signal transducing elements are assumed to be connected to the light-response pathway. The interplay of the light-response pathway with signaling cascades results in light-dependent regulation of primary and secondary metabolism, morphology, development, biocontrol activity, and virulence. The frequent use of fungi in biotechnology as well as analysis of fungi in the artificial environment of a laboratory therefore requires careful consideration of still operative evolutionary heritage of these organisms. This review summarizes the diverse effects of light on fungi and the mechanisms they apply to deal both with the information content and with the harmful properties of light. Additionally, the implications of the reaction of fungi to light in a laboratory environment for experimental work and industrial applications are discussed.
Collapse
|
47
|
Modeling of glucose-induced cAMP oscillations in pancreatic β cells: cAMP rocks when metabolism rolls. Biophys J 2016. [PMID: 26200880 DOI: 10.1016/j.bpj.2015.06.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Recent advances in imaging technology have revealed oscillations of cyclic adenosine monophosphate (cAMP) in insulin-secreting cells. These oscillations may be in phase with cytosolic calcium oscillations or out of phase. cAMP oscillations have previously been modeled as driven by oscillations in calcium, based on the known dependence of the enzymes that generate cAMP (adenylyl cyclase) and degrade it (phosphodiesterase). However, cAMP oscillations have also been reported to occur in the absence of calcium oscillations. Motivated by similarities between the properties of cAMP and metabolic oscillations in pancreatic β cells, we propose here that in addition to direct control by calcium, cAMP is controlled by metabolism. Specifically, we hypothesize that AMP inhibits adenylyl cyclase. We incorporate this hypothesis into the dual oscillator model for β cells, in which metabolic (glycolytic) oscillations cooperate with modulation of ion channels and metabolism by calcium. We show that the combination of oscillations in AMP and calcium in the dual oscillator model can account for the diverse oscillatory patterns that have been observed, as well as for experimental perturbations of those patterns. Predictions to further test the model are provided.
Collapse
|
48
|
Lai KP, Li JW, Tse ACK, Wang SY, Chan TF, Wu RSS. Differential responses of female and male brains to hypoxia in the marine medaka Oryzias melastigma. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2016; 172:36-43. [PMID: 26765084 DOI: 10.1016/j.aquatox.2015.12.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 12/23/2015] [Accepted: 12/24/2015] [Indexed: 05/26/2023]
Abstract
Hypoxia, an endocrine disruptor, affects synthesis and balance of sex steroid hormones, leading to reproductive impairment in both female and male fish. Cumulating reports demonstrated the alternation of hypothalamus-pituitary-gonad axis (HPG-axis) by hypoxia. However, the detail mechanism underlying how hypoxia may alter other brain functions remains largely unknown. In this report, we used marine medaka as a model and conducted a high-throughput RNA sequencing followed by bioinformatics analysis on hypoxia-exposed brain tissues, aiming to determine the change of transcriptional signature and to unravel the pathways that are induced by hypoxia. We found that hypoxia lead to dysregulation of brain functions (including synaptic transmission, axon guidance, potassium ion transport, neuron differentiation, and development of brain and pituitary gland), and also signaling pathways (e.g., gap junction, calcium signaling pathway, and GnRH signaling pathway). Our results further demonstrate gender-specific responses to hypoxia in female and male fish's brains, which provides novel insights into the mechanism underlying the hypoxia induced sex specific brain functions impairments.
Collapse
Affiliation(s)
- Keng-Po Lai
- School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, China; State Key Laboratory in Marine Pollution, Hong Kong SAR, China.
| | - Jing-Woei Li
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Anna Chung-Kwan Tse
- School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, China; State Key Laboratory in Marine Pollution, Hong Kong SAR, China.
| | - Simon Yuan Wang
- School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, China; State Key Laboratory in Marine Pollution, Hong Kong SAR, China.
| | - Ting-Fung Chan
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China; State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Rudolf Shiu-Sun Wu
- School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, China; State Key Laboratory in Marine Pollution, Hong Kong SAR, China.
| |
Collapse
|
49
|
Chen X, Dong G, Zheng C, Wang H, Yun W, Zhou X. A reduced susceptibility to chemoconvulsant stimulation in adenylyl cyclase 8 knockout mice. Epilepsy Res 2015; 119:24-9. [PMID: 26656781 DOI: 10.1016/j.eplepsyres.2015.11.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 10/18/2015] [Accepted: 11/09/2015] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Adenylyl cyclases (ACs) catalyze the synthesis of cAMP from ATP, and cAMP signaling affects a large number of neuronal processes. Ca(2+)-stimulated adenylyl cyclase 8 (AC8) expressed in the CNS plays a role in synaptic plasticity, drug addiction and ethanol sensitivity, and chronic pain. This study was to aim at examining the contributions of AC8 to epileptogenesis. METHODS In this study, we observed the seizure behavior induced by kainic acid (20 mg/kg or 30 mg/kg) or pilocarpine (350 mg/kg) in AC8 KO and wild-type mice. Next we injected kainic acid or pilocarpine to induce status epilepticus (SE), and examined neuronal degeneration (by Fluoro-Jade B staining) and mossy fiber sprouting (by Timm staining) 24h and 2 weeks after SE termination in the hippocampus, respectively. Finally, 15 min after intraperitoneal injection of kainic acid (30 mg/kg), we examined phosphor-ERK1/2 in the hippocampus by Western blot and immunochemistry staining. RESULTS We first observed that AC8 KO mutants display reduced susceptibility (including seizure latency and episodes) to two chemoconvulsants, kainic acid and pilocarpine. Moreover, we found that degenerative neurons and mossy fiber sprouting induced by chemoconvulsants were significant decreased in the hippocampus. Further, Western blot and immunochemistry analysis revealed that the MAPK signaling in the hippocampus was attenuated in kainic acid-injected AC8 KO mice. CONCLUSION AC8 is involved in epileptogenesis, and may serve as a potential target for the treatment of epilepsy.
Collapse
Affiliation(s)
- Xia Chen
- Laboratory of Neurological Diseases, Department of Neurology, Changzhou No. 2 People's Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu Province 213003, China
| | - Guoying Dong
- Laboratory of Neurological Diseases, Department of Neurology, Changzhou No. 2 People's Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu Province 213003, China
| | - Changhong Zheng
- Laboratory of Neurological Diseases, Department of Neurology, Changzhou No. 2 People's Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu Province 213003, China
| | - Hongbing Wang
- Department of Physiology and Neuroscience Program, Michigan State University, East Lansing, MI 48824, USA
| | - Wenwei Yun
- Laboratory of Neurological Diseases, Department of Neurology, Changzhou No. 2 People's Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu Province 213003, China.
| | - Xianju Zhou
- Laboratory of Neurological Diseases, Department of Neurology, Changzhou No. 2 People's Hospital, The Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu Province 213003, China.
| |
Collapse
|
50
|
Rangel-Barajas C, Coronel I, Florán B. Dopamine Receptors and Neurodegeneration. Aging Dis 2015; 6:349-68. [PMID: 26425390 DOI: 10.14336/ad.2015.0330] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 03/30/2015] [Indexed: 01/19/2023] Open
Abstract
Dopamine (DA) is one of the major neurotransmitters and participates in a number of functions such as motor coordination, emotions, memory, reward mechanism, neuroendocrine regulation etc. DA exerts its effects through five DA receptors that are subdivided in 2 families: D1-like DA receptors (D1 and D5) and the D2-like (D2, D3 and D4). All DA receptors are widely expressed in the central nervous system (CNS) and play an important role in not only in physiological conditions but also pathological scenarios. Abnormalities in the DAergic system and its receptors in the basal ganglia structures are the basis Parkinson's disease (PD), however DA also participates in other neurodegenerative disorders such as Huntington disease (HD) and multiple sclerosis (MS). Under pathological conditions reorganization of DAergic system has been observed and most of the times, those changes occur as a mechanism of compensation, but in some cases contributes to worsening the alterations. Here we review the changes that occur on DA transmission and DA receptors (DARs) at both levels expression and signals transduction pathways as a result of neurotoxicity, inflammation and in neurodegenerative processes. The better understanding of the role of DA receptors in neuropathological conditions is crucial for development of novel therapeutic approaches to treat alterations related to neurodegenerative diseases.
Collapse
Affiliation(s)
- Claudia Rangel-Barajas
- 1Department of Psychological and Brain Sciences Program in Neurosciences, Indiana University Bloomington, Bloomington, IN 47405, USA ; 2Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Israel Coronel
- 3Health Sciences Faculty, Anahuac University, Mexico Norte, State of Mexico, Mexico
| | - Benjamín Florán
- 4Department of Physiology, Biophysics and Neurosciences CINVESTAV-IPN, Mexico
| |
Collapse
|